Boston author turns real life into Hollywood-ready stories – Boston Herald

Boston author Ben Mezrich has a great track record when it comes to taking real events and turning them into made-for-Hollywood stories. His writing resume includes Bringing Down the House, which became the film 21, and The Accidental Billionaires, aka The Social Network on the big screen.

His latest, Woolly: The True Story of the Quest to Revive One of Historys Most Iconic Extinct Creatures is the saga of Harvard scientists attempting to bring back gigantic herbivores from extinction.

The Herald caught up with Mezrich in his Hub man cave/office.

HERALD: So this is where all the magic happens?

MEZRICH: Ive written all 18 of my books here thats why I can never leave. It looked like Charlie Sheens apartment before ... with black and leather ... Tonya (Mezrichs wife) helped me brighten it up.

HERALD: The view is stunning.

MEZRICH: The view is great ... You can see MIT from here, which served as inspiration when I wrote Bringing Down the House ... Theres Harvard ... Boston is the center of my writing career its where I do everything.

HERALD: Tell me about your latest book.

MEZRICH: Its about a Harvard lab and the eminent geneticist, George Church, who is just around the corner in his lab making a wooly mammoth. ...I Googled him and randomly wrote an email and he said come on in! So I embedded myself in his lab ... The idea that theres a wooly mammoth being made like a mile from here is just spectacular.

HERALD: Twentieth Century Fox is already making a movie about it will the movie come before an actual mammoth is born?

MEZRICH: We are only a few years away from an actual mammoth the fetus will be put in the womb of an elephant ... an elephant will give birth to one, as they are very closely related. The movie is already in the works! Marty Bowen (of Twilight fame) is producing it and Oscar Sharp ... who is like the next Spielberg ... will direct.

Check out our video with Mezrich and of our chat with the celeb author on bostonherald.com.

See more here:

Boston author turns real life into Hollywood-ready stories - Boston Herald

FDA Panel Recommends Approval for Gene-Altering Leukemia Treatment – New York Times

One of those patients, Emily Whitehead, now 12 and the first child ever given the altered cells, was at the meeting of the panel with her parents to advocate for approval of the drug that saved her life. In 2012, as a 6-year-old, she was treated in a study at the Childrens Hospital of Philadelphia. Severe side effects raging fever, crashing blood pressure, lung congestion nearly killed her. But she emerged cancer free, and has remained so.

We believe that when this treatment is approved it will save thousands of childrens lives around the world, Emilys father, Tom Whitehead, told the panel. I hope that someday all of you on the advisory committee can tell your families for generations that you were part of the process that ended the use of toxic treatments like chemotherapy and radiation as standard treatment, and turned blood cancers into a treatable disease that even after relapse most people survive.

The main evidence that Novartis presented to the F.D.A. came from a study of 63 patients who received the treatment from April 2015 to August 2016. Fifty-two of them, or 82.5 percent, went into remission a high rate for such a severe disease. Eleven others died.

Its a new world, an exciting therapy, said Dr. Gwen Nichols, the chief medical officer of the Leukemia and Lymphoma Society, which paid for some of the research that led to the treatment.

The next step, she said, will be to determine what we can combine it with and is there a way to use it in the future to treat patients with less disease, so that the immune system is in better shape and really able to fight. She added, This is the beginning of something big.

At the meeting, the panel of experts did not question the lifesaving potential of the treatment in hopeless cases. But they raised concerns about potentially life-threatening side effects short-term worries about acute reactions like those Emily experienced, and longer-term worries about whether the infused cells could, years later, cause secondary cancers or other problems.

Oncologists have learned how to treat the acute reactions, and so far, no long-term problems have been detected, but not enough time has passed to rule them out.

Patients who receive the treatment will be entered in a registry and tracked for 15 years.

Treatments involving live cells, known as biologics are generally far more difficult to manufacture than standard drugs, and the panelists also expressed concerns about whether Novartis would be able to produce consistent treatments and maintain quality control as it scaled up its operation.

Another parent at the meeting, Don McMahon, described his son Connors grueling 12 years with severe and relapsing leukemia, which started when he was 3. Mr. McMahon displayed painful photographs of Connor, bald and intubated during treatment. And he added that chemotherapy had left his son infertile.

A year ago, the family was preparing for a bone marrow transplant when they learned about the cell treatment, which Connor then underwent at Duke University. He has since returned to playing hockey. Compared with standard treatment, which required dozens of spinal taps and painful bone marrow tests, the T-cell treatment was far easier to tolerate, Mr. McMahon said, and he urged the panel to vote for approval.

A third parent, Amy Kappen, also recommended approval, even though her daughter, Sophia, 5, had died despite receiving the cell treatment. But it did relieve her symptoms and give her a few extra months. Sophias disease was far advanced, and Ms. Kappen thought that if the treatment could have been given sooner, Sophia might have survived.

We hope that more families have a longer time with their children fighting this evil disease, and our children deserve this chance, she said.

The treatment was developed by researchers at the University of Pennsylvania and licensed to Novartis.

Use will not be widespread at first because the disease is not common. It affects only 5,000 people a year, about 60 percent of them children and young adults. Most children are cured with standard treatments, but in 15 percent of cases like Emilys and Connors the disease does not respond, or it relapses.

Analysts predict that these individualized treatments could cost more than $300,000, but a spokeswoman for Novartis, Julie Masow, declined to specify a price.

Although the figure may seem high, people with cancer often endure years of expensive treatment and repeat hospital stays that can ultimately cost even more.

Because the treatment is complex and patients need expert care to manage the side effects, Novartis will initially limit its use to 30 or 35 medical centers where employees will be trained and approved to administer it, the company said.

As to whether the treatment, known as CTL019 or tisagenlecleucel (pronounced tis-a-gen-LEK-loo-sell), will be available in other countries, Ms. Masow said by email: Should CTL019 receive approval in the U.S., it will be the decision of the centers whether to receive international patients. We are working on bringing CTL019 to other countries around the world. She added that the company would file for approvals in the European Union later this year.

By late November 2016, 11 of the 52 patients in the study who went into remission relapsed. Twenty-nine were still in remission. Eleven others had further treatments, like bone marrow transplants. One patient was not available for assessment. Three who had relapses died, and one who did not relapse died from a new treatment given during remission. The median duration of remission is not known because it has not been reached: Some patients were still well when last checked.

Researchers are still debating about which patients can safely forgo further treatment, and which might need a bone marrow treatment to give the best chance of a cure.

The treatment requires removing millions of a patients T-cells a type of white blood cell often called soldiers of the immune system and genetically engineering them to kill cancer cells. The technique employs a disabled form of H.I.V., the virus that causes AIDS, to carry new genetic material into the T-cells to reprogram them. The process turbocharges the T-cells to attack B-cells, a normal part of the immune system that turn malignant in leukemia. The T-cells home in on a protein called CD-19 that is found on the surface of most B-cells.

The altered T-cells are then dripped back into the patients veins, where they multiply and start fighting the cancer.

Dr. Carl H. June, a leader of the University of Pennsylvania team that developed the treatment, calls the turbocharged cells serial killers. A single one can destroy up to 100,000 cancer cells.

Because the treatment destroys not only leukemic B-cells but also healthy ones, which help fight germs, patients need treatment to protect them from infection. So every few months they receive infusions of immune globulins.

In studies, the process of re-engineering T-cells for treatment sometimes took four months, and some patients were so sick that they died before their cells came back. At the meeting, Novartis said the turnaround time was now down to 22 days. The company also described bar-coding and other procedures used to keep from mixing up samples once the treatment is conducted on a bigger scale.

Michael Werner, a lawyer and expert on gene and cell technologies and regulation, and a partner at Holland and Knight in Washington, said that results so far proved that T-cell treatment works.

The fact that it can be done means more people will go into the field and more companies will start developing these products. He added, I think were in for really exciting times.

Katie Thomas contributed reporting.

A version of this article appears in print on July 13, 2017, on Page A1 of the New York edition with the headline: F.D.A. Panel Urges New Living Drug To Fight Cancer.

See the original post here:

FDA Panel Recommends Approval for Gene-Altering Leukemia Treatment - New York Times

Genetically engineered salmon is coming to America – The Week Magazine

Sign Up for

Our free email newsletters

On a hill above the cold waters around Prince Edward Island, technicians painstakingly create fertilized Atlantic salmon eggs that include growth-enhancing DNA from two other fish species. The eggs will be shipped to tanks in the high rainforest of Panama, where they will produce fish that mature far more quickly than normal farmed salmon.

More than 20 years after first seeking approval from the U.S. Food and Drug Administration, AquaBounty Technologies of Maynard, Massachusetts, plans to bring these "AquAdvantage" fish to the U.S. and Canadian markets next year. And in the small town of Albany, Indiana, workers will soon begin converting a land-based aquaculture facility to produce about 1,300 U.S. tons of these salmon annually, in the first U.S. facility to generate GE animals for human consumption.

The company also plans to open a second aquaculture facility at Prince Edward Island if it can rise above its latest round of legal battles and persuade grocery stores and restaurants to snap up the genetically engineered fish. Before the FDA cleared the salmon for consumption in 2015, in its first approval of GE animal protein as human food, it received 1.8 million messages opposing these fish. Perhaps more substantively, many outside researchers remain concerned about AquaBounty's plans.

Safety and nutrition

Aquaculture specialists generally aren't skeptical about whether the fish will be healthy to eat, although that's one issue hinted at in a lawsuit multiple organizations, including Friends of the Earth, have filed against the FDA. Dana Perls, senior food and technology campaigner with Friends of the Earth in Berkeley, California, says the FDA didn't fully examine questions about eating the salmon initially raised by Health Canada, that country's public health department including susceptibility to disease and potential allergic reactions.

"This is a poorly studied, risky, and unlabeled genetically engineered fish," she says, adding that more than 80 U.S. grocery chains have committed not to buy it. However, Health Canada eventually concluded that fillets derived from AquAdvantage salmon "are as safe and nutritious as fillets from current available farmed Atlantic salmon," and approved the fish for consumption in 2016.

"There's no reason to suspect these fish from a food safety perspective," says Cyr Couturier, chair of aquaculture programs at Memorial University's Marine Institute in St. John's, Newfoundland. "They have no unnatural products that humans wouldn't otherwise consume."

Similar transgenic salmon created by a decades-long Fisheries and Oceans Canada research program tested well within normal salmon variations, adds Robert Devlin, engineering research scientist at the agency in North Vancouver, British Columbia. But critics do raise two other main concerns about AquaBounty's quest: the economic sustainability of the land-based approach, and the environmental risk to ecosystems if the fish escape.

Fish on land

AquaBounty will raise its GE fish in land-based recirculating aquaculture systems, known as RAS basically huge aquaria designed to minimize water use, maximize resources and accommodate high stocking densities. "While farming salmon in sea cages is less expensive and less technologically complex than a land-based farm," the company's website points out, "sea cages are susceptible to a number of hazards such as violent storms, predators, harmful algal blooms, jellyfish attacks, fish escapes, and the transmission of pathogens and parasites from wild fish populations."

Given the potential opportunity to achieve greater production control and avoid some of the environmental concerns of sea farms, many RAS projects have launched around the world in the past decade. However, most of these projects are small, and many have failed or are struggling.

The big problem is cost. RAS facilities need much more capital than ocean farms with similar production rates, and they're expensive to operate.

"Land-based systems use a lot of freshwater, even though it's recirculated, and a lot of electricity," notes Couturier. Such systems "operate at an economic disadvantage because much of their cost goes toward creating growing conditions occurring naturally within the ocean," summed up one 2014 report that found producing Atlantic salmon in Nova Scotia would not be economically feasible.

AquaBounty, which is buying its Indiana plant from a collapsed RAS venture, expects to beat these odds mainly because its GE salmon reach market size in about half the time of normal farmed salmon in 1618 months rather than 2836 months, the company says. Ravenous as they are, with their growth hormones continually wired on, the fish still require about a quarter less feed than normal fish. (Although farmed salmon are very efficient at converting food to flesh a pound of feed converts close to a pound of flesh feed remains a major expense.)

The company also says that salmon in its RAS facilities won't need vaccines or antibiotics because it will tightly control conditions. However, "they will have some disease issues of course, as will any animal that's reared in high densities," Couturier predicts.

If AquaBounty can compete on cost, there will be some justification for promoting its product as "the world's most sustainable salmon." In addition to requiring less feed, growing fish in Indiana or Prince Edward Island can slash the high carbon costs of flying fish from Norway or Chile, two leading suppliers of farmed salmon in the U.S.

Still, says Couturier, "I wish them all the best, but I think it will be a small-scale niche for at least a decade."

Losing GE fish

Many aquaculture scientists remain uneasy about the environmental risk to wild ecosystems if transgenic fish slip out of their farms. Although other agencies will presumably be involved in assessing risk as the projects advance, "the FDA has no in-house capacity to evaluate or understand the ecological consequences of transgenics in an aquatic ecosystem," says Conner Bailey, professor emeritus of rural sociology at Auburn University in Alabama. "And once you get anything into an aquatic ecosystem, it's really hard to control."

AquaBounty's protection scheme begins with multiple levels of physical barriers in its RAS facilities. Additionally, the salmon are all female and "triploid" (their DNA is in three rather than two sets of chromosomes) so they can't reproduce. However, scientists say neither of these measures can be 100 percent effective at preventing transgenic fish from escaping, disrupting local ecosystems, and potentially breeding in the wild.

More generally, while AquaBounty is committed to land-based systems, there are concerns that it's also creating far more GE eggs than it needs for its own production. Other industry groups, such as the Atlantic Salmon Federation, worry that other producers AquaBounty sells to might not be so careful, or that other companies around the world might move ahead with similar projects but without the same precautions. And all bets on risk are off if GE fish are raised in the ocean, where fish routinely escape, sometimes in large numbers.

Devlin's group has extensively modeled the results of accidental releases, studying groups of transgenic and non-transgenic fish in "naturalized" aquatic test beds that are exposed to variations in conditions, such as food supply. Transgenic fish often behave quite differently, and the results have varied from peaceful coexistence to one experiment in which fully transgenic fish killed off all their competitors.

"In the multitude of different environments that exist in nature, the uncertainty is too great to make a reliable prediction of what the impact would be," he says.

GE or selective breeding?

Does the fast growth of AquAdvantage salmon justify taking on these unknown risks? Scientists point out that today's selective breeding research programs, built on genomics and other tools of modern biology, also have turbocharged fish development. "Some strains of rainbow trout, which have been selected for fast growth for 150 years, grow incredibly fast compared to wild-type fish," Devlin says. In fact, he says, his lab work across various species suggests that "the absolute fastest growth you can achieve either by domestication or by transgenesis seems to be very similar."

"Today's farmed salmon have had more than 10 generations of selection applied to them, and they are growing at more than double the rate compared to the 1970s," says Bjarne Gjerde, senior scientist at Nofima in Troms, Norway.

Farmed fish also must excel in many traits besides growth, such as disease resistance and food quality, he emphasizes. "Most of the traits we are breeding for are governed by many, many genes with small effects," he says. "That's a real challenge if you just want to take short cuts with genetic engineering."

When and if AquaBounty rises above all its challenges into a groundbreaking success in North America, the firm will send a signal around the world to unleash efforts for commercializing GE fish, observers say. Friends of the Earth's Perls remains hopeful that legal barriers and consumer boycotts will stop AquaBounty in its tracks. If not, "GE salmon could set a precedent to the approval of other GE animals in the pipeline, from fish to chickens, pigs, and cows," she says. "It is critical that we don't approve other GE animals without robust regulations and full environmental reviews to ensure that we're prioritizing human and environmental safety over profit."

"Fish are probably where transgenic animals will emerge, because it's much cheaper to maintain a herd of catfish or salmon than cattle or sheep or pigs," says Bailey.

This story was first published by Ensia, an environmental news magazine from the University of Minnesota.

This article originally appeared at PRI's The World.

More here:

Genetically engineered salmon is coming to America - The Week Magazine

Are Pink Pineapples Safe to Eat? – Observer

While the FDA has approved the new genetically modifiedfruit made by Del Monte, there really isnt any data about its safety. Unsplash/Pineapple Supply Co

#PinkPineapples may beinfiltrating your Instagram feed, but just how safe are they?

The truth is, we dont know. While the FDA has approved the new genetically modifiedfruit made by Del Monte, there really isnt any data about its safetybeyond what the companytold the FDA. According to NBC, Del Monte submitted information to the agency to demonstrate that the pink flesh pineapple is as safe and nutritious as its conventional counterparts.

But what constitutes safe? How do we know that in 10 or 20 yearsor after some length of time consuming these genetically modified foodsit wont cause a problem?What we do know is that genetic engineering changes the DNA of plants and crops in orderto make them heartier, more resistant to infections and herbicides and able toyield larger amounts of food to feed more people. This, of course, on the surface, sounds promisinguntil you dig deeper.

Genetic engineering creates changes at a cellular level that cannot be easily seen or understood. These changes have the potential for substantial health implications. Take allergies, for instance. If genes from a highly-allergenic plant are transferred to another plant, the new plant now carries that allergenic potential. This happened in 1996, when a Brazil nut gene was added to soybeans in order to increase its nutritional status.The gene from the Brazil nut codes for a protein that causes allergic reactions in people with Brazil nut allergies. By adding this to the soy, a number of people experienced allergic reactions to something they werent otherwise allergic to.

In order for the FDA to approve a genetically modified food, the GMO food must be substantially equivalent to the non-GMO foods. In other words, when it comes to how our bodies process the foods, the two should be almost identical. However, a recent study showed that GMO corn was not substantially equivalent tonon-GMO corn, even though the FDA approved it as such. The GMO corncontained higher levels of several proteins that are known to be toxic to humans. In general, we are seeing an increase in GMO plants that have toxic genes created for specific purposes.

Many plants are now genetically modified to internallyproduce their own pesticides, protecting them from the insects that might damage them. Butwhen these GMO plants are used to feed animals, the animals are at increased risk for diseases. A 2012 study in Norway showed that rats fed a diet of GMO corn with pest resistancegot fatter than rats fed non-GMO corn. The lead professor on that study explained that the rats that had fed on GMO corn were slightly larger, they ate slightly more, their intestines had a different microstructure, they were less able to digest proteins, and there were some changes to their immune system. Blood samples also showed some change in the blood. Isnt it interesting that these rats developed exactly what we are seeing in the explodingepidemics of obesity, metabolic syndrome and chronic diseases in humans?

Another takeaway from that study contradicts a long-held belief about the safety of genetically modified food. Proponents of GMO claim that the new genes introduced into the genetically modified food are harmless and are digested and eliminated throughthe gastrointestinal tract. But the 2012 Norwegian study showed that these new genes could be transferred through the intestinal wall into the blood, and were found in the liver and even in muscle tissue. This implies that foreign genes could assimilate into our own DNA, turning on or off genes that have important functions. This could have far reaching effects on things like the development of cancer, changes in growth or longevity, and even the development of diseases like diabetes.

We are also seeing an increase in GMO plants bred with resistance to the herbicide glyphosate, or Round Updesigned to protect plants from the weed killer used to kill everything around it. But instead of helping farmers, this increased the weeds resistance to weed killer creating what is known as superweeds. As a result, farmers have had to dramatically increase the amount of pesticides used to control the situation, driving up food cost but also increasing the toxic levels of pesticides in our environment. In addition, plants that are resistant to glyphosate have grown out of control as well. For example, genetically modified canola is spreading uncontrollably throughout California.

Like any new invention or discovery, the initial benefits of genetic engineeringseem exciting and life changing. With time, it is becoming clear that we are just at the tip of the GMO iceberg and need to brace ourselves for what is yet to come. Until we know more about the long-term effects, I would strongly recommendavoiding genetically modified foods whenever possible. For my family, I chooseorganic fruits,vegetables,grass-fed/pastured meat and eggs and I urge the same for my patients and for you.

Dr.TaniaDempseyMD is an expert in chronic disease, autoimmune disorders and mast cell activation syndrome. Dr.Dempseyreceived her MD from The Johns Hopkins University School of Medicine and her BS degree from Cornell University. She completed herResidency at NYU Medical Center/ Bellevue Hospital and then served as an attending physician at a large multi-specialty medical practice in White Plains, NY, before opening Armonk Integrative Medicine. Dr.Dempseyis sought after internationally for her knowledge of chronic immune dysregulation and MCAS.For more information, please visitwww.drtaniadempsey.com

Visit link:

Are Pink Pineapples Safe to Eat? - Observer

Can Genetic Engineering Put an End to Diamondback Moth Plague … – Growing Produce

Florida Struggling to Stay Afloat in Wake of Drought

USDA Invests $6.8 Million for Pollinator Health

The diamondback moth (DBM) is a pest of epic proportions to farmers worldwide. Growers of cabbage and collard crops know this all too well. Controlling the notorious DBM has become more difficult in recent times with incidence of pesticide resistance increasing. With that, the search for viable management continues to expand. For the last two years, Cornell scientists have been working with British-based biotech company Oxitec and testing its self-limiting gene capabilities on DBM in a protected environment. The team at Cornell has been waiting for a go-aheadfrom USDA to take the experiment out from behind closed doors and into one if its cabbage fields in Upstate New York. It looks like the wait is over.

USDAs Animal and Plant Health Inspection Service (APHIS) has officially announced the availability of a final environmental assessment and finding of no significant impact and will concurrently issue a permit for the field release of a genetically engineered DBM.

The pest moths are genetically engineered for repressible female lethality and to express red fluorescence as a marker. The purpose of the field release is to assess the feasibility and efficacy of these moths in reducing populations of DBM. Based on the finding of no significant impact, APHIS has determined that an environmental impact statement need not be prepared.

Notice of these actions will be published in theFederal Register.

The Northeast Organic Farming Association of New York (NOFA-NY) has since released a statement denouncing USDAs decision. A portion of the statement reads as follows: NOFA-NY considers the release of a novel genetically engineered organism to be a major activity with potentially significant and heretofore unknown health and environmental effects, said NOFA-NY Policy Advisor Liana Hoodes. It is now up to New York State Department of Environmental Conservation (DEC) to ensure the safety of its citizens before granting the necessary state permit. We call on the NYS DEC to require a full environmental impact statement and public hearings during a complete review under State Environmental Quality Review Act.

Read more here:

Can Genetic Engineering Put an End to Diamondback Moth Plague ... - Growing Produce

Writing the human genome – The Biological SCENE

[+]Enlarge

Credit: Will Ludwig/C&EN/Shutterstock

In brief

Synthetic biologists have been creating the genomes of organisms such as viruses and bacteria for the past 15 years. They aim to use these designer genetic codes to make cells capable of producing novel therapeutics and fuels. Now, some of these scientists have set their sights on synthesizing the human genomea vastly more complex genetic blueprint. Read on to learn about this initiative, called Genome Project-write, and the challenges researchers will faceboth technical and ethicalto achieve success.

Nineteenth-century novels are typically fodder for literature conferences, not scientific gatherings. Still, at a high-profile meeting of about 200 synthetic biologists in May, one presenter highlighted Mary Shelleys gothic masterpiece Frankenstein, which turns 200 next year.

Frankensteins monster, after all, is what many people think of when the possibility of human genetic engineering is raised, said University of Pennsylvania ethicist and historian Jonathan Moreno. The initiative being discussed at the New York City meetingGenome Project-write (GP-write)has been dogged by worries over creating unnatural beings. True, part of GP-write aims to synthesize from scratch all 23 chromosomes of the human genome and insert them into cells in the lab. But proponents of the project say theyre focused on decreasing the cost of synthesizing and assembling large amounts of DNA rather than on creating designer babies.

The overall project is still under development, and the projects members have not yet agreed on a specific road map for moving forward. Its also unclear where funding will come from.

What the members of GP-write do agree on is that creating a human genome from scratch is a tremendous scientific and engineering challenge that will hinge on developing new methods for synthesizing and delivering DNA. They will also need to get better at designing large groups of genes that work together in a predictable way, not to mention making sure that even larger assembliesgenomescan function.

GP-write consortium members argue that these challenges are the very thing that should move scientists to pick up the DNA pen and turn from sequence readers to writers. They believe writing the entire human genome is the only way to truly understand how it works. Many researchers quoted Richard Feynman during the meeting in May. The statement What I cannot create, I do not understand was found on the famed physicists California Institute of Technology blackboard after his death. I want to know the rules that make a genome tick, said Jef Boeke, one of GP-writes four coleaders, at the meeting.

To that end, Boeke and other GP-write supporters say the initiative will spur the development of new technologies for designing genomes with software and for synthesizing DNA. In turn, being better at designing and assembling genomes will yield synthetic cells capable of producing valuable fuels and drugs more efficiently. And turning to human genome synthesis will enable new cell therapies and other medical advances.

In 2010, researchers at the Venter Institute, including Gibson, demonstrated that a bacterial cell controlled by a synthetic genome was able to reproduce. Colonies formed by it and its sibling resembled a pair of blue eyes.

Credit: Science

Genome writers have already synthesized a few complete genomes, all of them much less complex than the human genome. For instance, in 2002, researchers chemically synthesized a DNA-based equivalent of the poliovirus RNA genome, which is only about 7,500 bases long. They then showed that this DNA copy could be transcribed by RNA polymerase to recapitulate the viral genome, which replicated itselfa demonstration of synthesizing what the authors called a chemical [C332,652H492,388N98,245O131,196P7,501S2,340] with a life cycle (Science 2002, DOI: 10.1126/science.1072266).

After tinkering with a handful of other viral genomes, in 2010, researchers advanced to bacteria, painstakingly assembling a Mycoplasma genome just over about a million bases in length and then transplanting it into a host cell.

Last year, researchers upped the ante further, publishing the design for an aggressively edited Escherichia coli genome measuring 3.97 million bases long (Science, DOI: 10.1126/science.aaf3639). GP-write coleader George Church and coworkers at Harvard used DNA-editing softwarea kind of Google Docs for writing genomesto make radical systematic changes. The so-called rE.coli-57 sequence, which the team is currently synthesizing, lacks seven codons (the three-base DNA words that code for particular amino acids) compared with the normal E. coli genome. The researchers replaced all 62,214 instances of those codons with DNA base synonyms to eliminate redundancy in the code.

Status report International teams of researchers have already synthesized six of yeast's 16 chromosomes, redesigning the organism's genome as part of the Sc2.0 project.

Bacterial genomes are no-frills compared with those of creatures in our domain, the eukaryotes. Bacterial genomes typically take the form of a single circular piece of DNA that floats freely around the cell. Eukaryotic cells, from yeast to plants to insects to people, confine their larger genomes within a cells nucleus and organize them in multiple bundles called chromosomes. An ongoing collaboration is now bringing genome synthesis to the eukaryote realm: Researchers are building a fully synthetic yeast genome, containing 17 chromosomes that range from about 1,800 to about 1.5 million bases long. Overall, the genome will contain more than 11 million bases.

The synthetic genomes and chromosomes already constructed by scientists are by no means simple, but to synthesize the human genome, scientists will have to address a whole other level of complexity. Our genome is made up of more than 3 billion bases across 23 paired chromosomes. The smallest human chromosome is number 21, at 46.7 million baseslarger than the smallest yeast chromosome. The largest, number 1, has nearly 249 million. Making a human genome will mean making much more DNA and solving a larger puzzle in terms of assembly and transfer into cells.

Today, genome-writing technology is in what Boeke, also the director for the Institute of Systems Genetics at New York University School of Medicine, calls the Gutenberg phase. (Johannes Gutenberg introduced the printing press in Europe in the 1400s.) Its still early days.

DNA synthesis companies routinely create fragments that are 100 bases long and then use enzymes to stitch them together to make sequences up to a few thousand bases long, about the size of a gene. Customers can put in orders for small bits of DNA, longer strands called oligos, and whole geneswhatever they needand companies will fabricate and mail the genetic material.

Although the technology that makes this mail-order system possible is impressive, its not prolific enough to make a human genome in a reasonable amount of time. Estimates vary on how long it would take to stitch together a more than 3 billion-base human genome and how much it would cost with todays methods. But the ballpark answer is about a decade and hundreds of millions of dollars.

Synthesis companies could help bring those figures down by moving past their current 100-base limit and creating longer DNA fragments. Some researchers and companies are moving in that direction. For example, synthesis firm Molecular Assemblies is developing an enzymatic process to write long stretches of DNA with fewer errors.

Synthesis speeds and prices have been improving rapidly, and researchers expect they will continue to do so. From my point of view, building DNA is no longer the bottleneck, says Daniel G. Gibson, vice president of DNA technology at Synthetic Genomics and an associate professor at the J. Craig Venter Institute (JCVI). Some way or another, if we need to build larger pieces of DNA, well do that.

Gibson isnt involved with GP-write. But his research showcases what is possible with todays toolseven if they are equivalent to Gutenbergs movable type. He has been responsible for a few of synthetic biologys milestones, including the development of one of the most commonly used genome-assembly techniques.

The Gibson method uses chemical means to join DNA fragments, yielding pieces thousands of bases long. For two fragments to connect, one must end with a 20- to 40-base sequence thats identical to the start of the next fragment. These overlapping DNA fragments can be mixed with a solution of three enzymesan exonuclease, a DNA polymerase, and a DNA ligasethat trim the 5 end of each fragment, overlap the pieces, and seal them together.

To make the first synthetic bacterial genome in 2008, that of Mycoplasma genitalium, Gibson and his colleagues at JCVI, where he was a postdoc at the time, started with his eponymous in vitro method. They synthesized more than 100 fragments of synthetic DNA, each about 5,000 bases long, and then harnessed the prodigious DNA-processing properties of yeast, introducing these large DNA pieces to yeast three or four at a time. The yeast used its own cellular machinery to bring the pieces together into larger sequences, eventually producing the entire Mycoplasma genome.

Next, the team had to figure out how to transplant this synthetic genome into a bacterial cell to create what the researchers called the first synthetic cell. The process is involved and requires getting the bacterial genome out of the yeast, then storing the huge, fragile piece of circular DNA in a protective agarose gel before melting it and mixing it with another species of Mycoplasma. As the bacterial cells fuse, some of them take in the synthetic genomes floating in solution. Then they divide to create three daughter cells, two containing the native genomes, and one containing the synthetic genome: the synthetic cell.

When Gibsons group at JCVI started building the synthetic cell in 2004, we didnt know what the limitations were, he says. So the scientists were cautious about overwhelming the yeast with too many DNA fragments, or pieces that were too long. Today, Gibson says he can bring together about 25 overlapping DNA fragments that are about 25,000 bases long, rather than three or four 5,000-base segments at a time.

Gibson expects that existing DNA synthesis and assembly methods havent yet been pushed to their limits. Yeast might be able to assemble millions of bases, not just hundreds of thousands, he says. Still, Gibson believes it would be a stretch to make a human genome with this technique.

One of the most ambitious projects in genome writing so far centers on that master DNA assembler, yeast. As part of the project, called Sc2.0 (a riff on the funguss scientific name, Saccharomyces cerevisiae), an international group of scientists is redesigning and building yeast one synthetic chromosome at a time. The yeast genome is far simpler than ours. But like us, yeasts are eukaryotes and have multiple chromosomes within their nuclei.

Synthetic biologists arent interested in rebuilding existing genomes by rote; they want to make changes so they can probe how genomes work and make them easier to build and reengineer for practical use. The main lesson learned from Sc2.0 so far, project scientists say, is how much the yeast chromosomes can be altered in the writing, with no apparent ill effects. Indeed, the Sc2.0 sequence is not a direct copy of the original. The synthetic genome has been reduced by about 8%. Overall, the research group will make 1.1 million bases worth of insertions, deletions, and changes to the yeast genome (Science 2017, DOI: 10.1126/science.aaf4557).

So far, says Boeke, whos also coleader of Sc2.0, teams have finished or almost finished the first draft of the organisms 16 chromosomes. Theyre also working on a neochromosome, one not found in normal yeast. In this chromosome, the designers have relocated all DNA coding for transfer RNA, which plays a critical role in protein assembly. The Sc2.0 group isolated these sequences because scientists predicted they would cause structural instability in the synthetic chromosomes, says Joel Bader, a computational biologist at Johns Hopkins University who leads the projects software and design efforts.

The team is making yeast cells with a new chromosome one at a time. The ultimate goal is to create a yeast cell that contains no native chromosomes and all 17 synthetic ones. To get there, the scientists are taking a relatively old-fashioned approach: breeding. So far, theyve made a yeast cell with three synthetic chromosomes and are continuing to breed it with strains containing the remaining ones. Once a new chromosome is in place, it requires some patching up because of recombination with the native chromosomes. Its a process, but it doesnt look like there are any significant barriers, Bader says. He estimates it will take another two to three years to produce cells with the entire Sc2.0 genome.

So far, even with these significant changes to the chromosomes, the yeast lives at no apparent disadvantage compared with yeast that has its original chromosomes. Its surprising how much you can torture the genome with no effect, Boeke says.

Boeke and Bader have founded a start-up company called Neochromosome that will eventually use Sc2.0 strains to produce large protein drugs, chemical precursors, and other biomolecules that are currently impossible to make in yeast or E. coli because the genetic pathways used to create them are too complex. With synthetic chromosomes well be able to make these large supportive pathways in yeast, Bader predicts.

Whether existing genome-engineering methods like those used in Sc2.0 will translate to humans is an open question.

Bader believes that yeast, so willing to take up and assemble large amounts of DNA, might serve as future human-chromosome producers, assembling genetic material that could then be transferred to other organisms, perhaps human cells. Transplanting large human chromosomes would be tricky, Synthetic Genomics Gibson says. First, the recipient cell must be prepped by somehow removing its native chromosome. Gibson expects physically moving the synthetic chromosome would also be difficult: Stretches of DNA larger than about 50,000 bases are fragile. You have to be very gentle so the chromosome doesnt breakonce its broken, its not going to be useful, he says. Some researchers are working on more direct methods for cell-to-cell DNA transfer, such as getting cells to fuse with one another.

Once the scientists solve the delivery challenge, the next question is whether the transplanted chromosome will function. Our genomes are patterned with methyl groups that silence regions of the genome and are wrapped around histone proteins that pack the long strands into a three-dimensional order in cells nuclei. If the synthetic chromosome doesnt have the appropriate methylation patterns, the right structure, it might not be recognized by the cell, Gibson says.

Biologists might sidestep these epigenetic and other issues by doing large-scale DNA assembly in human cells from the get-go. Ron Weiss, a synthetic biologist at Massachusetts Institute of Technology, is pushing the upper limits on this sort of approach. He has designed methods for inserting large amounts of DNA directly into human cells. Weiss endows human cells with large circuits, which are packages of engineered DNA containing groups of genes and regulatory machinery that will change a cells behavior.

In 2014, Weiss developed a landing pad method to insert about 64,000-base stretches of DNA into human and other mammalian cells. First, researchers use gene editing to create the landing pad, which is a set of markers at a designated spot on a particular chromosome where an enzyme called a recombinase will insert the synthetic genetic material. Then they string together the genes for a given pathway, along with their regulatory elements, add a matching recombinase site, and fashion this strand into a circular piece of DNA called a plasmid. The target cells are then incubated with the plasmid, take it up, and incorporate it at the landing site (Nucleic Acids Res. 2014, DOI: 10.1093/nar/gku1082).

This works, but its tedious. It takes about two weeks to generate these cell lines if youre doing well, and the payload only goes into a few of the cells, Weiss explains. Since his initial publication, he says, his team has been able to generate cells with three landing pads; that means they could incorporate a genetic circuit thats about 200,000 bases long.

Weiss doesnt see simple scale-up of the landing pad method as the way forward, though, even setting aside the tedium. He doesnt think the supersized circuits would even function in a human cell because he doesnt yet know how to design them.

The limiting factor in the size of the circuit is not the construction of DNA, but the design, Weiss says. Instead of working completely by trial and error, bioengineers use computer models to predict how synthetic circuits or genetic edits will work in living cells of any species. But the larger the synthetic element, the harder it is to know whether it will work in a real cell. And the more radical the deletion, the harder it is to foresee whether it will have unintended consequences and kill the cell. Researchers also have a hard time predicting the degree to which cells will express the genes in a complex synthetic circuita lot, a little, or not at all. Gene regulation in humans is not fully understood, and rewriting on the scale done in the yeast chromosome would have far less predictable outcomes.

Besides being willing to take up and incorporate DNA, yeast is relatively simple. Upstream from a yeast gene, biologists can easily find the promoter sequence that turns it on. In contrast, human genes are often regulated by elements found in distant regions of the genome. That means working out how to control large pathways is more difficult, and theres a greater risk that changing the genetic sequencesuch as deleting what looks like repetitive nonsensewill have unintended, currently unpredictable, consequences.

Gibson notes that even in the minimal cell, the organism with the simplest known genome on the planet, biologists dont know what one-third of the genes do. Moving from the simplest organism to humans is a leap into the unknown. One design flaw can change how the cell behaves or even whether the cells are viable, Gibson says. We dont have the design knowledge.

Many scientists believe this uncertainty about design is all the more reason to try writing human and other large genomes. People are entranced with the perfect, Harvards Church says. But engineering and medicine are about the pretty good. I learn much more by trying to make something than by observing it.

Others arent sure that the move from writing the yeast genome to writing the human genome is necessary, or ethical. When the project to write the human genome was made public in May 2016, the founders called it Human Genome Project-write. They held the first organizational meeting behind closed doors, with no journalists present. A backlash ensued.

In the magazine Cosmos, Stanford University bioengineer Drew Endy and Northwestern University ethicist Laurie Zoloth in May 2016 warned of unintended consequences of large-scale changes to the genome and of alienating the public, potentially putting at risk funding for the synthetic biology field at large. They wrote that the synthesis of less controversial and more immediately useful genomes along with greatly improved sub-genomic synthesis capacities should be pursued instead.

GP-write members seem to have taken such criticisms to heart, or come to a similar conclusion on their own. By this Mays conference, human was dropped from the projects name. Leaders emphasized that the human genome would be a subproject proceeding on a conservative timescale and that ethicists would be involved at every step along the way. We want to separate the overarching goal of technology development from the hot-button issue of human genome writing, Boeke explains.

Bringing the public on board with this kind of project can be difficult, says Alta Charo, a professor of law and bioethics at the University of Wisconsin, Madison, who is not involved with GP-write. Charo cochaired a National Academy of Sciences study on the ethics and governance of human gene editing, which was published in February.

She says the likelihood of positive outcomes, such as new therapies or advances in basic science, must be weighed against potential unintended consequences or unforeseen uses of genome writing. People see their basic values at stake in human genetic engineering. If scientists achieve their goalsmaking larger scale genetic engineering routine and more useful, and bringing it to the human genomemajor changes are possible to what Charo calls the fabric of our culture and society. People will have to decide whether they feel optimistic about that or not. (Charo does.)

Given humans cautiousness, Charo imagines in early times we might have decided against creating fire, saying, Lets live without that; we dont need to create this thing that might destroy us. People often see genetic engineering in extreme terms, as a fire that might illuminate human biology and light the way to new technologies, or one that will destroy us.

Charo says the GP-write plan to keep ethicists involved going forward is the right approach and that its difficult to make an ethical or legal call on the project until its leaders put forward a road map.

The group will announce a specific road map sometime this year, but it doesnt want to be restrictive ahead of time. You know when youre done reading something, Boeke said at the meeting in May. But writing has an artistic side to it, he added. You never know when youre done.

Katherine Bourzac is a freelance science writer based in San Francisco.

Excerpt from:

Writing the human genome - The Biological SCENE

Stanford’s Final Exams Pose Question About the Ethics of Genetic Engineering – Futurism

In BriefThe age of gene editing and creation will be upon us in thenext few decades, with the first lifeform having already beenprinted. Stanford University questions the ethics of prospectivestudents by asking a question we should all be thinking about. Stanfords Moral Pickle

When bioengineering students sit down to take their final exams for Stanford University,they are faced with a moral dilemma, as well as a series of grueling technical questions that are designed to sort the intellectual wheat from the less competent chaff:

If you and your future partner are planning to have kids, would you start saving money for college tuition, or for printing the genome of your offspring?

The question is a follow up to At what point will the cost of printing DNA to create a human equal the cost of teaching a student in Stanford? Both questions refer to the very real possibility that it may soon be in the realm of affordability to print off whatever stretch of DNA you so desire, using genetic sequencing and a machine capable of synthesizing the four building blocks of DNA A, C, G, and T into whatever order you desire.

The answer to the time question, by the way, is 19 years, given that the cost of tuition at Stanford remains at $50,000 and the price of genetic printing continues the 200-fold decrease that has occurred over the last 14 years. Precursory work has already been performed; a team lead by Craig Venter created the simplest life form ever known last year.

Stanfords moral question, though, is a little trickier. The question is part of a larger conundrum concerning humans interfering with their own biology; since the technology is developing so quickly, the issue is no longer whether we can or cant,but whether we should or shouldnt. The debate has two prongs: gene editing and life printing.

With the explosion of CRISPR technology many studies are due to start this year the ability to edit our genetic makeup will arrive soon. But how much should we manipulate our own genes? Should the technology be a reparative one, reserved for making sick humans healthy again, or should it be used to augment our current physical restrictions, making us bigger, faster, stronger, and smarter?

The question of printing life is similar in some respects; rather than altering organisms to have the desired genetic characteristics, we could print and culture them instead billions have already been invested. However, there is theadditional issue of playing God by sidestepping the methods of our reproduction that have existed since the beginning of life. Even if the ethical issue of creation was answered adequately, there are the further questions ofwho has the right to design life, what the regulations would be, and the potential restrictions on the technology based on cost; if its too pricey, gene editing could be reserved only for the rich.

It is vital to discuss the ethics of gene editing in order to ensure that the technology is not abused in the future. Stanfords question is praiseworthy because it makes todays students, who will most likely be spearheading the technologys developments, think about the consequences of their work.

Read more:

Stanford's Final Exams Pose Question About the Ethics of Genetic Engineering - Futurism

America’s First Free-Roaming Genetically Engineered Insects Are … – Gizmodo

Diamondback moths may be a mere half-inch in length, but their voracious appetite for Brussels sprouts, kale and cauliflower make them a major pain for farmers. This week, the U.S. Department of Agriculture approved a potential solution: moths genetically engineered to contain a special gene that makes them gradually die off. A field trial slated to take place in a small area of upstate New York will become the first wild release of an insect modified using genetic engineering in the US.

The moths have been engineered by the British biotech firm Oxitec, the same company that last year caused a stir with its plans to release genetically modified, Zika-fighting mosquitoes in the Florida Keys. The diamond back moths take a similar approach to the mosquitoes, modifying male mosquitoes to limit the population over time by passing on a gene to offspring when it mates with wild females that causes female moths to die before they reach maturity.

The technique is a riff on an approach used to manage agricultural pests since the 1950s, known as sterile insect technique. Using radiation, scientists made insects like the screwworm unable to produce viable offspring. By 1982, screwworm was eradicated from the US using this alternative to pesticides. In Silent Spring Rachel Carson suggested this approach was the solution to the dangers of harmful pesticides agricultural producers required to protect their crops. The problem was that it did not work on every insectin many cases, it simply left irradiated insects too weak to compete for mates with their healthier kin.

Diamondback moths are a sizable problem for farmers, and a problem thats growing as the moths develop resistance to traditional pesticides. They do about $5 billion in damage to cruciferous crops worldwide every year. In the upcoming trial, a team at Cornell University will oversee the release of the genetically engineered moths in a 10-acre field owned by Cornell in Geneva, New York.

After a review found that the field trial is unlikely to impact either the environment or humans, the USDA issued a permit that allows for the release of up to 30,000 moths per week over several months. It is caterpillars that damage crops, so the plan to release adult males that produce unviable offspring should not cause any additional crop damage. And any surviving moths will likely be killed off by pesticides or upstate New Yorks frigid winter, according to the report submitted to the USDA.

The plan to release modified mosquitoes in the Keys attracted much local ireafter initially getting the greenlight from the FDA, the project was ultimately stalled by a local vote and forced to find a new location for a trial.

In upstate New York, too, the moths have stirred up a debate over GMOs for the past several years, though the plan has not been met with quite the same level of opposition. The approval process through the USDA rather than the FDA, too, was much swifter.

In laboratory and greenhouse trials, the modified mosquito was reportedly effective in decreasing the overall population. But tests still need to determine how it will fare in open air.

Oxitec has released its engineered mosquitoes Brazil, Grand Cayman, and Panama, and still plans to go ahead with a field trial in the Keys. In December, the company announced plans for field trials of a genetically modified Mediterranean fruit fly in Western Australia. It is also working on genetically engineering several other agricultural pests, including Drosophila suzukii and the Olive fly.

Excerpt from:

America's First Free-Roaming Genetically Engineered Insects Are ... - Gizmodo

Genetically modified food is too advanced for its out-of-date regulations – The Hill (blog)

Last week, the USDA published a series ofquestionsseeking input to establish a National Bioengineered Food Disclosure Standard, as mandated by amendments to the Agricultural Marketing Act of 1946 that went into effect in July 2016.

TheNational Bioengineered Food Disclosure Standard Actrequires the Secretary of the Department of Agriculture to establish disclosure standards for bioengineered food. The Act preempts state-based labeling laws for genetically modified organisms (GMOs), such as those adopted inVermontlast year.

The USDA is considering public input on the disclosure standards untilJuly 17, 2017. Two key issues are under consideration. The first is whether certain genetic modifications should be treated as though they are found in nature for example, a mutation that naturally confers disease resistance in a crop. The second concerns what types of breeding techniques should be classified as conventional breeding among "conventional breeding" techniques are hybridization and the use of chemicals or radiation to introduce random genetic mutations.

These seemingly mundane questions strike at the heart of GMO controversies and implicate the use of breakthrough CRISPR gene editing technologies. Gene editing allows novel and precise genetic modifications to be introduced into crops and animals intended for human consumption. The answers to the USDA's questions are significant because the Disclosure Standard Act exempts from mandatory disclosure genetic modifications obtained without recombinant DNA (rDNA) techniques that can otherwise be found in nature.

Older genetic engineering methods relying on rDNA technology combine genes from foreign organisms and transfer them into plant or animal cells thus, creating "transgenic" organisms.

However, CRISPR gene editing need not rely on using any foreign DNA and can introduce genetic modifications that mirror those already found in nature. Unlike rDNA and conventional breeding methods, CRISPR technologies introduce genetic changes with far greater accuracy and precision.

In 2016, the USDAdeclined to regulatetwo CRISPR crops a mushroom and a waxy corn under regulations governing traditionalGMOs. But other regulatory agencies, including the FDA and EPA, have not yet made determinations on crops or animals modified with CRISPR technology, and uncertainty looms concerning the regulatory status of this new breed ofGMOs.

Opponents ofGMOs, who commonly argue thatGMOsare harmful to human health, decried the USDA's decision not to regulate CRISPR crops and argued thatpowerful corporations had found ways to circumvent the law through technical loopholes in outdated regulations.

Yet three decades of scientific research suggest that present-dayGMOcontroversies are not grounded in scientific fact. For instance, despite frequent rumors aboutGMO-induced cancers, a scientific consensus has now formed to support the health and environmental safety of genetically modified crops for animal and human consumption. That proposition is supported by investigations of theU.S. National Academies of Science, Engineering, and Medicineas well as scientific panels including the American Association for the Advancement of Science, the American Medical Association, the European Commission, and National Academies of Science in Australia, Brazil, China, France, Germany, India, the United Kingdom, and other countries.

In its rulemaking process, the USDA should rely upon science and facts. With regard to crops and animals with DNA altered through gene editing, rulemakers ought to distinguish among ways that CRISPR technology may be used to edit genes. For instance, CRISPR technology can be used as a DNA construct that is incorporated into the DNA of plant or animal cells, or as a preassembled RNA and protein complex.

How gene editing is carried out matters, because some methods appear to fall within the disclosure requirements while others do not. The law definesbioengineered foodas food that contains genetic material modified through in vitro rDNA techniques. Thus, under the Disclosure Standard Acts statutory constraints, CRISPR food created using DNA constructs that are incorporated into plant or animal cells would likely fall under the mandatory disclosures.

However, food derived from rDNA-free CRISPR gene editing using transient preassembled RNA and protein complexes should be excluded from the bioengineered food definition because such complexes are degraded shortly after gene editing takes place and do not insert themselves into the target organism DNA.

The nuances of ever-evolving biotechnological innovation highlight the complexity of our regulatory system and the need to modernize it. The National Bioengineered Food Disclosure Standard Act is just one of the latest pieces of that regulatory patchwork to emerge. Rules establishing bioengineered food disclosures should be coherent and science-based. Gene editing that uses no foreign DNA, is more precise than conventional breeding methods, and causes genetic modifications already found in nature should not be subject to onerous disclosure standards.

Paul Enrquez is a lawyer and scientist currently doing research in Structural & Molecular Biochemistry at North Carolina State University. His work focuses on the intersection of science and law and has been featured in both legal and scientific journals. He explores rising legal and regulatory issues concerning genome editing in crop production in depth and makes policy recommendations in his recently published article CRISPRGMOs.

The views expressed by contributors are their own and not the views of The Hill.

Read more here:

Genetically modified food is too advanced for its out-of-date regulations - The Hill (blog)

New Molecular Scalpel Acts as GPS to Improve Genetic Editing – Bioscience Technology

Researchers from the University of Copenhagen (Denmark), led by the Spanish researcher Guillermo Montoya, have discovered how Cpf1, a new molecular scissors unzip and cleave DNA. This member of the CRISPR-Cas family displays a high accuracy, capable of acting like a GPS in order to identify its destination within the intricate map of the genome. The high precision of Cpf1 will improve the use of this type of technology in repairing genetic damage and in other medical and biotechnological applications.

A scientific team from in the Novo Nordisk Foundation Center for Protein Research (NNF-CPR), at the University of Copenhagen, has succeeded in visualizing and describing how a new system for genome editing, known as Cpf1, works. This protein belongs to the Cas family and enables the cleavage of double stranded DNA, thus allowing the initiation of the genome modification process. The results of the study have been published in the journalNature.

Guillermo Montoya, a researcher in the fields of biochemistry and molecular biology who led the study, explains to SINC that the new molecular scissors "will enable us to more safely modify and edit the instructions written in the genome, due to the utmost precision of the target DNA sequence recognition".

The CRISPR Cas9 system for cutting and paste genome sequences is already being used to modify animal and plant genomes. Also to treat illnesses, such as cancer and retinal diseases, in humans and its applications are growing very fast.

X-Ray Crystallography Technique

Researchers across the world are trying to perfect this genome editing technique with the aim of making it yet more precise and efficient. To achieve this, they have also focused on other proteins that specifically cut DNA, such as Cpf1, whose manipulation can direct them to specific locations in the genome. Montoya's team has achieved this using an X-ray Crystallography to decipher the molecular mechanisms controlling this process.

"We radiated the crystals of the Cpf1 protein using X-rays to be able to observe its structure at atomic resolution, enabling us to see all its components," points out the co-author of this study. "X-ray diffraction is one of the main biophysical techniques used to elucidate biomolecular structures," he continues.

In his opinion, "the main advantage of Cpf1 lies in its high specificity and the cleaving mode of the DNA, since it is possible to create staggered ends with the new molecular scissors, instead of blunt-ended breaks as is the case with Cas9, which facilitates the insertion of a DNA sequence."

"The high precision of this protein recognising the DNA sequence on which it is going to act functions like a GPS, directing the Cpf1 system within the intricate map of the genome to identify its destination. In comparison with other proteins used for this purpose, it is also very versatile and easy to be reprogrammed," Montoya adds.

Genetic diseases and tumours

These properties make this system "particularly suitable for its use in the treatment of genetic diseases and tumours," he affirms.

The team has previously worked with the French biotechnology company Celletics on the use of meganucleases -other proteins that can be redesigned to cut the genome in a specific location- to treat certain types of leukemia.

The new technology "can also be used to modify microorganisms, with the aim of synthesising the metabolites required in the production of drugs and biofuels," adds Montoya.

This researcher, from Getxo (Biscay, Spain), says that there are many companies interested in this new technology. They are mostly from the biotechnology sector in the field of microorganism manipulation, but cannot be named due to confidentiality agreements.

Here is the original post:

New Molecular Scalpel Acts as GPS to Improve Genetic Editing - Bioscience Technology

Stop GM mustard release as it will harm farmers: scientists to PM – Livemint

New Delhi: Stating that genetically modified (GM) mustard crop, which is waiting for approval for commercial cultivation from Indias environment ministry , could harm farmers of the country, a group of scientists on Thursday wrote to Prime Minister Narendra Modi requesting him to stop the release of GM mustard.

The group that wrote to the prime minister on Thursday includes Dr Sharad Nimbalkar, former vice-chancellor of Panjabrao Agriculture University, Akola, in Maharashtra.

In their letter, the scientists said they are well informed that the 21st century is the era of science and technology and that technologies emerging through use of science cannot be neglected. However, it is very painful to see a few learned scientists in India, positioned in higher hierarchies in government administration, try to introduce technology harmful to Indian farmers and to our assets of biodiversity..., said the letter, which was reviewed by Mint.

Moreover, they have a hidden agenda to exhaust indigenous seeds of crops... with those under patent regimes of foreign multinationals. There is sufficient ground to doubt their integrity towards Indian farmers, who are being made more dependent on monopolised seeds under IPR regimes of foreign multinational companies, they added.

Indias regulator for GM productsthe Genetic Engineering Appraisal Committee (GEAC)on 11 May recommended approval for commercial production of GM mustard, developed by the Delhi University-based Centre for Genetic Manipulation of Crop Plants.

The application is currently awaiting a final nod from Union environment minister Harsh Vardhan. Faced with opposition from civil society groups, farmer groups, consumer bodies and others, the ministry has planned a series of meetings with opinion makers and stakeholders before he takes a final decision.

At present, cotton is the only GM crop allowed in India and if the environment ministry grants permission, GM mustard will become first GM food crop in India. In 2010, GEAC had cleared GM brinjal but the ministry did not clear it in the wake of protests from civil society groups.

The scientists warned that the technology used in preparing GM mustard could lead to indiscriminate use of herbicides, endangering indigenous varieties of mustard.

Alleging a nexus between promoters of GM mustard and GEAC, the scientists asked the prime minister to urgently intervene.

GM mustard is also facing resistance from groups like the Swadeshi Jagran Manch, an affiliate of the Rashtriya Swayamsevak Sangh (RSS), which is the ideological parent of the ruling Bharatiya Janata Party.

First Published: Thu, Jul 06 2017. 08 31 PM IST

See original here:

Stop GM mustard release as it will harm farmers: scientists to PM - Livemint

IARPA seeks tech to ID bioengineered life forms – FCW.com (blog)

IARPA seeks tech to ID bioengineered life forms

WHAT: A new bio-detection tech development effort to help defend against human engineered biological threats.

WHY: With advances in genetic engineering and gene editing, the intelligence community is concerned about possible threats from chimerical life forms.

The research arm of the intelligence community is hoping new bio-detection technology can be developed to help defend against human engineered biological threats.

That idea might sound a bit familiar to science fiction buffs.

It is reminiscent of the science fiction film "Blade Runner," an adaptation of Philp K. Dicks classic 1968 novel "Do Androids Dream of Electric Sheep?" In the story, special futuristic cops are charged with defending human civilization from the depredations of genetically engineered android super-soldiers dubbed replicants.

In a June 19 announcement, the Intelligence Advanced Research Projects Activity said it is looking for technology that can detect human engineered changes to natural biological systems.

Emerging genetic editing tools have the potential to aid in the development of new vaccines and pharmaceuticals and to create hardy strains of crops. However, in the wrong hands, these tools could also be used to warp organisms into deliberate weapons or be misused in ways that could "accidentally or deliberately" threaten national health, security or the economy, according to the solicitation.

IARPA said its Finding Engineering-Linked Indicators (FELIX) program looks to develop new tech that can spot genetically engineered changes within biological systems to spur "mitigation responses to unlawful or accidental release of organisms." IARPA said it wants to develop a suite of tools to detect a range of engineered bio-organisms from viruses, bacteria, insects, animals and plants that have been developed from natural organisms "that are either purposefully or accidentally developed and/or released with the potential to cause harm."

IARPA plans a proposers' day on July 27 before it sends out a formal solicitation for the technology.

IARPA said technologies it wants to discuss include novel methods and high throughput techniques in genomics, systems biology, bioinformatics and evolutionary biology.

The tools it's aiming to develop could find genetic signatures that haven't been accessible before with previous technologies, using data from multiple interrogation points, increasing sensitivity, improving the quality of the data and leveraging technologies that can increase throughput and reduce the complexity of sample analysis.

IARPA said it envisions FELIX development as a two-phase program. The first phase, it said, is to develop platforms and technologies that can be made general enough to detect "signatures" that would give away engineered biological systems and develop modeling and analysis of those indicators.

The second phase, IARPA said, will optimize the platform, analysis tools and technologies to detect increasingly complex and sophisticated changes in biological systems and find those engineered changes in a variety of organisms and sample types.

Click here to read the full announcement.

Posted by Mark Rockwell on Jul 05, 2017 at 12:33 PM

Link:

IARPA seeks tech to ID bioengineered life forms - FCW.com (blog)

‘Woolly’ Breathes New Life Into A Scientific Saga – NPR

In the winter of 1990, George Church and Ting Wu he resplendent in his bushy beard, she wearing a skirt, which she rarely did rode their bicycles to city hall in Cambridge, Mass., to be wed. For years they kept their marriage an open secret, and that relationship would have ramifications, both positive and otherwise, for their careers: They worked together in a Harvard lab, trying to unlock the secrets of DNA.

Ben Mezrich's new book, Woolly, is about science's attempt in recent years to use genetic engineering to revive the extinct woolly mammoth. But as with his previous bestselling works of narrative nonfiction such as Bringing Down the House, the basis of the film 21, and The Accidental Billionaires, the basis of the film The Social Network Woolly dwells on close-ups before zooming out to the big picture.

Church and Wu are two of the main characters in Mezrich's taut yet detailed dramatization. Theirs is a synergistic relationship, and while it would be an overreach to call Woolly a love story at heart, the couple's dynamic is one of the essential threads of Mezrich's story. By all accounts geniuses, the two form the nucleus of a group of Harvard scientists whose revolutionary research leads them to a staggering conclusion: They must use their knowledge and abilities to manipulate the genome of Mammuthus primigenius, the hairy pachyderm that perished from the face of the earth over 3,000 years ago.

Their reasons, as Mezrich spells out, are more than academic. By pioneering the methods it would take to clone a mammoth and gestate the fetus successfully in the womb of an elephant, Church, Wu, and crew would open the door to further efforts to revive extinct species and, through the impact these reintroduced species would have on the environment, to help reverse the damages that modern civilization has had on Earth's ecosystem and climate.

The Harvard group isn't the only one working toward this end. In Russia, the father-son team of Sergey and Nikita Zimov launch Pleistocene Park, a wildlife preserve on the steppes of Siberia, where the mammoth once freely roamed and where they could possibly roam once more.

It all sounds very Jurassic Park, of course, and Mezrich doesn't hesitate to draw that parallel. The hubris of such scientific endeavors, as well as the ethical issues involved, crop up in Woolly, although it's clear the author's sympathies lie with his subjects. Anecdotes like the wedding of Church and Wu form the backbone of the book, rather than serving as ornament. Mezrich's eye for characterization is as sharp as his ability to break down scientific jargon into easily digestible chunks.

The true protagonists of Mezrich's saga, though, are the great mammoths themselves. Through his fluid use of close perspective, poetic license, and present-tense recreations of past events not to mention his occasional speculation into the future the author dramatically illustrates his tale. It's paced like a thriller, with the frustrating politics of the research industry bleeding over into the maneuverings of capitalists who see dollar signs in investing in widespread genetic engineering. Mezrich also frequently reconstructs dialogue between the plot's players, which at times feels overly contrived and distracting.

Thankfully it's not enough to inhibit the intimate look into the lives of the men and women who are humbly and at time not so humbly hoping to put the power of creation at their fingertips. With all the passion and vision of the scientists seeking to bring the mammoth back to life, Woolly reanimates history and breathes new life into the narrative of nature.

Jason Heller is a senior writer at The A.V. Club, a Hugo Award-winning editor and author of the novel Taft 2012.

Read more from the original source:

'Woolly' Breathes New Life Into A Scientific Saga - NPR

The race to revive woolly mammoths using ancient DNA – CBS News

Like "Jurassic Park," what if you could use the science of DNA to resurrect long-extinct creatures that once roamed the earth?

Efforts to do that are actually underway.

Led by Dr. George Church "the Einstein of our times," according to author Ben Mezrich a lab at Harvard Medical School is working on bringing back the woolly mammoth through genetic engineering.

The process is detailed in Mezrich's new book, "Woolly: The True Story of the Quest to Revive One of History's Most Iconic Extinct Creatures," published by Atria Books, an imprint of Simon & Schuster which is a division of CBS. It's also being made into a movie.

"The woolly mammoths are coming up out of the ice. So the permafrost that is slowly getting warmer, these bodies are coming out and they're taking the genetic material and then they are synthesizing it and they're placing [it] into the cells of an Asian elephant so that an Asian elephant gives birth to a woolly mammoth," Mezrich said on "CBS This Morning: Saturday." "So essentially, you're recreating the mammoth using its relative that still exists today."

Mezrich likened the permafrost to "the ring of the world."

CBS News

"It's like a ticking time bomb. It contains within it more carbon than if we burned all the forests on Earth three times," Mezrich said. "And these Russian scientists, the Zimovs, have shown since the 80s that if you repopulate it with herbivores from the Pleistocene era -- and they're using tanks to mimic woolly mammoths and they're putting bison there -- they've managed to lower the temperature of the tundra by 15 degrees. So the goal is to put a herd of woolly mammoths in Siberian tundra to keep the permafrost from melting."

Mezrich said the woolly mammoths will help the world in an out-of-the-box way.

"I mean, elephants don't get cancer, which is very strange. Elephants have thousands and thousands of more cells than us. And why they don't get cancer is in their genes. If we can figure that out, we can use this genetic engineering to solve cancer," he said.

The author also addressed the ethical concerns related to these types of genetic engineering practices.

"The idea of playing God, the idea of making a mistake, of letting something out of the lab, these things come up. And that's why you need responsible scientists. Dr. George Church is an incredibly good person and you need people like that doing this because this box is open. The Pandora's box of this technology is here," Mezrich said. "There are labs all over the world not just making woolly mammoths but doing things that 10 years from now are going to have huge repercussions. So we want responsible scientists doing this."

2017 CBS Interactive Inc. All Rights Reserved.

Originally posted here:

The race to revive woolly mammoths using ancient DNA - CBS News

Scientific finding paves way for rice genetic engineering to develop efficient water storage – InterAksyon

Researchers at the Australian National University (ANU) have found a way to survive drought conditions 50 percent longer by leveraging chloroplasts to initiate the conservation of water by causing plant pores or stoma to close as temperatures rise.

The plant pumps water into the leaves, which, in turn, absorb carbon dioxide. Chlorophyll, water, carbon dioxide, and other food producing substances are available inside the chloroplast, and the entire process of photosynthesis takes place in the chloroplast.

Scientists observed that chloroplasts can sense drought stress and always activate a chemical that closes the plants pores or stomata to conserve water.

This finding was revealed in the article written by lead author Dr. Wannarat Pornsiriwong and others entitled A chloroplast retrograde signal, 3-phosphoadenosine5-phosphate, acts as a secondary messenger in abscisic acid signaling in stomatal closure and germination, in the peer-reviewed journal eLife on March 21, 2017.

The research team, led by Dr. Wannarat Pornsiriwong, Dr. Gonzalo Estavillo, Dr. Kai Chan and Dr. Barry Pogson from the Australian National University (ANU) Research School of Biology, found that chloroplasts, better known for their role in photosynthesis, play a role in regulating plant response during heat stress.

Chloroplasts are actually capable of sensing drought stress and telling the leaves to shut up and prevent water from being lost during drought stress, Dr. Pogson added.

So the chloroplasts are actually helping the plants to prevent losing too much water. We know how the drought alarm actually calls for help and we know how help comes in the form of closing pores on the leaves, he stressed.

Boosting the levels of this chloroplast signal also restores tolerance in drought-sensitive plants and extended their drought survival by about 50 per cent, Dr. Chan added.

By increasing the activity of the chloroplasts or stimulating this chemical signal in another way, plants could store water for a longer period and survive despite higher temperatures.

Through this specific function of chloroplasts, plant geneticists may employ genetic modification (GM) to develop plants with more spores or crops that have roots and stems big enough to store water the way pineapple, watermelon and turnips do.

This finding is significant as climatologists have predicted more intense global warming that could reduce rice yields.

The finding would also boost efforts by biotechnologists and plant breeders to coax rice varieties to use their chloroplasts efficiently to help the plants store water and thus support a bigger volume of rice grains in a panicle.

This basic scientific research has the potential to improve farming productivity in countries that suffer from drought stress, Dr. Pogson said.

If we can even alleviate drought stress a little, it would have a significant impact on our farmers and the economy, he added.

Excerpt from:

Scientific finding paves way for rice genetic engineering to develop efficient water storage - InterAksyon

Biotechnology confusion: Differences among GMOs, gene editing and genetic engineering – Genetic Literacy Project

Your body contains trillions of cells which make up the physical you. Each one of these cells has a blueprint that is completely unique to you, called yourDNA.

In order to read all that information on your DNA, we use machines that do gene sequencing. A gene is a distinct stretch of DNA that determines something about who you are. Gene sequencing is where we can go through and laboriously read every single character in your DNA and then store it in a big file.

What if we couldchange genes in order to start changing your attributes? This is now possible using a technology calledgene editing.This is where we are able to precisely snip sections of DNA from the strand and then replace them with our own snippets.

All these methods fall under the envelope of genetic engineering. Consequently, gene editing is just another form of genetic engineering.

Genetic engineering is the direct manipulation of an organisms DNA using any number of methods. GMO is the genetic modification of organisms. Its been around for a while and uses imprecise methods of genetic engineering. Gene editing is now a more precise method of genetic engineering which hopes to avoid any bad associations with GMO.

The GLP aggregated and excerpted this blog/article to reflect the diversity of news, opinion, and analysis. Read full, original post: GMO vs Gene Editing vs Genetic Engineering

Originally posted here:

Biotechnology confusion: Differences among GMOs, gene editing and genetic engineering - Genetic Literacy Project

Avoiding CRISPR-Mediated Gene-DriveEvolved Resistance in Mosquitoes – Genetic Engineering & Biotechnology News (blog)

Gene drives are used to bias genetic inheritance in favor of rapidly spreading, self-destructive genes and could be an environmentally friendly and cost-effective way to suppress populations of disease-spreading insects. The rise of CRISPR/Cas9 gene-editing technology has recently revolutionized gene-drive systems because it offers a rapid, efficient, and reliable way to make precise, targeted changes to the genome.

The new study based its calculations on past gene-drive findings that resulted in up to 99% of offspring inheriting the inserted gene. However, the few offspring that don't inherit the gene present a big problem for this technology. Since a fraction of these offspring is immune to the gene drive, any attempt to eliminate a mosquito species in this manner would result in a rapid rebound of those that are gene-drive immune. The impact of this resistance on the ability of gene drive to spread and suppress populations had previously been discussedbut had not been thoroughly evaluated.

The mathematical modeling that the investigators utilized found that the gene-driveevolved resistance would have a major impact on attempts to eliminate a mosquito species on a continent-wide scale. To address this issue, the research team devised a technique that they determined could potentially suppress mosquito species continent-wide.

Employing a strategy called multiplexing, which involves using one of the components of the CRISPR system, a gRNA, to target multiple locations in a gene at once, the research team suggested that the size of the population that could be suppressed increases exponentially with the number of these gRNAs utilized. It also shows that with four or five multiplexed gRNAs, a mosquito species could potentially be suppressed on a continental scale.

"Knowing that we can potentially overcome the issues of resistance through careful engineering and multiplexing is huge," noted senior study investigator Omar Akbari, Ph.D., assistant professor of entomology at UC Riverside.

The researchers demonstrated the technology was feasible using a fruit fly model. Now they are working to adapt this technology to the mosquito species that transmit malaria, dengue, and Zika.

"The potential of multiplexing is vast. With one gRNA, we could suppress a room of mosquitoes," Dr. Marshall concluded. With four, we could potentially suppress a continent and the diseases they transmit. But nature has a knack for finding a way around hurdles, so assessing that potential will require a lot more work."

More:

Avoiding CRISPR-Mediated Gene-DriveEvolved Resistance in Mosquitoes - Genetic Engineering & Biotechnology News (blog)

Genetic engineering tool generates antioxidant-rich purple rice – Phys.Org

June 27, 2017 A photograph of purple endosperm rice. Credit: Qinlong Zhu of the South China Agricultural University

Researchers in China have developed a genetic engineering approach capable of delivering many genes at once and used it to make rice endospermseed tissue that provides nutrients to the developing plant embryoproduce high levels of antioxidant-boosting pigments called anthocyanins. The resulting purple endosperm rice holds potential for decreasing the risk of certain cancers, cardiovascular disease, diabetes, and other chronic disorders. The work appears June 27th in the journal Molecular Plant.

"We have developed a highly efficient, easy-to-use transgene stacking system called TransGene Stacking II that enables the assembly of a large number of genes in single vectors for plant transformation," says senior study author Yao-Guang Liu of the South China Agricultural University. "We envisage that this vector system will have many potential applications in this era of synthetic biology and metabolic engineering."

To date, genetic engineering approaches have been used to develop rice enriched in beta-carotene and folate, but not anthocyanins. Although these health-promoting compounds are naturally abundant in some black and red rice varieties, they are absent in polished rice grains because the husk, bran, and germ have been removed, leaving only the endosperm.

Previous attempts to engineer anthocyanin production in rice have failed because the underlying biosynthesis pathway is highly complex, and it has been difficult to efficiently transfer many genes into plants.

To address this challenge, Liu and his colleagues first set out to identify the genes required to engineer anthocyanin production in the rice endosperm. To do so, they analyzed sequences of anthocyanin pathway genes in different rice varieties and pinpointed the defective genes in japonica and indica subspecies that do not produce anthocyanins.

Based on this analysis, they developed a transgene stacking strategy for expressing eight anthocyanin pathway genes specifically in the endosperm of the japonica and indica rice varieties. The resulting purple endosperm rice had high anthocyanin levels and antioxidant activity in the endosperm. "This is the first demonstration of engineering such a complex metabolic pathway in plants," Liu says.

In the future, this transgene stacking vector system could be used to develop plant bioreactors for the production of many other important nutrients and medicinal ingredients. For their own part, the researchers plan to evaluate the safety of purple endosperm rice as biofortified food, and they will also try to engineer the biosynthesis of anthocyanins in other crops to produce more purple endosperm cereals.

"Our research provides a high-efficiency vector system for stacking multiple genes for synthetic biology and makes it potentially feasible for engineering complex biosynthesis pathways in the endosperm of rice and other crop plants such as maize, wheat, and barley," Liu says.

Explore further: The origin and spread of 'Emperor's rice'

More information: Molecular Plant, Zhu et al.: "Development of ''Purple Endosperm Rice'' by Engineering Anthocyanin Biosynthesis in the Endosperm with a High-Efficiency Transgene Stacking System" http://www.cell.com/molecular-plant/abstract/S1674-2052(17)30140-5 , DOI: 10.1016/j.molp.2017.05.008

Journal reference: Molecular Plant

Provided by: Cell Press

Black rice has a rich cultural history; called "Forbidden" or "Emperor's" rice, it was reserved for the Emperor in ancient China and used as a tribute food. In the time since, it remained popular in certain regions of China ...

We've all heard examples of animal altruism: Dogs caring for orphaned kittens, chimps sharing food or dolphins nudging injured mates to the surface. Now, a study led by the University of Colorado Boulder suggests some plants ...

(Phys.org)Hybrids of many plant and animal species and subspecies are sterile, and a group of researchers in China have now identified the genes that operate to make crossbred rice sterile.

The origins of rice have been cast in a new light by research publishing in the open-access journal PLoS Genetics on June 9, 2011. By reconciling two theories, the authors show that the domestication of rice occurred at least ...

Anthocyanins, plant pigments known for their health-promoting properties, are in demand for medicinal and industrial uses. Anthocyanins have become sought-after natural products, but the small number of plants that naturally ...

Anthocyanins, pigments that give plants their red, blue, or purple hues, are not typically produced in citrus fruits grown under tropical or subtropical conditions. Now, scientists have genetically engineered a lime that ...

When insects skip the light fandango their romantic foreplay often involves some pretty crazy things like hypnotic dance moves and flashy colors. In some species it ends with a complex ejaculate package that does more than ...

A study of yeast reveals new mechanism that allows cells to adapt to environmental changes more rapidly by accelerating genetic changes around genes that boost fitness, publishing 27 June in the open access journal PLOS Biology, ...

In the fight against the viruses that invade everyday life, seeing and understanding the battleground is essential. Scientists at the Morgridge Institute for Research have, for the first time, imaged molecular structures ...

Researchers from the University of Zurich and the University Hospital Zurich have discovered the protein that enables natural embryonic stem cells to form all body cells. In the case of embryonic stem cells maintained in ...

For us humans, it goes without saying that we reward others as an indication of the gratitude we feel towards them. Scientists from the Max Planck Institutes for Evolutionary Anthropology and for Mathematics in the Sciences ...

Researchers in China have developed a genetic engineering approach capable of delivering many genes at once and used it to make rice endospermseed tissue that provides nutrients to the developing plant embryoproduce ...

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

Read the original:

Genetic engineering tool generates antioxidant-rich purple rice - Phys.Org

Simple, affordable tests can prevent genetic disorders – Khaleej Times

Premarital tests are so important, but what's most important is for couples to understand the consequences of their union

Over 400 genetic disorders are present among the UAE's populace, however, its continued spread can be prevented by simple and affordable tests, some costing as little as Dh30, according to local health experts.

Dr Ebtehaj Al Anizi, Obstetrics and Gynaecology specialist, told Khaleej Times that genetic disorder prevention tests are crucial, especially when it comes to consanguineous marriages. However, she stressed that not all couples take the results, nor the doctor's advice seriously, leading to devastating consequences for their offspring's health.

"As marriage between relatives is common in the region, so is the risk of genetic diseases. A lot of high risk couples do no listen to us when advised not to go ahead with the marriage," Dr Al Anizi said.

Dr Al Anizi said the most common genetic disorders in the UAE include thalassemia, sickle cell anaemia, G6PD deficiency, cystic fibrosis and haemophilia. Raising awareness about the suffering of future children who could inherit the disease is vital. "If both parents are thalassemia minor, then their offspring, who will have thalassemia major, will suffer greatly. The child will have chronic anaemia, and their lives will depend on frequent blood transfusions."

Dr Al Anizi said this is when doctors often advise couples to not get married. "These kids suffer a lot: they have anaemia, fatigue, the structure of their faces and bones change, they have enlargement in the belly, and unfortunately, often die before they reach 10."

She said sick cell disease can also be prevented in the next generation, by testing both parties. "If the child inherits this disease, it really is a crisis for the entire family."

Premarital tests important

Dr Al Anizi stressed that common health problems, including a severe drop in oxygen levels, body and chest pain, as well as fatigue, will arise. "Children are often admitted to the emergency department and need heavy medication and painkillers. The pain is intolerable.

"Premarital tests are so important, but what's most important is for couples to understand the consequences of their union."

Dr Mariam Mater, founder and chairperson of the UAE Genetic Diseases Association (UAE GDA), said prevention tests are accessible in the UAE and are cost effective in the long run. "The cost of genetic screening of thalassemia is Dh120, and the cost of treating a patient is Dh35,000 per annum."

She stressed that the risk of neural tube defects, which are also common in the UAE, can also be prevented by a course of folic acid, which costs approximately Dh30, whereas treating a neural tube defects case costs a whopping Dh2.7million.

"Prevention is the key in reducing the impact of genetic disorders, socially and economically, and is a long-term sustainable solution, especially in a country like ours where close to 60 per cent of the population is under 30 years of age."

Genetric engineering helps

Genetic engineering could help couples in the UAE conceive healthy children. Dr Rashmi Mathai, Internal Medicine specialist, Universal Hospital, said a recent research in the US revealed that genetic engineering may help curb the manifestation of genetic disorders in the next generation, which will be a huge relief for couples in the UAE, if made available in the country.

"A patient carrying a mutation that carries a genetic disease, such as cystic fibrosis, may simultaneously carry a mutation in another gene that buffers the effect of the harmful gene. This buffer gene unravels a whole new treatment modality for those carrying genetic diseases.

"This type of genetic engineering will certainly help a lot of couples if it comes to the UAE," Dr Mathai said.

However, Dr Mathai pointed out that couples in the UAE cannot depend on waiting to see what the future of genetic engineering unravels, and must thus opt for what is already available, to help prevent the increasing genetic diseases statistics. "The closer the blood relation between the spouses, the higher the chance the children will carry the disease. Public awareness and premarital counselling are the most important aspects, because it could help save lives."

jasmine@khaleejtimes.com

MORE FROM News

With over 200 nationalities residing in the UAE, the country is a... READ MORE

Children also breathe faster, hence are more susceptible to illnesses ... READ MORE

With the three-week programme, students and parents managed to create ... READ MORE

Among other side effects of black henna are blisters, redness, cuts,... READ MORE

MORE FROM Khaleej Times

The police have detained several friends and acquaintances of the... READ MORE

The number of companies and agencies reportedly affected by the... READ MORE

The citizens of Switzerland and the United Kingdom are also part of... READ MORE

The amendments also permit no more than 50 per cent glass tinting for ... READ MORE

Continue reading here:

Simple, affordable tests can prevent genetic disorders - Khaleej Times

HIV fix: Can gene editing work alongside the virus to provide a cure? – Genetic Literacy Project

Gene therapy and management of human immunodeficiency virus (HIV) infection haveeventful histories going back several decades. Both are saving lives today and both are innovating in ways that will lead soon to a convergence. In the years to come, gene therapy and other therapeutic genetic engineering modalities will be used against HIV. The conventional strategy for gene therapy is addition of new genes to a patients genome. ButHIV requires new approaches inspired by anti viral tricks from nature if we hope to reduce the damage being done to third-world nations by the virus.

The currentstrategy in gene therapy is based onlimiting,replacing or supplementing defective genes. This is a workable approach for recessive genetic diseases. However, right around the corner is the option of deleting genes, and subsequently line-item editing of genes that are too large to fit inside AAV, a virus that gene therapists commonly use as a carrier for genetic payloads. Both gene deletion and line-item editing of larger genes can be achieved using CRISPR genome editing carried inside an AAV. But ironically, the HIV itself could work better as a carrier for anti-HIV gene therapy. At the same time, modulating the immune response by suppressing gene activity without deleting the relevant gene is showing promise in laboratory studies. This approach utilizes a non-CRISPR genetic engineering tool, RNA interference (RNAi).

As gene therapy has evolved, scientists have anticipated its eventual use as an HIV treatment. Butwhy consider gene therapy against an infectious disease? For inherited enzyme deficiencies like cystic fibrosis and Tay-Sachs disease, and for cancer, the potential for gene therapy seems fairly intuitive. But a well-known feature of infectious diseases is their ability to generate an immune response. When exposed to a foreign agent, our bodies launch a T-cell and antibody response. For the vast majority of diseases that impacted human mortality at the dawn of the 20th century, medical researchers were able to boost the immune response with vaccines. Polio, diphtheria, measles all the major infectious killers that plagued our ancestors are prevented today. But a minority of microbial nemeses have evolved particularly devious tricks. Mycobacterium tuberculosis is one of them; so is the Plasmodium parasite that causes malaria. They hide in human tissues, so vaccine development has been particularly challenging.

HIV has multiple ways to evade both the sensors and armaments of the human immune system. Once it enters human cells as a retrovirus, it normally embeds itself into the human genome through a reversal of the whats called the genetic dogma. The latter refers to the passage of genetic information only in one direction, from DNA to RNA to proteins. In violation of the genetic dogma, an enzyme called reverse transcriptase enables HIV and other retroviruses to create DNA from RNA sequences that are carried in the viral genome. This makes removal of HIV analogous to the task of getting toothpaste back into the tube; theoretically, its possible, but snazzy techniques are required.

In gene therapy 1.0, only new genes can be added, so that cannot help with HIV. But the advent of CRISPR in 2012 has addedthe prospect of using gene therapy to delete genes, although one would still need an innovative tactic for identifying those sequences and weeding them out. RNAi is another genetic engineering modality, one that Caltech HIV researcher andNobel laureate David Baltimore believes entails particular potential. The target for RNAi is messenger RNA (mRNA), the sub-type of RNA that carries the genetic sequence transcribed from a DNA gene. In RNAi, special molecules are slipped into cells to suppress activity of mRNA strands.

Research published recently in the online journal PLOS ONE implies a possible new strategy. The study suggests that a strain of HIV called HIV-1 works by hijacking species-specific adaptations that nature has evolved. This makes the virus infectious to some ape species, but not others. CRISPR-based gene therapy we might call it gene therapy 2.0, or RNAi, potentially can incorporate the species-dependent mechanisms employed by HIV itself. This means either eliminating HIV from a patients genome, or rendering the virus neutral by blocking mRNA that is made from viral instructions.

The challenge is figuring out whichgenes should be targeted with this method. The answer could come from a minority of humans with a genetic deficiency that makes them resist HIV infection. To cause disease, HIV requires a certain protein on the surface of the immune systems T-cells. People who cannot make that protein because the copy of the gene from both mother and father are defective are protected from HIV. Those who have one normal gene copy and one defective copy are partially protected. Thus, use of RNAi against the mRNA made from this gene should be protective as well.

These strategies can lead to a sea change in the fight against the HIV pandemic, particularly in developing countries. In developed countries, when diagnosed at an early stage, HIV is not a death sentence, but staying alive requires constant vigilance and treatment. Similar to diabetes, survival depends on monitoring a patients health, adjusting drug treatment accordingly, and managing complications aggressively. Gold standard treatment consists of drug combination therapy, which means using multiple classes of medications, each that attacks the virus through a different mechanism.

Earlier, we mentioned the enzyme reverse transcriptase and how it enables HIV to incorporate its genes into the human genome by causing DNA sequences to be created using RNA sequences. The enzyme is encoded by the virus own genes. Inside the virus, those viral genes exist as RNA sequences, not DNA sequences, and they include the genetic sequence that creates the enzyme. But in the 1990s, researchers began using drugs that inhibit reverse transcriptase and showed that they can slow the onset of acquired immunodeficiency syndrome (AIDS) in HIV-infected patients. Today, there several reverse transcriptase inhibitor drugs, divided into two classes that work differently.

Physicians also have another drug class at their disposal, protease inhibitors, which interfere with another process thats vital to HIVs ability to reproduce itself. When patients are given both reverse transcriptase inhibitors (often more than one type) plus protease inhibitors, survival can continue for many years, and improves still more when an additional class of drugs is added. This is the current paradigm, but, like diabetes therapy, it keeps patients alive without curing them.

Combination therapy is extremely expensive and requires excellent patient compliance, and also societal support. All of these factors make it particularly challenging in developing countries, where HIV infection therefore has a much worse prognosis than it has here in North America, or in Europe.

The prospect of a high-tech approach like gene therapy solving a major public health issue like HIV may sound far-fetched. After all, high-tech solutions generally take hold in developed countries first. The same pattern is likely with HIV. Developed, rich countries will have the treatment first. But if it does work and eliminates the virus, it wont require the kind of followup and constant vigilance thats central to standard combination drug therapy.

David Warmflash is an astrobiologist, physician and science writer. Follow @CosmicEvolution to read what he is saying on Twitter.

Go here to see the original:

HIV fix: Can gene editing work alongside the virus to provide a cure? - Genetic Literacy Project