Page 40«..1020..39404142..5060..»

Category Archives: Genetic Engineering

Genetic engineering of animals: Ethical issues, including …

Posted: November 1, 2021 at 6:39 am

Can Vet J. 2011 May; 52(5): 544550.

Canadian Council on Animal Care, 1510-130 Albert Street, Ottawa, Ontario K1P 5G4 (Ormandy, Dale, Griffin); The University of British Columbia, Animal Welfare Program, 2357 Main Mall, Vancouver, British Columbia V6T 1Z4 (Ormandy)

The genetic engineering of animals has increased significantly in recent years, and the use of this technology brings with it ethical issues, some of which relate to animal welfare defined by the World Organisation for Animal Health as the state of the animalhow an animal is coping with the conditions in which it lives (1). These issues need to be considered by all stakeholders, including veterinarians, to ensure that all parties are aware of the ethical issues at stake and can make a valid contribution to the current debate regarding the creation and use of genetically engineered animals. In addition, it is important to try to reflect societal values within scientific practice and emerging technology, especially publicly funded efforts that aim to provide societal benefits, but that may be deemed ethically contentious. As a result of the extra challenges that genetically engineered animals bring, governing bodies have started to develop relevant policies, often calling for increased vigilance and monitoring of potential animal welfare impacts (2). Veterinarians can play an important role in carrying out such monitoring, especially in the research setting when new genetically engineered animal strains are being developed.

Several terms are used to describe genetically engineered animals: genetically modified, genetically altered, genetically manipulated, transgenic, and biotechnology-derived, amongst others. In the early stages of genetic engineering, the primary technology used was transgenesis, literally meaning the transfer of genetic material from one organism to another. However, with advances in the field, new technology emerged that did not necessarily require transgenesis: recent applications allow for the creation of genetically engineered animals via the deletion of genes, or the manipulation of genes already present. To reflect this progress and to include those animals that are not strictly transgenic, the umbrella term genetically engineered has been adopted into the guidelines developed by the Canadian Council on Animal Care (CCAC). For clarity, in the new CCAC guidelines on: genetically-engineered animals used in science (currently in preparation) the CCAC offers the following definition of a genetically engineered animal: an animal that has had a change in its nuclear or mitochondrial DNA (addition, deletion, or substitution of some part of the animals genetic material or insertion of foreign DNA) achieved through a deliberate human technological intervention. Those animals that have undergone induced mutations (for example, by chemicals or radiation as distinct from spontaneous mutations that naturally occur in populations) and cloned animals are also considered to be genetically engineered due to the direct intervention and planning involved in creation of these animals.

Cloning is the replication of certain cell types from a parent cell, or the replication of a certain part of the cell or DNA to propagate a particular desirable genetic trait. There are 3 types of cloning: DNA cloning, therapeutic cloning, and reproductive cloning (3). For the purposes of this paper, the term cloning is used to refer to reproductive cloning, as this is the most likely to lead to animal welfare issues. Reproductive cloning is used if the intention is to generate an animal that has the same nuclear DNA as another currently, or previously existing animal. The process used to generate this type of cloned animal is called somatic cell nuclear transfer (SCNT) (4).

During the development of the CCAC guidelines on: genetically- engineered animals used in science, some key ethical issues, including animal welfare concerns, were identified: 1) invasiveness of procedures; 2) large numbers of animals required; 3) unanticipated welfare concerns; and 4) how to establish ethical limits to genetic engineering (see Ethical issues of genetic engineering). The different applications of genetically engineered animals are presented first to provide context for the discussion.

Genetic engineering technology has numerous applications involving companion, wild, and farm animals, and animal models used in scientific research. The majority of genetically engineered animals are still in the research phase, rather than actually in use for their intended applications, or commercially available.

By inserting genes from sea anemone and jellyfish, zebrafish have been genetically engineered to express fluorescent proteins hence the commonly termed GloFish. GloFish began to be marketed in the United States in 2003 as ornamental pet fish; however, their sale sparked controversial ethical debates in California the only US state to prohibit the sale of GloFish as pets (5). In addition to the insertion of foreign genes, gene knock-out techniques are also being used to create designer companion animals. For example, in the creation of hypoallergenic cats some companies use genetic engineering techniques to remove the gene that codes for the major cat allergen Fel d1: (http://www.felixpets.com/technology.html).

Companion species have also been derived by cloning. The first cloned cat, CC, was created in 2002 (6). At the time, the ability to clone mammals was a coveted prize, and after just a few years scientists created the first cloned dog, Snuppy (7).

With the exception of a couple of isolated cases, the genetically engineered pet industry is yet to move forward. However, it remains feasible that genetically engineered pets could become part of day-to-day life for practicing veterinarians, and there is evidence that clients have started to enquire about genetic engineering services, in particular the cloning of deceased pets (5).

The primary application of genetic engineering to wild species involves cloning. This technology could be applied to either extinct or endangered species; for example, there have been plans to clone the extinct thylacine and the woolly mammoth (5). Holt et al (8) point out that, As many conservationists are still suspicious of reproductive technologies, it is unlikely that cloning techniques would be easily accepted. Individuals involved in field conservation often harbour suspicions that hi-tech approaches, backed by high profile publicity would divert funding away from their own efforts. However, cloning may prove to be an important tool to be used alongside other forms of assisted reproduction to help retain genetic diversity in small populations of endangered species.

As reviewed by Laible (9), there is an assorted range of agricultural livestock applications [for genetic engineering] aimed at improving animal productivity; food quality and disease resistance; and environmental sustainability. Productivity of farm animal species can be increased using genetic engineering. Examples include transgenic pigs and sheep that have been genetically altered to express higher levels of growth hormone (9).

Genetically engineered farm animals can be created to enhance food quality (9). For example, pigs have been genetically engineered to express the 12 fatty acid desaturase gene (from spinach) for higher levels of omega-3, and goats have been genetically engineered to express human lysozyme in their milk. Such advances may add to the nutritional value of animal-based products.

Farm species may be genetically engineered to create disease-resistant animals (9). Specific examples include conferring immunity to offspring via antibody expression in the milk of the mother; disruption of the virus entry mechanism (which is applicable to diseases such as pseudorabies); resistance to prion diseases; parasite control (especially in sheep); and mastitis resistance (particularly in cattle).

Genetic engineering has also been applied with the aim of reducing agricultural pollution. The best-known example is the EnviropigTM; a pig that is genetically engineered to produce an enzyme that breaks down dietary phosphorus (phytase), thus limiting the amount of phosphorus released in its manure (9).

Despite resistance to the commercialization of genetically engineered animals for food production, primarily due to lack of support from the public (10), a recent debate over genetically engineered AquAdvantageTM Atlantic salmon may result in these animals being introduced into commercial production (11).

Effort has also been made to generate genetically engineered farm species such as cows, goats, and sheep that express medically important proteins in their milk. According to Dyck et al (12), transgenic animal bioreactors represent a powerful tool to address the growing need for therapeutic recombinant proteins. In 2006, ATryn became the first therapeutic protein produced by genetically engineered animals to be approved by the Food and Drug Administration (FDA) of the United States. This product is used as a prophylactic treatment for patients that have hereditary antithrombin deficiency and are undergoing surgical procedures.

Biomedical applications of genetically engineered animals are numerous, and include understanding of gene function, modeling of human disease to either understand disease mechanisms or to aid drug development, and xenotransplantation.

Through the addition, removal, or alteration of genes, scientists can pinpoint what a gene does by observing the biological systems that are affected. While some genetic alterations have no obvious effect, others may produce different phenotypes that can be used by researchers to understand the function of the affected genes. Genetic engineering has enabled the creation of human disease models that were previously unavailable. Animal models of human disease are valuable resources for understanding how and why a particular disease develops, and what can be done to halt or reverse the process. As a result, efforts have focused on developing new genetically engineered animal models of conditions such as Alzheimers disease, amyotrophic lateral sclerosis (ALS), Parkinsons disease, and cancer. However, as Wells (13) points out: these [genetically engineered animal] models do not always accurately reflect the human condition, and care must be taken to understand the limitation of such models.

The use of genetically engineered animals has also become routine within the pharmaceutical industry, for drug discovery, drug development, and risk assessment. As discussed by Rudmann and Durham (14): Transgenic and knock out mouse models are extremely useful in drug discovery, especially when defining potential therapeutic targets for modifying immune and inflammatory responsesSpecific areas for which [genetically engineered animal models] may be useful are in screening for drug induced immunotoxicity, genotoxicity, and carcinogenicity, and in understanding toxicity related drug metabolizing enzyme systems.

Perhaps the most controversial use of genetically engineered animals in science is to develop the basic research on xenotrans-plantation that is, the transplant of cells, tissues, or whole organs from animal donors into human recipients. In relation to organ transplants, scientists have developed a genetically engineered pig with the aim of reducing rejection of pig organs by human recipients (15). This particular application of genetic engineering is currently at the basic research stage, but it shows great promise in alleviating the long waiting lists for organ transplants, as the number of people needing transplants currently far outweighs the number of donated organs. However, as a direct result of public consultation, a moratorium is currently in place preventing pig organ transplantation from entering a clinical trial phase until the public is assured that the potential disease transfer from pigs to humans can be satisfactorily managed (16). According to Health Canada, xenotransplantation is currently not prohibited in Canada. However, the live cells and organs from animal sources are considered to be therapeutic products (drugs or medical devices)No clinical trial involving xenotransplantation has yet been approved by Health Canada (see http://www.hc-sc.gc.ca for details).

Ethical issues, including concerns for animal welfare, can arise at all stages in the generation and life span of an individual genetically engineered animal. The following sections detail some of the issues that have arisen during the peer-driven guidelines development process and associated impact analysis consultations carried out by the CCAC. The CCAC works to an accepted ethic of animal use in science, which includes the principles of the Three Rs (Reduction of animal numbers, Refinement of practices and husbandry to minimize pain and distress, and Replacement of animals with non-animal alternatives wherever possible) (17). Together the Three Rs aim to minimize any pain and distress experienced by the animals used, and as such, they are considered the principles of humane experimental technique. However, despite the steps taken to minimize pain and distress, there is evidence of public concerns that go beyond the Three Rs and animal welfare regarding the creation and use of genetically engineered animals (18).

The generation of a new genetically engineered line of animals often involves the sacrifice of some animals and surgical procedures (for example, vasectomy, surgical embryo transfer) on others. These procedures are not unique to genetically engineered animals, but they are typically required for their production.

During the creation of new genetically engineered animals (particularly mammalian species) oocyte and blastocyst donor females may be induced to superovulate via intraperitoneal or subcutaneous injection of hormones; genetically engineered embryos may be surgically implanted to female recipients; males may be surgically vasectomized under general anesthesia and then used to induce pseudopregnancy in female embryo recipients; and all offspring need to be genotyped, which is typically performed by taking tissue samples, sometimes using tail biopsies or ear notching (19). However, progress is being made to refine the genetic engineering techniques that are applied to mammals (mice in particular) so that less invasive methods are feasible. For example, typical genetic engineering procedures require surgery on the recipient female so that genetically engineered embryos can be implanted and can grow to full term; however, a technique called non-surgical embryo transfer (NSET) acts in a similar way to artificial insemination, and removes the need for invasive surgery (20). Other refinements include a method referred to as deathless transgenesis, which involves the introduction of DNA into the sperm cells of live males and removes the need to euthanize females in order to obtain germ line transmission of a genetic alteration; and the use of polymerase chain reaction (PCR) for genotyping, which requires less tissue than Southern Blot Analysis (20).

Many of the embryos that undergo genetic engineering procedures do not survive, and of those that do survive only a small proportion (between 1% to 30%) carry the genetic alteration of interest (19). This means that large numbers of animals are produced to obtain genetically engineered animals that are of scientific value, and this contradicts efforts to minimize animal use. In addition, the advancement of genetic engineering technologies in recent years has lead to a rapid increase in the number and varieties of genetically engineered animals, particularly mice (21). Although the technology is continually being refined, current genetic engineering techniques remain relatively inefficient, with many surplus animals being exposed to harmful procedures. One key refinement and reduction effort is the preservation of genetically engineered animal lines through the freezing of embryos or sperm (cryopreservation), which is particularly important for those lines with the potential to experience pain and distress (22).

As mentioned, the number of research projects creating and/or using genetically engineered animals worldwide has increased in the past decade (21). In Canada, the CCACs annual data on the numbers of animals used in science show an increase in Category D procedures (procedures with the potential to cause moderate to severe pain and distress) at present the creation of a new genetically engineered animal line is a Category D procedure (23). The data also show an increase in the use of mice (24), which are currently the most commonly used species for genetic engineering, making up over 90% of the genetically engineered animals used in research and testing (21). This rise in animal use challenges the Three Rs principle of Reduction (17). It has been reasoned that once created, the use of genetically engineered animals will reduce the total number of animals used in any given experiment by providing novel and more accurate animal models, especially in applications such as toxicity testing (25). However, the greater variety of available applications, and the large numbers of animals required for the creation and maintenance of new genetically engineered strains indicate that there is still progress to be made in implementation of the Three Rs principle of Reduction in relation to the creation and use of genetically engineered animals (21).

Little data has been collected on the net welfare impacts to genetically engineered animals or to those animals required for their creation, and genetic engineering techniques have been described as both unpredictable and inefficient (19). The latter is due, in part, to the limitations in controlling the integration site of foreign DNA, which is inherent in some genetic engineering techniques (such as pro-nuclear microinjection). In such cases, scientists may generate several independent lines of genetically engineered animals that differ only in the integration site (26), thereby further increasing the numbers of animals involved. This conflicts with efforts to adhere to the principles of the Three Rs, specifically Reduction. With other, more refined techniques that allow greater control of DNA integration (for example, gene targeting), unexpected outcomes are attributed to the unpredictable interaction of the introduced DNA with host genes. These interactions also vary with the genetic background of the animal, as has frequently been observed in genetically engineered mice (27). Interfering with the genome by inserting or removing fragments of DNA may result in alteration of the animals normal genetic homeostasis, which can be manifested in the behavior and well-being of the animals in unpredictable ways. For example, many of the early transgenic livestock studies produced animals with a range of unexpected side effects including lameness, susceptibility to stress, and reduced fertility (9).

A significant limitation of current cloning technology is the prospect that cloned offspring may suffer some degree of abnormality. Studies have revealed that cloned mammals may suffer from developmental abnormalities, including extended gestation; large birth weight; inadequate placental formation; and histological effects in organs and tissues (for example, kidneys, brain, cardiovascular system, and muscle). One annotated review highlights 11 different original research articles that documented the production of cloned animals with abnormalities occurring in the developing embryo, and suffering for the newborn animal and the surrogate mother (28).

Genetically engineered animals, even those with the same gene manipulation, can exhibit a variety of phenotypes; some causing no welfare issues, and some causing negative welfare impacts. It is often difficult to predict the effects a particular genetic modification can have on an individual animal, so genetically engineered animals must be monitored closely to mitigate any unanticipated welfare concerns as they arise. For newly created genetically engineered animals, the level of monitoring needs to be greater than that for regular animals due to the lack of predictability. Once a genetically engineered animal line is established and the welfare concerns are known, it may be possible to reduce the levels of monitoring if the animals are not exhibiting a phenotype that has negative welfare impacts. To aid this monitoring process, some authors have called for the implementation of a genetically engineered animal passport that accompanies an individual animal and alerts animal care staff to the particular welfare needs of that animal (29). This passport document is also important if the intention is to breed from the genetically engineered animal in question, so the appropriate care and husbandry can be in place for the offspring.

With progress in genetic engineering techniques, new methods (30,31) may substantially reduce the unpredictability of the location of gene insertion. As a result, genetic engineering procedures may become less of a welfare concern over time.

As pointed out by Lassen et al (32), Until recently the main limits [to genetic engineering] were technical: what it is possible to do. Now scientists are faced with ethical limits as well: what it is acceptable to do (emphasis theirs). Questions regarding whether it is acceptable to make new transgenic animals go beyond consideration of the Three Rs, animal health, and animal welfare, and prompt the discussion of concepts such as intrinsic value, integrity, and naturalness (33).

When discussing the nature of an animal, it may be useful to consider the Aristotelian concept of telos, which describes the essence and purpose of a creature (34). Philosopher Bernard Rollin applied this concept to animal ethics as follows: Though [telos] is partially metaphysical (in defining a way of looking at the world), and partially empirical (in that it can and will be deepened and refined by increasing empirical knowledge), it is at root a moral notion, both because it is morally motivated and because it contains the notion of what about an animal we ought to at least try to respect and accommodate (emphasis Rollins) (34). Rollin has also argued that as long as we are careful to accommodate the animals interests when we alter an animals telos, it is morally permissible. He writes, given a telos, we should respect the interests which flow from it. This principle does not logically entail that we cannot modify the telos and thereby generate different or alternative interests (34).

Views such as those put forward by Rollin have been argued against on the grounds that health and welfare (or animal interests) may not be the only things to consider when establishing ethical limits. Some authors have made the case that genetic engineering requires us to expand our existing notions of animal ethics to include concepts of the intrinsic value of animals (35), or of animal integrity or dignity (33). Veerhoog argues that, we misuse the word telos when we say that human beings can change the telos of an animal or create a new telos that is to say animals have intrinsic value, which is separate from their value to humans. It is often on these grounds that people will argue that genetic engineering of animals is morally wrong. For example, in a case study of public opinion on issues related to genetic engineering, participants raised concerns about the nature of animals and how this is affected (negatively) by genetic engineering (18).

An alternative view put forward by Schicktanz (36) argues that it is the human-animal relationship that may be damaged by genetic engineering due to the increasingly imbalanced distribution of power between humans and animals. This imbalance is termed asymmetry and it is raised alongside ambivalence as a concern regarding modern human-animal relationships. By using genetically engineered animals as a case study, Schicktanz (36) argues that genetic engineering presents a troubling shift for all human-animal relationships.

Opinions regarding whether limits can, or should, be placed on genetic engineering are often dependent on peoples broader worldview. For some, the genetic engineering of animals may not put their moral principles at risk. For example, this could perhaps be because genetic engineering is seen as a logical continuation of selective breeding, a practice that humans have been carrying out for years; or because human life is deemed more important than animal life. So if genetic engineering creates animals that help us to develop new human medicine then, ethically speaking, we may actually have a moral obligation to create and use them; or because of an expectation that genetic engineering of animals can help reduce experimental animal numbers, thus implementing the accepted Three Rs framework.

For others, the genetic engineering of animals may put their moral principles at risk. For example costs may always be seen to outweigh benefits because the ultimate cost is the violation of species integrity and disregard for the inherent value of animals. Some may view telos as something that cannot or should not be altered, and therefore altering the telos of an animal would be morally wrong. Some may see genetic engineering as exaggerating the imbalance of power between humans and animals, whilst others may fear that the release of genetically engineered animals will upset the natural balance of the ecosystem. In addition, there may be those who feel strongly opposed to certain applications of genetic engineering, but more accepting of others. For example, recent evidence suggests that people may be more accepting of biomedical applications than those relating to food production (37).

Such underlying complexity of views regarding genetic engineering makes the setting of ethical limits difficult to achieve, or indeed, even discuss. However, progress needs to be made on this important issue, especially for those genetically engineered species that are intended for life outside the research laboratory, where there may be less careful oversight of animal welfare. Consequently, limits to genetic engineering need to be established using the full breadth of public and expert opinion. This highlights the importance for veterinarians, as animal health experts, to be involved in the discussion.

Genetic engineering also brings with it concerns over intellectual property, and patenting of created animals and/or the techniques used to create them. Preserving intellectual property can breed a culture of confidentiality within the scientific community, which in turn limits data and animal sharing. Such limits to data and animal sharing may create situations in which there is unnecessary duplication of genetically engineered animal lines, thereby challenging the principle of Reduction. Indeed, this was a concern that was identified in a recent workshop on the creation and use of genetically engineered animals in science (20).

It should be noted that no matter what the application of genetically engineered animals, there are restrictions on the methods of their disposal once they have been euthanized. The reason for this is to restrict the entry of genetically engineered animal carcasses into the natural ecosystem until the long-term effects and risks are better understood. Environment Canada (http://www.ec.gc.ca/) and Health Canada (http://www.hc-sc.gc.ca/) offer specific guidelines in this regard.

As genetically engineered animals begin to enter the commercial realm, it will become increasingly important for veterinarians to inform themselves about any special care and management required by these animals. As animal health professionals, veterinarians can also make important contributions to policy discussions related to the oversight of genetic engineering as it is applied to animals, and to regulatory proceedings for the commercial use of genetically engineered animals.

It is likely that public acceptance of genetically engineered animal products will be an important step in determining when and what types of genetically engineered animals will appear on the commercial market, especially those animals used for food production. Veterinarians may also be called on to inform the public about genetic engineering techniques and any potential impacts to animal welfare and food safety. Consequently, for the discussion regarding genetically engineered animals to progress effectively, veterinarians need to be aware of the current context in which genetically engineered animals are created and used, and to be aware of the manner in which genetic engineering technology and the animals derived from it may be used in the future.

Genetic engineering techniques can be applied to a range of animal species, and although many genetically engineered animals are still in the research phase, there are a variety of intended applications for their use. Although genetic engineering may provide substantial benefits in areas such as biomedical science and food production, the creation and use of genetically engineered animals not only challenge the Three Rs principles, but may also raise ethical issues that go beyond considerations of animal health, animal welfare, and the Three Rs, opening up issues relating to animal integrity and/or dignity. Consequently, even if animal welfare can be satisfactorily safeguarded, intrinsic ethical concerns about the genetic engineering of animals may be cause enough to restrict certain types of genetically engineered animals from reaching their intended commercial application. Given the complexity of views regarding genetic engineering, it is valuable to involve all stakeholders in discussions about the applications of this technology.

The authors thank the members of the Canadian Veterinary Medicine Association Animal Welfare Committee for their comments on the draft, and Dr. C. Schuppli for her insight on how the issues discussed may affect veterinarians.

Use of this article is limited to a single copy for personal study. Anyone interested in obtaining reprints should contact the CVMA office (gro.vmca-amvc@nothguorbh) for additional copies or permission to use this material elsewhere.

See the original post:
Genetic engineering of animals: Ethical issues, including ...

Posted in Genetic Engineering | Comments Off on Genetic engineering of animals: Ethical issues, including …

CRISPR: A game-changing genetic engineering technique …

Posted: at 6:39 am

Have you heard? A revolution has seized the scientific community. Within only a few years, research labs worldwide have adopted a new technology that facilitates making specific changes in the DNA of humans, other animals, and plants. Compared to previous techniques for modifying DNA, this new approach is much faster and easier. This technology is referred to as CRISPR, and it has changed not only the way basic research is conducted, but also the way we can now think about treating diseases [1,2].

CRISPR is an acronym for Clustered Regularly Interspaced Short Palindromic Repeat. This name refers to the unique organization of short, partially palindromic repeated DNA sequences found in the genomes of bacteria and other microorganisms. While seemingly innocuous, CRISPR sequences are a crucial component of the immune systems [3] of these simple life forms. The immune system is responsible for protecting an organisms health and well-being. Just like us, bacterial cells can be invaded by viruses, which are small, infectious agents. If a viral infection threatens a bacterial cell, the CRISPR immune system can thwart the attack by destroying the genome of the invading virus [4]. The genome of the virus includes genetic material that is necessary for the virus to continue replicating. Thus, by destroying the viral genome, the CRISPR immune system protects bacteria from ongoing viral infection.

Figure 1 ~ The steps of CRISPR-mediated immunity. CRISPRs are regions in the bacterial genome that help defend against invading viruses. These regions are composed of short DNA repeats (black diamonds) and spacers (colored boxes). When a previously unseen virus infects a bacterium, a new spacer derived from the virus is incorporated amongst existing spacers. The CRISPR sequence is transcribed and processed to generate short CRISPR RNA molecules. The CRISPR RNA associates with and guides bacterial molecular machinery to a matching target sequence in the invading virus. The molecular machinery cuts up and destroys the invading viral genome. Figure adapted from Molecular Cell 54, April 24, 2014 [5].

Interspersed between the short DNA repeats of bacterial CRISPRs are similarly short variable sequences called spacers (FIGURE 1). These spacers are derived from DNA of viruses that have previously attacked the host bacterium [3]. Hence, spacers serve as a genetic memory of previous infections. If another infection by the same virus should occur, the CRISPR defense system will cut up any viral DNA sequence matching the spacer sequence and thus protect the bacterium from viral attack. If a previously unseen virus attacks, a new spacer is made and added to the chain of spacers and repeats.

The CRISPR immune system works to protect bacteria from repeated viral attack via three basic steps [5]:

Step 1) Adaptation DNA from an invading virus is processed into short segments that are inserted into the CRISPR sequence as new spacers.

Step 2) Production of CRISPR RNA CRISPR repeats and spacers in the bacterial DNA undergo transcription, the process of copying DNA into RNA (ribonucleic acid). Unlike the double-chain helix structure of DNA, the resulting RNA is a single-chain molecule. This RNA chain is cut into short pieces called CRISPR RNAs.

Step 3) Targeting CRISPR RNAs guide bacterial molecular machinery to destroy the viral material. Because CRISPR RNA sequences are copied from the viral DNA sequences acquired during adaptation, they are exact matches to the viral genome and thus serve as excellent guides.

The specificity of CRISPR-based immunity in recognizing and destroying invading viruses is not just useful for bacteria. Creative applications of this primitive yet elegant defense system have emerged in disciplines as diverse as industry, basic research, and medicine.

In Industry

The inherent functions of the CRISPR system are advantageous for industrial processes that utilize bacterial cultures. CRISPR-based immunity can be employed to make these cultures more resistant to viral attack, which would otherwise impede productivity. In fact, the original discovery of CRISPR immunity came from researchers at Danisco, a company in the food production industry [2,3]. Danisco scientists were studying a bacterium called Streptococcus thermophilus, which is used to make yogurts and cheeses. Certain viruses can infect this bacterium and damage the quality or quantity of the food. It was discovered that CRISPR sequences equipped S. thermophilus with immunity against such viral attack. Expanding beyond S. thermophilus to other useful bacteria, manufacturers can apply the same principles to improve culture sustainability and lifespan.

In the Lab

Beyond applications encompassing bacterial immune defenses, scientists have learned how to harness CRISPR technology in the lab [6] to make precise changes in the genes of organisms as diverse as fruit flies, fish, mice, plants and even human cells. Genes are defined by their specific sequences, which provide instructions on how to build and maintain an organisms cells. A change in the sequence of even one gene can significantly affect the biology of the cell and in turn may affect the health of an organism. CRISPR techniques allow scientists to modify specific genes while sparing all others, thus clarifying the association between a given gene and its consequence to the organism.

Rather than relying on bacteria to generate CRISPR RNAs, scientists first design and synthesize short RNA molecules that match a specific DNA sequencefor example, in a human cell. Then, like in the targeting step of the bacterial system, this guide RNA shuttles molecular machinery to the intended DNA target. Once localized to the DNA region of interest, the molecular machinery can silence a gene or even change the sequence of a gene (Figure 2)! This type of gene editing can be likened to editing a sentence with a word processor to delete words or correct spelling mistakes. One important application of such technology is to facilitate making animal models with precise genetic changes to study the progress and treatment of human diseases.

Figure 2 ~ Gene silencing and editing with CRISPR. Guide RNA designed to match the DNA region of interest directs molecular machinery to cut both strands of the targeted DNA. During gene silencing, the cell attempts to repair the broken DNA, but often does so with errors that disrupt the geneeffectively silencing it. For gene editing, a repair template with a specified change in sequence is added to the cell and incorporated into the DNA during the repair process. The targeted DNA is now altered to carry this new sequence.

In Medicine

With early successes in the lab, many are looking toward medical applications of CRISPR technology. One application is for the treatment of genetic diseases. The first evidence that CRISPR can be used to correct a mutant gene and reverse disease symptoms in a living animal was published earlier this year [7]. By replacing the mutant form of a gene with its correct sequence in adult mice, researchers demonstrated a cure for a rare liver disorder that could be achieved with a single treatment. In addition to treating heritable diseases, CRISPR can be used in the realm of infectious diseases, possibly providing a way to make more specific antibiotics that target only disease-causing bacterial strains while sparing beneficial bacteria [8]. A recent SITN Waves article discusses how this technique was also used to make white blood cells resistant to HIV infection [9].

Of course, any new technology takes some time to understand and perfect. It will be important to verify that a particular guide RNA is specific for its target gene, so that the CRISPR system does not mistakenly attack other genes. It will also be important to find a way to deliver CRISPR therapies into the body before they can become widely used in medicine. Although a lot remains to be discovered, there is no doubt that CRISPR has become a valuable tool in research. In fact, there is enough excitement in the field to warrant the launch of several Biotech start-ups that hope to use CRISPR-inspired technology to treat human diseases [8].

Ekaterina Pak is a Ph.D. student in the Biological and Biomedical Sciences program at Harvard Medical School.

1. Palca, J. A CRISPR way to fix faulty genes. (26 June 2014) NPR < http://www.npr.org/blogs/health/2014/06/26/325213397/a-crispr-way-to-fix-faulty-genes> [29 June 2014]

2. Pennisi, E. The CRISPR Craze. (2013) Science, 341 (6148): 833-836.

3. Barrangou, R., Fremaux, C., Deveau, H., Richards, M., Boyaval, P., Moineau, S., Romero, D.A., and Horvath, P. (2007). CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 17091712.

4. Brouns, S.J., Jore, M.M., Lundgren, M., Westra, E.R., Slijkhuis, R.J., Snijders, A.P., Dickman, M.J., Makarova, K.S., Koonin, E.V., and van der Oost, J. (2008). Small CRISPR RNAs guide antiviral defense in prokaryotes. Science 321, 960964.

5. Barrangou, R. and Marraffini, L. CRISPR-Cas Systems: Prokaryotes Upgrade to Adaptive Immunity (2014). Molecular Cell 54, 234-244.

6. Jinkek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. (2012) 337(6096):816-21.

7. CRISPR reverses disease symptoms in living animals for first time. (31 March 2014). Genetic Engineering and Biotechnology News. <http://www.genengnews.com/gen-news-highlights/crispr-reverses-disease-symptoms-in-living-animals-for-first-time/81249682/> [27 July 2014]

8. Pollack, A. A powerful new way to edit DNA. (3 March 2014). NYTimes < http://www.nytimes.com/2014/03/04/health/a-powerful-new-way-to-edit-dna.html?_r=0> [16 July 2014]

9. Gene editing technique allows for HIV resistance? <http://sitn.hms.harvard.edu/flash/waves/2014/gene-editing-technique-allows-for-hiv-resistance/> [13 June 2014]

Read the original post:
CRISPR: A game-changing genetic engineering technique ...

Posted in Genetic Engineering | Comments Off on CRISPR: A game-changing genetic engineering technique …

Genetics in fiction – Wikipedia

Posted: at 6:39 am

Aspects of genetics including mutation, hybridisation, cloning, genetic engineering, and eugenics have appeared in fiction since the 19th century.

Genetics is a young science, having started in 1900 with the rediscovery of Gregor Mendel's study on the inheritance of traits in pea plants. During the 20th century it developed to create new sciences and technologies including molecular biology, DNA sequencing, cloning, and genetic engineering. The ethical implications were brought into focus with the eugenics movement.

Since then, many science fiction novels and films have used aspects of genetics as plot devices, often taking one of two routes: a genetic accident with disastrous consequences; or, the feasibility and desirability of a planned genetic alteration. The treatment of science in these stories has been uneven and often unrealistic. The film Gattaca did attempt to portray science accurately but was criticised by scientists.

Modern genetics began with the work of the monk Gregor Mendel in the 19th century, on the inheritance of traits in pea plants. Mendel found that visible traits, such as whether peas were round or wrinkled, were inherited discretely, rather than by blending the attributes of the two parents.[1] In 1900, Hugo de Vries and other scientists rediscovered Mendel's research; William Bateson coined the term "genetics" for the new science, which soon investigated a wide range of phenomena including mutation (inherited changes caused by damage to the genetic material), genetic linkage (when some traits are to some extent inherited together), and hybridisation (crosses of different species).[2]

Eugenics, the production of better human beings by selective breeding, was named and advocated by Charles Darwin's cousin, the scientist Francis Galton, in 1883. It had both a positive aspect, the breeding of more children with high intelligence and good health; and a negative aspect, aiming to suppress "race degeneration" by preventing supposedly "defective" families with attributes such as profligacy, laziness, immoral behaviour and a tendency to criminality from having children.[3][4]

Molecular biology, the interactions and regulation of genetic materials, began with the identification in 1944 of DNA as the main genetic material;[5] the genetic code and the double helix structure of DNA was determined by James Watson and Francis Crick in 1953.[6][7] DNA sequencing, the identification of an exact sequence of genetic information in an organism, was developed in 1977 by Frederick Sanger.[8]

Genetic engineering, the modification of the genetic material of a live organism, became possible in 1972 when Paul Berg created the first recombinant DNA molecules (artificially assembled genetic material) using viruses.[9]

Cloning, the production of genetically identical organisms from some chosen starting point, was shown to be practicable in a mammal with the creation of Dolly the sheep from an ordinary body cell in 1996 at the Roslin Institute.[10]

Mutation and hybridisation are widely used in fiction, starting in the 19th century with science fiction works such as Mary Shelley's 1818 novel Frankenstein and H. G. Wells's 1896 The Island of Dr Moreau.[11]

In her 1977 Biological Themes in Modern Science Fiction, Helen Parker identified two major types of story: "genetic accident", the uncontrolled, unexpected and disastrous alteration of a species;[12][13] and "planned genetic alteration", whether controlled by humans or aliens, and the question of whether that would be either feasible or desirable.[12][13] In science fiction up to the 1970s, the genetic changes were brought about by radiation, breeding programmes, or manipulation with chemicals or surgery (and thus, notes Lars Schmeink, not necessarily by strictly genetic means).[13] Examples include The Island of Dr Moreau with its horrible manipulations; Aldous Huxley's 1932 Brave New World with a breeding programme; and John Taine's 1951 Seeds of Life, using radiation to create supermen.[13] After the discovery of the double helix and then recombinant DNA, genetic engineering became the focus for genetics in fiction, as in books like Brian Stableford's tale of a genetically modified society in his 1998 Inherit the Earth, or Michael Marshall Smith's story of organ farming in his 1997 Spares.[13]

Comic books have imagined mutated superhumans with extraordinary powers. The DC Universe (from 1939) imagines "metahumans"; the Marvel Universe (from 1961) calls them "mutants", while the Wildstorm (from 1992) and Ultimate Marvel (20002015) Universes name them "posthumans".[14] Stan Lee introduced the concept of mutants in the Marvel X-Men books in 1963; the villain Magneto declares his plan to "make Homo sapiens bow to Homo superior!", implying that mutants will be an evolutionary step up from current humanity. Later, the books speak of an X-gene that confers powers from puberty onwards. X-men powers include telepathy, telekinesis, healing, strength, flight, time travel, and the ability to emit blasts of energy. Marvel's god-like Celestials are later (1999) said to have visited Earth long ago and to have modified human DNA to enable mutant powers.[15]

James Blish's 1952 novel Titan's Daughter (in Kendell Foster Crossen's Future Tense collection) featured stimulated polyploidy (giving organisms multiple sets of genetic material, something that can create new species in a single step), based on spontaneous polyploidy in flowering plants, to create humans with more than normal height, strength, and lifespans.[16]

Cloning, too, is a familiar plot device. Aldous Huxley's 1931 dystopian novel Brave New World imagines the in vitro cloning of fertilised human eggs.[17][18] Huxley was influenced by J. B. S. Haldane's 1924 non-fiction book Daedalus; or, Science and the Future, which used the Greek myth of Daedalus to symbolise the coming revolution in genetics; Haldane predicted that humans would control their own evolution through directed mutation and in vitro fertilisation.[19] Cloning was explored further in stories such as Poul Anderson's 1953 UN-Man.[20] In his 1976 novel, The Boys from Brazil, Ira Levin describes the creation of 96 clones of Adolf Hitler, replicating for all of them the rearing of Hitler (including the death of his father at age 13), with the goal of resurrecting Nazism. In his 1990 novel Jurassic Park, Michael Crichton imagined the recovery of the complete genome of a dinosaur from fossil remains, followed by its use to recreate living animals of an extinct species.[11]

Cloning is a recurring theme in science fiction films like Jurassic Park (1993), Alien Resurrection (1997), The 6th Day (2000), Resident Evil (2002), Star Wars: Episode II (2002) and The Island (2005). The process of cloning is represented variously in fiction. Many works depict the artificial creation of humans by a method of growing cells from a tissue or DNA sample; the replication may be instantaneous, or take place through slow growth of human embryos in artificial wombs. In the long-running British television series Doctor Who, the Fourth Doctor and his companion Leela were cloned in a matter of seconds from DNA samples ("The Invisible Enemy", 1977) and thenin an apparent homage to the 1966 film Fantastic Voyageshrunk to microscopic size in order to enter the Doctor's body to combat an alien virus. The clones in this story are short-lived, and can only survive a matter of minutes before they expire.[21] Films such as The Matrix and Star Wars: Episode II Attack of the Clones have featured human foetuses being cultured on an industrial scale in enormous tanks.[22]

Cloning humans from body parts is a common science fiction trope, one of several genetics themes parodied in Woody Allen's 1973 comedy Sleeper, where an attempt is made to clone an assassinated dictator from his disembodied nose.[23]

Genetic engineering features in many science fiction stories.[16] Films such as The Island (2005) and Blade Runner (1982) bring the engineered creature to confront the person who created it or the being it was cloned from, a theme seen in some film versions of Frankenstein. Few films have informed audiences about genetic engineering as such, with the exception of the 1978 The Boys from Brazil and the 1993 Jurassic Park, both of which made use of a lesson, a demonstration, and a clip of scientific film.[11][24] In 1982, Frank Herbert's novel The White Plague described the deliberate use of genetic engineering to create a pathogen which specifically killed women.[16] Another of Herbert's creations, the Dune series of novels, starting with Dune in 1965, emphasises genetics. It combines selective breeding by a powerful sisterhood, the Bene Gesserit, to produce a supernormal male being, the Kwisatz Haderach, with the genetic engineering of the powerful but despised Tleilaxu.[25]

Genetic engineering methods are weakly represented in film; Michael Clark, writing for The Wellcome Trust, calls the portrayal of genetic engineering and biotechnology "seriously distorted"[24] in films such as Roger Spottiswoode's 2000 The 6th Day, which makes use of the trope of a "vast clandestine laboratory ... filled with row upon row of 'blank' human bodies kept floating in tanks of nutrient liquid or in suspended animation". In Clark's view, the biotechnology is typically "given fantastic but visually arresting forms" while the science is either relegated to the background or fictionalised to suit a young audience.[24]

Eugenics plays a central role in films such as Andrew Niccol's 1997 Gattaca, the title alluding to the letters G, A, T, C for guanine, adenine, thymine, and cytosine, the four nucleobases of DNA. Genetic engineering of humans is unrestricted, resulting in genetic discrimination, loss of diversity, and adverse effects on society. The film explores the ethical implications; the production company, Sony Pictures, consulted with a gene therapy researcher, French Anderson, to ensure that the portrayal of science was realistic, and test-screened the film with the Society of Mammalian Cell Biologists and the American National Human Genome Research Institute before its release. This care did not prevent researchers from attacking the film after its release. Philim Yam of Scientific American called it "science bashing"; in Nature Kevin Davies called it a ""surprisingly pedestrian affair"; and the molecular biologist Lee Silver described the film's extreme genetic determinism as "a straw man".[26][27]

The geneticist Dan Koboldt observes that while science and technology play major roles in fiction, from fantasy and science fiction to thrillers, the representation of science in both literature and film is often unrealistic.[28] In Koboldt's view, genetics in fiction is frequently oversimplified, and some myths are common and need to be debunked. For example, the Human Genome Project has not (he states) immediately led to a Gattaca world, as the relationship between genotype and phenotype is not straightforward. People do differ genetically, but only very rarely because they are missing a gene that other people have: people have different alleles of the same genes. Eye and hair colour are controlled not by one gene each, but by multiple genes. Mutations do occur, but they are rare: people are 99.99% identical genetically, the 3 million differences between any two people being dwarfed by the hundreds of millions of DNA bases which are identical; nearly all DNA variants are inherited, not acquired afresh by mutation. And, Koboldt writes, believable scientists in fiction should know their knowledge is limited.[29]

Originally posted here:
Genetics in fiction - Wikipedia

Posted in Genetic Engineering | Comments Off on Genetics in fiction – Wikipedia

CRISPR Revolution: Do We Need Tighter Gene-Editing Regulations? No – American Council on Science and Health

Posted: at 6:39 am

Life goes on as gene-edited foods begin to hit the market. Japanese consumers have recently startedbuying tomatoes that fight high blood pressure, and Americans have been consuming soy engineered to produce high amounts of heart-healthy oils for a little over two years. Few people noticed these developments because, as scientists have said for a long time, the safety profile of a crop is not dictated by the breeding method that produced it. For all intents and purposes, it seems that food-safety regulators have done a reasonablejob of safeguarding public health against whatever hypothetical risks gene editing may pose.

But this has not stopped critics of genetic engineering from advocating for more federal oversight of CRISPR and othertechniquesused to make discrete changes to the genomes of plants, animals and other organisms we use for food or medicine. Over at The Conversation, a team of scientists recently made the case for tighter rules in Calling the latest gene technologies natural is a semantic distraction they must still be regulated.

Many scientists have defended gene editing, in part, by arguing that it simply mimics nature. A mutation that boosts the nutrient content of rice, for example, is the same whether it was induced by a plant breeder or some natural phenomenon. Indeed, the DNA of plants and animals we eat contains untold numbers of harmless, naturally occurringmutations. But The Conversation authors will have none of this:

Unfortunately, the risks from technology dont disappear by calling it natural... Proponents of deregulation of gene technology use the naturalness argument to make their case. But we argue this is not a good basis for deciding whether a technology should be regulated.

They have written a very long peer-reviewed article outlining a regulatory framework based on "scale of use."The ideais that the more widely a technology is implemented, the greater risk it may pose to human health and the environment, which necessitates regulatory "control points" to ensure its safe use. It's an interesting proposal, but it's plagued by several serious flaws.

Where's the data?

The most significant issue with a scale-based regulatory approachis that it's a reaction to risks that have never materialized. This isn't to say that a potentially harmful genetically engineered organism will never be commercialized. But if we're going to upend our biotechnology regulatory framework, we need to do so based on real-world evidence. Some experts have actually argued, based on decades of safety data, that the US over-regulates biotech products. As biologist and ACSHadvisorDr. Henry Miller and legal scholar John Cohrssen wrote recently in Nature:

After 35 years of real-world experience with genetically engineered plants and microorganisms, and countless risk-assessment experiments, it is past time to reevaluate the rationale for, and the costs and benefits of, the case-by-case reviews of genetically engineered products now required by the US Environmental Protection Agency (EPA), US Department of Agriculture (USDA) and US Food and Drug Administration (FDA).

The problem with scale

Real-world data aside for the moment, there are some theoretical problems with the scalabilitymodel as well. Theargument assumes thatrisks associated with gene editing proliferate as use of the technology expands, because each gene edit carries a certain level of risk. This is a false assumption, as plant geneticist Kevin Folta pointed out on a recent episode of the podcast we co-host (21 minute mark).

Scientists have a variety of tools with which to monitor and limit the effects of specific gene edits. For example, proteins known as anti-CRISPRs can be utilized to halt the gene-editing machinery so it makes only the changes we want it to. University of Toronto biochemist Karen Maxwell has explained how this could work in practice:

In genome editing applications, anti-CRISPRs may provide a valuable 'off switch for Cas9 activity for therapeutic uses and gene drives. One concern of CRISPR-Cas gene editing technology is the limited ability to control its activity after it has been delivered to the cell . which can lead to off-target mutations. Anti-CRISPRs can potentially be exploited to target Cas9 activity to particular tissues or organs, to particular points of the cell cycle, or to limit the amount of time it is active

Suffice it to say that these and other safeguards significantly alter the risk equation and weaken concerns about a gene-edits-gone-wild scenario. Parenthetically, scientists design these sorts of preventative measures as they develop more genetic engineering applications for widespread use. This is why the wide variety of cars in production today have safety features that would have been unheard of in years past.

Absurdity alert: The A-Bomb analogy

To bolster their argument, The Conversation authors made the following analogy:

Imagine if other technologies with the capacity to harm were governed by resemblance to nature. Should we deregulate nuclear bombs because the natural decay chain of uranium-238 also produces heat, gamma radiation and alpha and beta particles? We inherently recognize the fallacy of this logic. The technology risk equation is more complicated than a supercilious 'its just like nature' argument

If someone has to resort to this kind of rhetoric, the chances are excellent that their argument is weak. Fat Man and Little Boy, the bombs dropped on Japan in 1945, didn't destroy two cities because a nuclear physicist in New Mexico made a technical mistake. These weapons are designed to wreak havoc. Tomatoes bred to produce more of an amino acid, in contrast, are not.

The point of arguing that gene-editing techniques mimic natural processes isn't to assert that natural stuff is good; therefore, gene editing is also good. Instead, the point is to illustrate that inducing mutations in the genomes of plants and animals is not novel or uniquely risky. Even the overpriced products marketed as all-natural have been improved by mutations resulting from many years of plant breeding.

Nonetheless, some scientists have argued that reframing the gene-editing conversation in terms of risk vs benefit would be a smarter approach than making comparisons to nature. I agree with them, so let's start now. The benefits of employing gene editing to improve our food supply and treat disease far outweigh the potential risks, which we can mitigate. Very little about modern life is naturaland it's time we all got over it.

More:
CRISPR Revolution: Do We Need Tighter Gene-Editing Regulations? No - American Council on Science and Health

Posted in Genetic Engineering | Comments Off on CRISPR Revolution: Do We Need Tighter Gene-Editing Regulations? No – American Council on Science and Health

Empowering algae to shape the future of bioenergy – ASU Now

Posted: at 6:39 am

Humble microalgae may seem minor at first glance, but when optimally farmed and converted into biofuels, the potential of this renewable resource to combat climate change is anything but insignificant.

Through the extraction of lipids, they can be converted into biofuels. And, like plants, photosynthesizing algae absorb carbon dioxide, or CO2, and release oxygen into the atmosphere. But algae can do that at much faster rates and higher efficiencies than plants, and they dont need arable land or even fresh water to grow, which has sustainability scientists and engineers intrigued. Taylor Weiss (left), co-PI and assistant professor of environmental and resource management at the Polytechnic School, and Duane Barbano (right), a biological design PhD candidate in the School for Engineering of Matter, Transport and Energy, are farming algae in a pond at the Arizona Center for Algae Technology and Innovation on ASU's Polytechnic campus. Photo by Deanna Dent/ASU Download Full Image

On a small scale, converting algae into biofuels can be fairly straightforward. However, for an algal system to be sustainable, scalable and economical, it must be able to deliver and utilize CO2 efficiently.

A new U.S. Department of Energy grant awarded to the Arizona Center for Algae Technology and Innovation, or AzCATI, will investigate novel methods of CO2 sourcing, delivery and absorption with the goal of promoting algae resiliency and pathways to large-scale biomass production and eventual conversion into low-carbon biofuels an alternative to petroleum.

This initiative is especially important in reducing the carbon footprint of the transportation sector specifically airplanes and ships which accounts for approximately 30% of total U.S. energy consumption and generates the largest share of the countrys greenhouse gas emissions.

AzCATI, located on 4 acres of Arizona State Universitys Polytechnic campus, is home to one of the countrys largest and most comprehensive algae test-bed facilities. In partnership with researchers from all over the world, AzCATI has been investigating algal technology since its establishment in 2010 and has since attracted more than $45 million in federal, state and private funding.

AzCATI will receive $3.2 million for this DOE-supported effort out of a total $34 million in funding for 11 industry- and university-led projects to support the high-impact research and development of biofuels, biopower and bioproducts.

John McGowen, a portfolio manager for research in the Knowledge Enterprise at ASU, will lead the project. He says that about 80% of algae funding at ASU is from the DOE.

We are essentially a national test bed, the longest-running, continually funded outdoor cultivation test bed in the country that isnt commercial. With our experienced faculty, staff, upwards of 30 students and unique testing abilities, we are set up to test new technologies, break them and move on, or improve them and make breakthroughs.

McGowen was one of AzCATIs first researchers and has witnessed the evolution of algae research over the past 11 years.

He explains that the high levels of oils and carbohydrates and proteins created by algae are refined and used as various forms of biofuels and valuable bioproducts.

McGowen says its important to know that the high-density algae needed to create biofuel cant be grown naturally in the environment because of current CO2 levels in the atmosphere, and that they need an additional source delivered directly to them to be viable for this purpose.

A trifecta of research objectives will define AzCATIs three-year DOE project, titled, Direct Air Capture Integration With Algae Carbon Biocatalysis. Researchers at AzCATI will model a novel technology called passive-direct air capture, or PDAC, developed by Klaus Lackner, a professor at the School of Sustainable Engineering and the Built Environment, one of the seven Ira A. Fulton Schools of Engineering at ASU.

Coinciding research entails precisely delivering the CO2 product derived from PDAC to the algae for optimal absorption and low product loss, followed by improving the algaes ability to assimilate CO2 for more resilient and robust ponds.

The goal of PDAC is to offer a sustainable and efficient supply of self-sourced CO2 from the atmosphere versus conventionally purchasing costly CO2 from the merchant market. It also may help in shifting the paradigm on the cost of CO2, McGowen says. This method of CO2 sourcing would remove the necessity for algae to be co-located near a point source emitter, such as a power plant or a CO2 pipeline, meaning they could potentially grow anywhere at scale an essential step in large-scale biofuel production.

The collaboration of key partners will make this concept a reality. Carbon Collect Limited, which has licensed technology developed by Lackner and the Center for Negative Carbon Emissions at ASU, has commercialized PDAC through the development of MechanicalTrees, which according to their website are a thousand times more efficient than natural trees at removing CO2 from the air.

AzCATI will leverage Carbon Collects installation in Tempe, Arizona, and use the CO2 generated from their MechanicalTrees. It will be transported in truckloads to AzCATI and will serve as the main CO2 source for their research, meaning there will be two wholly completed unique test-bed facilities at ASU directly interacting with each other, says Taylor Weiss, co-PI and assistant professor of environmental and resource management at The Polytechnic School.

In this case, the MechanicalTrees arent in close proximity to the algae ponds at AzCATI, requiring the need for CO2 transportation. However, in theory, strategically placing a cluster of MechanicalTrees on an algae crop would offer a continuous and unlimited source of CO2, achieving a self-sustaining crop wherever it makes sense to grow it, McGowen says.

The most promising locations possessing both the water resources and ideal climate for high-productivity algae cultivation are not near pipeline infrastructure, nor do they have the available land, he says. This is where the need for PDAC technology becomes apparent.

Weiss says that even with a sustainably sourced supply of CO2 through PDAC, there remain additional challenges in achieving high productivity, including how efficiently you can deliver that CO2 into the culture and how efficiently the algae can actually convert that CO2 into the most ideal form, in particular for biofuels.

Additional research partners the National Renewable Energy Laboratory, or NREL, and Burge Environmentalwill assist in taking on these challenges. They will offer expertise in innovative CO2 delivery and biocatalysis or supporting the CO2 uptake within algae cells, as well as providing support in genetically engineering algae to give them the ability to assimilate CO2 to improve the microbial ecology within a pond to enable robust outdoor cultivation.

Weiss believes that NRELs expertise will not only improve the efficiency of CO2 dissolution into the culture once it has been captured by PDAC, but will also leverage years of experience building a genetic engineering toolkit to enhance the rate of CO2 uptake by the algae cells.

McGowen and Weiss say that using algae for atmospheric CO2 mitigation to combat climate change is a promising pathway. They also think that algae are only part of the toolbox when it comes to decarbonizing the atmosphere, and they hope to see other technologies and innovations work in tandem with algae to make significant breakthroughs.

This investigation is about redirecting the CO2 within the cell into different forms of more valuable carbon products, while eliminating environmental threats to the algae that contribute to lower output, Weiss says. We look forward to putting this technology into action and empowering algae to reach their full potential.

Here is the original post:
Empowering algae to shape the future of bioenergy - ASU Now

Posted in Genetic Engineering | Comments Off on Empowering algae to shape the future of bioenergy – ASU Now

Five companies that underwent major rebrands – The National

Posted: at 6:39 am

Facebook on Thursday announced it is changing its name to Meta, joining a long list of well-known companies that have undergone major rebrands.

The new name is meant to reflect the technology company's shifting focus on its virtual "metaverse" world, which chief executive Mark Zuckerberg showcased during Facebook's annual conference on virtual and augmented reality.

Here are five other companies that taken on new names:

After changing its name to Alphabet in 2015, a new "slimmed-down" Google allowed investors to focus on the core search business. AP

Search behemoth Google worth more than $400 billion at the time shockingly announced in 2015 that it was changing its name to Alphabet, a technology conglomerate.

We liked the name 'Alphabet' because it means a collection of letters that represent language, one of humanity's most important innovations, and is the core of how we index with Google search, former chief executive Larry Page said in a blog post.

The slimmed-down Google allowed investors to focus on the strengths of the core search business.

Alphabet would take on some of the riskier ventures including genetic engineering and self-driving cars.

Dunkin' Donuts launched the tagline America runs on Dunkin' in 2006 and, after months of testing over a decade later, dropped Donuts from its name and logo.

Growing pressure from coffee chains and changes in Americans' eating habits led the company to shift its focus to drinks, which Dunkin' Brands chief executive David Hoffmann said has a higher margin for profit than its food.

The 2018 rebrand, complete with a new, modern logo, was meant to reflect a more streamlined concept.

By simplifying and modernising our name, while still paying homage to our heritage, we have an opportunity to create an incredible new energy for Dunkin," Mr Hoffman said.

But doughnuts are still on the menu and the chain sells billions of the pastries every year.

In 2002, the world's biggest wrestling company was forced to change its name after a legal battle.

The World Wrestling Federation, as it was then known, found itself in trouble with the World Wildlife Fund. The wilderness preservation charity had branded itself under the same abbreviation, WWF, 18 years before the Federation.

The World Wildlife Fund in 1994 insisted the Federation sign a legal document ensuring the wrestling company would limit its use of WWF outside of North America. In return, the Fund would not press further charges.

But the wrestling company largely ignored the agreement and continued to brand itself as WWF worldwide, going so far as to register a web domain nearly identical with that of the Fund. Following the wrestling boom of the late 1990s, Federation chief Vince McMachon's company landed in hot water again.

The charity successfully sued the Federation in 2000, forcing Mr McMahon to rebrand his wrestling empire.

Mr McMahon changed the wrestling company's name in 2002 to World Wrestling Entertainment, where it eventually came to be known simply as WWE.

The logo of Exxon Mobil Corporation is shown on a monitor above the floor of the New York Stock Exchange. Reuters

Famed entrepreneur John D Rockefeller's Standard Oil company once controlled more than 90 per cent of oil production in the US. As a result, an antitrust suit was filed in 1906, with the company accused of raising prices where it had a monopoly and slashing prices where it faced competition.

The oil company was broken up into 34 different companies in 1911, primarily based on geographical region. Two of these successor companies are now the largest oil companies in the US: Chevron and ExxonMobil.

In 2000, Chevron acquired Texaco in a deal valued at $45 billion, becoming ChevronTexaco only to drop Texaco from its name a few years later.

A year earlier, two of Standard Oil's largest offshoots reunited in a blockbuster merger.

Exxon, part of the Standard Oil New Jersey branch, signed a $75.3bn merger agreement with the New York successor, Mobil. Following this merger, the company rebranded itself as ExxonMobil and is now Standard Oil's largest direct descendant.

Disgraced US cyclist Lance Armstrong stepped down from his role as chairman at his foundation after being stripped of his seven Tour de France titles. AP

Following the biggest doping scandal in cycling history, the Lance Armstrong Foundation changed its name to Livestrong in 2012 to distance itself from the disgraced American cyclist.

Armstrong founded the charity in 1997 after he was diagnosed with testicular cancer and before his first Tour de France title.

In October 2012, Armstrong announced he was stepping down as chairman of the foundation after the International Cycling Union stripped him of his seven Tour de France wins.

That followed an earlier report from the US Anti-Doping Agency accusing him of running the most sophisticated, professionalised and successful doping programme that sport has ever seen".

The foundation soon changed its name to Livestrong the word inscribed on its signature yellow wristbands.

All of us especially Lance wanted Livestrong to have a presence that was bigger than its founder, board member Mark McKinnon said.

Updated: October 28th 2021, 8:57 PM

See the original post:
Five companies that underwent major rebrands - The National

Posted in Genetic Engineering | Comments Off on Five companies that underwent major rebrands – The National

Global CRISPR Cas9 Market to 2027 Industry Perspective, Comprehensive Analysis, and Forecast Chip Design Magazine – Chip Design Magazine

Posted: at 6:39 am

The CRISPR Cas9 market report contains a detailed focused scene in which major players(Caribou Biosciences, Editas Medicine, Intellia Therapeutics, Mirus Bio, Integrated DNA Technologies (IDT), Horizon Discovery Group, Takara Bio, Thermo Fisher Scientific, Agilent Technologies, Cellecta, Merck, GeneCopoeia, CRISPR Therapeutics, GenScript)are profiled. Various companies engaged with the CRISPR Cas9 Market studies. TheCRISPR Cas9 market research reportgives a worldwide viewpoint. This can bolster the end consumer in making the right decision eventually leading to the growth of the CRISPR Cas9 market.

The report gives a forward-looking viewpoint on different driving and limiting factors needed for thedevelopment of the CRISPR Cas9 market. It gives a forecast based on how the market is assumed to increase. Their general organization review, major financial aspects, key advancements, weighted SWOT examination, land spread, developments, and processes are studied and have been competently mentioned in the CRISPR Cas9 market report.

Get Free PDF Sample Copy of the Report (Including Full TOC, List of Tables & Figures, Chart and Covid-19 Impact Analysis):https://www.marketresearchstore.com/sample/crispr-cas9-market-798239

This report studies the CRISPR Cas9 market based on its classifications. In addition to this, major regions (North America, Latin America, Europe, Asia Pacific, the Middle East, and Africa) are also studies via this report. This report offers a detailed examination of the market by studying aggressive factors of the CRISPR Cas9 market. It supports in identifying the major product sectors and their outlook in the years to come.

Leading Major Players included in the CRISPR Cas9 market reports are :

Caribou Biosciences, Editas Medicine, Intellia Therapeutics, Mirus Bio, Integrated DNA Technologies (IDT), Horizon Discovery Group, Takara Bio, Thermo Fisher Scientific, Agilent Technologies, Cellecta, Merck, GeneCopoeia, CRISPR Therapeutics, GenScriptand More

Market Segment By Type:

Genome Editing, Genetic engineering, gRNA Database/Gene Librar, CRISPR Plasmid, Human Stem Cells, Genetically Modified Organisms/Crops, Cell Line Engineering

Market Segment By Application:

Biotechnology Companies, Pharmaceutical Companies, Academic Institutes, Research and Development Institutes

The foundation of the CRISPR Cas9 market is also mentioned in the report that can allow the consumers in applying primary techniques to gain a competitive advantage. Such a far-reaching and in-depth analysis provides the crucial extension with key recommendations and straight moderate review. This can be used to enhance the present position and design future extensions in a specific area in the CRISPR Cas9 market.

Do Inquiry here for more analysis:https://www.marketresearchstore.com/inquiry/crispr-cas9-market-798239

Key Region & Countries:

Important Key Points of CRISPR Cas9 Market:

Years considered for this report:

Key questions answered in this report:

Imperative regions worldwide are studied and the patterns, drivers, advancements, difficulties, and restrictions impacting the CRISPR Cas9 market growth over these essential geologies are taken into considerations. A study of the impact of government policies and strategies on the processes of the CRISPR Cas9 market is also added to offer an in the general summary of the CRISPR Cas9 markets future.

For More Information with full TOC:https://www.marketresearchstore.com/market-insights/crispr-cas9-market-798239

NOTE:Our research team is examining the Covid-19 impact on various industry verticals and Country Level impact for a better analysis of markets. The 2021 newest edition of this report is entitled to render extra information on the current situation, the strike of the economy, and COVID-19 impact on the overall industry.

Original post:
Global CRISPR Cas9 Market to 2027 Industry Perspective, Comprehensive Analysis, and Forecast Chip Design Magazine - Chip Design Magazine

Posted in Genetic Engineering | Comments Off on Global CRISPR Cas9 Market to 2027 Industry Perspective, Comprehensive Analysis, and Forecast Chip Design Magazine – Chip Design Magazine

Novavax Files for Authorization of its COVID-19 Vaccine in the United Kingdom – PRNewswire

Posted: at 6:39 am

GAITHERSBURG, Md., Oct. 27, 2021 /PRNewswire/ --Novavax,Inc. (Nasdaq: NVAX), a biotechnology company dedicated to developing and commercializing next-generation vaccines for serious infectious diseases, today announced the completion of its rolling regulatory submission to the U.K. Medicines and Healthcare products Regulatory Agency (MHRA) for authorization of its COVID-19 vaccine candidate. The company's application for Conditional Marketing Authorization (CMA) marks the first submission for authorization of a protein-based COVID-19 vaccine in the United Kingdom.

"This submission brings Novavax significantly closer to delivering millions of doses of the first protein-based COVID-19 vaccine, built on a proven, well-understood vaccine platform that demonstrated high efficacy against multiple strains of the coronavirus," said Stanley C. Erck, President and Chief Executive Officer, Novavax. "We look forward to MHRA's review and will be prepared to deliver vaccine doses following what we anticipate will be a positive decision. We thank the clinical trial participants and trial sites in the United Kingdom, as well as the U.K. Vaccines Taskforce, for their support and vital contributions to this program."

Novavax has now completed the submission of all modules required by MHRA for the regulatory review of NVX-CoV2373, the company's recombinant nanoparticle protein-based COVID-19 vaccine with Matrix-M adjuvant. This includes preclinical, clinical, and chemistry, manufacturing and controls (CMC) data. Clinical data from a pivotal Phase 3 trial of 15,000 volunteers in the U.K. was submitted to MHRA earlier this yearin which NVX-CoV2373 demonstrated efficacy of 96.4% against the original virus strain, 86.3% against the Alpha (B.1.1.7) variant and 89.7% efficacy overall, as well as a favorable safety and tolerability profile. The submission also includes data from PREVENT-19, a 30,000-person trial in the U.S. and Mexico, which demonstrated 100% protection against moderate and severe disease and 90.4% efficacy overall. NVX-CoV2373 was generally well-tolerated and elicited a robust antibody response.

Novavax expects to complete additional regulatory filings in key markets, including Europe, Canada, Australia, New Zealand, the World Health Organization and other markets around the world shortly following the U.K. submission. In the U.S., Novavax expects to submit the complete package to the FDA by the end of the year. The company continues to work closely with governments, regulatory authorities and non-governmental organizations (NGOs) in its commitment to ensuring equitable global access to its COVID-19 vaccine.

"The submission to MHRA leverages our manufacturing partnership with the Serum Institute of India, the world's largest supplier of COVID-19 vaccines," said Rick Crowley, Executive Vice President, Chief Operations Officer, Novavax. "In the near future, we expect to supplement this filing with supply from our global supply chain."

Clickhereto view multimedia content, including B-roll and other resources that accompany this press release.

About NVX-CoV2373NVX-CoV2373 is a protein-based vaccine candidate engineered from the genetic sequence of the first strain of SARS-CoV-2, the virus that causes COVID-19 disease. NVX-CoV2373 was created using Novavax' recombinant nanoparticle technology to generate antigen derived from the coronavirus spike (S) protein and is formulated with Novavax' patented saponin-based Matrix-M adjuvant to enhance the immune response and stimulate high levels of neutralizing antibodies. NVX-CoV2373 contains purified protein antigen and can neither replicate nor can it cause COVID-19.

Novavax' COVID-19 vaccine is packaged as a ready-to-use liquid formulation in a vial containing ten doses. The vaccination regimen calls for two 0.5 ml doses (5 microgram antigen and 50 microgram Matrix-Madjuvant) given intramuscularly 21 days apart. The vaccine is stored at 2- 8Celsius, enabling the use of existing vaccine supply and cold chain channels.

About Matrix-M AdjuvantNovavax' patented saponin-based Matrix-M adjuvant has demonstrated a potent and well-tolerated effect by stimulating the entry of antigen-presenting cells into the injection site and enhancing antigen presentation in local lymph nodes, boosting immune response.

About NovavaxNovavax, Inc.(Nasdaq: NVAX) is a biotechnology company that promotes improved health globally through the discovery, development and commercialization of innovative vaccines to prevent serious infectious diseases. The company's proprietary recombinant technology platform combines the power and speed of genetic engineering to efficiently produce highly immunogenic nanoparticles designed to address urgent global health needs.Novavaxis conducting late-stage clinical trials for NVX-CoV2373, its vaccine candidate against SARS-CoV-2, the virus that causes COVID-19. NanoFlu, its quadrivalent influenza nanoparticle vaccine, met all primary objectives in its pivotal Phase 3 clinical trial in older adults. Both vaccine candidates incorporateNovavax' proprietary saponin-based Matrix-M adjuvant to enhance the immune response and stimulate high levels of neutralizing antibodies.

For more information, visitwww.novavax.comand connect with us on TwitterandLinkedIn.

Forward-Looking StatementsStatements herein relating to the future of Novavax, its operating plans and prospects, its partnerships, the ongoing development of NVX-CoV2373 and other Novavax vaccine product candidates, the scope, timing and outcome of future regulatory filings and actions and the preparedness of Novavax to deliver vaccine doses are forward-looking statements. Novavax cautions that these forward-looking statements are subject to numerous risks and uncertainties that could cause actual results to differ materially from those expressed or implied by such statements. These risks and uncertainties include challenges satisfying, alone or together with partners, various safety, efficacy, and product characterization requirements, including those related to process qualification and assay validation, necessary to satisfy applicable regulatory authorities; difficulty obtaining scarce raw materials and supplies; resource constraints, including human capital and manufacturing capacity, on the ability of Novavax to pursue planned regulatory pathways; challenges meeting contractual requirements under agreements with multiple commercial, governmental, and other entities; and those other risk factors identified in the "Risk Factors" and "Management's Discussion and Analysis of Financial Condition and Results of Operations" sections of Novavax' Annual Report on Form 10-K for the year ended December 31, 2020 and subsequent Quarterly Reports on Form 10-Q, as filed with the Securities and Exchange Commission (SEC). We caution investors not to place considerable reliance on forward-looking statements contained in this press release. You are encouraged to read our filings with the SEC, available at http://www.sec.gov and http://www.novavax.com, for a discussion of these and other risks and uncertainties. The forward-looking statements in this press release speak only as of the date of this document, and we undertake no obligation to update or revise any of the statements. Our business is subject to substantial risks and uncertainties, including those referenced above. Investors, potential investors, and others should give careful consideration to these risks and uncertainties.

Contacts:

InvestorsNovavax, Inc.Erika Schultz| 240-268-2022[emailprotected]

Solebury TroutAlexandra Roy| 617-221-9197[emailprotected]

MediaAlison Chartan| 240-720-7804Laura KeenanLindsey | 202-709-7521[emailprotected]

SOURCE Novavax, Inc.

http://www.novavax.com

See more here:
Novavax Files for Authorization of its COVID-19 Vaccine in the United Kingdom - PRNewswire

Posted in Genetic Engineering | Comments Off on Novavax Files for Authorization of its COVID-19 Vaccine in the United Kingdom – PRNewswire

UKZN and UN agency partnership paves the way for access to medicine – Mail and Guardian

Posted: at 6:39 am

The International Centre for Genetic Engineering and Biotechnology (ICGEB) and the University of KwaZulu-Natal (UKZN) this week signed a ground-breaking agreement that will see the university partnering with local companies to develop advanced biotherapeutics to be used in the treatment of various conditions including diabetes, arthritis, cancer and others.

The ICGEB was created by the United Nations in 1983 to facilitate biotechnology developments in the developing world. The organisations council of scientific advisors comprises the worlds leading scientists, among them Nobel prize-winners for medicine.

The ICGEB has three global centres: one is in Cape Town, and the others are in Trieste in Italy and New Delhi in India.

ICGEB Director-General Dr Lawrence Banks said during the signing ceremony that the partnership with UKZN is perfect, as the two organisations have lots of shared values and aims.

Dr Banks said the cornerstone of the collaboration is to ensure that state-of-the-art technology and science can bring benefits for all people in the world. He said this should begin with education, which forms the core mandate of the UKZN.

He said they must ensure that nobody is left behind in the partnership, and that this will be done in practical ways. He said they will ensure that they have fellowship programmes that will bring people not only from South Africa, but also from across the continent to work on state-of-the-art programmes within the life sciences.

What we do is not only for South Africa, but for the entire continent, said Dr Banks. He emphasised that the partnership must ensure that the fruits of modern biotechnology reach the people who need it.

At the end of the day, you can have wonderful therapeutics, but if its not affordable to the people its a complete waste of time, he said, adding that the partnership with UKZN is fundamental in bringing this about.

Dr Phil Mjwara, Director-General of the Department of Science and Innovation, said the partnership was in line with the White Paper on Science, Technology and Innovation adopted by the government in 2019.

The White Paper introduced a number of policy shifts, which relate to, among others, increasing the focus on inclusivity, transformation and linkages in the NSI; enhancing the innovation culture in society and government and improving policy coherence and budget co-ordination across government.

UKZNs Deputy Vice-Chancellor of Research and Innovation, Professor Mosa Moshabela, and the Dean of the School of Clinical Medicine, Professor Ncoza Dlova, will be responsible for conducting clinical trials within the next year.

The partnership is set to allow poor people to access expensive life saving medicines for the first time. The collaboration will be facilitated by AfricaBio through its President Dr Nhlanhla Msomi.

AfricaBio is an independent non-profit stakeholders association which represents the interests of all stakeholders involved in the biotechnology sector throughout Africa. It focuses on agriculture, health, industrial, environmental and marine biotech.

Dr Thami Chiliza, Microbiologist at the School of Life Sciences, UKZN, and a stakeholder of AfricaBio, said the partnership with the ICGEB will ensure that expertise rubs off onto students in an easier way, exposes them to what is out in there in the world of science, and contributes to job creation.

I really believe this will allow students to gain more exposure and experience in terms of the biotechnology sector, said Dr Chiliza.

It is expected that the collaboration will soon include other partnerships with the universities of Limpopo, Venda and Walter Sisulu.

Researcher Dr Thandeka Khoza said the partnership fits in with the UKZNs mission statement and goals, which include achievement of excellence in teaching and learning, excellence and high impact in research, innovation and entrepreneurship.

She said the university has various innovative research projects lined up that can offer various solutions to various diseases, and they have identified products from natural products from plants for use in cancer and TB research.

If we have ICGEB on board, these projects can move faster towards the project development stage so we can have a wide door of opportunities for all members of the university, said Dr Khoza.

What it does is, it bridges the gap between academic research and product-driven or industry-based research, which is what we are at this point in time, gearing ourselves towards and also attracting skills that position us for such research. So, we are confident that in no time our research will be applied research, and it will also be cost effective. We are going to have graduates that are fit for purpose, she said.

UKZN Deputy Vice-Chancellor Professor Mosa Moshabela said the partnership underlined the reality that institutions must work together: Ivory towers have to come to an end; we need to flatten the hierarchy, we have to get into equal partnerships with different stakeholders, we have to create a culture of sharing, and we must have the humility to learn from others. There is no way we can advance by working in isolation.

See original here:
UKZN and UN agency partnership paves the way for access to medicine - Mail and Guardian

Posted in Genetic Engineering | Comments Off on UKZN and UN agency partnership paves the way for access to medicine – Mail and Guardian

How biological detective work can reveal who engineered a virus – Vox.com

Posted: October 24, 2021 at 10:49 am

SARS-CoV-2, the virus that causes Covid-19, wasnt intentionally created in a lab. We dont have much evidence one way or the other whether its emergence into the world was the result of a lab accident or a natural jump from animal to human, but we know for sure that the virus is not the product of deliberate gene editing in a lab.

How do we know that? Bioengineering leaves traces characteristic patterns in the RNA, the genetic code of a virus, that come from splicing in genes from elsewhere. And investigations by researchers have definitively shown that the novel coronavirus behind Covid-19 doesnt bear the hallmarks of such manipulation.

That fact about bioengineered viruses raises an interesting question: What if those traces that gene editing leave behind were more like fingerprints? That is, what if its possible not just to tell if a virus was engineered but precisely where it was engineered?

Thats the idea behind genetic engineering attribution: the effort to develop tools that let us look at a genetically engineered sequence and determine which lab developed it. A big international contest among researchers earlier this year demonstrates that the technology is within our reach though itll take lots of refining to move from impressive contest results to tools we can reliably use for bio detective work.

The contest, the Genetic Engineering Attribution Challenge, was sponsored by some of the leading bioresearch labs in the world. The idea was to challenge teams to develop techniques in genetic engineering attribution. The most successful entrants in the competition could predict, using machine-learning algorithms, which lab produced a certain genetic sequence with more than 80 percent accuracy, according to a new preprint summing up the results of the contest.

This may seem technical, but it could actually be fairly consequential in the effort to make the world safe from a type of threat we should all be more attuned to post-pandemic: bioengineered weapons and leaks of bioengineered viruses.

One of the challenges of preventing bioweapon research and deployment is that perpetrators can remain hidden its difficult to find the source of a killer virus and hold them accountable.

But if its widely known that bioweapons can immediately and verifiably be traced right back to a bad actor, that could be a valuable deterrent.

Its also extremely important for biosafety more broadly. If an engineered virus is accidentally leaked, tools like these would allow us to identify where they leaked from and know what labs are doing genetic engineering work with inadequate safety procedures.

Hundreds of design choices go into genetic engineering: what genes you use, what enzymes you use to connect them together, what software you use to make those decisions for you, computational immunologist Will Bradshaw, a co-author on the paper, told me.

The enzymes that people use to cut up the DNA cut in different patterns and have different error profiles, Bradshaw says. You can do that in the same way that you can recognize handwriting.

Because different researchers with different training and different equipment have their own distinctive tells, its possible to look at a genetically engineered organism and guess who made it at least if youre using machine-learning algorithms.

The algorithms that are trained to do this work are fed data on more than 60,000 genetic sequences different labs produced. The idea is that, when fed an unfamiliar sequence, the algorithms are able to predict which of the labs theyve encountered (if any) likely produced it.

A year ago, researchers at altLabs, the Johns Hopkins Center for Health Security, and other top bioresearch programs collaborated on the challenge, organizing a competition to find the best approaches to this biological forensics problem. The contest attracted intense interest from academics, industry professionals, and citizen scientists one member of a winning team was a kindergarten teacher. Nearly 300 teams from all over the world submitted at least one machine-learning system for identifying the lab of origin of different sequences.

In that preprint paper (which is still undergoing peer review), the challenges organizers summarize the results: The competitors collectively took a big step forward on this problem. Winning teams achieved dramatically better results than any previous attempt at genetic engineering attribution, with the top-scoring team and all-winners ensemble both beating the previous state-of-the-art by over 10 percentage points, the paper notes.

The big picture is that researchers, aided by machine-learning systems, are getting really good at finding the lab that built a given plasmid, or a specific DNA strand used in gene manipulation.

The top-performing teams had 95 percent accuracy at naming a plasmids creator by one metric called top 10 accuracy meaning if the algorithm identifies 10 candidate labs, the true lab is one of them. They had 82 percent top 1 accuracy that is, 82 percent of the time, the lab they identified as the likely designer of that bioengineered plasmid was, in fact, the lab that designed it.

Top 1 accuracy is showy, but for biological detective work, top 10 accuracy is nearly as good: If you can narrow down the search for culprits to a small number of labs, you can then use other approaches to identify the exact lab.

Theres still a lot of work to do. The competition looked at only simple engineered plasmids; ideally, wed have approaches that work for fully engineered viruses and bacteria. And the competition didnt look at adversarial examples, where researchers deliberately try to conceal the fingerprints of their lab on their work.

Knowing which lab produced a bioweapon can protect us in three ways, biosecurity researchers argued in Nature Communications last year.

First, knowledge of who was responsible can inform response efforts by shedding light on motives and capabilities, and so mitigate the events consequences. That is, figuring out who built something will also give us clues about the goals they might have had and the risk we might be facing.

Second, obviously, it allows the world to sanction and stop any lab or government that is producing bioweapons in violation of international law.

And third, the article argues, hopefully, if these capabilities are widely known, they make the use of bioweapons much less appealing in the first place.

But the techniques have more mundane uses as well.

Bradshaw told me he envisions applications of the technology could be used to find accidental lab leaks, identify plagiarism in academic papers, and protect biological intellectual property and those applications will validate and extend the tools for the really critical uses.

Its worth repeating that SARS-CoV-2 was not an engineered virus. But the past year and a half should have us all thinking about how devastating pandemic disease can be and about whether the precautions being taken by research labs and governments are really adequate to prevent the next pandemic.

The answer, to my mind, is that were not doing enough, but more sophisticated biological forensics could certainly help. Genetic engineering attribution is still a new field. With more effort, itll likely be possible to one day make attribution possible on a much larger scale and to do it for viruses and bacteria. That could make for a much safer future.

A version of this story was initially published in the Future Perfect newsletter. Sign up here to subscribe!

More:
How biological detective work can reveal who engineered a virus - Vox.com

Posted in Genetic Engineering | Comments Off on How biological detective work can reveal who engineered a virus – Vox.com

Page 40«..1020..39404142..5060..»