Tag Archives: DNA

Nanoparticle computing

I’m fascinated with this news and I’m pretty sure it’s my first exposure to nanoparticle computing so am quite excited about this ‘discovery of mine’.

A February 25, 2019 news item on Nanowerk announces the research from Korean scientists,

Computation is a ubiquitous concept in physical sciences, biology, and engineering, where it provides many critical capabilities. Historically, there have been ongoing efforts to merge computation with “unusual” matters across many length scales, from microscopic droplets (Science 315, 832, 2007) to DNA nanostructures (Science 335, 831, 2012; Nat. Chem. 9, 1056, 2017) and molecules (Science 266, 1021, 1994; Science 314, 1585, 2006; Nat. Nanotech. 2, 399, 2007; Nature 375, 368, 2011).

However, the implementation of complex computation in particle systems, especially in nanoparticles, remains challenging, despite a wide range of potential applications that would benefit from algorithmically controlling their unique and potentially useful intrinsic features (such as photonic, plasmonic, catalytic, photothermal, optoelectronic, electrical, magnetic and material properties) without human interventions.

This challenge is not due to the lack of sophistication in the the current state-of-the-art of stimuli-responsive nanoparticles, many of which can conceptually function as elementary logic gates. This is mostly due to the lack of scalable architectures that would enable systematic integration and wiring of the gates into a large integrated circuit.

Previous approaches are limited to (i) demonstrating one simple logic operation per test tube or (ii) relying on complicated enzyme-based molecular circuits in solution. It should be also noted that modular and scalable aspects are key challenges in DNA computing for practical and widespread use.

A February 23, 2019 Seoul National University press release on EurekAlert, which originated the news items, dives into more detail,

In nature, the cell membrane is analogous to a circuit board, as it organizes a wide range of biological nanostructures (e.g. proteins) as (computational) units and allows them to dynamically interact with each other on the fluidic 2D surface to carry out complex functions as a network and often induce signaling intracellular signaling cascades. For example, the membrane proteins on the membrane take chemical/physical cues as inputs (e.g. binding with chemical agents, mechanical stimuli) and change their conformations and/or dimerize as outputs. Most importantly, such biological “computing” processes occur in a massively parallel fashion. Information processing on living cell membranes is a key to how biological systems adapt to changes in external environments.

This manuscript reports the development of a nanoparticle-lipid bilayer hybrid-based computing platform termed lipid nanotablet (LNT), in which nanoparticles, each programmed with surface chemical ligands (DNA in this case), are tethered to a supported lipid bilayer to carry out computation. Taking inspirations from parallel computing processes on cellular membranes, we exploited supported lipid bilayers (SLBs)–synthetic mimics for cell surfaces–as chemical circuit boards to construct nanoparticle circuits. This “nano?bio” computing, which occurs at the interface of nanostructures and biomolecules, translates molecular information in solution (input) into dynamic assembly/disassembly of nanoparticles on a lipid bilayer (output).

We introduced two types of nanoparticles to a lipid bilayer that differ in mobility: mobile Nano-Floaters and immobile Nano-Receptors. Due to high mobility, floaters actively interact with receptors across space and time, functioning as active units of computation. The nanoparticles are functionalized with specially designed DNA [deoxyribonucleic acid] ligands, and the surface ligands render receptor-floater interactions programmable, thereby transforming a pair of receptor and floater into a logic gate. A nanoparticle logic gate takes DNA strands in solution as inputs and generates nanoparticle assembly or disassembly events as outputs. The nanoparticles and their interactions can be imaged and tracked by dark-field microscopy with single-nanoparticle resolution because of strong and stable scattering signals from plasmonic nanoparticles. Using this approach (termed “interface programming”), we first demonstrated that a pair of nanoparticles (that is, two nanoparticles on a lipid bilayer) can carry out AND, OR, INHIBIT logic operations and take multiple inputs (fan-in) and generate multiple outputs (fan-out). Also, multiple logic gates can be modularly wired with AND or OR logic via floaters, as the mobility of floaters enables the information cascade among several nanoparticle logic gates. We termed this strategy “network programming.” By combining these two strategies (interfacial and network programming), we were able to implement complex logic circuits such as multiplexer.

The most important contributions of our paper are the conceptual one and the major advances in modular and scalable molecular computing (DNA computing in this case). LNT platform, for the first time, introduces the idea of using lipid bilayer membranes as key components for information processing. As the two-dimensional (2D) fluidic lipid membrane is bio-compatible and chemically modifiable, any nanostructures can be potentially introduced and used as computing units. When tethered to the lipid bilayer “chip”, these nanostructures can be visualized and become controllable at the single-particle level; this dimensionality reduction, bringing the nanostructures from freely diffusible solution phase (3D) to fluidic membrane (2D), transforms a collection of nanostructures into a programmable, analyzable reaction network. Moreover, we also developed a digitized imaging method and software for quantitative and massively parallel analysis of interacting nanoparticles. In addition, LNT platform provides many practical merits to current state-of-the-art in molecular computing and nanotechnology. On LNT platforms, a network of nanoparticles (each with unique and beneficial properties) can be design to autonomously respond to molecular information; such capability to algorithmically control nanoparticle networks will be very useful for addressing many challenges with molecular computing and developing new computing platforms. As the title of our manuscript suggests, this nano-bio computing will lead to exciting opportunities in biocomputation, nanorobotics, DNA nanotechnology, artificial bio-interfaces, smart biosensors, molecular diagnostics, and intelligent nanomaterials. In summary, the operating and design principles of lipid nanotablet platform are as follows:

(1) LNT uses single nanoparticles as units of computation. By tracking numerous nanoparticles and their actions with dark-field microscopy at the single-particle level, we could treat a single nanoparticle as a two-state device representing a bit. A nanoparticle provides a discrete, in situ optical readout of its interaction (e.g. association or dissociation) with another particle as an output of logic computation.

(2) Nanoparticles on LNT function as Boolean logic gates. We exploited the programmable bonding interaction within particle-particle interfaces to transform two interacting nanoparticles into a Boolean logic gate. The gate senses single-stranded DNA as inputs and triggers an assembly or disassembly reaction of the pair as an output. We demonstrated two-input AND, two-input OR and INHIBIT logic operations, and fan-in/fan-out of logic gates.

(3) LNT enables modular wiring of multiple nanoparticle logic gates into a combinational circuit. We exploited parallel, single-particle imaging to program nanoparticle networks and thereby wire multiple logic gates into a combinational circuit. We demonstrate a multiplexer MUX2to1 circuit built from the network wiring rules.

Here’s a link to and a citation for the team’s latest paper,

Nano-bio-computing lipid nanotablet by Jinyoung Seo, Sungi Kim, Ha H. Park, Da Yeon Choi, and Jwa-Min Nam. Science Advances 22 Feb 2019: Vol. 5, no. 2, eaau2124 DOI: 10.1126/sciadv.aau2124

This paper appears to be open access.

The Backstreet Boys sing genetics (not really) but their latest album is called “DNA”

Other that the promotional artwork, cover art and the title, the Backstreet Boys pop band does not seem to have taken science or DNA (deoxyribonucleic acid)/genetics to heart in their latest oeuvre. As for what chickens have to do with it, I I gather this is some sort of humorous nod to a past hit song. Still, I am weirdly fascinated by this January 25, 2019 video news item on Billboard,

Having looked at the list of songs on the DNA album (they’re listed in the Billboard news item where they’ve embedded audio samples), I can’t find anything that suggests an interest in genetics but perhaps you can: Don’t Go Breaking My Heart? Nobody Else? Breathe? New Love? Passionate? Is It Just Me? Chances? No Place? Chateau? The Way It Was? Just Like You Like it? OK? Anyone who can figure out how the songs relate to DNA, please let me know in the Comments.

Frankly, that’s as much analysis as I can offer on the topic. Thankfully, Karen James (an independent educator, researcher, and consultant in molecular biology) has written a February 5, 2019 article (I Want DNA That Way; The Backstreet Boys’ new album and tour features a very old-school depiction of DNA) for slate.com where she unpacks the imagery in the promotional material and on the cover (Note: Links have been removed),

The Backstreet Boys are back. Credit: Dennis Leupold [downloaded from https://slate.com/technology/2019/02/backstreet-boys-dna-album-cover-gene-sequencing.html]

The Backstreet Boys released a new album. I never thought I’d start a science article—or any article—with that sentence, but here we are.

We are here because the promotional artwork for the album (above) is a photograph of the boy band (man band?) lit by a projection of DNA bands. The image, and the album’s title, DNA, jumped out of my Twitter timeline because I’m a geneticist, I work with DNA, and I’ve seen countless images just like it in textbooks and research articles. I’ve even made them myself in the lab.

What struck me as funny (both funny-ha-ha and funny-odd) is that the lab methods that could have produced this image are old—older even than the Backstreet Boys’ first album. One of the methods—called Sanger sequencing—was published in 1977, making it even older than two of the Backstreet Boys themselves, scientist Kristy Lamb pointed out. Genetics is a particularly fast-moving science. New technologies are constantly emerging and eclipsing prior ones. Yet this 40-year-old imagery persists, and not just in the promotional artwork for DNA. Just do a Google image search for “DNA sequencing” and you’ll see plenty of images like this mixed in with the double helices and long GATTACA readouts.

After her description of Sanger sequencing James offers another ‘sequencing’ possibility, almost as old as the Sanger technique,

Careful readers might have noticed that I suggested there was more than one method that produces images like this. At first glance, I thought the projection in the Backstreet Boys’ publicity photo was modified from an image made with Sanger sequencing. But when I looked again in preparation for writing this article, I had second thoughts. Why aren’t the lanes clustered in groups of four? Why are some of the bands in adjacent lanes the same size? (They shouldn’t be if you’re doing Sanger sequencing.) It could be that the photo was heavily modified with individual lanes copied and pasted. Indeed, some of the lanes are even identical to each other (*suppresses fake ivory tower scoff*).

Or it could be that this image was made with another old method: DNA fingerprinting. Made famous in so many crime TV shows, DNA fingerprinting was invented in 1984 by Alec Jeffreys, who, though he did not win a Nobel Prize, was made a knight of the British Empire for his contribution to science, among many other prestigious awards, which is nice.

I suspect the Backstreet Boys weren’t going for a tongue-in-cheek reference to their own advancing age. While today’s DNA sequencing methods produce images that scarcely resemble those produced by Sanger sequencing and DNA fingerprinting, the old-school imagery is still everywhere. The Backstreet Boys’ promotional team probably just went with a stock image that looked compelling and worked well as a projection.

James returns to her theme, why use imagery associated with outdated techniques? (Note: Links have been removed),

But that doesn’t answer the real question: Why is 40-year-old imagery still so ubiquitous? As science writer and editor Stephanie Keep tweeted, one reason may be that, despite its age, the Sanger method is still taught in high school classrooms: “It’s so visual and intuitive.” It’s true. When I teach students about DNA sequencing, I always start with Sanger sequencing and use that as the basis for explaining newer technologies, adding more complexity as I go, following the historical timeline.

Another reason the old imagery is still in use may be that the images produced by newer, so-called next-generation sequencing methods aren’t visually scored by a scientist sitting at a lab bench, but by computers. As such, the images themselves often go unseen by human eyes [emphasis mine], despite their colorful beauty.

Interesting, eh? The latest imagery is not seen by human eyes. So the newest imagery is intended for machines. James presents an example of the ‘new’ imagery,

An image generated using a next-generation DNA sequencing method.. Credit: Illumina [downloaded from https://slate.com/technology/2019/02/backstreet-boys-dna-album-cover-gene-sequencing.html]

According to James, this image was not easily obtained according to one of her tweets. [https://twitter.com/kejames/status/1092888034322845696] So, big thanks to Illumina (there’s also a Wikipedia entry about the company). Getting back to James’ and her article, she asks why the band titled their latest album, DNA,

But why did the Backstreet Boys call their album DNA in the first place? The official RCA Records press release announcing the album says, “BSB analyzed their individual DNA profiles to see what crucial element each member represents in the groups DNA.” It links to a YouTube video that supposedly explains “how their individual strains, when brought together, create the unstoppable and legendary Backstreet Boys.”

The video is a futuristic, spy movie–esque montage, complete with a computerized female voice describing the various characteristics of each Backstreet Boy. Reader, I confess: I cringed. There were so many tropes and misconceptions about DNA packed into the 83-second video, I would have to write a follow-up to this just to explore them. The cringeworthiness doesn’t end there, though. The cover of DNA has each Backstreet Boy on his own spiral staircase.

The staircases are surely meant to evoke the structure of DNA: the famous double helix. But there’s a problem, as the social media account for the journal Genome Biology tweeted: The staircases are spiraling in the wrong direction. DNA is usually right-handed. If you stick out your right thumb, your fingers will naturally curl in a right-handed spiral as you move your hand in the direction your thumb is pointing. The Backstreet Boys’ staircases are left-handed.

Here’s the promotional trailer for DNA,

It’s everything James says it is. As for those wrongly spiraling DNA staircases,

RCA Records [downloaded from https://slate.com/technology/2019/02/backstreet-boys-dna-album-cover-gene-sequencing.html]

Thank you to Karen James for this illuminating article. If you have time, I encourage you to read her piece in its entirety:
I Want DNA That Way; The Backstreet Boys’ new album and tour features a very old-school depiction of DNA.

As for why the Backstreet Boys called their album DNA and you likely guessed. it would seem to be a promotional gimmick meant to leverage the perceived interest in commercial DNA testing by companies such as 23andMe and Ancestry, amongst others.

Why not monetize your DNA for 2019?

I’m not a big fan of DNA (deoxyribonucleic acid) companies that promise to tell you about your ancestors and, depending on the kit, predisposition to certain health issues as per their reports about your genetic code. (I regularly pray no one in my family has decided to pay one of these companies to analyze their spit.)

During Christmas season 2018, the DNA companies (23andMe and Ancestry) advertised special prices so you could gift someone in your family with a kit. All this corporate largesse may not be wholly in service of the Christmas spirit. After all, there’s money to be made once they’ve gotten your sample.

Monetizing your DNA in 2016

I don’t know when 23andMe started selling DNA information or if any similar company predated their efforts but this June 21, 2016 article by Antonio Regalado for MIT (Massachusetts Institute of Technology) Review offers the earliest information I found,

“Welcome to You.” So says the genetic test kit that 23andMe will send to your home. Pay $199, spit in a tube, and several weeks later you’ll get a peek into your DNA. Have you got the gene for blond hair? Which of 36 disease risks could you pass to a child?

Run by entrepreneur Anne Wojcicki, the ex-wife of Google founder Sergey Brin, and until last year housed alongside the Googleplex, the company created a test that has been attacked by regulators and embraced by a curious public. It remains, nine years after its introduction, the only one of its kind sold directly to consumers. 23andMe has managed to amass a collection of DNA information about 1.2 million people, which last year began to prove its value when the company revealed it had sold access to the data to more than 13 drug companies. One, Genentech, anted up $10 million for a look at the genes of people with Parkinson’s disease.

That means 23andMe is monetizing DNA rather the way Facebook makes money from our “likes.” What’s more, it gets its customers to pay for the privilege. That idea so appeals to investors that they have valued the still-unprofitable company at over $1 billion. “Money follows data,” says Barbara Evans, a legal scholar at the University of Houston, who studies personal genetics. “It takes a lot of labor and capital to get that information in a form that is useful.”

Monetizing your DNA in 2018 and privacy concerns

Starting with Adele Peters’ December 13, 2018 article for Fast Company (Note: A link has been removed),

When 23andMe made a $300 million deal with GlaxoSmithKline [GSK] in July[2018]–so the pharmaceutical giant could access a vast store of genetic data as it works on new drugs–the consumers who actually provided that data didn’t get a cut of the proceeds. A new health platform is taking a different approach: If you choose to share your own DNA data or other health records, you’ll get company shares that will later pay you dividends if that data is sold.

Before getting to the start-up that would allow you rather than a company to profit or at least somewhat monetize your DNA, I’m including a general overview of the July 2018 GSK/23andMe deal in Jamie Ducharme’s July 26, 2018 article for TIME (Note: Links have been removed),

Consumer genetic testing company 23andMe announced on Wednesday [July 25, 2018] that GlaxoSmithKline purchased a $300 million stake in the company, allowing the pharmaceutical giant to use 23andMe’s trove of genetic data to develop new drugs — and raising new privacy concerns for consumers

The “collaboration” is a way to make “novel treatments and cures a reality,” 23andMe CEO Anne Wojcicki said in a company blog post. But, though it isn’t 23andMe’s first foray into drug discovery, the deal doesn’t seem quite so simple to some medical experts — or some of the roughly 5 million 23andMe customers who have sent off tubes of their spit in exchange for ancestry and health insights

Perhaps the most obvious issue is privacy, says Peter Pitts, president of the Center for Medicine in the Public Interest, a non-partisan non-profit that aims to promote patient-centered health care.

“If people are concerned about their social security numbers being stolen, they should be concerned about their genetic information being misused,” Pitts says. “This information is never 100% safe. The risk is magnified when one organization shares it with a second organization. When information moves from one place to another, there’s always a chance for it to be intercepted by unintended third parties.

That risk is real, agrees Dr. Arthur Caplan, head of the division of medical ethics at the New York University School of Medicine. Caplan says that any genetic privacy concerns also extend to your blood relatives, who likely did not consent to having their DNA tested — echoing some of the questions that arose after law enforcement officials used a genealogy website to find and arrest the suspected Golden State Killer in April [2018].

“A lot of people paid money to 23andMe to get their ancestry determined — fun, recreational stuff,” Caplan says. “Even though they may have signed a thing saying, ‘I’m okay if you use this information for medical research,’ I’m not sure they understood what that really meant. I’m not sure they understood that it meant, ‘Yes, we’ll go to Glaxo, and that’s where we’re really going to make a lot of money off of you.’”

A 23andMe spokesperson told TIME that data privacy is a “top priority” for the company, emphasizing that customer data isn’t used in research without consent, and that GlaxoSmithKline will only receive “summary statistics from analyses 23andMe conducts so that no single individual can be identified.”

Yes the data is supposed to be stripped of identifying information but given how many times similar claims about geolocation data have been disproved, I am skeptical. DJ Pangburn’s September 26, 2017 article (Even This Data Guru Is Creeped Out By What Anonymous Location Data Reveals About Us) for Fast Company illustrate the fragility of ‘anonymized data’,

… as a number of studies have shown, even when it’s “anonymous,” stripped of so-called personally identifiable information, geographic data can help create a detailed portrait of a person and, with enough ancillary data, identify them by name

Curious to see this kind of data mining in action, I emailed Gilad Lotan, now vice president of BuzzFeed’s data science team. He agreed to look at a month’s worth of two different users’ anonymized location data, and to come up with individual profiles that were as accurate as possible

The results, produced in just a few days’ time, range from the expected to the surprisingly revealing, and demonstrate just how “anonymous” data can identify individuals.

Last fall Lotan taught a class at New York University on surveillance that kicked off with an assignment like the one I’d given him: link anonymous location data with other data sets–from LinkedIn, Facebook, home registration and mortgage records, and other online data.
“It’s not hard to figure out who this [unnamed] person is,” says Lotan. In class, students found that tracking location data around holidays proved to be the easiest way to determine who, exactly, the data belonged to. “Basically,” he says, “visits to private homes that are owned and publicly registered.”

In 2013, researchers at MIT and the Université Catholique de Louvain in Belgium published a paper reporting on 15 months of study of human mobility data for over 1.5 million individuals. What they found is that only four spatio-temporal points are required to “uniquely identify 95% of the individuals.” The researchers concluded that there was very little privacy even in raw location data. Four years later, their calls for policies rectifying concerns about location tracking have fallen largely on deaf ears.

Getting back to DNA, there was also some concern at Fox News,

Other than warnings, I haven’t seen much about any possible legislation regarding DNA and privacy in either Canada or the US.

Now, let’s get to how you can monetize your self.

Me making money off me

I’ve found two possibilities for an individual who wants to consider monetizing their own DNA.

Health shares

Adele Peters’ December 13, 2018 article describes a start-up company and the model they’re proposing to allow you profit from your own DNA (Note: Links have been removed),

“You can’t say data is valuable and then take that data away from everybody,” says Dawn Barry, president and cofounder of LunaPBC, the public benefit corporation that manages the community-owned platform, called LunaDNA, which recently got SEC approval to recognize health data as currency. “What we’re finding is that [our early adopters are] very excited about the transparency of this model–that when we all come together and create value, that value flows down to the individuals who shared their data.

The platform shares some anonymized data with nonprofits, such as foundations that study rare diseases. In that case, money wouldn’t initially change hands, but “there could be intellectual property that at some point in time is monetized, and the community would share in that,” says Bob Kain, CEO and cofounder of LunaPBC. “When we have enough data in the near future, then we’ll work with pharmaceutical companies, for instance, to drive discovery for those companies. And they will pay market rates.

The company doesn’t offer DNA analysis itself, but chose to focus on data management. If you’ve sent a tube of spit to 23andMe, AncestryDNA, MyHeritage, or FamilyTree DNA, you can contribute that data to LunaDNA and get shares. (If you’d rather not let the original testing company keep your data, you can also separately take the steps to delete it.

“We looked at a number of different models to enable people to have ownership, including cryptocurrency, which is a proxy for ownership, too,” says Kain. “Cryptocurrency is hard to understand for most people, and right now, the regulatory landscape is blurry. So we thought, to move forward, we’d go with something much more traditional and easy to understand, and that is stock shares, basically.

For sharing targeted genes, you get 10 shares. For sharing your whole genome, you get 300 shares. At the moment, that’s not worth very much–the valuation takes into account the risk that the data might not be monetized, and the fact that the startup isn’t the exclusive owner of your data. The SEC filing says that the estimated fair market value of a whole genome is only $21. Some other health information is worth far less; 20 days of data from a fitness tracker garners two shares, valued at 14¢. But as more people contribute data, the research value of the whole database (and dividends) will increase. If the shareholders ever decided to sell the company itself, they would also make money that way.

Luna’s is a very interesting approach and I encourage you to read the December 13, 2018 article in its entirety.

Blockchain and crypto me

At least one effort to introduce blockchain/cryptocurrency technology to the process for monetizing your DNA garnered a lot of attention in February 2018.

A February 8, 2018 article by Eric Rosenbaum for CNBC (a US cable tv channel) explores an effort by George Church (Note: Links have been removed),

It’s probably wise to be skeptical of anyone who says they have a new idea for a blockchain-based company, or worse still, a company changing its business model to focus on the crypto world. That ice tea company that shifted its model to the blockchain, or Kodak saying its road back to riches was managing photo rights using a blockchain system. Raise eyebrow, or move directly onto outright shake of head

However, when a world renown Harvard geneticist announces he’s launching a blockchain-based start-up, it merits some attention. And it’s not the crypto-angle itself that might make you do a double-take, but the assets that will be managed, and exchanged, using digital currency: your DNA

Harvard University genetics guru George Church — one of the scientists at the forefront of the CRISPR genetic engineering revolution — announced on Wednesday a start-up, Nebula Genomics, that will use the blockchain to not only allow individuals to share their personal genome for research purposes, but retain ownership and monetize their DNA through trading of a custom digital currency.

The genomics revolution has been exponentially advanced by drastic reductions in cost. As Nebula noted in a white paper explaining its business model, the first human genome was sequenced in 2001 at a cost of $3 billion. Today, human genome sequencing costs less than $1,000, and in a few years the price will drop below $100

In fact, some big Silicon Valley start-ups, led by 23andMe, have capitalized on this rapid advance and already offer personal DNA testing kits for around $100 (sometimes with discounts even less)

Nebula took direct aim at 23andMe in its white paper, and one reason why it can offer genetic testing for less

“Today, 23andMe (23andme.com) and Ancestry (ancestry.com) are the two leading personal genomics companies. Both use DNA microarray-based genotyping for their genetic tests. It is an outdated and significantly less powerful alternative to DNA sequencing. Instead of sequencing continuous stretches of DNA, genotyping identifies single letters spaced at approximately regular intervals across the genome. …

Outdated genetic tests? Interesting, eh? Zoë Corbyn provides more information about Church’s plans in her February 18, 2018 article for the Guardian,

“Under the current system, personal genomics companies effectively own your personal genomics data, and you don’t see any benefit at all,” says Grishin [Dennis Grishin, Nebula co-founder]. “We want to eliminate the middleman.

Although the aim isn’t to provide a get-rich-quick scheme, the company believes there is potential for substantial returns. Though speculative, its modelling suggests that someone in the US could earn up to 50 times the cost of sequencing their genome – about $50,000 at current rates – taking into account both what could be made from a lifetime of renting out their genetic data, and reductions in medical bills if the results throw up a potentially preventable disease

The startup also thinks it can solve the problem of the dearth of genetic data researchers have to draw on, due to individuals – put off by cost or privacy concerns – not getting sequenced.

Payouts when you grant access to your genome would come in the form of Nebula tokens, the company’s cryptocurrency, and companies would need to buy tokens from the startup to pay people whose data they wanted to access. Though the value of a token is yet to be set and the number of tokens defined, it might, for example, take one Nebula token to get your genome sequenced. An individual new to the system could begin to earn fractions of a token by taking part in surveys about their heath posted by prospective data buyers. When someone had earned enough, they could get sequenced and begin renting out their data and amassing tokens. Alternatively, if an individual wasn’t yet sequenced they may find data buyers willing to pay for or subsidise their genome sequencing in exchange for access to it. “Potentially you wouldn’t have to pay out of pocket for the sequencing of your genome,” says Grishin.

In all cases, stress Grishin and Obbad [Kamal Obbad, Nebula co-founder], the sequence would belong to the individual, so they could rent it out over and over, including to multiple companies simultaneously. And the data buyer would never take ownership or possession of it – rather, it would be stored by the individual (for example in their computer or on their Dropbox account) with Nebula then providing a secure computation platform on which the data buyer could compute on the data. “You stay in control of your data and you can share it securely with who you want to,” explains Obbad. Nebula makes money not by taking any transaction fee but by being a participant providing computing and storage services. The cryptocurrency would be able to be cashed out for real money via existing cryptocurrency exchanges.

Hopefully, Luna and Nebula, as well as, any competitors in this race to allow individuals to monetize their own DNA will have excellent security.

For the curious, you can find Luna here and Nebula here.Note: I am not endorsing either company or any others mentioned here. This posting is strictly informational.

First CRISPR gene-edited babies? Ethics and the science story

Scientists, He Jiankui and Michael Deem, may have created the first human babies born after being subjected to CRISPR (clustered regularly interspaced short palindromic repeats) gene editing.  At this point, no one is entirely certain that these babies  as described actually exist since the information was made public in a rather unusual (for scientists) fashion.

The news broke on Sunday, November 25, 2018 through a number of media outlets none of which included journals associated with gene editing or high impact journals such as Cell, Nature, or Science.The news broke in MIT Technology Review and in Associated Press. Plus, this all happened just before the Second International Summit on Human Genome Editing (Nov. 27 – 29, 2018) in Hong Kong. He Jiankui was scheduled to speak today, Nov. 27, 2018.

Predictably, this news has caused quite a tizzy.

Breaking news

Antonio Regalado broke the news in a November 25, 2018  article for MIT [Massachusetts Institute of Technology] Technology Review (Note: Links have been removed),

According to Chinese medical documents posted online this month (here and here), a team at the Southern University of Science and Technology, in Shenzhen, has been recruiting couples in an effort to create the first gene-edited babies. They planned to eliminate a gene called CCR5 in hopes of rendering the offspring resistant to HIV, smallpox, and cholera.

The clinical trial documents describe a study in which CRISPR is employed to modify human embryos before they are transferred into women’s uteruses.

The scientist behind the effort, He Jiankui, did not reply to a list of questions about whether the undertaking had produced a live birth. Reached by telephone, he declined to comment.

However, data submitted as part of the trial listing shows that genetic tests have been carried out on fetuses as late as 24 weeks, or six months. It’s not known if those pregnancies were terminated, carried to term, or are ongoing.

Apparently He changed his mind because Marilynn Marchione in a November 26, 2018 article for the Associated Press confirms the news,

A Chinese researcher claims that he helped make the world’s first genetically edited babies — twin girls born this month whose DNA he said he altered with a powerful new tool capable of rewriting the very blueprint of life.

If true, it would be a profound leap of science and ethics.

A U.S. scientist [Dr. Michael Deem] said he took part in the work in China, but this kind of gene editing is banned in the United States because the DNA changes can pass to future generations and it risks harming other genes.

Many mainstream scientists think it’s too unsafe to try, and some denounced the Chinese report as human experimentation.

There is no independent confirmation of He’s claim, and it has not been published in a journal, where it would be vetted by other experts. He revealed it Monday [November 26, 2018] in Hong Kong to one of the organizers of an international conference on gene editing that is set to begin Tuesday [November 27, 2018], and earlier in exclusive interviews with The Associated Press.

“I feel a strong responsibility that it’s not just to make a first, but also make it an example,” He told the AP. “Society will decide what to do next” in terms of allowing or forbidding such science.

Some scientists were astounded to hear of the claim and strongly condemned it.

It’s “unconscionable … an experiment on human beings that is not morally or ethically defensible,” said Dr. Kiran Musunuru, a University of Pennsylvania gene editing expert and editor of a genetics journal.

“This is far too premature,” said Dr. Eric Topol, who heads the Scripps Research Translational Institute in California. “We’re dealing with the operating instructions of a human being. It’s a big deal.”

However, one famed geneticist, Harvard University’s George Church, defended attempting gene editing for HIV, which he called “a major and growing public health threat.”

“I think this is justifiable,” Church said of that goal.

h/t Cale Guthrie Weissman’s Nov. 26, 2018 article for Fast Company.

Diving into more detail

Ed Yong in a November 26, 2018 article for The Atlantic provides more details about the claims (Note: Links have been removed),

… “Two beautiful little Chinese girls, Lulu and Nana, came crying into the world as healthy as any other babies a few weeks ago,” He said in the first of five videos, posted yesterday {Nov. 25, 2018] to YouTube [link provided at the end of this section of the post]. “The girls are home now with their mom, Grace, and dad, Mark.” The claim has yet to be formally verified, but if true, it represents a landmark in the continuing ethical and scientific debate around gene editing.

Late last year, He reportedly enrolled seven couples in a clinical trial, and used their eggs and sperm to create embryos through in vitro fertilization. His team then used CRISPR to deactivate a single gene called CCR5 in the embryos, six of which they then implanted into mothers. CCR5 is a protein that the HIV virus uses to gain entry into human cells; by deactivating it, the team could theoretically reduce the risk of infection. Indeed, the fathers in all eight couples were HIV-positive.

Whether the experiment was successful or not, it’s intensely controversial. Scientists have already begun using CRISPR and other gene-editing technologies to alter human cells, in attempts to treat cancers, genetic disorders, and more. But in these cases, the affected cells stay within a person’s body. Editing an embryo [it’s often called, germline editing] is very different: It changes every cell in the body of the resulting person, including the sperm or eggs that would pass those changes to future generations. Such work is banned in many European countries, and prohibited in the United States. “I understand my work will be controversial, but I believe families need this technology and I’m willing to take the criticism for them,” He said.

“Was this a reasonable thing to do? I would say emphatically no,” says Paula Cannon of the University of Southern California. She and others have worked on gene editing, and particularly on trials that knock out CCR5 as a way to treat HIV. But those were attempts to treat people who were definitively sick and had run out of other options. That wasn’t the case with Nana and Lulu.

“The idea that being born HIV-susceptible, which is what the vast majority of humans are, is somehow a disease state that requires the extraordinary intervention of gene editing blows my mind,” says Cannon. “I feel like he’s appropriating this potentially valuable therapy as a shortcut to doing something in the sphere of gene editing. He’s either very naive or very cynical.”

“I want someone to make sure that it has happened,” says Hank Greely, an ethicist at Stanford University. If it hasn’t, that “would be a pretty bald-faced fraud,” but such deceptions have happened in the past. “If it is true, I’m disappointed. It’s reckless on safety grounds, and imprudent and stupid on social grounds.” He notes that a landmark summit in 2015 (which included Chinese researchers) and a subsequent major report from the National Academies of Science, Engineering, and Medicine both argued that “public participation should precede any heritable germ-line editing.” That is: Society needs to work out how it feels about making gene-edited babies before any babies are edited. Absent that consensus, He’s work is “waving a red flag in front of a bull,” says Greely. “It provokes not just the regular bio-Luddites, but also reasonable people who just wanted to talk it out.”

Societally, the creation of CRISPR-edited babies is a binary moment—a Rubicon that has been crossed. But scientifically, the devil is in the details, and most of those are still unknown.

CRISPR is still inefficient. [emphasis mine] The Chinese teams who first used it to edit human embryos only did so successfully in a small proportion of cases, and even then, they found worrying levels of “off-target mutations,” where they had erroneously cut parts of the genome outside their targeted gene. He, in his video, claimed that his team had thoroughly sequenced Nana and Lulu’s genomes and found no changes in genes other than CCR5.

That claim is impossible to verify in the absence of a peer-reviewed paper, or even published data of any kind. “The paper is where we see whether the CCR5 gene was properly edited, what effect it had at the cellular level, and whether [there were] any off-target effects,” said Eric Topol of the Scripps Research Institute. “It’s not just ‘it worked’ as a binary declaration.”

In the video, He said that using CRISPR for human enhancement, such as enhancing IQ or selecting eye color, “should be banned.” Speaking about Nana and Lulu’s parents, he said that they “don’t want a designer baby, just a child who won’t suffer from a disease that medicine can now prevent.”

But his rationale is questionable. Huang [Junjiu Huang of Sun Yat-sen University ], the first Chinese researcher to use CRISPR on human embryos, targeted the faulty gene behind an inherited disease called beta thalassemia. Mitalipov, likewise, tried to edit a gene called MYBPC3, whose faulty versions cause another inherited disease called hypertrophic cardiomyopathy (HCM). Such uses are still controversial, but they rank among the more acceptable applications for embryonic gene editing as ways of treating inherited disorders for which treatments are either difficult or nonexistent.

In contrast, He’s team disableda normal gene in an attempt to reduce the risk of a disease that neither child had—and one that can be controlled. There are already ways of preventing fathers from passing HIV to their children. There are antiviral drugs that prevent infections. There’s safe-sex education. “This is not a plague for which we have no tools,” says Cannon.

As Marilynn Marchione of the AP reports, early tests suggest that He’s editing was incomplete [emphasis mine], and at least one of the twins is a mosaic, where some cells have silenced copies of CCR5 and others do not. If that’s true, it’s unlikely that they would be significantly protected from HIV. And in any case, deactivating CCR5 doesn’t confer complete immunity, because some HIV strains can still enter cells via a different protein called CXCR4.

Nana and Lulu might have other vulnerabilities. …

It is also unclear if the participants in He’s trial were fully aware of what they were signing up for. [emphasis mine] The team’s informed-consent document describes their work as an “AIDS vaccine development project,” and while it describes CRISPR gene editing, it does so in heavily technical language. It doesn’t mention any of the risks of disabling CCR5, and while it does note the possibility of off-target effects, it also says that the “project team is not responsible for the risk.”

He owns two genetics companies, and his collaborator, Michael Deem of Rice University,  [emphasis mine] holds a small stake in, and sits on the advisory board of, both of them. The AP’s Marchione reports, “Both men are physics experts with no experience running human clinical trials.” [emphasis mine]

Yong’s article is well worth reading in its entirety. As for YouTube, here’s The He Lab’s webpage with relevant videos.

Reactions

Gina Kolata, Sui-Lee Wee, and Pam Belluck writing in a Nov. 26, 2018 article for the New York Times chronicle some of the response to He’s announcement,

It is highly unusual for a scientist to announce a groundbreaking development without at least providing data that academic peers can review. Dr. He said he had gotten permission to do the work from the ethics board of the hospital Shenzhen Harmonicare, but the hospital, in interviews with Chinese media, denied being involved. Cheng Zhen, the general manager of Shenzhen Harmonicare, has asked the police to investigate what they suspect are “fraudulent ethical review materials,” according to the Beijing News.

The university that Dr. He is attached to, the Southern University of Science and Technology, said Dr. He has been on no-pay leave since February and that the school of biology believed that his project “is a serious violation of academic ethics and academic norms,” according to the state-run Beijing News.

In a statement late on Monday, China’s national health commission said it has asked the health commission in southern Guangdong province to investigate Mr. He’s claims.

“I think that’s completely insane,” said Shoukhrat Mitalipov, director of the Center for Embryonic Cell and Gene Therapy at Oregon Health and Science University. Dr. Mitalipov broke new ground last year by using gene editing to successfully remove a dangerous mutation from human embryos in a laboratory dish. [I wrote a three-part series about CRISPR, which included what was then the latest US news, Mitalipov’s announcement, along with a roundup of previous work in China. Links are at the end of this section.’

Dr. Mitalipov said that unlike his own work, which focuses on editing out mutations that cause serious diseases that cannot be prevented any other way, Dr. He did not do anything medically necessary. There are other ways to prevent H.I.V. infection in newborns.

Just three months ago, at a conference in late August on genome engineering at Cold Spring Harbor Laboratory in New York, Dr. He presented work on editing the CCR₅ gene in the embryos of nine couples.

At the conference, whose organizers included Jennifer Doudna, one of the inventors of Crispr technology, Dr. He gave a careful talk about something that fellow attendees considered squarely within the realm of ethically approved research. But he did not mention that some of those embryos had been implanted in a woman and could result in genetically engineered babies.

“What we now know is that as he was talking, there was a woman in China carrying twins,” said Fyodor Urnov, deputy director of the Altius Institute for Biomedical Sciences and a visiting researcher at the Innovative Genomics Institute at the University of California. “He had the opportunity to say ‘Oh and by the way, I’m just going to come out and say it, people, there’s a woman carrying twins.’”

“I would never play poker against Dr. He,” Dr. Urnov quipped.

Richard Hynes, a cancer researcher at the Massachusetts Institute of Technology, who co-led an advisory group on human gene editing for the National Academy of Sciences and the National Academy of Medicine, said that group and a similar organization in Britain had determined that if human genes were to be edited, the procedure should only be done to address “serious unmet needs in medical treatment, it had to be well monitored, it had to be well followed up, full consent has to be in place.”

It is not clear why altering genes to make people resistant to H.I.V. is “a serious unmet need.” Men with H.I.V. do not infect embryos. …

Dr. He got his Ph.D., from Rice University, in physics and his postdoctoral training, at Stanford, was with Stephen Quake, a professor of bioengineering and applied physics who works on sequencing DNA, not editing it.

Experts said that using Crispr would actually be quite easy for someone like Dr. He.

After coming to Shenzhen in 2012, Dr. He, at age 28, established a DNA sequencing company, Direct Genomics, and listed Dr. Quake on its advisory board. But, in a telephone interview on Monday, Dr. Quake said he was never associated with the company.

Deem, the US scientist who worked in China with He is currently being investigated (from a Nov. 26, 2018 article by Andrew Joseph in STAT),

Rice University said Monday that it had opened a “full investigation” into the involvement of one of its faculty members in a study that purportedly resulted in the creation of the world’s first babies born with edited DNA.

Michael Deem, a bioengineering professor at Rice, told the Associated Press in a story published Sunday that he helped work on the research in China.

Deem told the AP that he was in China when participants in the study consented to join the research. Deem also said that he had “a small stake” in and is on the scientific advisory boards of He’s two companies.

Megan Molteni in a Nov. 27, 2018 article for Wired admits she and her colleagues at the magazine may have dismissed CRISPR concerns about designer babies prematurely while shedding more light on this  latest development (Note: Links have been removed),

We said “don’t freak out,” when scientists first used Crispr to edit DNA in non-viable human embryos. When they tried it in embryos that could theoretically produce babies, we said “don’t panic.” Many years and years of boring bench science remain before anyone could even think about putting it near a woman’s uterus. Well, we might have been wrong. Permission to push the panic button granted.

Late Sunday night, a Chinese researcher stunned the world by claiming to have created the first human babies, a set of twins, with Crispr-edited DNA….

What’s perhaps most strange is not that He ignored global recommendations on conducting responsible Crispr research in humans. He also ignored his own advice to the world—guidelines that were published within hours of his transgression becoming public.

On Monday, He and his colleagues at Southern University of Science and Technology, in Shenzhen, published a set of draft ethical principles “to frame, guide, and restrict clinical applications that communities around the world can share and localize based on religious beliefs, culture, and public-health challenges.” Those principles included transparency and only performing the procedure when the risks are outweighed by serious medical need.

The piece appeared in the The Crispr Journal, a young publication dedicated to Crispr research, commentary, and debate. Rodolphe Barrangou, the journal’s editor in chief, where the peer-reviewed perspective appeared, says that the article was one of two that it had published recently addressing the ethical concerns of human germline editing, the other by a bioethicist at the University of North Carolina. Both papers’ authors had requested that their writing come out ahead of a major gene editing summit taking place this week in Hong Kong. When half-rumors of He’s covert work reached Barrangou over the weekend, his team discussed pulling the paper, but ultimately decided that there was nothing too solid to discredit it, based on the information available at the time.

Now Barrangou and his team are rethinking that decision. For one thing, He did not disclose any conflicts of interest, which is standard practice among respectable journals. It’s since become clear that not only is He at the helm of several genetics companies in China, He was actively pursuing controversial human research long before writing up a scientific and moral code to guide it.“We’re currently assessing whether the omission was a matter of ill-management or ill-intent,” says Barrangou, who added that the journal is now conducting an audit to see if a retraction might be warranted. …

“There are all sorts of questions these issues raise, but the most fundamental is the risk-benefit ratio for the babies who are going to be born,” says Hank Greely, an ethicist at Stanford University. “And the risk-benefit ratio on this stinks. Any institutional review board that approved it should be disbanded if not jailed.”

Reporting by Stat indicates that He may have just gotten in over his head and tried to cram a self-guided ethics education into a few short months. The young scientist—records indicate He is just 34—has a background in biophysics, with stints studying in the US at Rice University and in bioengineer Stephen Quake’s lab at Stanford. His resume doesn’t read like someone steeped deeply in the nuances and ethics of human research. Barrangou says that came across in the many rounds of edits He’s framework went through.

… China’s central government in Beijing has yet to come down one way or another. Condemnation would make He a rogue and a scientific outcast. Anything else opens the door for a Crispr IVF cottage industry to emerge in China and potentially elsewhere. “It’s hard to imagine this was the only group in the world doing this,” says Paul Knoepfler, a stem cell researcher at UC Davis who wrote a book on the future of designer babies called GMO Sapiens. “Some might say this broke the ice. Will others forge ahead and go public with their results or stop what they’re doing and see how this plays out?”

Here’s some of the very latest information with the researcher attempting to explain himself.

What does He have to say?

After He’s appearance at the Second International Summit on Human Genome Editing today, Nov. 27, 2018, David Cyranoski produced this article for Nature,

He Jiankui, the Chinese scientist who claims to have helped produce the first people born with edited genomes — twin girls — appeared today at a gene-editing summit in Hong Kong to explain his experiment. He gave his talk amid threats of legal action and mounting questions, from the scientific community and beyond, about the ethics of his work and the way in which he released the results.

He had never before presented his work publicly outside of a handful of videos he posted on YouTube. Scientists welcomed the fact that he appeared at all — but his talk left many hungry for more answers, and still not completely certain that He has achieved what he claims.

“There’s no reason not to believe him,” says Robin Lovell-Badge, a developmental biologist at the Francis Crick Institute in London. “I’m just not completely convinced.”

Lovell-Badge, like others at the conference, says that an independent body should confirm the test results by performing an in-depth comparison of the parents’ and childrens’ genes.

Many scientists faulted He for a lack of transparency and the seemingly cavalier nature in which he embarked on such a landmark, and potentially risky, project.

“I’m happy he came but I was really horrified and stunned when he described the process he used,” says Jennifer Doudna, a biochemist at the University of California, Berkeley and a pioneer of the CRISPR/Cas-9 gene-editing technique that He used. “It was so inappropriate on so many levels.”

He seemed shaky approaching the stage and nervous during the talk. “I think he was scared,” says Matthew Porteus, who researches genome-editing at Stanford University in California and co-hosted a question-and-answer session with He after his presentation. Porteus attributes this either to the legal pressures that He faces or the mounting criticism from the scientists and media he was about to address.

He’s talk leaves a host of other questions unanswered, including whether the prospective parents were properly informed of the risks; why He selected CCR5 when there are other, proven ways to prevent HIV; why he chose to do the experiment with couples in which the fathers have HIV, rather than mothers who have a higher chance of passing the virus on to their children; and whether the risks of knocking out CCR5 — a gene normally present in people, which could have necessary but still unknown functions — outweighed the benefits in this case.

In the discussion following He’s talk, one scientist asked why He proceeded with the experiments despite the clear consensus among scientists worldwide that such research shouldn’t be done. He didn’t answer the question.

He’s attempts to justify his actions mainly fell flat. In response to questions about why the science community had not been informed of the experiments before the first women were impregnated, he cited presentations that he gave last year at meetings at the University of California, Berkeley, and at the Cold Spring Harbor Laboratory in New York. But Doudna, who organized the Berkeley meeting, says He did not present anything that showed he was ready to experiment in people. She called his defence “disingenuous at best”.

He also said he discussed the human experiment with unnamed scientists in the United States. But Porteus says that’s not enough for such an extraordinary experiment: “You need feedback not from your two closest friends but from the whole community.” …

Pressure was mounting on He ahead of the presentation. On 27 November, the Chinese national health commission ordered the Guangdong health commission, in the province where He’s university is located, to investigate.

On the same day, the Chinese Academy of Sciences issued a statement condemning his work, and the Genetics Society of China and the Chinese Society for Stem Cell Research jointly issued a statement saying the experiment “violates internationally accepted ethical principles regulating human experimentation and human rights law”.

The hospital cited in China’s clinical-trial registry as the that gave ethical approval for He’s work posted a press release on 27 November saying it did not give any approval. It questioned the signatures on the approval form and said that the hospital’s medical-ethics committee never held a meeting related to He’s research. The hospital, which itself is under investigation by the Shenzhen health authorities following He’s revelations, wrote: “The Company does not condone the means of the Claimed Project, and has reservations as to the accuracy, reliability and truthfulness of its contents and results.”

He has not yet responded to requests for comment on these statements and investigations, nor on why the hospital was listed in the registry and the claim of apparent forged signatures.

Alice Park’s Nov. 26, 2018 article for Time magazine includes an embedded video of He’s Nov. 27, 2018 presentation at the summit meeting.

What about the politics?

Mara Hvistendahl’s Nov. 27, 2018 article about this research for Slate.com poses some geopolitical questions (Note: Links have been removed),

The informed consent agreement for He Jiankui’s experiment describes it as an “AIDS vaccine development project” and used highly technical language to describe the procedure that patients would undergo. If the reality for some Chinese patients is that such agreements are glossed over, densely written, or never read, the reality for some researchers working in the country is that the appeal of cutting-edge trials is too great to resist. It is not just Chinese scientists who can be blinded by the lure of quick breakthroughs. Several of the most notable breaches of informed consent on the mainland have involved Western researchers or co-authors. … When people say that the usual rules don’t apply in China, they are really referring to authoritarian science, not some alternative communitarian ethics.

For the many scientists in China who adhere to recognized international standards, the incident comes as a disgrace. He Jiankui now faces an ethics investigation from provincial health authorities, and his institution, Southern University of Science and Technology, was quick to issue a statement noting that He was on unpaid leave. …

It would seem that US [and from elsewhere]* scientists wanting to avoid pesky ethics requirements in the US have found that going to China could be the answer to their problems. I gather it’s not just big business that prefers deregulated environments.

Guillaume Levrier’s  (he’ studying for a PhD at the Universté Sorbonne Paris Cité) November 16, 2018 essay for The Conversation sheds some light on political will and its impact on science (Note: Links have been removed),

… China has entered a “genome editing” race among great scientific nations and its progress didn’t come out of nowhere. China has invested heavily in the natural-sciences sector over the past 20 years. The Ninth Five-Year Plan (1996-2001) mentioned the crucial importance of biotechnologies. The current Thirteenth Five-Year Plan is even more explicit. It contains a section dedicated to “developing efficient and advanced biotechnologies” and lists key sectors such as “genome-editing technologies” intended to “put China at the bleeding edge of biotechnology innovation and become the leader in the international competition in this sector”.

Chinese embryo research is regulated by a legal framework, the “technical norms on human-assisted reproductive technologies”, published by the Science and Health Ministries. The guidelines theoretically forbid using sperm or eggs whose genome have been manipulated for procreative purposes. However, it’s hard to know how much value is actually placed on this rule in practice, especially in China’s intricate institutional and political context.

In theory, three major actors have authority on biomedical research in China: the Science and Technology Ministry, the Health Ministry, and the Chinese Food and Drug Administration. In reality, other agents also play a significant role. Local governments interpret and enforce the ministries’ “recommendations”, and their own interpretations can lead to significant variations in what researchers can and cannot do on the ground. The Chinese National Academy of Medicine is also a powerful institution that has its own network of hospitals, universities and laboratories.

Another prime actor is involved: the health section of the People’s Liberation Army (PLA), which has its own biomedical faculties, hospitals and research labs. The PLA makes its own interpretations of the recommendations and has proven its ability to work with the private sector on gene editing projects. …

One other thing from Levrier’s essay,

… And the media timing is just a bit too perfect, …

Do read the essay; there’s a twist at the end.

Final thoughts and some links

If I read this material rightly, there are suspicions there may be more of this work being done in China and elsewhere. In short, we likely don’t have the whole story.

As for the ethical issues, this is a discussion among experts only, so far. The great unwashed (thee and me) are being left at the wayside. Sure, we’ll be invited to public consultations, one day,  after the big decisions have been made.

Anyone who’s read up on the history of science will tell you this kind of breach is very common at the beginning. Richard Holmes’  2008 book, ‘The Age of Wonder: How the Romantic Generation Discovered the Beauty and Terror of Science’ recounts stories of early scientists (European science) who did crazy things. Some died, some shortened their life spans; and, some irreversibly damaged their health.  They also experimented on other people. Informed consent had not yet been dreamed up.

In fact, I remember reading somewhere that the largest human clinical trial in history was held in Canada. The small pox vaccine was highly contested in the US but the Canadian government thought it was a good idea so they offered US scientists the option of coming here to vaccinate Canadian babies. This was in the 1950s and the vaccine seems to have been administered almost universally. That was a lot of Canadian babies. Thankfully, it seems to have worked out but it does seem mind-boggling today.

For all the indignation and shock we’re seeing, this is not the first time nor will it be the last time someone steps over a line in order to conduct scientific research. And, that is the eternal problem.

Meanwhile I think some of the real action regarding CRISPR and germline editing is taking place in the field (pun!) of agriculture:

My Nov. 27, 2018 posting titled: ‘Designer groundcherries by CRISPR (clustered regularly interspaced short palindromic repeats)‘ and a more disturbing Nov. 27, 2018 post titled: ‘Agriculture and gene editing … shades of the AquAdvantage salmon‘. That second posting features a company which is trying to sell its gene-editing services to farmers who would like cows that  never grow horns and pigs that never reach puberty.

Then there’s this ,

The Genetic Revolution‘, a documentary that offers relatively up-to-date information about gene editing, which was broadcast on Nov. 11, 2018 as part of The Nature of Things series on CBC (Canadian Broadcasting Corporation).

My July 17, 2018 posting about research suggesting that scientists hadn’t done enough research on possible effects of CRISPR editing titled: ‘The CRISPR ((clustered regularly interspaced short palindromic repeats)-CAS9 gene-editing technique may cause new genetic damage kerfuffle’.

My 2017 three-part series on CRISPR and germline editing:

CRISPR and editing the germline in the US (part 1 of 3): In the beginning

CRISPR and editing the germline in the US (part 2 of 3): ‘designer babies’?

CRISPR and editing the germline in the US (part 3 of 3): public discussions and pop culture

There you have it.

Added on November 30, 2018: David Cyanowski has written one final article (Nov. 30, 2018 for Nature) about He and the Second International Summit on Human Genome Editing. He did not make his second scheduled appearance at the summit, returning to China before the summit concluded. He was rebuked in a statement produced by the Summit’s organizing committee at the end of the three-day meeting. The situation with regard to his professional status in China is ambiguous. Cyanowski ends his piece with the information that the third summit will take place in London (likely in the UK) in 2021. I encourage you to read Cyanowski’s Nov. 30, 2018 article in its entirety; it’s not long.

Added on Dec. 3, 2018: The story continues. Ed Yong has written a summary of the issues to date in a Dec. 3, 2018 article for The Atlantic (even if you know the story ift’s eyeopening to see all the parts put together.

J. Benjamin Hurlbut, Associate Professor of Life Sciences at Arizona State University (ASU) and Jason Scott Robert, Director of the Lincoln Center for Applied Ethics at Arizona State University have written a provocative (and true) Dec. 3, 2018 essay titled, CRISPR babies raise an uncomfortable reality – abiding by scientific standards doesn’t guarantee ethical research, for The Conversation. h/t phys.org

*[and from elsewhere] added January 17, 2019.

Added on January 23, 2019: He has been fired by his university (Southern University of Science and Technology in Shenzhen) as announced on January 21, 2019.  David Cyranoski provides a details accounting in his January 22, 2019 article for Nature.

I found it at the movies: a commentary on/review of “Films from the Future”

Kudos to anyone who recognized the reference to Pauline Kael (she changed film criticism forever) and her book “I Lost it at the Movies.” Of course, her book title was a bit of sexual innuendo, quite risqué for an important film critic in 1965 but appropriate for a period (the 1960s) associated with a sexual revolution. (There’s more about the 1960’s sexual revolution in the US along with mention of a prior sexual revolution in the 1920s in this Wikipedia entry.)

The title for this commentary is based on an anecdote from Dr. Andrew Maynard’s (director of the Arizona State University [ASU] Risk Innovation Lab) popular science and technology book, “Films from the Future: The Technology and Morality of Sci-Fi Movies.”

The ‘title-inspiring’ anecdote concerns Maynard’s first viewing of ‘2001: A Space Odyssey, when as a rather “bratty” 16-year-old who preferred to read science fiction, he discovered new ways of seeing and imaging the world. Maynard isn’t explicit about when he became a ‘techno nerd’ or how movies gave him an experience books couldn’t but presumably at 16 he was already gearing up for a career in the sciences. That ‘movie’ revelation received in front of a black and white television on January 1,1982 eventually led him to write, “Films from the Future.” (He has a PhD in physics which he is now applying to the field of risk innovation. For a more detailed description of Dr. Maynard and his work, there’s his ASU profile webpage and, of course, the introduction to his book.)

The book is quite timely. I don’t know how many people have noticed but science and scientific innovation is being covered more frequently in the media than it has been in many years. Science fairs and festivals are being founded on what seems to be a daily basis and you can now find science in art galleries. (Not to mention the movies and television where science topics are covered in comic book adaptations, in comedy, and in standard science fiction style.) Much of this activity is centered on what’s called ’emerging technologies’. These technologies are why people argue for what’s known as ‘blue sky’ or ‘basic’ or ‘fundamental’ science for without that science there would be no emerging technology.

Films from the Future

Isn’t reading the Table of Contents (ToC) the best way to approach a book? (From Films from the Future; Note: The formatting has been altered),

Table of Contents
Chapter One
In the Beginning 14
Beginnings 14
Welcome to the Future 16
The Power of Convergence 18
Socially Responsible Innovation 21
A Common Point of Focus 25
Spoiler Alert 26
Chapter Two
Jurassic Park: The Rise of Resurrection Biology 27
When Dinosaurs Ruled the World 27
De-Extinction 31
Could We, Should We? 36
The Butterfly Effect 39
Visions of Power 43
Chapter Three
Never Let Me Go: A Cautionary Tale of Human Cloning 46
Sins of Futures Past 46
Cloning 51
Genuinely Human? 56
Too Valuable to Fail? 62
Chapter Four
Minority Report: Predicting Criminal Intent 64
Criminal Intent 64
The “Science” of Predicting Bad Behavior 69
Criminal Brain Scans 74
Machine Learning-Based Precognition 77
Big Brother, Meet Big Data 79
Chapter Five
Limitless: Pharmaceutically-enhanced Intelligence 86
A Pill for Everything 86
The Seduction of Self-Enhancement 89
Nootropics 91
If You Could, Would You? 97
Privileged Technology 101
Our Obsession with Intelligence 105
Chapter Six
Elysium: Social Inequity in an Age of Technological
Extremes 110
The Poor Shall Inherit the Earth 110
Bioprinting Our Future Bodies 115
The Disposable Workforce 119
Living in an Automated Future 124
Chapter Seven
Ghost in the Shell: Being Human in an
Augmented Future 129
Through a Glass Darkly 129
Body Hacking 135
More than “Human”? 137
Plugged In, Hacked Out 142
Your Corporate Body 147
Chapter Eight
Ex Machina: AI and the Art of Manipulation 154
Plato’s Cave 154
The Lure of Permissionless Innovation 160
Technologies of Hubris 164
Superintelligence 169
Defining Artificial Intelligence 172
Artificial Manipulation 175
Chapter Nine
Transcendence: Welcome to the Singularity 180
Visions of the Future 180
Technological Convergence 184
Enter the Neo-Luddites 190
Techno-Terrorism 194
Exponential Extrapolation 200
Make-Believe in the Age of the Singularity 203
Chapter Ten
The Man in the White Suit: Living in a Material World 208
There’s Plenty of Room at the Bottom 208
Mastering the Material World 213
Myopically Benevolent Science 220
Never Underestimate the Status Quo 224
It’s Good to Talk 227
Chapter Eleven
Inferno: Immoral Logic in an Age of
Genetic Manipulation 231
Decoding Make-Believe 231
Weaponizing the Genome 234
Immoral Logic? 238
The Honest Broker 242
Dictating the Future 248
Chapter Twelve
The Day After Tomorrow: Riding the Wave of
Climate Change 251
Our Changing Climate 251
Fragile States 255
A Planetary “Microbiome” 258
The Rise of the Anthropocene 260
Building Resiliency 262
Geoengineering the Future 266
Chapter Thirteen
Contact: Living by More than Science Alone 272
An Awful Waste of Space 272
More than Science Alone 277
Occam’s Razor 280
What If We’re Not Alone? 283
Chapter Fourteen
Looking to the Future 288
Acknowledgments 293

The ToC gives the reader a pretty clue as to where the author is going with their book and Maynard explains how he chose his movies in his introductory chapter (from Films from the Future),

“There are some quite wonderful science fiction movies that didn’t make the cut because they didn’t fit the overarching narrative (Blade Runner and its sequel Blade Runner 2049, for instance, and the first of the Matrix trilogy). There are also movies that bombed with the critics, but were included because they ably fill a gap in the bigger story around emerging and converging technologies. Ultimately, the movies that made the cut were chosen because, together, they create an overarching narrative around emerging trends in biotechnologies, cybertechnologies, and materials-based technologies, and they illuminate a broader landscape around our evolving relationship with science and technology. And, to be honest, they are all movies that I get a kick out of watching.” (p. 17)

Jurassic Park (Chapter Two)

Dinosaurs do not interest me—they never have. Despite my profound indifference I did see the movie, Jurassic Park, when it was first released (someone talked me into going). And, I am still profoundly indifferent. Thankfully, Dr. Maynard finds meaning and a connection to current trends in biotechnology,

Jurassic Park is unabashedly a movie about dinosaurs. But it’s also a movie about greed, ambition, genetic engineering, and human folly—all rich pickings for thinking about the future, and what could possibly go wrong. (p. 28)

What really stands out with Jurassic Park, over twenty-five years later, is how it reveals a very human side of science and technology. This comes out in questions around when we should tinker with technology and when we should leave well enough alone. But there is also a narrative here that appears time and time again with the movies in this book, and that is how we get our heads around the sometimes oversized roles mega-entrepreneurs play in dictating how new tech is used, and possibly abused. These are all issues that are just as relevant now as they were in 1993, and are front and center of ensuring that the technologyenabled future we’re building is one where we want to live, and not one where we’re constantly fighting for our lives.  (pp. 30-1)

He also describes a connection to current trends in biotechnology,

De-Extinction

In a far corner of Siberia, two Russians—Sergey Zimov and his son Nikita—are attempting to recreate the Ice Age. More precisely, their vision is to reconstruct the landscape and ecosystem of northern Siberia in the Pleistocene, a period in Earth’s history that stretches from around two and a half million years ago to eleven thousand years ago. This was a time when the environment was much colder than now, with huge glaciers and ice sheets flowing over much of the Earth’s northern hemisphere. It was also a time when humans
coexisted with animals that are long extinct, including saber-tooth cats, giant ground sloths, and woolly mammoths.

The Zimovs’ ambitions are an extreme example of “Pleistocene rewilding,” a movement to reintroduce relatively recently extinct large animals, or their close modern-day equivalents, to regions where they were once common. In the case of the Zimovs, the
father-and-son team believe that, by reconstructing the Pleistocene ecosystem in the Siberian steppes and elsewhere, they can slow down the impacts of climate change on these regions. These areas are dominated by permafrost, ground that never thaws through
the year. Permafrost ecosystems have developed and survived over millennia, but a warming global climate (a theme we’ll come back to in chapter twelve and the movie The Day After Tomorrow) threatens to catastrophically disrupt them, and as this happens, the impacts
on biodiversity could be devastating. But what gets climate scientists even more worried is potentially massive releases of trapped methane as the permafrost disappears.

Methane is a powerful greenhouse gas—some eighty times more effective at exacerbating global warming than carbon dioxide— and large-scale releases from warming permafrost could trigger catastrophic changes in climate. As a result, finding ways to keep it in the ground is important. And here the Zimovs came up with a rather unusual idea: maintaining the stability of the environment by reintroducing long-extinct species that could help prevent its destruction, even in a warmer world. It’s a wild idea, but one that has some merit.8 As a proof of concept, though, the Zimovs needed somewhere to start. And so they set out to create a park for deextinct Siberian animals: Pleistocene Park.9

Pleistocene Park is by no stretch of the imagination a modern-day Jurassic Park. The dinosaurs in Hammond’s park date back to the Mesozoic period, from around 250 million years ago to sixty-five million years ago. By comparison, the Pleistocene is relatively modern history, ending a mere eleven and a half thousand years ago. And the vision behind Pleistocene Park is not thrills, spills, and profit, but the serious use of science and technology to stabilize an increasingly unstable environment. Yet there is one thread that ties them together, and that’s using genetic engineering to reintroduce extinct species. In this case, the species in question is warm-blooded and furry: the woolly mammoth.

The idea of de-extinction, or bringing back species from extinction (it’s even called “resurrection biology” in some circles), has been around for a while. It’s a controversial idea, and it raises a lot of tough ethical questions. But proponents of de-extinction argue
that we’re losing species and ecosystems at such a rate that we can’t afford not to explore technological interventions to help stem the flow.

Early approaches to bringing species back from the dead have involved selective breeding. The idea was simple—if you have modern ancestors of a recently extinct species, selectively breeding specimens that have a higher genetic similarity to their forebears can potentially help reconstruct their genome in living animals. This approach is being used in attempts to bring back the aurochs, an ancestor of modern cattle.10 But it’s slow, and it depends on
the fragmented genome of the extinct species still surviving in its modern-day equivalents.

An alternative to selective breeding is cloning. This involves finding a viable cell, or cell nucleus, in an extinct but well-preserved animal and growing a new living clone from it. It’s definitely a more appealing route for impatient resurrection biologists, but it does mean getting your hands on intact cells from long-dead animals and devising ways to “resurrect” these, which is no mean feat. Cloning has potential when it comes to recently extinct species whose cells have been well preserved—for instance, where the whole animal has become frozen in ice. But it’s still a slow and extremely limited option.

Which is where advances in genetic engineering come in.

The technological premise of Jurassic Park is that scientists can reconstruct the genome of long-dead animals from preserved DNA fragments. It’s a compelling idea, if you think of DNA as a massively long and complex instruction set that tells a group of biological molecules how to build an animal. In principle, if we could reconstruct the genome of an extinct species, we would have the basic instruction set—the biological software—to reconstruct
individual members of it.

The bad news is that DNA-reconstruction-based de-extinction is far more complex than this. First you need intact fragments of DNA, which is not easy, as DNA degrades easily (and is pretty much impossible to obtain, as far as we know, for dinosaurs). Then you
need to be able to stitch all of your fragments together, which is akin to completing a billion-piece jigsaw puzzle without knowing what the final picture looks like. This is a Herculean task, although with breakthroughs in data manipulation and machine learning,
scientists are getting better at it. But even when you have your reconstructed genome, you need the biological “wetware”—all the stuff that’s needed to create, incubate, and nurture a new living thing, like eggs, nutrients, a safe space to grow and mature, and so on. Within all this complexity, it turns out that getting your DNA sequence right is just the beginning of translating that genetic code into a living, breathing entity. But in some cases, it might be possible.

In 2013, Sergey Zimov was introduced to the geneticist George Church at a conference on de-extinction. Church is an accomplished scientist in the field of DNA analysis and reconstruction, and a thought leader in the field of synthetic biology (which we’ll come
back to in chapter nine). It was a match made in resurrection biology heaven. Zimov wanted to populate his Pleistocene Park with mammoths, and Church thought he could see a way of
achieving this.

What resulted was an ambitious project to de-extinct the woolly mammoth. Church and others who are working on this have faced plenty of hurdles. But the technology has been advancing so fast that, as of 2017, scientists were predicting they would be able to reproduce the woolly mammoth within the next two years.

One of those hurdles was the lack of solid DNA sequences to work from. Frustratingly, although there are many instances of well preserved woolly mammoths, their DNA rarely survives being frozen for tens of thousands of years. To overcome this, Church and others
have taken a different tack: Take a modern, living relative of the mammoth, and engineer into it traits that would allow it to live on the Siberian tundra, just like its woolly ancestors.

Church’s team’s starting point has been the Asian elephant. This is their source of base DNA for their “woolly mammoth 2.0”—their starting source code, if you like. So far, they’ve identified fifty plus gene sequences they think they can play with to give their modern-day woolly mammoth the traits it would need to thrive in Pleistocene Park, including a coat of hair, smaller ears, and a constitution adapted to cold.

The next hurdle they face is how to translate the code embedded in their new woolly mammoth genome into a living, breathing animal. The most obvious route would be to impregnate a female Asian elephant with a fertilized egg containing the new code. But Asian elephants are endangered, and no one’s likely to allow such cutting edge experimentation on the precious few that are still around, so scientists are working on an artificial womb for their reinvented woolly mammoth. They’re making progress with mice and hope to crack the motherless mammoth challenge relatively soon.

It’s perhaps a stretch to call this creative approach to recreating a species (or “reanimation” as Church refers to it) “de-extinction,” as what is being formed is a new species. … (pp. 31-4)

This selection illustrates what Maynard does so very well throughout the book where he uses each film as a launching pad for a clear, readable description of relevant bits of science so you understand why the premise was likely, unlikely, or pure fantasy while linking it to contemporary practices, efforts, and issues. In the context of Jurassic Park, Maynard goes on to raise some fascinating questions such as: Should we revive animals rendered extinct (due to obsolescence or inability to adapt to new conditions) when we could develop new animals?

General thoughts

‘Films for the Future’ offers readable (to non-scientific types) science, lively writing, and the occasional ‘memorish’ anecdote. As well, Dr. Maynard raises the curtain on aspects of the scientific enterprise that most of us do not get to see.  For example, the meeting  between Sergey Zimov and George Church and how it led to new ‘de-extinction’ work’. He also describes the problems that the scientists encountered and are encountering. This is in direct contrast to how scientific work is usually presented in the news media as one glorious breakthrough after the next.

Maynard does discuss the issues of social inequality and power and ownership. For example, who owns your transplant or data? Puzzlingly, he doesn’t touch on the current environment where scientists in the US and elsewhere are encouraged/pressured to start up companies commercializing their work.

Nor is there any mention of how universities are participating in this grand business experiment often called ‘innovation’. (My March 15, 2017 posting describes an outcome for the CRISPR [gene editing system] patent fight taking place between Harvard University’s & MIT’s [Massachusetts Institute of Technology] Broad Institute vs the University of California at Berkeley and my Sept. 11, 2018 posting about an art/science exhibit in Vancouver [Canada] provides an update for round 2 of the Broad Institute vs. UC Berkeley patent fight [scroll down about 65% of the way.) *To read about how my ‘cultural blindness’ shows up here scroll down to the single asterisk at the end.*

There’s a foray through machine-learning and big data as applied to predictive policing in Maynard’s ‘Minority Report’ chapter (my November 23, 2017 posting describes Vancouver’s predictive policing initiative [no psychics involved], the first such in Canada). There’s no mention of surveillance technology, which if I recall properly was part of the future environment, both by the state and by corporations. (Mia Armstrong’s November 15, 2018 article for Slate on Chinese surveillance being exported to Venezuela provides interesting insight.)

The gaps are interesting and various. This of course points to a problem all science writers have when attempting an overview of science. (Carl Zimmer’s latest, ‘She Has Her Mother’s Laugh: The Powers, Perversions, and Potential of Heredity’] a doorstopping 574 pages, also has some gaps despite his focus on heredity,)

Maynard has worked hard to give an comprehensive overview in a remarkably compact 279 pages while developing his theme about science and the human element. In other words, science is not monolithic; it’s created by human beings and subject to all the flaws and benefits that humanity’s efforts are always subject to—scientists are people too.

The readership for ‘Films from the Future’ spans from the mildly interested science reader to someone like me who’s been writing/blogging about these topics (more or less) for about 10 years. I learned a lot reading this book.

Next time, I’m hopeful there’ll be a next time, Maynard might want to describe the parameters he’s set for his book in more detail that is possible in his chapter headings. He could have mentioned that he’s not a cinéaste so his descriptions of the movies are very much focused on the story as conveyed through words. He doesn’t mention colour palates, camera angles, or, even, cultural lenses.

Take for example, his chapter on ‘Ghost in the Shell’. Focused on the Japanese animation film and not the live action Hollywood version he talks about human enhancement and cyborgs. The Japanese have a different take on robots, inanimate objects, and, I assume, cyborgs than is found in Canada or the US or Great Britain, for that matter (according to a colleague of mine, an Englishwoman who lived in Japan for ten or more years). There’s also the chapter on the Ealing comedy, The Man in The White Suit, an English film from the 1950’s. That too has a cultural (as well as, historical) flavour but since Maynard is from England, he may take that cultural flavour for granted. ‘Never let me go’ in Chapter Two was also a UK production, albeit far more recent than the Ealing comedy and it’s interesting to consider how a UK production about cloning might differ from a US or Chinese or … production on the topic. I am hearkening back to Maynard’s anecdote about movies giving him new ways of seeing and imagining the world.

There’s a corrective. A couple of sentences in Maynard’s introductory chapter cautioning that in depth exploration of ‘cultural lenses’ was not possible without expanding the book to an unreadable size followed by a sentence in each of the two chapters that there are cultural differences.

One area where I had a significant problem was with regard to being “programmed” and having  “instinctual” behaviour,

As a species, we are embarrassingly programmed to see “different” as “threatening,” and to take instinctive action against it. It’s a trait that’s exploited in many science fiction novels and movies, including those in this book. If we want to see the rise of increasingly augmented individuals, we need to be prepared for some social strife. (p. 136)

These concepts are much debated in the social sciences and there are arguments for and against ‘instincts regarding strangers and their possible differences’. I gather Dr. Maynard hies to the ‘instinct to defend/attack’ school of thought.

One final quandary, there was no sex and I was expecting it in the Ex Machina chapter, especially now that sexbots are about to take over the world (I exaggerate). Certainly, if you’re talking about “social strife,” then sexbots would seem to be fruitful line of inquiry, especially when there’s talk of how they could benefit families (my August 29, 2018 posting). Again, there could have been a sentence explaining why Maynard focused almost exclusively in this chapter on the discussions about artificial intelligence and superintelligence.

Taken in the context of the book, these are trifling issues and shouldn’t stop you from reading Films from the Future. What Maynard has accomplished here is impressive and I hope it’s just the beginning.

Final note

Bravo Andrew! (Note: We’ve been ‘internet acquaintances/friends since the first year I started blogging. When I’m referring to him in his professional capacity, he’s Dr. Maynard and when it’s not strictly in his professional capacity, it’s Andrew. For this commentary/review I wanted to emphasize his professional status.)

If you need to see a few more samples of Andrew’s writing, there’s a Nov. 15, 2018 essay on The Conversation, Sci-fi movies are the secret weapon that could help Silicon Valley grow up and a Nov. 21, 2018 article on slate.com, The True Cost of Stain-Resistant Pants; The 1951 British comedy The Man in the White Suit anticipated our fears about nanotechnology. Enjoy.

****Added at 1700 hours on Nov. 22, 2018: You can purchase Films from the Future here.

*Nov. 23, 2018: I should have been more specific and said ‘academic scientists’. In Canada, the great percentage of scientists are academic. It’s to the point where the OECD (Organization for Economic Cooperation and Development) has noted that amongst industrialized countries, Canada has very few industrial scientists in comparison to the others.

Café Scientifique Vancouver (Canada) talk on August 28th 2018: Getting the message: What is gene expression and why does it matter?

Here’s more about the latest Café Scientifique talk from an August  22, 2018 announcement received via email,

Our next café will happen on TUESDAY, AUGUST 28TH at 7:30PM in the back
room at YAGGER'S DOWNTOWN (433 W Pender [St., Vancouver]). Our speaker for the
evening will be DR. KATIE MARSHALL from the Department of Zoology at
UBC [University of British Columbia]. Her topic will be:

GETTING THE MESSAGE: WHAT IS GENE EXPRESSION AND WHY DOES IT MATTER?

Many of us think that DNA is like a light switch; you have a particular
sequence of base pairs or a particular chromosome, and these directly
cause a large change in biological functioning. But the truth is that
any given gene can be up or downregulated through a dizzying array of
biochemical “dimmer switches” that finely control how much that
particular gene is expressed. Understanding how this works is key to
answering questions like “How does a sequence of base pairs in DNA
become a whole organism?” and “Why is it that every cell has the
same DNA sequence but different function?”. We’ll chat about the
advances in computing needed to answer these questions, the importance
of gene expression in disease, and how this science can help us
understand social issues better too.

I wasn’t able to find out too much more about Dr. Katie but there is this profile page on the UBC Zoology Department website,

The long-term goal of my research is to understand how abiotic stress filters through physiology to shape species abundance and distribution. While abiotic stressors such as temperature have been used very successfully to predict population growth, distribution, and diversity of insect species, integration of the mechanisms of how these stressors are experienced by individuals from alteration of physiology through to fitness impacts has lagged. Inclusion of these mechanisms is crucial for accurate modelling predictions of individual (and therefore population-level) responses. My research to date has focused on how the impact of frequency of stress (rather than the duration or intensity of stress) is a superior predictor of both survival and reproductive success , and used insect cold tolerance as a model system.

At UBC I’ll be focusing on the cold tolerance and cryobiology of invertebrates in the intertidal. These organisms face freezing stress through the winter, yet remarkably little is known about how they do so. I’ll also be investigating plasticity in cold tolerance by looking for interactive effects of ocean acidification and community composition on thermal tolerance.

Enjoy!

Being smart about using artificial intelligence in the field of medicine

Since my August 20, 2018 post featured an opinion piece about the possibly imminent replacement of radiologists with artificial intelligence systems and the latest research about employing them for diagnosing eye diseases, it seems like a good time to examine some of the mythology embedded in the discussion about AI and medicine.

Imperfections in medical AI systems

An August 15, 2018 article for Slate.com by W. Nicholson Price II (who teaches at the University of Michigan School of Law; in addition to his law degree he has a PhD in Biological Sciences from Columbia University) begins with the peppy, optimistic view before veering into more critical territory (Note: Links have been removed),

For millions of people suffering from diabetes, new technology enabled by artificial intelligence promises to make management much easier. Medtronic’s Guardian Connect system promises to alert users 10 to 60 minutes before they hit high or low blood sugar level thresholds, thanks to IBM Watson, “the same supercomputer technology that can predict global weather patterns.” Startup Beta Bionics goes even further: In May, it received Food and Drug Administration approval to start clinical trials on what it calls a “bionic pancreas system” powered by artificial intelligence, capable of “automatically and autonomously managing blood sugar levels 24/7.”

An artificial pancreas powered by artificial intelligence represents a huge step forward for the treatment of diabetes—but getting it right will be hard. Artificial intelligence (also known in various iterations as deep learning and machine learning) promises to automatically learn from patterns in medical data to help us do everything from managing diabetes to finding tumors in an MRI to predicting how long patients will live. But the artificial intelligence techniques involved are typically opaque. We often don’t know how the algorithm makes the eventual decision. And they may change and learn from new data—indeed, that’s a big part of the promise. But when the technology is complicated, opaque, changing, and absolutely vital to the health of a patient, how do we make sure it works as promised?

Price describes how a ‘closed loop’ artificial pancreas with AI would automate insulin levels for diabetic patients, flaws in the automated system, and how companies like to maintain a competitive advantage (Note: Links have been removed),

[…] a “closed loop” artificial pancreas, where software handles the whole issue, receiving and interpreting signals from the monitor, deciding when and how much insulin is needed, and directing the insulin pump to provide the right amount. The first closed-loop system was approved in late 2016. The system should take as much of the issue off the mind of the patient as possible (though, of course, that has limits). Running a close-loop artificial pancreas is challenging. The way people respond to changing levels of carbohydrates is complicated, as is their response to insulin; it’s hard to model accurately. Making it even more complicated, each individual’s body reacts a little differently.

Here’s where artificial intelligence comes into play. Rather than trying explicitly to figure out the exact model for how bodies react to insulin and to carbohydrates, machine learning methods, given a lot of data, can find patterns and make predictions. And existing continuous glucose monitors (and insulin pumps) are excellent at generating a lot of data. The idea is to train artificial intelligence algorithms on vast amounts of data from diabetic patients, and to use the resulting trained algorithms to run a closed-loop artificial pancreas. Even more exciting, because the system will keep measuring blood glucose, it can learn from the new data and each patient’s artificial pancreas can customize itself over time as it acquires new data from that patient’s particular reactions.

Here’s the tough question: How will we know how well the system works? Diabetes software doesn’t exactly have the best track record when it comes to accuracy. A 2015 study found that among smartphone apps for calculating insulin doses, two-thirds of the apps risked giving incorrect results, often substantially so. … And companies like to keep their algorithms proprietary for a competitive advantage, which makes it hard to know how they work and what flaws might have gone unnoticed in the development process.

There’s more,

These issues aren’t unique to diabetes care—other A.I. algorithms will also be complicated, opaque, and maybe kept secret by their developers. The potential for problems multiplies when an algorithm is learning from data from an entire hospital, or hospital system, or the collected data from an entire state or nation, not just a single patient. …

The [US Food and Drug Administraiont] FDA is working on this problem. The head of the agency has expressed his enthusiasm for bringing A.I. safely into medical practice, and the agency has a new Digital Health Innovation Action Plan to try to tackle some of these issues. But they’re not easy, and one thing making it harder is a general desire to keep the algorithmic sauce secret. The example of IBM Watson for Oncology has given the field a bit of a recent black eye—it turns out that the company knew the algorithm gave poor recommendations for cancer treatment but kept that secret for more than a year. …

While Price focuses on problems with algorithms and with developers and their business interests, he also hints at some of the body’s complexities.

Can AI systems be like people?

Susan Baxter, a medical writer with over 20 years experience, a PhD in health economics, and author of countless magazine articles and several books, offers a more person-centered approach to the discussion in her July 6, 2018 posting on susanbaxter.com,

The fascination with AI continues to irk, given that every second thing I read seems to be extolling the magic of AI and medicine and how It Will Change Everything. Which it will not, trust me. The essential issue of illness remains perennial and revolves around an individual for whom no amount of technology will solve anything without human contact. …

But in this world, or so we are told by AI proponents, radiologists will soon be obsolete. [my August 20, 2018 post] The adaptational learning capacities of AI mean that reading a scan or x-ray will soon be more ably done by machines than humans. The presupposition here is that we, the original programmers of this artificial intelligence, understand the vagaries of real life (and real disease) so wonderfully that we can deconstruct these much as we do the game of chess (where, let’s face it, Big Blue ate our lunch) and that analyzing a two-dimensional image of a three-dimensional body, already problematic, can be reduced to a series of algorithms.

Attempting to extrapolate what some “shadow” on a scan might mean in a flesh and blood human isn’t really quite the same as bishop to knight seven. Never mind the false positive/negatives that are considered an acceptable risk or the very real human misery they create.

Moravec called it

It’s called Moravec’s paradox, the inability of humans to realize just how complex basic physical tasks are – and the corresponding inability of AI to mimic it. As you walk across the room, carrying a glass of water, talking to your spouse/friend/cat/child; place the glass on the counter and open the dishwasher door with your foot as you open a jar of pickles at the same time, take a moment to consider just how many concurrent tasks you are doing and just how enormous the computational power these ostensibly simple moves would require.

Researchers in Singapore taught industrial robots to assemble an Ikea chair. Essentially, screw in the legs. A person could probably do this in a minute. Maybe two. The preprogrammed robots took nearly half an hour. And I suspect programming those robots took considerably longer than that.

Ironically, even Elon Musk, who has had major production problems with the Tesla cars rolling out of his high tech factory, has conceded (in a tweet) that “Humans are underrated.”

I wouldn’t necessarily go that far given the political shenanigans of Trump & Co. but in the grand scheme of things I tend to agree. …

Is AI going the way of gene therapy?

Susan draws a parallel between the AI and medicine discussion with the discussion about genetics and medicine (Note: Links have been removed),

On a somewhat similar note – given the extent to which genetics discourse has that same linear, mechanistic  tone [as AI and medicine] – it turns out all this fine talk of using genetics to determine health risk and whatnot is based on nothing more than clever marketing, since a lot of companies are making a lot of money off our belief in DNA. Truth is half the time we don’t even know what a gene is never mind what it actually does;  geneticists still can’t agree on how many genes there are in a human genome, as this article in Nature points out.

Along the same lines, I was most amused to read about something called the Super Seniors Study, research following a group of individuals in their 80’s, 90’s and 100’s who seem to be doing really well. Launched in 2002 and headed by Angela Brooks Wilson, a geneticist at the BC [British Columbia] Cancer Agency and SFU [Simon Fraser University] Chair of biomedical physiology and kinesiology, this longitudinal work is examining possible factors involved in healthy ageing.

Turns out genes had nothing to do with it, the title of the Globe and Mail article notwithstanding. (“Could the DNA of these super seniors hold the secret to healthy aging?” The answer, a resounding “no”, well hidden at the very [end], the part most people wouldn’t even get to.) All of these individuals who were racing about exercising and working part time and living the kind of life that makes one tired just reading about it all had the same “multiple (genetic) factors linked to a high probability of disease”. You know, the gene markers they tell us are “linked” to cancer, heart disease, etc., etc. But these super seniors had all those markers but none of the diseases, demonstrating (pretty strongly) that the so-called genetic links to disease are a load of bunkum. Which (she said modestly) I have been saying for more years than I care to remember. You’re welcome.

The fundamental error in this type of linear thinking is in allowing our metaphors (genes are the “blueprint” of life) and propensity towards social ideas of determinism to overtake common sense. Biological and physiological systems are not static; they respond to and change to life in its entirety, whether it’s diet and nutrition to toxic or traumatic insults. Immunity alters, endocrinology changes, – even how we think and feel affects the efficiency and effectiveness of physiology. Which explains why as we age we become increasingly dissimilar.

If you have the time, I encourage to read Susan’s comments in their entirety.

Scientific certainties

Following on with genetics, gene therapy dreams, and the complexity of biology, the June 19, 2018 Nature article by Cassandra Willyard (mentioned in Susan’s posting) highlights an aspect of scientific research not often mentioned in public,

One of the earliest attempts to estimate the number of genes in the human genome involved tipsy geneticists, a bar in Cold Spring Harbor, New York, and pure guesswork.

That was in 2000, when a draft human genome sequence was still in the works; geneticists were running a sweepstake on how many genes humans have, and wagers ranged from tens of thousands to hundreds of thousands. Almost two decades later, scientists armed with real data still can’t agree on the number — a knowledge gap that they say hampers efforts to spot disease-related mutations.

In 2000, with the genomics community abuzz over the question of how many human genes would be found, Ewan Birney launched the GeneSweep contest. Birney, now co-director of the European Bioinformatics Institute (EBI) in Hinxton, UK, took the first bets at a bar during an annual genetics meeting, and the contest eventually attracted more than 1,000 entries and a US$3,000 jackpot. Bets on the number of genes ranged from more than 312,000 to just under 26,000, with an average of around 40,000. These days, the span of estimates has shrunk — with most now between 19,000 and 22,000 — but there is still disagreement (See ‘Gene Tally’).

… the inconsistencies in the number of genes from database to database are problematic for researchers, Pruitt says. “People want one answer,” she [Kim Pruitt, a genome researcher at the US National Center for Biotechnology Information {NCB}] in Bethesda, Maryland] adds, “but biology is complex.”

I wanted to note that scientists do make guesses and not just with genetics. For example, Gina Mallet’s 2005 book ‘Last Chance to Eat: The Fate of Taste in a Fast Food World’ recounts the story of how good and bad levels of cholesterol were established—the experts made some guesses based on their experience. That said, Willyard’s article details the continuing effort to nail down the number of genes almost 20 years after the human genome project was completed and delves into the problems the scientists have uncovered.

Final comments

In addition to opaque processes with developers/entrepreneurs wanting to maintain their secrets for competitive advantages and in addition to our own poor understanding of the human body (how many genes are there anyway?), there are same major gaps (reflected in AI) in our understanding of various diseases. Angela Lashbrook’s August 16, 2018 article for The Atlantic highlights some issues with skin cancer and shade of your skin (Note: Links have been removed),

… While fair-skinned people are at the highest risk for contracting skin cancer, the mortality rate for African Americans is considerably higher: Their five-year survival rate is 73 percent, compared with 90 percent for white Americans, according to the American Academy of Dermatology.

As the rates of melanoma for all Americans continue a 30-year climb, dermatologists have begun exploring new technologies to try to reverse this deadly trend—including artificial intelligence. There’s been a growing hope in the field that using machine-learning algorithms to diagnose skin cancers and other skin issues could make for more efficient doctor visits and increased, reliable diagnoses. The earliest results are promising—but also potentially dangerous for darker-skinned patients.

… Avery Smith, … a software engineer in Baltimore, Maryland, co-authored a paper in JAMA [Journal of the American Medical Association] Dermatology that warns of the potential racial disparities that could come from relying on machine learning for skin-cancer screenings. Smith’s co-author, Adewole Adamson of the University of Texas at Austin, has conducted multiple studies on demographic imbalances in dermatology. “African Americans have the highest mortality rate [for skin cancer], and doctors aren’t trained on that particular skin type,” Smith told me over the phone. “When I came across the machine-learning software, one of the first things I thought was how it will perform on black people.”

Recently, a study that tested machine-learning software in dermatology, conducted by a group of researchers primarily out of Germany, found that “deep-learning convolutional neural networks,” or CNN, detected potentially cancerous skin lesions better than the 58 dermatologists included in the study group. The data used for the study come from the International Skin Imaging Collaboration, or ISIC, an open-source repository of skin images to be used by machine-learning algorithms. Given the rise in melanoma cases in the United States, a machine-learning algorithm that assists dermatologists in diagnosing skin cancer earlier could conceivably save thousands of lives each year.

… Chief among the prohibitive issues, according to Smith and Adamson, is that the data the CNN relies on come from primarily fair-skinned populations in the United States, Australia, and Europe. If the algorithm is basing most of its knowledge on how skin lesions appear on fair skin, then theoretically, lesions on patients of color are less likely to be diagnosed. “If you don’t teach the algorithm with a diverse set of images, then that algorithm won’t work out in the public that is diverse,” says Adamson. “So there’s risk, then, for people with skin of color to fall through the cracks.”

As Adamson and Smith’s paper points out, racial disparities in artificial intelligence and machine learning are not a new issue. Algorithms have mistaken images of black people for gorillas, misunderstood Asians to be blinking when they weren’t, and “judged” only white people to be attractive. An even more dangerous issue, according to the paper, is that decades of clinical research have focused primarily on people with light skin, leaving out marginalized communities whose symptoms may present differently.

The reasons for this exclusion are complex. According to Andrew Alexis, a dermatologist at Mount Sinai, in New York City, and the director of the Skin of Color Center, compounding factors include a lack of medical professionals from marginalized communities, inadequate information about those communities, and socioeconomic barriers to participating in research. “In the absence of a diverse study population that reflects that of the U.S. population, potential safety or efficacy considerations could be missed,” he says.

Adamson agrees, elaborating that with inadequate data, machine learning could misdiagnose people of color with nonexistent skin cancers—or miss them entirely. But he understands why the field of dermatology would surge ahead without demographically complete data. “Part of the problem is that people are in such a rush. This happens with any new tech, whether it’s a new drug or test. Folks see how it can be useful and they go full steam ahead without thinking of potential clinical consequences. …

Improving machine-learning algorithms is far from the only method to ensure that people with darker skin tones are protected against the sun and receive diagnoses earlier, when many cancers are more survivable. According to the Skin Cancer Foundation, 63 percent of African Americans don’t wear sunscreen; both they and many dermatologists are more likely to delay diagnosis and treatment because of the belief that dark skin is adequate protection from the sun’s harmful rays. And due to racial disparities in access to health care in America, African Americans are less likely to get treatment in time.

Happy endings

I’ll add one thing to Price’s article, Susan’s posting, and Lashbrook’s article about the issues with AI , certainty, gene therapy, and medicine—the desire for a happy ending prefaced with an easy solution. If the easy solution isn’t possible accommodations will be made but that happy ending is a must. All disease will disappear and there will be peace on earth. (Nod to Susan Baxter and her many discussions with me about disease processes and happy endings.)

The solutions, for the most part, are seen as technological despite the mountain of evidence suggesting that technology reflects our own imperfect understanding of health and disease therefore providing what is at best an imperfect solution.

Also, we tend to underestimate just how complex humans are not only in terms of disease and health but also with regard to our skills, understanding, and, perhaps not often enough, our ability to respond appropriately in the moment.

There is much to celebrate in what has been accomplished: no more black death, no more smallpox, hip replacements, pacemakers, organ transplants, and much more. Yes, we should try to improve our medicine. But, maybe alongside the celebration we can welcome AI and other technologies with a lot less hype and a lot more skepticism.

The CRISPR ((clustered regularly interspaced short palindromic repeats)-CAS9 gene-editing technique may cause new genetic damage kerfuffle

Setting the stage

Not unexpectedly, CRISPR-Cas9  or clustered regularly interspaced short palindromic repeats-CRISPR-associated protein 9 can be dangerous as these scientists note in a July 16, 2018 news item on phys.org,

Scientists at the Wellcome Sanger Institute have discovered that CRISPR/Cas9 gene editing can cause greater genetic damage in cells than was previously thought. These results create safety implications for gene therapies using CRISPR/Cas9 in the future as the unexpected damage could lead to dangerous changes in some cells.

Reported today (16 July 2018) in the journal Nature Biotechnology, the study also revealed that standard tests for detecting DNA changes miss finding this genetic damage, and that caution and specific testing will be required for any potential gene therapies.

This CRISPR-Cas9 image reminds me of popcorn,

CRISPR-associated protein Cas9 (white) from Staphylococcus aureus based on Protein Database ID 5AXW. Credit: Thomas Splettstoesser (Wikipedia, CC BY-SA 4.0)[ downloaded from https://phys.org/news/2018-07-genome-crisprcas9-gene-higher-thought.html#jCp]

A July 16, 2018 Wellcome Sanger Institute press release (also on EurekAlert), which originated the news item, offers a little more explanation,

CRISPR/Cas9 is one of the newest genome editing tools. It can alter sections of DNA in cells by cutting at specific points and introducing changes at that location. Already extensively used in scientific research, CRISPR/Cas9 has also been seen as a promising way to create potential genome editing treatments for diseases such as HIV, cancer or sickle cell disease. Such therapeutics could inactivate a disease-causing gene, or correct a genetic mutation. However, any potential treatments would have to prove that they were safe.

Previous research had not shown many unforeseen mutations from CRISPR/Cas9 in the DNA at the genome editing target site. To investigate this further the researchers carried out a full systematic study in both mouse and human cells and discovered that CRISPR/Cas9 frequently caused extensive mutations, but at a greater distance from the target site.

The researchers found many of the cells had large genetic rearrangements such as DNA deletions and insertions. These could lead to important genes being switched on or off, which could have major implications for CRISPR/Cas9 use in therapies. In addition, some of these changes were too far away from the target site to be seen with standard genotyping methods.

Prof Allan Bradley, corresponding author on the study from the Wellcome Sanger Institute, said: “This is the first systematic assessment of unexpected events resulting from CRISPR/Cas9 editing in therapeutically relevant cells, and we found that changes in the DNA have been seriously underestimated before now. It is important that anyone thinking of using this technology for gene therapy proceeds with caution, and looks very carefully to check for possible harmful effects.”

Michael Kosicki, the first author from the Wellcome Sanger Institute, said: “My initial experiment used CRISPR/Cas9 as a tool to study gene activity, however it became clear that something unexpected was happening. Once we realised the extent of the genetic rearrangements we studied it systematically, looking at different genes and different therapeutically relevant cell lines, and showed that the CRISPR/Cas9 effects held true.”

The work has implications for how CRISPR/Cas9 is used therapeutically and is likely to re-spark researchers’ interest in finding alternatives to the standard CRISPR/Cas9 method for gene editing.

Prof Maria Jasin, an independent researcher from Memorial Slone Kettering Cancer Centre, New York, who was not involved in the study said: “This study is the first to assess the repertoire of genomic damage arising at a CRISPR/Cas9 cleavage site. While it is not known if genomic sites in other cell lines will be affected in the same way, this study shows that further research and specific testing is needed before CRISPR/Cas9 is used clinically.”

For anyone who’d like to better understand the terms gene editing and CRISPR-Cas9, the Wellcome Sanger Institute provides these explanatory webpages, What is genome editing? and What is CRISPR-Cas9?

For the more advanced, here’s a link and a citation for the paper,

Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements by Michael Kosicki, Kärt Tomberg, & Allan Bradley. Nature Biotechnology DOI: https://doi.org/10.1038/nbt.4192 Published 16 July 2018

This paper appears to be open access.

The kerfuffle

It seems this news has affected the CRISPR market. From a July 16, 2018 article by Cale Guthrie Weissman for Fast Company,

… CRISPR could unknowingly delete or alter non-targeted genes, which could lead to myriad unintended consequences. This is especially frightening, since the technology is going to be used in human clinical trials.

Meanwhile, other scientists working with CRISPR are trying to downplay the findings, telling STAT [a life sciences and business journalism website] that there have been no reported adverse effects similar to what the study describes. The news, however, has brought about a market reaction–at least three publicly traded companies that focus on CRISPR-based therapies are in stock nosedive. Crispr Therapeutics is down by over 6%; Editas fell by over 3%; and Intellia Therapeutics dropped by over 5%. [emphasis mine]

Damage control

Gaetan Burgio (geneticist, Australian National University)  in a July 16, 2018 essay on phys.org (originating from The Conversation) suggests some calm (Note: Links have been removed),

But a new study has called into question the precision of the technique [CRISPR gene editing technology].

The hope for gene editing is that it will be able to cure and correct diseases. To date, many successes have been reported, including curing deafness in mice, and in altering cells to cure cancer.

Some 17 clinical trials in human patients are registered [emphasis mine] testing gene editing on leukaemias, brain cancers and sickle cell anaemia (where red blood cells are misshaped, causing them to die). Before implementing CRISPR technology in clinics to treat cancer or congenital disorders, we must address whether the technique is safe and accurate.

There are a few options for getting around this problem. One option is to isolate the cells we wish to edit from the body and reinject only the ones we know have been correctly edited.

For example, lymphocytes (white blood cells) that are crucial to killing cancer cells could be taken out of the body, then modified using CRISPR to heighten their cancer-killing properties. The DNA of these cells could be sequenced in detail, and only the cells accurately and specifically gene-modified would be selected and delivered back into the body to kill the cancer cells.

While this strategy is valid for cells we can isolate from the body, some cells, such as neurons and muscles, cannot be removed from the body. These types of cells might not be suitable for gene editing using Cas9 scissors.

Fortunately, researchers have discovered other forms of CRISPR systems that don’t require the DNA to be cut. Some CRISPR systems only cut the RNA, not the DNA (DNA contains genetic instructions, RNA convey the instructions on how to synthesise proteins).

As RNA [ribonucleic acid] remains in our cells only for a specific period of time before being degraded, this would allow us to control the timing and duration of the CRISPR system delivery and reverse it (so the scissors are only functional for a short period of time).

This was found to be successful for dementia in mice. Similarly, some CRISPR systems simply change the letters of the DNA, rather than cutting them. This was successful for specific mutations causing diseases such as hereditary deafness in mice.

I agree with Burgio’s conclusion (not included here) that we have a lot more to learn and I can’t help wondering why there are 17 registered human clinical trials at this point.

CRISPR-Cas12a as a new diagnostic tool

Similar to Cas9, Cas12a is has an added feature as noted in this February 15, 2018 news item on ScienceDaily,

Utilizing an unsuspected activity of the CRISPR-Cas12a protein, researchers created a simple diagnostic system called DETECTR to analyze cells, blood, saliva, urine and stool to detect genetic mutations, cancer and antibiotic resistance and also diagnose bacterial and viral infections. The scientists discovered that when Cas12a binds its double-stranded DNA target, it indiscriminately chews up all single-stranded DNA. They then created reporter molecules attached to single-stranded DNA to signal when Cas12a finds its target.

A February 15, 2018 University of California at Berkeley (UC Berkeley) news release by Robert Sanders and which originated the news item, provides more detail and history,

CRISPR-Cas12a, one of the DNA-cutting proteins revolutionizing biology today, has an unexpected side effect that makes it an ideal enzyme for simple, rapid and accurate disease diagnostics.

blood in test tube

(iStock)

Cas12a, discovered in 2015 and originally called Cpf1, is like the well-known Cas9 protein that UC Berkeley’s Jennifer Doudna and colleague Emmanuelle Charpentier turned into a powerful gene-editing tool in 2012.

CRISPR-Cas9 has supercharged biological research in a mere six years, speeding up exploration of the causes of disease and sparking many potential new therapies. Cas12a was a major addition to the gene-cutting toolbox, able to cut double-stranded DNA at places that Cas9 can’t, and, because it leaves ragged edges, perhaps easier to use when inserting a new gene at the DNA cut.

But co-first authors Janice Chen, Enbo Ma and Lucas Harrington in Doudna’s lab discovered that when Cas12a binds and cuts a targeted double-stranded DNA sequence, it unexpectedly unleashes indiscriminate cutting of all single-stranded DNA in a test tube.

Most of the DNA in a cell is in the form of a double-stranded helix, so this is not necessarily a problem for gene-editing applications. But it does allow researchers to use a single-stranded “reporter” molecule with the CRISPR-Cas12a protein, which produces an unambiguous fluorescent signal when Cas12a has found its target.

“We continue to be fascinated by the functions of bacterial CRISPR systems and how mechanistic understanding leads to opportunities for new technologies,” said Doudna, a professor of molecular and cell biology and of chemistry and a Howard Hughes Medical Institute investigator.

DETECTR diagnostics

The new DETECTR system based on CRISPR-Cas12a can analyze cells, blood, saliva, urine and stool to detect genetic mutations, cancer and antibiotic resistance as well as diagnose bacterial and viral infections. Target DNA is amplified by RPA to make it easier for Cas12a to find it and bind, unleashing indiscriminate cutting of single-stranded DNA, including DNA attached to a fluorescent marker (gold star) that tells researchers that Cas12a has found its target.

The UC Berkeley researchers, along with their colleagues at UC San Francisco, will publish their findings Feb. 15 [2018] via the journal Science’s fast-track service, First Release.

The researchers developed a diagnostic system they dubbed the DNA Endonuclease Targeted CRISPR Trans Reporter, or DETECTR, for quick and easy point-of-care detection of even small amounts of DNA in clinical samples. It involves adding all reagents in a single reaction: CRISPR-Cas12a and its RNA targeting sequence (guide RNA), fluorescent reporter molecule and an isothermal amplification system called recombinase polymerase amplification (RPA), which is similar to polymerase chain reaction (PCR). When warmed to body temperature, RPA rapidly multiplies the number of copies of the target DNA, boosting the chances Cas12a will find one of them, bind and unleash single-strand DNA cutting, resulting in a fluorescent readout.

The UC Berkeley researchers tested this strategy using patient samples containing human papilloma virus (HPV), in collaboration with Joel Palefsky’s lab at UC San Francisco. Using DETECTR, they were able to demonstrate accurate detection of the “high-risk” HPV types 16 and 18 in samples infected with many different HPV types.

“This protein works as a robust tool to detect DNA from a variety of sources,” Chen said. “We want to push the limits of the technology, which is potentially applicable in any point-of-care diagnostic situation where there is a DNA component, including cancer and infectious disease.”

The indiscriminate cutting of all single-stranded DNA, which the researchers discovered holds true for all related Cas12 molecules, but not Cas9, may have unwanted effects in genome editing applications, but more research is needed on this topic, Chen said. During the transcription of genes, for example, the cell briefly creates single strands of DNA that could accidentally be cut by Cas12a.

The activity of the Cas12 proteins is similar to that of another family of CRISPR enzymes, Cas13a, which chew up RNA after binding to a target RNA sequence. Various teams, including Doudna’s, are developing diagnostic tests using Cas13a that could, for example, detect the RNA genome of HIV.

infographic about DETECTR system

(Infographic by the Howard Hughes Medical Institute)

These new tools have been repurposed from their original role in microbes where they serve as adaptive immune systems to fend off viral infections. In these bacteria, Cas proteins store records of past infections and use these “memories” to identify harmful DNA during infections. Cas12a, the protein used in this study, then cuts the invading DNA, saving the bacteria from being taken over by the virus.

The chance discovery of Cas12a’s unusual behavior highlights the importance of basic research, Chen said, since it came from a basic curiosity about the mechanism Cas12a uses to cleave double-stranded DNA.

“It’s cool that, by going after the question of the cleavage mechanism of this protein, we uncovered what we think is a very powerful technology useful in an array of applications,” Chen said.

Here’s a link to and a citation for the paper,

CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity by Janice S. Chen, Enbo Ma, Lucas B. Harrington, Maria Da Costa, Xinran Tian, Joel M. Palefsky, Jennifer A. Doudna. Science 15 Feb 2018: eaar6245 DOI: 10.1126/science.aar6245

This paper is behind a paywall.