fbpx
Wikipedia

Genetically modified organism

A genetically modified organism (GMO) is any organism whose genetic material has been altered using genetic engineering techniques. The exact definition of a genetically modified organism and what constitutes genetic engineering varies, with the most common being an organism altered in a way that "does not occur naturally by mating and/or natural recombination".[1] A wide variety of organisms have been genetically modified (GM), including animals, plants, and microorganisms.

Genetic modification can include the introduction of new genes or enhancing, altering, or knocking out endogenous genes. In some genetic modifications, genes are transferred within the same species, across species (creating transgenic organisms), and even across kingdoms.

Creating a genetically modified organism is a multi-step process. Genetic engineers must isolate the gene they wish to insert into the host organism and combine it with other genetic elements, including a promoter and terminator region and often a selectable marker. A number of techniques are available for inserting the isolated gene into the host genome. Recent advancements using genome editing techniques, notably CRISPR, have made the production of GMOs much simpler. Herbert Boyer and Stanley Cohen made the first genetically modified organism in 1973, a bacterium resistant to the antibiotic kanamycin. The first genetically modified animal, a mouse, was created in 1974 by Rudolf Jaenisch, and the first plant was produced in 1983. In 1994, the Flavr Savr tomato was released, the first commercialized genetically modified food. The first genetically modified animal to be commercialized was the GloFish (2003) and the first genetically modified animal to be approved for food use was the AquAdvantage salmon in 2015.

Bacteria are the easiest organisms to engineer and have been used for research, food production, industrial protein purification (including drugs), agriculture, and art. There is potential to use them for environmental purposes or as medicine. Fungi have been engineered with much the same goals. Viruses play an important role as vectors for inserting genetic information into other organisms. This use is especially relevant to human gene therapy. There are proposals to remove the virulent genes from viruses to create vaccines. Plants have been engineered for scientific research, to create new colors in plants, deliver vaccines, and to create enhanced crops. Genetically modified crops are publicly the most controversial GMOs, in spite of having the most human health and environmental benefits.[2] The majority are engineered for herbicide tolerance or insect resistance. Golden rice has been engineered with three genes that increase its nutritional value. Other prospects for GM crops are as bioreactors for the production of biopharmaceuticals, biofuels, or medicines.

Animals are generally much harder to transform and the vast majority are still at the research stage. Mammals are the best model organisms for humans, making ones genetically engineered to resemble serious human diseases important to the discovery and development of treatments. Human proteins expressed in mammals are more likely to be similar to their natural counterparts than those expressed in plants or microorganisms. Livestock is modified with the intention of improving economically important traits such as growth rate, quality of meat, milk composition, disease resistance, and survival. Genetically modified fish are used for scientific research, as pets, and as a food source. Genetic engineering has been proposed as a way to control mosquitos, a vector for many deadly diseases. Although human gene therapy is still relatively new, it has been used to treat genetic disorders such as severe combined immunodeficiency and Leber's congenital amaurosis.

Many objections have been raised over the development of GMOs, particularly their commercialization. Many of these involve GM crops and whether food produced from them is safe and what impact growing them will have on the environment. Other concerns are the objectivity and rigor of regulatory authorities, contamination of non-genetically modified food, control of the food supply, patenting of life, and the use of intellectual property rights. Although there is a scientific consensus that currently available food derived from GM crops poses no greater risk to human health than conventional food, GM food safety is a leading issue with critics. Gene flow, impact on non-target organisms, and escape are the major environmental concerns. Countries have adopted regulatory measures to deal with these concerns. There are differences in the regulation for the release of GMOs between countries, with some of the most marked differences occurring between the US and Europe. Key issues concerning regulators include whether GM food should be labeled and the status of gene-edited organisms.

Definition

The definition of a genetically modified organism (GMO) is not clear and varies widely between countries, international bodies, and other communities. At its broadest, the definition of a GMO can include anything that has had its genes altered, including by nature.[3][4] Taking a less broad view, it can encompass every organism that has had its genes altered by humans, which would include all crops and livestock. In 1993, the Encyclopedia Britannica defined genetic engineering as "any of a wide range of techniques ... among them artificial insemination, in vitro fertilization (e.g., 'test-tube' babies), sperm banks, cloning, and gene manipulation."[5] The European Union (EU) included a similarly broad definition in early reviews, specifically mentioning GMOs being produced by "selective breeding and other means of artificial selection"[6] These definitions were promptly adjusted with a number of exceptions added as the result of pressure from scientific and farming communities, as well as developments in science. The EU definition later excluded traditional breeding, in vitro fertilization, induction of polyploidy, mutation breeding, and cell fusion techniques that do not use recombinant nucleic acids or a genetically modified organism in the process.[7][8][9]

Another approach was the definition provided by the Food and Agriculture Organization, the World Health Organization, and the European Commission, stating that the organisms must be altered in a way that does "not occur naturally by mating and/or natural recombination".[10][11][12] Progress in science, such as the discovery of horizontal gene transfer being a relatively common natural phenomenon, further added to the confusion on what "occurs naturally", which led to further adjustments and exceptions.[13] There are examples of crops that fit this definition, but are not normally considered GMOs.[14] For example, the grain crop triticale was fully developed in a laboratory in 1930 using various techniques to alter its genome.[15]

Genetically engineered organism (GEO) can be considered a more precise term compared to GMO when describing organisms' genomes that have been directly manipulated with biotechnology.[16][8] The Cartagena Protocol on Biosafety used the synonym living modified organism (LMO) in 2000 and defined it as "any living organism that possesses a novel combination of genetic material obtained through the use of modern biotechnology."[17] Modern biotechnology is further defined as "In vitro nucleic acid techniques, including recombinant deoxyribonucleic acid (DNA) and direct injection of nucleic acid into cells or organelles, or fusion of cells beyond the taxonomic family."[18]

Originally, the term GMO was not commonly used by scientists to describe genetically engineered organisms until after usage of GMO became common in popular media.[19] The United States Department of Agriculture (USDA) considers GMOs to be plants or animals with heritable changes introduced by genetic engineering or traditional methods, while GEO specifically refers to organisms with genes introduced, eliminated, or rearranged using molecular biology, particularly recombinant DNA techniques, such as transgenesis.[20]

The definitions focus on the process more than the product, which means there could be GMOS and non-GMOs with very similar genotypes and phenotypes.[21][22] This has led scientists to label it as a scientifically meaningless category,[23] saying that it is impossible to group all the different types of GMOs under one common definition.[24] It has also caused issues for organic institutions and groups looking to ban GMOs.[25][26] It also poses problems as new processes are developed. The current definitions came in before genome editing became popular and there is some confusion as to whether they are GMOs. The EU has adjudged that they are[27] changing their GMO definition to include "organisms obtained by mutagenesis", but has excluded them from regulation based on their "long safety record" and that they have been "conventionally been used in a number of applications".[9] In contrast the USDA has ruled that gene edited organisms are not considered GMOs.[28]

Even greater inconsistency and confusion is associated with various "Non-GMO" or "GMO-free" labeling schemes in food marketing, where even products such as water or salt, which do not contain any organic substances and genetic material (and thus cannot be genetically modified by definition), are being labeled to create an impression of being "more healthy".[29][30][31]

Production

 
A gene gun uses biolistics to insert DNA into plant tissue.

Creating a genetically modified organism (GMO) is a multi-step process. Genetic engineers must isolate the gene they wish to insert into the host organism. This gene can be taken from a cell[32] or artificially synthesized.[33] If the chosen gene or the donor organism's genome has been well studied it may already be accessible from a genetic library. The gene is then combined with other genetic elements, including a promoter and terminator region and a selectable marker.[34]

A number of techniques are available for inserting the isolated gene into the host genome. Bacteria can be induced to take up foreign DNA, usually by exposed heat shock or electroporation.[35] DNA is generally inserted into animal cells using microinjection, where it can be injected through the cell's nuclear envelope directly into the nucleus, or through the use of viral vectors.[36] In plants the DNA is often inserted using Agrobacterium-mediated recombination,[37][38] biolistics[39] or electroporation.

As only a single cell is transformed with genetic material, the organism must be regenerated from that single cell. In plants this is accomplished through tissue culture.[40][41] In animals it is necessary to ensure that the inserted DNA is present in the embryonic stem cells.[37] Further testing using PCR, Southern hybridization, and DNA sequencing is conducted to confirm that an organism contains the new gene.[42]

Traditionally the new genetic material was inserted randomly within the host genome. Gene targeting techniques, which creates double-stranded breaks and takes advantage on the cells natural homologous recombination repair systems, have been developed to target insertion to exact locations. Genome editing uses artificially engineered nucleases that create breaks at specific points. There are four families of engineered nucleases: meganucleases,[43][44] zinc finger nucleases,[45][46] transcription activator-like effector nucleases (TALENs),[47][48] and the Cas9-guideRNA system (adapted from CRISPR).[49][50] TALEN and CRISPR are the two most commonly used and each has its own advantages.[51] TALENs have greater target specificity, while CRISPR is easier to design and more efficient.[51]

History

 
Herbert Boyer (pictured) and Stanley Cohen created the first genetically modified organism in 1973.

Humans have domesticated plants and animals since around 12,000 BCE, using selective breeding or artificial selection (as contrasted with natural selection).[52]: 25  The process of selective breeding, in which organisms with desired traits (and thus with the desired genes) are used to breed the next generation and organisms lacking the trait are not bred, is a precursor to the modern concept of genetic modification.[53]: 1 [54]: 1  Various advancements in genetics allowed humans to directly alter the DNA and therefore genes of organisms. In 1972, Paul Berg created the first recombinant DNA molecule when he combined DNA from a monkey virus with that of the lambda virus.[55][56]

Herbert Boyer and Stanley Cohen made the first genetically modified organism in 1973.[57] They took a gene from a bacterium that provided resistance to the antibiotic kanamycin, inserted it into a plasmid and then induced other bacteria to incorporate the plasmid. The bacteria that had successfully incorporated the plasmid was then able to survive in the presence of kanamycin.[58] Boyer and Cohen expressed other genes in bacteria. This included genes from the toad Xenopus laevis in 1974, creating the first GMO expressing a gene from an organism of a different kingdom.[59]

 
In 1974, Rudolf Jaenisch created the first genetically modified animal.

In 1974, Rudolf Jaenisch created a transgenic mouse by introducing foreign DNA into its embryo, making it the world's first transgenic animal.[60][61] However it took another eight years before transgenic mice were developed that passed the transgene to their offspring.[62][63] Genetically modified mice were created in 1984 that carried cloned oncogenes, predisposing them to developing cancer.[64] Mice with genes removed (termed a knockout mouse) were created in 1989. The first transgenic livestock were produced in 1985[65] and the first animal to synthesize transgenic proteins in their milk were mice in 1987.[66] The mice were engineered to produce human tissue plasminogen activator, a protein involved in breaking down blood clots.[67]

In 1983, the first genetically engineered plant was developed by Michael W. Bevan, Richard B. Flavell and Mary-Dell Chilton. They infected tobacco with Agrobacterium transformed with an antibiotic resistance gene and through tissue culture techniques were able to grow a new plant containing the resistance gene.[68] The gene gun was invented in 1987, allowing transformation of plants not susceptible to Agrobacterium infection.[69] In 2000, Vitamin A-enriched golden rice was the first plant developed with increased nutrient value.[70]

In 1976, Genentech, the first genetic engineering company was founded by Herbert Boyer and Robert Swanson; a year later, the company produced a human protein (somatostatin) in E. coli. Genentech announced the production of genetically engineered human insulin in 1978.[71] The insulin produced by bacteria, branded Humulin, was approved for release by the Food and Drug Administration in 1982.[72] In 1988, the first human antibodies were produced in plants.[73] In 1987, a strain of Pseudomonas syringae became the first genetically modified organism to be released into the environment[74] when a strawberry and potato field in California were sprayed with it.[75]

The first genetically modified crop, an antibiotic-resistant tobacco plant, was produced in 1982.[76] China was the first country to commercialize transgenic plants, introducing a virus-resistant tobacco in 1992.[77] In 1994, Calgene attained approval to commercially release the Flavr Savr tomato, the first genetically modified food.[78] Also in 1994, the European Union approved tobacco engineered to be resistant to the herbicide bromoxynil, making it the first genetically engineered crop commercialized in Europe.[79] An insect resistant Potato was approved for release in the US in 1995,[80] and by 1996 approval had been granted to commercially grow 8 transgenic crops and one flower crop (carnation) in 6 countries plus the EU.[81]

In 2010, scientists at the J. Craig Venter Institute announced that they had created the first synthetic bacterial genome. They named it Synthia and it was the world's first synthetic life form.[82][83]

The first genetically modified animal to be commercialized was the GloFish, a Zebra fish with a fluorescent gene added that allows it to glow in the dark under ultraviolet light.[84] It was released to the US market in 2003.[85] In 2015, AquAdvantage salmon became the first genetically modified animal to be approved for food use.[86] Approval is for fish raised in Panama and sold in the US.[86] The salmon were transformed with a growth hormone-regulating gene from a Pacific Chinook salmon and a promoter from an ocean pout enabling it to grow year-round instead of only during spring and summer.[87]

Bacteria

 
 
Left: Bacteria transformed with pGLO under ambient light
Right: Bacteria transformed with pGLO visualized under ultraviolet light

Bacteria were the first organisms to be genetically modified in the laboratory, due to the relative ease of modifying their chromosomes.[88] This ease made them important tools for the creation of other GMOs. Genes and other genetic information from a wide range of organisms can be added to a plasmid and inserted into bacteria for storage and modification. Bacteria are cheap, easy to grow, clonal, multiply quickly and can be stored at −80 °C almost indefinitely. Once a gene is isolated it can be stored inside the bacteria, providing an unlimited supply for research.[89] A large number of custom plasmids make manipulating DNA extracted from bacteria relatively easy.[90]

Their ease of use has made them great tools for scientists looking to study gene function and evolution. The simplest model organisms come from bacteria, with most of our early understanding of molecular biology coming from studying Escherichia coli.[91] Scientists can easily manipulate and combine genes within the bacteria to create novel or disrupted proteins and observe the effect this has on various molecular systems. Researchers have combined the genes from bacteria and archaea, leading to insights on how these two diverged in the past.[92] In the field of synthetic biology, they have been used to test various synthetic approaches, from synthesizing genomes to creating novel nucleotides.[93][94][95]

Bacteria have been used in the production of food for a long time, and specific strains have been developed and selected for that work on an industrial scale. They can be used to produce enzymes, amino acids, flavorings, and other compounds used in food production. With the advent of genetic engineering, new genetic changes can easily be introduced into these bacteria. Most food-producing bacteria are lactic acid bacteria, and this is where the majority of research into genetically engineering food-producing bacteria has gone. The bacteria can be modified to operate more efficiently, reduce toxic byproduct production, increase output, create improved compounds, and remove unnecessary pathways.[96] Food products from genetically modified bacteria include alpha-amylase, which converts starch to simple sugars, chymosin, which clots milk protein for cheese making, and pectinesterase, which improves fruit juice clarity.[97] The majority are produced in the US and even though regulations are in place to allow production in Europe, as of 2015 no food products derived from bacteria are currently available there.[98]

Genetically modified bacteria are used to produce large amounts of proteins for industrial use. The bacteria are generally grown to a large volume before the gene encoding the protein is activated. The bacteria are then harvested and the desired protein purified from them.[99] The high cost of extraction and purification has meant that only high value products have been produced at an industrial scale.[100] The majority of these products are human proteins for use in medicine.[101] Many of these proteins are impossible or difficult to obtain via natural methods and they are less likely to be contaminated with pathogens, making them safer.[99] The first medicinal use of GM bacteria was to produce the protein insulin to treat diabetes.[102] Other medicines produced include clotting factors to treat hemophilia,[103] human growth hormone to treat various forms of dwarfism,[104][105] interferon to treat some cancers, erythropoietin for anemic patients, and tissue plasminogen activator which dissolves blood clots.[99] Outside of medicine they have been used to produce biofuels.[106] There is interest in developing an extracellular expression system within the bacteria to reduce costs and make the production of more products economical.[100]

With a greater understanding of the role that the microbiome plays in human health, there is a potential to treat diseases by genetically altering the bacteria to, themselves, be therapeutic agents. Ideas include altering gut bacteria so they destroy harmful bacteria, or using bacteria to replace or increase deficient enzymes or proteins. One research focus is to modify Lactobacillus, bacteria that naturally provide some protection against HIV, with genes that will further enhance this protection. If the bacteria do not form colonies inside the patient, the person must repeatedly ingest the modified bacteria in order to get the required doses. Enabling the bacteria to form a colony could provide a more long-term solution, but could also raise safety concerns as interactions between bacteria and the human body are less well understood than with traditional drugs. There are concerns that horizontal gene transfer to other bacteria could have unknown effects. As of 2018 there are clinical trials underway testing the efficacy and safety of these treatments.[107]

For over a century, bacteria have been used in agriculture. Crops have been inoculated with Rhizobia (and more recently Azospirillum) to increase their production or to allow them to be grown outside their original habitat. Application of Bacillus thuringiensis (Bt) and other bacteria can help protect crops from insect infestation and plant diseases. With advances in genetic engineering, these bacteria have been manipulated for increased efficiency and expanded host range. Markers have also been added to aid in tracing the spread of the bacteria. The bacteria that naturally colonize certain crops have also been modified, in some cases to express the Bt genes responsible for pest resistance. Pseudomonas strains of bacteria cause frost damage by nucleating water into ice crystals around themselves. This led to the development of ice-minus bacteria, which have the ice-forming genes removed. When applied to crops they can compete with the non-modified bacteria and confer some frost resistance.[108]

 
This artwork is made with bacteria modified to express 8 different colors of fluorescent proteins.

Other uses for genetically modified bacteria include bioremediation, where the bacteria are used to convert pollutants into a less toxic form. Genetic engineering can increase the levels of the enzymes used to degrade a toxin or to make the bacteria more stable under environmental conditions.[109] Bioart has also been created using genetically modified bacteria. In the 1980s artist Jon Davis and geneticist Dana Boyd converted the Germanic symbol for femininity (ᛉ) into binary code and then into a DNA sequence, which was then expressed in Escherichia coli.[110] This was taken a step further in 2012, when a whole book was encoded onto DNA.[111] Paintings have also been produced using bacteria transformed with fluorescent proteins.[110]

Viruses

Viruses are often modified so they can be used as vectors for inserting genetic information into other organisms. This process is called transduction and if successful the recipient of the introduced DNA becomes a GMO. Different viruses have different efficiencies and capabilities. Researchers can use this to control for various factors; including the target location, insert size, and duration of gene expression. Any dangerous sequences inherent in the virus must be removed, while those that allow the gene to be delivered effectively are retained.[112]

While viral vectors can be used to insert DNA into almost any organism it is especially relevant for its potential in treating human disease. Although primarily still at trial stages,[113] there has been some successes using gene therapy to replace defective genes. This is most evident in curing patients with severe combined immunodeficiency rising from adenosine deaminase deficiency (ADA-SCID),[114] although the development of leukemia in some ADA-SCID patients[115] along with the death of Jesse Gelsinger in a 1999 trial set back the development of this approach for many years.[116] In 2009, another breakthrough was achieved when an eight-year-old boy with Leber's congenital amaurosis regained normal eyesight[116] and in 2016 GlaxoSmithKline gained approval to commercialize a gene therapy treatment for ADA-SCID.[114] As of 2018, there are a substantial number of clinical trials underway, including treatments for hemophilia, glioblastoma, chronic granulomatous disease, cystic fibrosis and various cancers.[115]

The most common virus used for gene delivery comes from adenoviruses as they can carry up to 7.5 kb of foreign DNA and infect a relatively broad range of host cells, although they have been known to elicit immune responses in the host and only provide short term expression. Other common vectors are adeno-associated viruses, which have lower toxicity and longer-term expression, but can only carry about 4kb of DNA.[115] Herpes simplex viruses make promising vectors, having a carrying capacity of over 30kb and providing long term expression, although they are less efficient at gene delivery than other vectors.[117] The best vectors for long term integration of the gene into the host genome are retroviruses, but their propensity for random integration is problematic. Lentiviruses are a part of the same family as retroviruses with the advantage of infecting both dividing and non-dividing cells, whereas retroviruses only target dividing cells. Other viruses that have been used as vectors include alphaviruses, flaviviruses, measles viruses, rhabdoviruses, Newcastle disease virus, poxviruses, and picornaviruses.[115]

Most vaccines consist of viruses that have been attenuated, disabled, weakened or killed in some way so that their virulent properties are no longer effective. Genetic engineering could theoretically be used to create viruses with the virulent genes removed. This does not affect the viruses infectivity, invokes a natural immune response and there is no chance that they will regain their virulence function, which can occur with some other vaccines. As such they are generally considered safer and more efficient than conventional vaccines, although concerns remain over non-target infection, potential side effects and horizontal gene transfer to other viruses.[118] Another potential approach is to use vectors to create novel vaccines for diseases that have no vaccines available or the vaccines that do not work effectively, such as AIDS, malaria, and tuberculosis.[119] The most effective vaccine against Tuberculosis, the Bacillus Calmette–Guérin (BCG) vaccine, only provides partial protection. A modified vaccine expressing a M tuberculosis antigen is able to enhance BCG protection.[120] It has been shown to be safe to use at phase II trials, although not as effective as initially hoped.[121] Other vector-based vaccines have already been approved and many more are being developed.[119]

Another potential use of genetically modified viruses is to alter them so they can directly treat diseases. This can be through expression of protective proteins or by directly targeting infected cells. In 2004, researchers reported that a genetically modified virus that exploits the selfish behavior of cancer cells might offer an alternative way of killing tumours.[122][123] Since then, several researchers have developed genetically modified oncolytic viruses that show promise as treatments for various types of cancer.[124][125][126][127][128] In 2017, researchers genetically modified a virus to express spinach defensin proteins. The virus was injected into orange trees to combat citrus greening disease that had reduced orange production by 70% since 2005.[129]

Natural viral diseases, such as myxomatosis and rabbit hemorrhagic disease, have been used to help control pest populations. Over time the surviving pests become resistant, leading researchers to look at alternative methods. Genetically modified viruses that make the target animals infertile through immunocontraception have been created in the laboratory[130] as well as others that target the developmental stage of the animal.[131] There are concerns with using this approach regarding virus containment[130] and cross species infection.[132] Sometimes the same virus can be modified for contrasting purposes. Genetic modification of the myxoma virus has been proposed to conserve European wild rabbits in the Iberian peninsula and to help regulate them in Australia. To protect the Iberian species from viral diseases, the myxoma virus was genetically modified to immunize the rabbits, while in Australia the same myxoma virus was genetically modified to lower fertility in the Australian rabbit population.[133]

Outside of biology scientists have used a genetically modified virus to construct a lithium-ion battery and other nanostructured materials. It is possible to engineer bacteriophages to express modified proteins on their surface and join them up in specific patterns (a technique called phage display). These structures have potential uses for energy storage and generation, biosensing and tissue regeneration with some new materials currently produced including quantum dots, liquid crystals, nanorings and nanofibres.[134] The battery was made by engineering M13 bacteriaophages so they would coat themselves in iron phosphate and then assemble themselves along a carbon nanotube. This created a highly conductive medium for use in a cathode, allowing energy to be transferred quickly. They could be constructed at lower temperatures with non-toxic chemicals, making them more environmentally friendly.[135]

Fungi

Fungi can be used for many of the same processes as bacteria. For industrial applications, yeasts combine the bacterial advantages of being a single-celled organism that is easy to manipulate and grow with the advanced protein modifications found in eukaryotes. They can be used to produce large complex molecules for use in food, pharmaceuticals, hormones, and steroids.[136] Yeast is important for wine production and as of 2016 two genetically modified yeasts involved in the fermentation of wine have been commercialized in the United States and Canada. One has increased malolactic fermentation efficiency, while the other prevents the production of dangerous ethyl carbamate compounds during fermentation.[96] There have also been advances in the production of biofuel from genetically modified fungi.[137]

Fungi, being the most common pathogens of insects, make attractive biopesticides. Unlike bacteria and viruses they have the advantage of infecting the insects by contact alone, although they are out competed in efficiency by chemical pesticides. Genetic engineering can improve virulence, usually by adding more virulent proteins,[138] increasing infection rate or enhancing spore persistence.[139] Many of the disease carrying vectors are susceptible to entomopathogenic fungi. An attractive target for biological control are mosquitos, vectors for a range of deadly diseases, including malaria, yellow fever and dengue fever. Mosquitos can evolve quickly so it becomes a balancing act of killing them before the Plasmodium they carry becomes the infectious disease, but not so fast that they become resistant to the fungi. By genetically engineering fungi like Metarhizium anisopliae and Beauveria bassiana to delay the development of mosquito infectiousness the selection pressure to evolve resistance is reduced.[140] Another strategy is to add proteins to the fungi that block transmission of malaria[140] or remove the Plasmodium altogether.[141]

Agaricus bisporus the common white button mushroom, has been gene edited to resist browning, giving it a longer shelf life. The process used CRISPR to knock out a gene that encodes polyphenol oxidase. As it didn't introduce any foreign DNA into the organism it was not deemed to be regulated under existing GMO frameworks and as such is the first CRISPR-edited organism to be approved for release.[142] This has intensified debates as to whether gene-edited organisms should be considered genetically modified organisms[143] and how they should be regulated.[144]

Plants

 
Tissue culture used to regenerate Arabidopsis thaliana

Plants have been engineered for scientific research, to display new flower colors, deliver vaccines, and to create enhanced crops. Many plants are pluripotent, meaning that a single cell from a mature plant can be harvested and under the right conditions can develop into a new plant. This ability can be taken advantage of by genetic engineers; by selecting for cells that have been successfully transformed in an adult plant a new plant can then be grown that contains the transgene in every cell through a process known as tissue culture.[145]

Much of the advances in the field of genetic engineering has come from experimentation with tobacco. Major advances in tissue culture and plant cellular mechanisms for a wide range of plants has originated from systems developed in tobacco.[146] It was the first plant to be altered using genetic engineering and is considered a model organism for not only genetic engineering, but a range of other fields.[147] As such the transgenic tools and procedures are well established making tobacco one of the easiest plants to transform.[148] Another major model organism relevant to genetic engineering is Arabidopsis thaliana. Its small genome and short life cycle makes it easy to manipulate and it contains many homologs to important crop species.[149] It was the first plant sequenced, has a host of online resources available and can be transformed by simply dipping a flower in a transformed Agrobacterium solution.[150]

In research, plants are engineered to help discover the functions of certain genes. The simplest way to do this is to remove the gene and see what phenotype develops compared to the wild type form. Any differences are possibly the result of the missing gene. Unlike mutagenisis, genetic engineering allows targeted removal without disrupting other genes in the organism.[145] Some genes are only expressed in certain tissues, so reporter genes, like GUS, can be attached to the gene of interest allowing visualization of the location.[151] Other ways to test a gene is to alter it slightly and then return it to the plant and see if it still has the same effect on phenotype. Other strategies include attaching the gene to a strong promoter and see what happens when it is overexpressed, forcing a gene to be expressed in a different location or at different developmental stages.[145]

 
Suntory "blue" rose

Some genetically modified plants are purely ornamental. They are modified for flower color, fragrance, flower shape and plant architecture.[152] The first genetically modified ornamentals commercialized altered color.[153] Carnations were released in 1997, with the most popular genetically modified organism, a blue rose (actually lavender or mauve) created in 2004.[154] The roses are sold in Japan, the United States, and Canada.[155][156] Other genetically modified ornamentals include Chrysanthemum and Petunia.[152] As well as increasing aesthetic value there are plans to develop ornamentals that use less water or are resistant to the cold, which would allow them to be grown outside their natural environments.[157]

It has been proposed to genetically modify some plant species threatened by extinction to be resistant to invasive plants and diseases, such as the emerald ash borer in North American and the fungal disease, Ceratocystis platani, in European plane trees.[158] The papaya ringspot virus devastated papaya trees in Hawaii in the twentieth century until transgenic papaya plants were given pathogen-derived resistance.[159] However, genetic modification for conservation in plants remains mainly speculative. A unique concern is that a transgenic species may no longer bear enough resemblance to the original species to truly claim that the original species is being conserved. Instead, the transgenic species may be genetically different enough to be considered a new species, thus diminishing the conservation worth of genetic modification.[158]

Crops

 
Wild type peanut (top) and transgenic peanut with Bacillus thuringiensis gene added (bottom) exposed to cornstalk borer larva

Genetically modified crops are genetically modified plants that are used in agriculture. The first crops developed were used for animal or human food and provide resistance to certain pests, diseases, environmental conditions, spoilage or chemical treatments (e.g. resistance to a herbicide). The second generation of crops aimed to improve the quality, often by altering the nutrient profile. Third generation genetically modified crops could be used for non-food purposes, including the production of pharmaceutical agents, biofuels, and other industrially useful goods, as well as for bioremediation.[160]

 
Kenyans examining insect-resistant transgenic Bacillus thuringiensis (Bt) corn

There are three main aims to agricultural advancement; increased production, improved conditions for agricultural workers and sustainability. GM crops contribute by improving harvests through reducing insect pressure, increasing nutrient value and tolerating different abiotic stresses. Despite this potential, as of 2018, the commercialized crops are limited mostly to cash crops like cotton, soybean, maize and canola and the vast majority of the introduced traits provide either herbicide tolerance or insect resistance.[160] Soybeans accounted for half of all genetically modified crops planted in 2014.[161] Adoption by farmers has been rapid, between 1996 and 2013, the total surface area of land cultivated with GM crops increased by a factor of 100.[162] Geographically though the spread has been uneven, with strong growth in the Americas and parts of Asia and little in Europe and Africa.[160] Its socioeconomic spread has been more even, with approximately 54% of worldwide GM crops grown in developing countries in 2013.[162] Although doubts have been raised,[163] most studies have found growing GM crops to be beneficial to farmers through decreased pesticide use as well as increased crop yield and farm profit.[164][165][166]

The majority of GM crops have been modified to be resistant to selected herbicides, usually a glyphosate or glufosinate based one. Genetically modified crops engineered to resist herbicides are now more available than conventionally bred resistant varieties;[167] in the USA 93% of soybeans and most of the GM maize grown is glyphosate tolerant.[168] Most currently available genes used to engineer insect resistance come from the Bacillus thuringiensis bacterium and code for delta endotoxins. A few use the genes that encode for vegetative insecticidal proteins.[169] The only gene commercially used to provide insect protection that does not originate from B. thuringiensis is the Cowpea trypsin inhibitor (CpTI). CpTI was first approved for use cotton in 1999 and is currently undergoing trials in rice.[170][171] Less than one percent of GM crops contained other traits, which include providing virus resistance, delaying senescence and altering the plants composition.[161]

 
Golden rice compared to white rice

Golden rice is the most well known GM crop that is aimed at increasing nutrient value. It has been engineered with three genes that biosynthesise beta-carotene, a precursor of vitamin A, in the edible parts of rice.[70] It is intended to produce a fortified food to be grown and consumed in areas with a shortage of dietary vitamin A,[172] a deficiency which each year is estimated to kill 670,000 children under the age of 5[173] and cause an additional 500,000 cases of irreversible childhood blindness.[174] The original golden rice produced 1.6μg/g of the carotenoids, with further development increasing this 23 times.[175] It gained its first approvals for use as food in 2018.[176]

Plants and plant cells have been genetically engineered for production of biopharmaceuticals in bioreactors, a process known as pharming. Work has been done with duckweed Lemna minor,[177] the algae Chlamydomonas reinhardtii[178] and the moss Physcomitrella patens.[179][180] Biopharmaceuticals produced include cytokines, hormones, antibodies, enzymes and vaccines, most of which are accumulated in the plant seeds. Many drugs also contain natural plant ingredients and the pathways that lead to their production have been genetically altered or transferred to other plant species to produce greater volume.[181] Other options for bioreactors are biopolymers[182] and biofuels.[183] Unlike bacteria, plants can modify the proteins post-translationally, allowing them to make more complex molecules. They also pose less risk of being contaminated.[184] Therapeutics have been cultured in transgenic carrot and tobacco cells,[185] including a drug treatment for Gaucher's disease.[186]

Vaccine production and storage has great potential in transgenic plants. Vaccines are expensive to produce, transport, and administer, so having a system that could produce them locally would allow greater access to poorer and developing areas.[181] As well as purifying vaccines expressed in plants it is also possible to produce edible vaccines in plants. Edible vaccines stimulate the immune system when ingested to protect against certain diseases. Being stored in plants reduces the long-term cost as they can be disseminated without the need for cold storage, don't need to be purified, and have long term stability. Also being housed within plant cells provides some protection from the gut acids upon digestion. However the cost of developing, regulating, and containing transgenic plants is high, leading to most current plant-based vaccine development being applied to veterinary medicine, where the controls are not as strict.[187]

Genetically modified crops have been proposed as one of the ways to reduce farming-related CO2 emissions due to higher yield, reduced use of pesticides, reduced use of tractor fuel and no tillage. According to a 2021 study, in EU alone widespread adoption of GE crops would reduce greenhouse gas emissions by 33 million tons of CO2 equivalent or 7.5% of total farming-related emissions.[188]

Animals

The vast majority of genetically modified animals are at the research stage with the number close to entering the market remaining small.[189] As of 2018 only three genetically modified animals have been approved, all in the USA. A goat and a chicken have been engineered to produce medicines and a salmon has increased its own growth.[190] Despite the differences and difficulties in modifying them, the end aims are much the same as for plants. GM animals are created for research purposes, production of industrial or therapeutic products, agricultural uses, or improving their health. There is also a market for creating genetically modified pets.[191]

Mammals

 
Some chimeras, like the blotched mouse shown, are created through genetic modification techniques like gene targeting.

The process of genetically engineering mammals is slow, tedious, and expensive. However, new technologies are making genetic modifications easier and more precise.[192] The first transgenic mammals were produced by injecting viral DNA into embryos and then implanting the embryos in females.[60] The embryo would develop and it would be hoped that some of the genetic material would be incorporated into the reproductive cells. Then researchers would have to wait until the animal reached breeding age and then offspring would be screened for the presence of the gene in every cell. The development of the CRISPR-Cas9 gene editing system as a cheap and fast way of directly modifying germ cells, effectively halving the amount of time needed to develop genetically modified mammals.[193]

A porcine model of hemophilia A

Mammals are the best models for human disease, making genetic engineered ones vital to the discovery and development of cures and treatments for many serious diseases. Knocking out genes responsible for human genetic disorders allows researchers to study the mechanism of the disease and to test possible cures. Genetically modified mice have been the most common mammals used in biomedical research, as they are cheap and easy to manipulate. Pigs are also a good target as they have a similar body size and anatomical features, physiology, pathophysiological response and diet.[194] Nonhuman primates are the most similar model organisms to humans, but there is less public acceptance towards using them as research animals.[195] In 2009, scientists announced that they had successfully transferred a gene into a primate species (marmosets) for the first time.[196][197] Their first research target for these marmosets was Parkinson's disease, but they were also considering amyotrophic lateral sclerosis and Huntington's disease.[198]

Human proteins expressed in mammals are more likely to be similar to their natural counterparts than those expressed in plants or microorganisms. Stable expression has been accomplished in sheep, pigs, rats and other animals. In 2009, the first human biological drug produced from such an animal, a goat, was approved. The drug, ATryn, is an anticoagulant which reduces the probability of blood clots during surgery or childbirth and is extracted from the goat's milk.[199] Human alpha-1-antitrypsin is another protein that has been produced from goats and is used in treating humans with this deficiency.[200] Another medicinal area is in creating pigs with greater capacity for human organ transplants (xenotransplantation). Pigs have been genetically modified so that their organs can no longer carry retroviruses[201] or have modifications to reduce the chance of rejection.[202][203] Pig lungs from genetically modified pigs are being considered for transplantation into humans.[204][205] There is even potential to create chimeric pigs that can carry human organs.[194][206]

Livestock are modified with the intention of improving economically important traits such as growth-rate, quality of meat, milk composition, disease resistance and survival. Animals have been engineered to grow faster, be healthier[207] and resist diseases.[208] Modifications have also improved the wool production of sheep and udder health of cows.[189] Goats have been genetically engineered to produce milk with strong spiderweb-like silk proteins in their milk.[209] A GM pig called Enviropig was created with the capability of digesting plant phosphorus more efficiently than conventional pigs.[210][211] They could reduce water pollution since they excrete 30 to 70% less phosphorus in manure.[210][212] Dairy cows have been genetically engineered to produce milk that would be the same as human breast milk.[213] This could potentially benefit mothers who cannot produce breast milk but want their children to have breast milk rather than formula.[214][215] Researchers have also developed a genetically engineered cow that produces allergy-free milk.[216]

 
Mice expressing the green fluorescent protein

Scientists have genetically engineered several organisms, including some mammals, to include green fluorescent protein (GFP), for research purposes.[217] GFP and other similar reporting genes allow easy visualization and localization of the products of the genetic modification.[218] Fluorescent pigs have been bred to study human organ transplants, regenerating ocular photoreceptor cells, and other topics.[219] In 2011, green-fluorescent cats were created to help find therapies for HIV/AIDS and other diseases[220] as feline immunodeficiency virus is related to HIV.[221]

There have been suggestions that genetic engineering could be used to bring animals back from extinction. It involves changing the genome of a close living relative to resemble the extinct one and is currently being attempted with the passenger pigeon.[222] Genes associated with the woolly mammoth have been added to the genome of an African Elephant, although the lead researcher says he has no intention of creating live elephants and transferring all the genes and reversing years of genetic evolution is a long way from being feasible.[223][224] It is more likely that scientists could use this technology to conserve endangered animals by bringing back lost diversity or transferring evolved genetic advantages from adapted organisms to those that are struggling.[225]

Humans

Gene therapy[226] uses genetically modified viruses to deliver genes which can cure disease in humans. Although gene therapy is still relatively new, it has had some successes. It has been used to treat genetic disorders such as severe combined immunodeficiency,[227] and Leber's congenital amaurosis.[228] Treatments are also being developed for a range of other currently incurable diseases, such as cystic fibrosis,[229] sickle cell anemia,[230] Parkinson's disease,[231][232] cancer,[233][234][235] diabetes,[236] heart disease[237] and muscular dystrophy.[238] These treatments only effect somatic cells, meaning any changes would not be inheritable. Germline gene therapy results in any change being inheritable, which has raised concerns within the scientific community.[239][240]

In 2015, CRISPR was used to edit the DNA of non-viable human embryos.[241][242] In November 2018, He Jiankui announced that he had edited the genomes of two human embryos, in an attempt to disable the CCR5 gene, which codes for a receptor that HIV uses to enter cells. He said that twin girls, Lulu and Nana, had been born a few weeks earlier and that they carried functional copies of CCR5 along with disabled CCR5 (mosaicism) and were still vulnerable to HIV. The work was widely condemned as unethical, dangerous, and premature.[243]

Fish

When exposed to 13 °C water the zebrafish modified to express a carp creatine kinase (right) maintained swimming behavior, while wild type zebrafish (left) could not.[244]

Genetically modified fish are used for scientific research, as pets and as a food source. Aquaculture is a growing industry, currently providing over half the consumed fish worldwide.[245] Through genetic engineering it is possible to increase growth rates, reduce food intake, remove allergenic properties, increase cold tolerance and provide disease resistance. Fish can also be used to detect aquatic pollution or function as bioreactors.[246]

Several groups have been developing zebrafish to detect pollution by attaching fluorescent proteins to genes activated by the presence of pollutants. The fish will then glow and can be used as environmental sensors.[247][248] The GloFish is a brand of genetically modified fluorescent zebrafish with bright red, green, and orange fluorescent color. It was originally developed by one of the groups to detect pollution, but is now part of the ornamental fish trade, becoming the first genetically modified animal to become publicly available as a pet when in 2003 it was introduced for sale in the USA.[249]

GM fish are widely used in basic research in genetics and development. Two species of fish, zebrafish and medaka, are most commonly modified because they have optically clear chorions (membranes in the egg), rapidly develop, and the one-cell embryo is easy to see and microinject with transgenic DNA.[250] Zebrafish are model organisms for developmental processes, regeneration, genetics, behavior, disease mechanisms and toxicity testing.[251] Their transparency allows researchers to observe developmental stages, intestinal functions and tumour growth.[252][253] The generation of transgenic protocols (whole organism, cell or tissue specific, tagged with reporter genes) has increased the level of information gained by studying these fish.[254]

GM fish have been developed with promoters driving an over-production of growth hormone for use in the aquaculture industry to increase the speed of development and potentially reduce fishing pressure on wild stocks. This has resulted in dramatic growth enhancement in several species, including salmon,[255] trout[256] and tilapia.[257] AquaBounty Technologies, a biotechnology company, have produced a salmon (called AquAdvantage salmon) that can mature in half the time as wild salmon.[258] It obtained regulatory approval in 2015, the first non-plant GMO food to be commercialized.[259] As of August 2017, GMO salmon is being sold in Canada.[260] Sales in the US started in May 2021.[261]

Insects

Overexpression of methyl-CpG–binding protein 2 in Drosophila impairs climbing ability (right) compared to the control group (left).[262]

In biological research, transgenic fruit flies (Drosophila melanogaster) are model organisms used to study the effects of genetic changes on development.[263] Fruit flies are often preferred over other animals due to their short life cycle and low maintenance requirements. They also have a relatively simple genome compared to many vertebrates, with typically only one copy of each gene, making phenotypic analysis easy.[264] Drosophila have been used to study genetics and inheritance, embryonic development, learning, behavior, and aging.[265] The discovery of transposons, in particular the p-element, in Drosophila provided an early method to add transgenes to their genome, although this has been taken over by more modern gene-editing techniques.[266]

Due to their significance to human health, scientists are looking at ways to control mosquitoes through genetic engineering. Malaria-resistant mosquitoes have been developed in the laboratory by inserting a gene that reduces the development of the malaria parasite[267] and then use homing endonucleases to rapidly spread that gene throughout the male population (known as a gene drive).[268][269] This approach has been taken further by using the gene drive to spread a lethal gene.[270][271] In trials the populations of Aedes aegypti mosquitoes, the single most important carrier of dengue fever and Zika virus, were reduced by between 80% and by 90%.[272][273][271] Another approach is to use a sterile insect technique, whereby males genetically engineered to be sterile out compete viable males, to reduce population numbers.[274]

Other insect pests that make attractive targets are moths. Diamondback moths cause US$4 to $5 billion of damage each year worldwide.[275] The approach is similar to the sterile technique tested on mosquitoes, where males are transformed with a gene that prevents any females born from reaching maturity.[276] They underwent field trials in 2017.[275] Genetically modified moths have previously been released in field trials.[277] In this case a strain of pink bollworm that were sterilized with radiation were genetically engineered to express a red fluorescent protein making it easier for researchers to monitor them.[278]

Silkworm, the larvae stage of Bombyx mori, is an economically important insect in sericulture. Scientists are developing strategies to enhance silk quality and quantity. There is also potential to use the silk producing machinery to make other valuable proteins.[279] Proteins currently developed to be expressed by silkworms include; human serum albumin, human collagen α-chain, mouse monoclonal antibody and N-glycanase.[280] Silkworms have been created that produce spider silk, a stronger but extremely difficult to harvest silk,[281] and even novel silks.[282]

Other

 
Frog expressing green fluorescent protein

Systems have been developed to create transgenic organisms in a wide variety of other animals. Chickens have been genetically modified for a variety of purposes. This includes studying embryo development,[283] preventing the transmission of bird flu[284] and providing evolutionary insights using reverse engineering to recreate dinosaur-like phenotypes.[285] A GM chicken that produces the drug Kanuma, an enzyme that treats a rare condition, in its egg passed US regulatory approval in 2015.[286] Genetically modified frogs, in particular Xenopus laevis and Xenopus tropicalis, are used in developmental biology research. GM frogs can also be used as pollution sensors, especially for endocrine disrupting chemicals.[287] There are proposals to use genetic engineering to control cane toads in Australia.[288][289]

The nematode Caenorhabditis elegans is one of the major model organisms for researching molecular biology.[290] RNA interference (RNAi) was discovered in C. elegans[291] and could be induced by simply feeding them bacteria modified to express double stranded RNA.[292] It is also relatively easy to produce stable transgenic nematodes and this along with RNAi are the major tools used in studying their genes.[293] The most common use of transgenic nematodes has been studying gene expression and localization by attaching reporter genes. Transgenes can also be combined with RNAi techniques to rescue phenotypes, study gene function, image cell development in real time or control expression for different tissues or developmental stages.[293] Transgenic nematodes have been used to study viruses,[294] toxicology,[295] diseases,[296][297] and to detect environmental pollutants.[298]

 
Transgenic Hydra expressing green fluorescent protein

The gene responsible for albinism in sea cucumbers has been found and used to engineer white sea cucumbers, a rare delicacy. The technology also opens the way to investigate the genes responsible for some of the cucumbers more unusual traits, including hibernating in summer, eviscerating their intestines, and dissolving their bodies upon death.[299] Flatworms have the ability to regenerate themselves from a single cell.[300] Until 2017 there was no effective way to transform them, which hampered research. By using microinjection and radiation scientists have now created the first genetically modified flatworms.[301] The bristle worm, a marine annelid, has been modified. It is of interest due to its reproductive cycle being synchronized with lunar phases, regeneration capacity and slow evolution rate.[302] Cnidaria such as Hydra and the sea anemone Nematostella vectensis are attractive model organisms to study the evolution of immunity and certain developmental processes.[303] Other animals that have been genetically modified include snails,[304] geckos, turtles,[305] crayfish, oysters, shrimp, clams, abalone[306] and sponges.[307]

Regulation

Genetically modified organisms are regulated by government agencies. This applies to research as well as the release of genetically modified organisms, including crops and food. The development of a regulatory framework concerning genetic engineering began in 1975, at Asilomar, California. The Asilomar meeting recommended a set of guidelines regarding the cautious use of recombinant technology and any products resulting from that technology.[308] The Cartagena Protocol on Biosafety was adopted on 29 January 2000 and entered into force on 11 September 2003.[309] It is an international treaty that governs the transfer, handling, and use of genetically modified organisms.[310] One hundred and fifty-seven countries are members of the Protocol and many use it as a reference point for their own regulations.[311]

Universities and research institutes generally have a special committee that is responsible for approving any experiments that involve genetic engineering. Many experiments also need permission from a national regulatory group or legislation. All staff must be trained in the use of GMOs and all laboratories must gain approval from their regulatory agency to work with GMOs.[312] The legislation covering GMOs are often derived from regulations and guidelines in place for the non-GMO version of the organism, although they are more severe.[313] There is a near-universal system for assessing the relative risks associated with GMOs and other agents to laboratory staff and the community. They are assigned to one of four risk categories based on their virulence, the severity of the disease, the mode of transmission, and the availability of preventive measures or treatments. There are four biosafety levels that a laboratory can fall into, ranging from level 1 (which is suitable for working with agents not associated with disease) to level 4 (working with life-threatening agents). Different countries use different nomenclature to describe the levels and can have different requirements for what can be done at each level.[313]

 
A label marking this peanut butter as being non-GMO
 
Detail of a French cheese box declaring "GMO-free" production (i.e., below 0.9%)

There are differences in the regulation for the release of GMOs between countries, with some of the most marked differences occurring between the US and Europe.[314] Regulation varies in a given country depending on the intended use of the products of the genetic engineering. For example, a crop not intended for food use is generally not reviewed by authorities responsible for food safety.[315] Some nations have banned the release of GMOs or restricted their use, and others permit them with widely differing degrees of regulation.[316][317][318][319] In 2016, thirty eight countries officially ban or prohibit the cultivation of GMOs and nine (Algeria, Bhutan, Kenya, Kyrgyzstan, Madagascar, Peru, Russia, Venezuela and Zimbabwe) ban their importation.[320] Most countries that do not allow GMO cultivation do permit research using GMOs.[321] Despite regulation, illegal releases have sometimes occurred, due to weakness of enforcement.[8]

The European Union (EU) differentiates between approval for cultivation within the EU and approval for import and processing.[322] While only a few GMOs have been approved for cultivation in the EU a number of GMOs have been approved for import and processing.[323] The cultivation of GMOs has triggered a debate about the market for GMOs in Europe.[324] Depending on the coexistence regulations, incentives for cultivation of GM crops differ.[325] The US policy does not focus on the process as much as other countries, looks at verifiable scientific risks and uses the concept of substantial equivalence.[326] Whether gene edited organisms should be regulated the same as genetically modified organism is debated. USA regulations sees them as separate and does not regulate them under the same conditions, while in Europe a GMO is any organism created using genetic engineering techniques.[28]

One of the key issues concerning regulators is whether GM products should be labeled. The European Commission says that mandatory labeling and traceability are needed to allow for informed choice, avoid potential false advertising[327] and facilitate the withdrawal of products if adverse effects on health or the environment are discovered.[328] The American Medical Association[329] and the American Association for the Advancement of Science[330] say that absent scientific evidence of harm even voluntary labeling is misleading and will falsely alarm consumers. Labeling of GMO products in the marketplace is required in 64 countries.[331] Labeling can be mandatory up to a threshold GM content level (which varies between countries) or voluntary. In Canada and the US labeling of GM food is voluntary,[332] while in Europe all food (including processed food) or feed which contains greater than 0.9% of approved GMOs must be labeled.[333] In 2014, sales of products that had been labeled as non-GMO grew 30 percent to $1.1 billion.[334]

Controversy

There is controversy over GMOs, especially with regard to their release outside laboratory environments. The dispute involves consumers, producers, biotechnology companies, governmental regulators, non-governmental organizations, and scientists. Many of these concerns involve GM crops and whether food produced from them is safe and what impact growing them will have on the environment. These controversies have led to litigation, international trade disputes, and protests, and to restrictive regulation of commercial products in some countries.[335] Most concerns are around the health and environmental effects of GMOs. These include whether they may provoke an allergic reaction, whether the transgenes could transfer to human cells, and whether genes not approved for human consumption could outcross into the food supply.[336]

 
A protester advocating for the labeling of GMOs

There is a scientific consensus[337][338][339][340] that currently available food derived from GM crops poses no greater risk to human health than conventional food,[341][342][343][344][345] but that each GM food needs to be tested on a case-by-case basis before introduction.[346][347][348] Nonetheless, members of the public are much less likely than scientists to perceive GM foods as safe.[349][350][351][352] The legal and regulatory status of GM foods varies by country, with some nations banning or restricting them, and others permitting them with widely differing degrees of regulation.[353][354][355][356]

As late as the 1990s gene flow into wild populations was thought to be unlikely and rare, and if it were to occur, easily eradicated. It was thought that this would add no additional environmental costs or risks – no effects were expected other than those already caused by pesticide applications.[357] However, in the decades since, several such examples have been observed. Gene flow between GM crops and compatible plants, along with increased use of broad-spectrum herbicides,[358] can increase the risk of herbicide resistant weed populations.[359] Debate over the extent and consequences of gene flow intensified in 2001 when a paper was published showing transgenes had been found in landrace maize in Mexico, the crop's center of diversity.[360][361] Gene flow from GM crops to other organisms has been found to generally be lower than what would occur naturally.[362] In order to address some of these concerns some GMOs have been developed with traits to help control their spread. To prevent the genetically modified salmon inadvertently breeding with wild salmon, all the fish raised for food are females, triploid, 99% are reproductively sterile, and raised in areas where escaped salmon could not survive.[363][364] Bacteria have also been modified to depend on nutrients that cannot be found in nature,[365] and genetic use restriction technology has been developed, though not yet marketed, that causes the second generation of GM plants to be sterile.[366]

Other environmental and agronomic concerns include a decrease in biodiversity, an increase in secondary pests (non-targeted pests) and evolution of resistant insect pests.[367][368][369] In the areas of China and the US with Bt crops the overall biodiversity of insects has increased and the impact of secondary pests has been minimal.[370] Resistance was found to be slow to evolve when best practice strategies were followed.[370] The impact of Bt crops on beneficial non-target organisms became a public issue after a 1999 paper suggested they could be toxic to monarch butterflies. Follow up studies have since shown that the toxicity levels encountered in the field were not high enough to harm the larvae.[371]

Accusations that scientists are "playing God" and other religious issues have been ascribed to the technology from the beginning.[372] With the ability to genetically engineer humans now possible there are ethical concerns over how far this technology should go, or if it should be used at all.[373] Much debate revolves around where the line between treatment and enhancement is and whether the modifications should be inheritable.[374] Other concerns include contamination of the non-genetically modified food supply,[375][376] the rigor of the regulatory process,[377][378] consolidation of control of the food supply in companies that make and sell GMOs,[379] exaggeration of the benefits of genetic modification,[380] or concerns over the use of herbicides with glyphosate.[381] Other issues raised include the patenting of life[382] and the use of intellectual property rights.[383]

There are large differences in consumer acceptance of GMOs, with Europeans more likely to view GM food negatively than North Americans.[384] GMOs arrived on the scene as the public confidence in food safety, attributed to recent food scares such as Bovine spongiform encephalopathy and other scandals involving government regulation of products in Europe, was low.[385] This along with campaigns run by various non-governmental organizations (NGO) have been very successful in blocking or limiting the use of GM crops.[386] NGOs like the Organic Consumers Association, the Union of Concerned Scientists,[387][388][389] Greenpeace and other groups have said that risks have not been adequately identified and managed[390] and that there are unanswered questions regarding the potential long-term impact on human health from food derived from GMOs. They propose mandatory labeling[391][392] or a moratorium on such products.[379][377][393]

References

  1. ^ "Food, genetically modified". www.who.int. Retrieved 15 August 2023.
  2. ^ Smyth SJ (April 2020). "The human health benefits from GM crops". Plant Biotechnology Journal. 18 (4): 887–888. doi:10.1111/pbi.13261. PMC 7061863. PMID 31544299.
  3. ^ Chilton MD (4 October 2016). "Nature, The First Creator of GMOs". Forbes. Retrieved 4 January 2019.
  4. ^ Blakemore E. "The First GMO Is 8,000 Years Old". Smithsonian. Retrieved 5 January 2019.
  5. ^ The new encyclopaedia Britannica (15th ed.). Chicago: Encyclopaedia Britannica. 1993. pp. 178. ISBN 0-85229-571-5. OCLC 27665641.
  6. ^ Staff Economic Impacts of Genetically Modified Crops on the Agri-Food Sector; p. 42 Glossary – Term and Definitions 14 May 2013 at the Wayback Machine The European Commission Directorate-General for Agriculture, "Genetic engineering: The manipulation of an organism's genetic endowment by introducing or eliminating specific genes through modern molecular biology techniques. A broad definition of genetic engineering also includes selective breeding and other means of artificial selection", Retrieved 5 November 2012
  7. ^ The European Parliament and the council of the European Union (12 March 2001). "Directive on the release of genetically modified organisms (GMOs) Directive 2001/18/EC ANNEX I A". Official Journal of the European Communities.
  8. ^ a b c Freedman W (27 August 2018). "6 ~ Evolution". Environmental Science – a Canadian perspective (6 ed.). Dalhousie University.
  9. ^ a b "Organisms obtained by mutagenesis are GMOs and are, in principle, subject to the obligations laid down by the GMO Directive" (PDF). curia.europa.eu. (PDF) from the original on 25 July 2018. Retrieved 5 January 2019.
  10. ^ "Section 2: Description and Definitions". www.fao.org. Retrieved 3 January 2019.
  11. ^ "Frequently asked questions on genetically modified foods". WHO. Retrieved 3 January 2019.
  12. ^ "The EU Legislation on GMOs – An Overview". EU Science Hub – European Commission. 29 June 2010. Retrieved 3 January 2019.
  13. ^ "GMOs and Horizontal Gene Transfer". NeuroLogica Blog. 13 October 2016. Retrieved 9 July 2021.
  14. ^ Zhang C, Wohlhueter R, Zhang H (September 2016). "Genetically modified foods: A critical review of their promise and problems". Food Science and Human Wellness. 5 (3): 116–123. doi:10.1016/j.fshw.2016.04.002.
  15. ^ Oliver MJ (2014). "Why we need GMO crops in agriculture". Missouri Medicine. 111 (6): 492–507. PMC 6173531. PMID 25665234.
  16. ^ Center for Food Safety and Applied Nutrition. "Food from Genetically Engineered Plants – Consumer Info About Food from Genetically Engineered Plants". www.fda.gov. Retrieved 8 January 2019.
  17. ^ Secretariat of the Convention on Biological Diversity. Montreal: 2000. The Cartagena Protocol on Biosafety to the Convention on Biological Diversity.
  18. ^ "Frequently Asked Questions (FAQs) on the Cartagena Protocol". The Biosafety Clearing-House (BCH). 29 February 2012. Retrieved 3 January 2019.
  19. ^ "What Is the Difference Between Genetically Modified Organisms and Genetically Engineered Organisms?". agbiotech.ces.ncsu.edu. Retrieved 8 January 2019.
  20. ^ "Agricultural Biotechnology Glossary | USDA". www.usda.gov. Retrieved 8 January 2019.
  21. ^ Colombo L (2007). "The semantics of the term 'genetically modified organism' // Genetic impact of aquaculture activities on native populations". Genimpact Final Scientific Report (E U Contract N. RICA-CT -2005-022802): 123–125.
  22. ^ Chassy BM (2007). "The History and Future of GMOs in Food and Agriculture". Cereal Foods World. doi:10.1094/cfw-52-4-0169. ISSN 0146-6283.
  23. ^ "Why the term GMO is 'scientifically meaningless'". Public Radio International. Retrieved 5 January 2019.
  24. ^ Tagliabue G (September 2015). "The nonsensical GMO pseudo-category and a precautionary rabbit hole". Nature Biotechnology. 33 (9): 907–908. doi:10.1038/nbt.3333. PMID 26348954. S2CID 205281930.
  25. ^ "National Organic Standards Board Materials/GMO Subcommittee Second Discussion Document on Excluded Methods Terminology" (PDF). United States Department of Agriculture. 22 August 2014. (PDF) from the original on 2 October 2015. Retrieved 4 January 2019.
  26. ^ "Here's Why You Should Vote Against Measure P, Even If You Hate GMOs". Lost Coast Outpost. Retrieved 4 January 2019.
  27. ^ Neslen A (25 July 2018). "Gene-edited plants and animals are GM foods, EU court rules". The Guardian. ISSN 0261-3077. Retrieved 5 January 2019.
  28. ^ a b "A CRISPR definition of genetic modification". Nature Plants. 4 (5): 233. May 2018. doi:10.1038/s41477-018-0158-1. PMID 29725105.
  29. ^ "Viewpoint: Non-GMO salt exploits Americans' scientific illiteracy". Genetic Literacy Project. 1 June 2018. Retrieved 9 July 2021.
  30. ^ Knutson J (28 May 2018). "A sad day for our society when salt is labeled non-GMO". Agweek. Retrieved 9 July 2021.
  31. ^ "Non GMO salt? Water? Food companies exploit GMO free labels, misleading customers, promoting misinformation". Genetic Literacy Project. 24 August 2015. Retrieved 9 July 2021.
  32. ^ Nicholl DS (29 May 2008). An Introduction to Genetic Engineering. Cambridge University Press. p. 34. ISBN 978-1-139-47178-7.
  33. ^ Liang J, Luo Y, Zhao H (2011). "Synthetic biology: putting synthesis into biology". Wiley Interdisciplinary Reviews: Systems Biology and Medicine. 3 (1): 7–20. doi:10.1002/wsbm.104. PMC 3057768. PMID 21064036.
  34. ^ Berg P, Mertz JE (January 2010). "Personal reflections on the origins and emergence of recombinant DNA technology". Genetics. 184 (1): 9–17. doi:10.1534/genetics.109.112144. PMC 2815933. PMID 20061565.
  35. ^ Rahimzadeh M, Sadeghizadeh M, Najafi F, Arab S, Mobasheri H (December 2016). "Impact of heat shock step on bacterial transformation efficiency". Molecular Biology Research Communications. 5 (4): 257–261. PMC 5326489. PMID 28261629.
  36. ^ Chen I, Dubnau D (March 2004). "DNA uptake during bacterial transformation". Nature Reviews. Microbiology. 2 (3): 241–9. doi:10.1038/nrmicro844. PMID 15083159. S2CID 205499369.
  37. ^ a b National Research Council (US) Committee on Identifying and Assessing Unintended Effects of Genetically Engineered Foods on Human Health (1 January 2004). Methods and Mechanisms for Genetic Manipulation of Plants, Animals, and Microorganisms. National Academies Press (US).
  38. ^ Gelvin SB (March 2003). "Agrobacterium-mediated plant transformation: the biology behind the 'gene-jockeying' tool". Microbiology and Molecular Biology Reviews. 67 (1): 16–37, table of contents. doi:10.1128/MMBR.67.1.16-37.2003. PMC 150518. PMID 12626681.
  39. ^ Head G, Hull RH, Tzotzos GT (2009). Genetically Modified Plants: Assessing Safety and Managing Risk. London: Academic Press. p. 244. ISBN 978-0-12-374106-6.
  40. ^ Tuomela M, Stanescu I, Krohn K (October 2005). "Validation overview of bio-analytical methods". Gene Therapy. 12 (S1): S131-8. doi:10.1038/sj.gt.3302627. PMID 16231045. S2CID 23000818.
  41. ^ Narayanaswamy S (1994). Plant Cell and Tissue Culture. Tata McGraw-Hill Education. pp. vi. ISBN 978-0-07-460277-5.
  42. ^ Setlow JK (31 October 2002). Genetic Engineering: Principles and Methods. Springer Science & Business Media. p. 109. ISBN 978-0-306-47280-0.
  43. ^ Grizot S, Smith J, Daboussi F, Prieto J, Redondo P, Merino N, Villate M, Thomas S, Lemaire L, Montoya G, Blanco FJ, Pâques F, Duchateau P (September 2009). "Efficient targeting of a SCID gene by an engineered single-chain homing endonuclease". Nucleic Acids Research. 37 (16): 5405–19. doi:10.1093/nar/gkp548. PMC 2760784. PMID 19584299.
  44. ^ Gao H, Smith J, Yang M, Jones S, Djukanovic V, Nicholson MG, West A, Bidney D, Falco SC, Jantz D, Lyznik LA (January 2010). "Heritable targeted mutagenesis in maize using a designed endonuclease". The Plant Journal. 61 (1): 176–87. doi:10.1111/j.1365-313X.2009.04041.x. PMID 19811621.
  45. ^ Townsend JA, Wright DA, Winfrey RJ, Fu F, Maeder ML, Joung JK, Voytas DF (May 2009). "High-frequency modification of plant genes using engineered zinc-finger nucleases". Nature. 459 (7245): 442–5. Bibcode:2009Natur.459..442T. doi:10.1038/nature07845. PMC 2743854. PMID 19404258.
  46. ^ Shukla VK, Doyon Y, Miller JC, DeKelver RC, Moehle EA, Worden SE, Mitchell JC, Arnold NL, Gopalan S, Meng X, Choi VM, Rock JM, Wu YY, Katibah GE, Zhifang G, McCaskill D, Simpson MA, Blakeslee B, Greenwalt SA, Butler HJ, Hinkley SJ, Zhang L, Rebar EJ, Gregory PD, Urnov FD (May 2009). "Precise genome modification in the crop species Zea mays using zinc-finger nucleases". Nature. 459 (7245): 437–41. Bibcode:2009Natur.459..437S. doi:10.1038/nature07992. PMID 19404259. S2CID 4323298.
  47. ^ Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hummel A, Bogdanove AJ, Voytas DF (October 2010). "Targeting DNA double-strand breaks with TAL effector nucleases". Genetics. 186 (2): 757–61. doi:10.1534/genetics.110.120717. PMC 2942870. PMID 20660643.
  48. ^ Li T, Huang S, Jiang WZ, Wright D, Spalding MH, Weeks DP, Yang B (January 2011). "TAL nucleases (TALNs): hybrid proteins composed of TAL effectors and FokI DNA-cleavage domain". Nucleic Acids Research. 39 (1): 359–72. doi:10.1093/nar/gkq704. PMC 3017587. PMID 20699274.
  49. ^ Esvelt KM, Wang HH (2013). "Genome-scale engineering for systems and synthetic biology". Molecular Systems Biology. 9: 641. doi:10.1038/msb.2012.66. PMC 3564264. PMID 23340847.
  50. ^ Tan WS, Carlson DF, Walton MW, Fahrenkrug SC, Hackett PB (2012). "Precision editing of large animal genomes". Advances in Genetics Volume 80. Vol. 80. pp. 37–97. doi:10.1016/B978-0-12-404742-6.00002-8. ISBN 978-0-12-404742-6. PMC 3683964. PMID 23084873.
  51. ^ a b Malzahn A, Lowder L, Qi Y (24 April 2017). "Plant genome editing with TALEN and CRISPR". Cell & Bioscience. 7: 21. doi:10.1186/s13578-017-0148-4. PMC 5404292. PMID 28451378.
  52. ^ Kingsbury N (2009). Hybrid: The History and Science of Plant Breeding. University of Chicago Press. ISBN 978-0-226-43705-7.
  53. ^ Clive Root (2007). Domestication. Greenwood Publishing Groups.
  54. ^ Zohary D, Hopf M, Weiss E (2012). Domestication of Plants in the Old World: The Origin and Spread of Plants in the Old World. Oxford University Press.
  55. ^ Jackson DA, Symons RH, Berg P (October 1972). "Biochemical method for inserting new genetic information into DNA of Simian Virus 40: circular SV40 DNA molecules containing lambda phage genes and the galactose operon of Escherichia coli". Proceedings of the National Academy of Sciences of the United States of America. 69 (10): 2904–9. Bibcode:1972PNAS...69.2904J. doi:10.1073/pnas.69.10.2904. PMC 389671. PMID 4342968.
  56. ^ Sateesh MK (25 August 2008). Bioethics And Biosafety. I. K. International Pvt Ltd. pp. 456–. ISBN 978-81-906757-0-3. Retrieved 27 March 2013.
  57. ^ Zhang C, Wohlhueter R, Zhang H (2016). "Genetically modified foods: A critical review of their promise and problems". Food Science and Human Wellness. 5 (3): 116–123. doi:10.1016/j.fshw.2016.04.002.
  58. ^ Russo E (January 2003). "The birth of biotechnology". Nature. 421 (6921): 456–7. Bibcode:2003Natur.421..456R. doi:10.1038/nj6921-456a. PMID 12540923.
  59. ^ Morrow JF, Cohen SN, Chang AC, Boyer HW, Goodman HM, Helling RB (May 1974). "Replication and transcription of eukaryotic DNA in Escherichia coli". Proceedings of the National Academy of Sciences of the United States of America. 71 (5): 1743–7. Bibcode:1974PNAS...71.1743M. doi:10.1073/pnas.71.5.1743. PMC 388315. PMID 4600264.
  60. ^ a b Jaenisch R, Mintz B (April 1974). "Simian virus 40 DNA sequences in DNA of healthy adult mice derived from preimplantation blastocysts injected with viral DNA". Proceedings of the National Academy of Sciences of the United States of America. 71 (4): 1250–4. Bibcode:1974PNAS...71.1250J. doi:10.1073/pnas.71.4.1250. PMC 388203. PMID 4364530.
  61. ^ "'Any idiot can do it.' Genome editor CRISPR could put mutant mice in everyone's reach". Science | AAAS. 2 November 2016. Retrieved 2 December 2016.
  62. ^ Gordon JW, Ruddle FH (December 1981). "Integration and stable germ line transmission of genes injected into mouse pronuclei". Science. 214 (4526): 1244–6. Bibcode:1981Sci...214.1244G. doi:10.1126/science.6272397. PMID 6272397.
  63. ^ Costantini F, Lacy E (November 1981). "Introduction of a rabbit beta-globin gene into the mouse germ line". Nature. 294 (5836): 92–4. Bibcode:1981Natur.294...92C. doi:10.1038/294092a0. PMID 6945481. S2CID 4371351.
  64. ^ Hanahan D, Wagner EF, Palmiter RD (September 2007). "The origins of oncomice: a history of the first transgenic mice genetically engineered to develop cancer". Genes & Development. 21 (18): 2258–70. doi:10.1101/gad.1583307. PMID 17875663.
  65. ^ Brophy B, Smolenski G, Wheeler T, Wells D, L'Huillier P, Laible G (February 2003). "Cloned transgenic cattle produce milk with higher levels of beta-casein and kappa-casein". Nature Biotechnology. 21 (2): 157–62. doi:10.1038/nbt783. PMID 12548290. S2CID 45925486.
  66. ^ Clark AJ (July 1998). "The mammary gland as a bioreactor: expression, processing, and production of recombinant proteins". Journal of Mammary Gland Biology and Neoplasia. 3 (3): 337–50. doi:10.1023/a:1018723712996. PMID 10819519.
  67. ^ Gordon K, Lee E, Vitale JA, Smith AE, Westphal H, Hennighausen L (1987). "Production of human tissue plasminogen activator in transgenic mouse milk. 1987". Biotechnology. 24 (11): 425–8. doi:10.1038/nbt1187-1183. PMID 1422049. S2CID 3261903.
  68. ^ Bevan MW, Flavell RB, Chilton MD (1983). "A chimaeric antibiotic resistance gene as a selectable marker for plant cell transformation. 1983". Nature. 304 (5922): 184. Bibcode:1983Natur.304..184B. doi:10.1038/304184a0. S2CID 28713537.
  69. ^ Jinturkar KA, Rathi MN, Misra A (2011). "Gene Delivery Using Physical Methods". Challenges in Delivery of Therapeutic Genomics and Proteomics. pp. 83–126. doi:10.1016/b978-0-12-384964-9.00003-7. ISBN 978-0-12-384964-9.
  70. ^ a b Ye X, Al-Babili S, Klöti A, Zhang J, Lucca P, Beyer P, Potrykus I (January 2000). "Engineering the provitamin A (beta-carotene) biosynthetic pathway into (carotenoid-free) rice endosperm". Science. 287 (5451): 303–5. Bibcode:2000Sci...287..303Y. doi:10.1126/science.287.5451.303. PMID 10634784. S2CID 40258379.
  71. ^ Goeddel DV, Kleid DG, Bolivar F, Heyneker HL, Yansura DG, Crea R, Hirose T, Kraszewski A, Itakura K, Riggs AD (January 1979). "Expression in Escherichia coli of chemically synthesized genes for human insulin". Proceedings of the National Academy of Sciences of the United States of America. 76 (1): 106–10. Bibcode:1979PNAS...76..106G. doi:10.1073/pnas.76.1.106. PMC 382885. PMID 85300.
  72. ^ . Time. 15 November 1982. Archived from the original on 27 October 2011. Retrieved 17 July 2010.
  73. ^ Horn ME, Woodard SL, Howard JA (May 2004). "Plant molecular farming: systems and products". Plant Cell Reports. 22 (10): 711–20. doi:10.1007/s00299-004-0767-1. PMC 7079917. PMID 14997337.
  74. ^ BBC News 14 June 2002 GM crops: A bitter harvest?
  75. ^ Maugh, Thomas H. II (9 June 1987). "Altered Bacterium Does Its Job: Frost Failed to Damage Sprayed Test Crop, Company Says". Los Angeles Times.
  76. ^ Fraley RT, Rogers SG, Horsch RB, Sanders PR, Flick JS, Adams SP, Bittner ML, Brand LA, Fink CL, Fry JS, Galluppi GR, Goldberg SB, Hoffmann NL, Woo SC (August 1983). "Expression of bacterial genes in plant cells". Proceedings of the National Academy of Sciences of the United States of America. 80 (15): 4803–7. Bibcode:1983PNAS...80.4803F. doi:10.1073/pnas.80.15.4803. PMC 384133. PMID 6308651.
  77. ^ James, Clive (1997). "Global Status of Transgenic Crops in 1997" (PDF). ISAAA Briefs No. 5.: 31. (PDF) from the original on 16 January 2009.
  78. ^ Bruening G, Lyons JM (2000). "The case of the FLAVR SAVR tomato". California Agriculture. 54 (4): 6–7. doi:10.3733/ca.v054n04p6.
  79. ^ Debora MacKenzie (18 June 1994). "Transgenic tobacco is European first". New Scientist.
  80. ^ Genetically Altered Potato Ok'd For Crops Lawrence Journal-World. 6 May 1995
  81. ^ James C (1996). "Global Review of the Field Testing and Commercialization of Transgenic Plants: 1986 to 1995" (PDF). The International Service for the Acquisition of Agri-biotech Applications. (PDF) from the original on 16 June 2010. Retrieved 17 July 2010.
  82. ^ Gibson DG, Glass JI, Lartigue C, Noskov VN, Chuang RY, Algire MA, Benders GA, Montague MG, Ma L, Moodie MM, Merryman C, Vashee S, Krishnakumar R, Assad-Garcia N, Andrews-Pfannkoch C, Denisova EA, Young L, Qi ZQ, Segall-Shapiro TH, Calvey CH, Parmar PP, Hutchison CA, Smith HO, Venter JC (July 2010). "Creation of a bacterial cell controlled by a chemically synthesized genome". Science. 329 (5987): 52–6. Bibcode:2010Sci...329...52G. doi:10.1126/science.1190719. PMID 20488990. S2CID 7320517.
  83. ^ Sample I (20 May 2010). "Craig Venter creates synthetic life form". guardian.co.uk. London.
  84. ^ Vàzquez-Salat N, Salter B, Smets G, Houdebine LM (1 November 2012). "The current state of GMO governance: are we ready for GM animals?". Biotechnology Advances. Special issue on ACB 2011. 30 (6): 1336–43. doi:10.1016/j.biotechadv.2012.02.006. PMID 22361646.
  85. ^ "Glowing fish to be first genetically changed pet". CNN. 21 November 2003. Retrieved 25 December 2018.
  86. ^ a b Pollack A (19 November 2015). "Genetically Engineered Salmon Approved for Consumption". The New York Times. ISSN 0362-4331. Archived from the original on 2 January 2022. Retrieved 27 January 2019.
  87. ^ Bodnar A (October 2010). (PDF). ISB News Report. Archived from the original (PDF) on 8 March 2021. Retrieved 22 January 2016.
  88. ^ Melo EO, Canavessi AM, Franco MM, Rumpf R (March 2007). "Animal transgenesis: state of the art and applications" (PDF). Journal of Applied Genetics. 48 (1): 47–61. doi:10.1007/BF03194657. PMID 17272861. S2CID 24578435.
  89. ^ . www.learner.org. Archived from the original on 3 December 2019. Retrieved 18 August 2017.
  90. ^ Fan M, Tsai J, Chen B, Fan K, LaBaer J (March 2005). "A central repository for published plasmids". Science. 307 (5717): 1877. doi:10.1126/science.307.5717.1877a. PMID 15790830. S2CID 27404861.
  91. ^ Cooper GM (2000). "Cells As Experimental Models". The Cell: A Molecular Approach (2nd ed.).
  92. ^ Patel P (June 2018). "Microbe Mystery". Scientific American. 319 (1): 18. Bibcode:2018SciAm.319a..18P. doi:10.1038/scientificamerican0718-18a. PMID 29924081. S2CID 49310760.
  93. ^ Arpino JA, Hancock EJ, Anderson J, Barahona M, Stan GB, Papachristodoulou A, Polizzi K (July 2013). "Tuning the dials of Synthetic Biology". Microbiology. 159 (Pt 7): 1236–53. doi:10.1099/mic.0.067975-0. PMC 3749727. PMID 23704788.
  94. ^ Pollack A (7 May 2014). "Researchers Report Breakthrough in Creating Artificial Genetic Code". The New York Times. Archived from the original on 2 January 2022. Retrieved 7 May 2014.
  95. ^ Malyshev DA, Dhami K, Lavergne T, Chen T, Dai N, Foster JM, Corrêa IR, Romesberg FE (May 2014). "A semi-synthetic organism with an expanded genetic alphabet". Nature. 509 (7500): 385–8. Bibcode:2014Natur.509..385M. doi:10.1038/nature13314. PMC 4058825. PMID 24805238.
  96. ^ a b Kärenlampi SO, von Wright AJ (1 January 2016). "Genetically Modified Microorganisms". Encyclopedia of Food and Health. Encyclopedia of Food and Health. pp. 211–216. doi:10.1016/B978-0-12-384947-2.00356-1. ISBN 978-0-12-384953-3.
  97. ^ Panesar, Pamit et al. (2010) Enzymes in Food Processing: Fundamentals and Potential Applications, Chapter 10, I K International Publishing House, ISBN 978-93-80026-33-6
  98. ^ Blair R, Regenstein JM (3 August 2015). Genetic Modification and Food Quality: A Down to Earth Analysis. John Wiley & Sons. pp. 20–24. ISBN 978-1-118-75641-6.
  99. ^ a b c Jumba M (2009). Genetically Modified Organisms the Mystery Unraveled. Durham: Eloquent Books. pp. 51–54. ISBN 978-1-60911-081-9.
  100. ^ a b Zhou Y, Lu Z, Wang X, Selvaraj JN, Zhang G (February 2018). "Genetic engineering modification and fermentation optimization for extracellular production of recombinant proteins using Escherichia coli". Applied Microbiology and Biotechnology. 102 (4): 1545–1556. doi:10.1007/s00253-017-8700-z. PMID 29270732. S2CID 253769838.
  101. ^ Leader B, Baca QJ, Golan DE (January 2008). "Protein therapeutics: a summary and pharmacological classification". Nature Reviews. Drug Discovery. A guide to drug discovery. 7 (1): 21–39. doi:10.1038/nrd2399. PMID 18097458. S2CID 3358528.
  102. ^ Walsh G (April 2005). "Therapeutic insulins and their large-scale manufacture". Applied Microbiology and Biotechnology. 67 (2): 151–9. doi:10.1007/s00253-004-1809-x. PMID 15580495. S2CID 5986035.
  103. ^ Pipe SW (May 2008). "Recombinant clotting factors". Thrombosis and Haemostasis. 99 (5): 840–50. doi:10.1160/TH07-10-0593. PMID 18449413. S2CID 2701961.
  104. ^ Bryant J, Baxter L, Cave CB, Milne R (July 2007). Bryant J (ed.). "Recombinant growth hormone for idiopathic short stature in children and adolescents" (PDF). The Cochrane Database of Systematic Reviews (3): CD004440. doi:10.1002/14651858.CD004440.pub2. PMID 17636758.
  105. ^ Baxter L, Bryant J, Cave CB, Milne R (January 2007). Bryant J (ed.). "Recombinant growth hormone for children and adolescents with Turner syndrome" (PDF). The Cochrane Database of Systematic Reviews (1): CD003887. doi:10.1002/14651858.CD003887.pub2. PMID 17253498.
  106. ^ Summers, Rebecca (24 April 2013). "Bacteria churn out first ever petrol-like biofuel". New Scientist, Retrieved 27 April 2013
  107. ^ Reardon S (June 2018). "Genetically modified bacteria enlisted in fight against disease". Nature. 558 (7711): 497–498. Bibcode:2018Natur.558..497R. doi:10.1038/d41586-018-05476-4. PMID 29946090.
  108. ^ Amarger N (November 2002). "Genetically modified bacteria in agriculture". Biochimie. 84 (11): 1061–72. doi:10.1016/s0300-9084(02)00035-4. PMID 12595134.
  109. ^ Sharma B, Dangi AK, Shukla P (March 2018). "Contemporary enzyme based technologies for bioremediation: A review". Journal of Environmental Management. 210: 10–22. doi:10.1016/j.jenvman.2017.12.075. PMID 29329004.
  110. ^ a b Yetisen AK, Davis J, Coskun AF, Church GM, Yun SH (December 2015). "Bioart". Trends in Biotechnology. 33 (12): 724–734. doi:10.1016/j.tibtech.2015.09.011. PMID 26617334. S2CID 259584956.
  111. ^ Church GM, Gao Y, Kosuri S (September 2012). "Next-generation digital information storage in DNA". Science. 337 (6102): 1628. Bibcode:2012Sci...337.1628C. doi:10.1126/science.1226355. PMID 22903519.
  112. ^ Baldo A, van den Akker E, Bergmans HE, Lim F, Pauwels K (December 2013). "General considerations on the biosafety of virus-derived vectors used in gene therapy and vaccination". Current Gene Therapy. 13 (6): 385–94. doi:10.2174/15665232113136660005. PMC 3905712. PMID 24195604.
  113. ^ "Is gene therapy available to treat my disorder?". Genetics Home Reference. Retrieved 14 December 2018.
  114. ^ a b Aiuti A, Roncarolo MG, Naldini L (June 2017). "ex vivo gene therapy in Europe: paving the road for the next generation of advanced therapy medicinal products". EMBO Molecular Medicine. 9 (6): 737–740. doi:10.15252/emmm.201707573. PMC 5452047. PMID 28396566.
  115. ^ a b c d Lundstrom K (May 2018). "Viral Vectors in Gene Therapy". Diseases. 6 (2): 42. doi:10.3390/diseases6020042. PMC 6023384. PMID 29883422.
  116. ^ a b Sheridan C (February 2011). "Gene therapy finds its niche". Nature Biotechnology. 29 (2): 121–8. doi:10.1038/nbt.1769. PMID 21301435. S2CID 5063701.
  117. ^ Manservigi R, Epstein AL, Argnani R, Marconi P (2013). HSV as a Vector in Vaccine Development and Gene Therapy. Landes Bioscience.
  118. ^ Chan VS (November 2006). "Use of genetically modified viruses and genetically engineered virus-vector vaccines: environmental effects". Journal of Toxicology and Environmental Health. Part A. 69 (21): 1971–7. doi:10.1080/15287390600751405. PMID 16982535. S2CID 41198650.
  119. ^ a b Ramezanpour B, Haan I, Osterhaus A, Claassen E (December 2016). "Vector-based genetically modified vaccines: Exploiting Jenner's legacy". Vaccine. 34 (50): 6436–6448. doi:10.1016/j.vaccine.2016.06.059. PMC 7115478. PMID 28029542.
  120. ^ Tameris MD, Hatherill M, Landry BS, Scriba TJ, Snowden MA, Lockhart S, Shea JE, McClain JB, Hussey GD, Hanekom WA, Mahomed H, McShane H (March 2013). "Safety and efficacy of MVA85A, a new tuberculosis vaccine, in infants previously vaccinated with BCG: a randomized, placebo-controlled phase 2b trial". Lancet. 381 (9871): 1021–8. doi:10.1016/S0140-6736(13)60177-4. PMC 5424647. PMID 23391465.
  121. ^ Delany I, Rappuoli R, De Gregorio E (June 2014). "Vaccines for the 21st century". EMBO Molecular Medicine. 6 (6): 708–20. doi:10.1002/emmm.201403876. PMC 4203350. PMID 24803000.
  122. ^ Bhattacharya S. "Genetically-modified virus explodes cancer cells". New Scientist.
  123. ^ Khamsi R. "GM virus shrinks cancer tumours in humans". New Scientist.
  124. ^ Leja J, Yu D, Nilsson B, Gedda L, Zieba A, Hakkarainen T, Åkerström G, Öberg K, Giandomenico V, Essand M (November 2011). "Oncolytic adenovirus modified with somatostatin motifs for selective infection of neuroendocrine tumor cells". Gene Therapy. 18 (11): 1052–62. doi:10.1038/gt.2011.54. PMID 21490682. S2CID 22520171.
  125. ^ Perett, Linda (30 June 2011) Measles viruses genetically modified to treat ovarian cancer National Cancer Institute, Benchmarks, Retrieved 5 September 2012
  126. ^ Breitbach CJ, Thorne SH, Bell JC, Kirn DH (July 2012). "Targeted and armed oncolytic poxviruses for cancer: the lead example of JX-594". Current Pharmaceutical Biotechnology. 13 (9): 1768–72. doi:10.2174/138920112800958922. PMID 21740365.
  127. ^ Beasley, Deena (31 August 2011) Cancer-fighting virus shown to target tumors alone Reuters Science, Retrieved 5 September 2012
  128. ^ Garber K (March 2006). "China approves world's first oncolytic virus therapy for cancer treatment". Journal of the National Cancer Institute. 98 (5): 298–300. doi:10.1093/jnci/djj111. PMID 16507823.
  129. ^ Molteni M (12 April 2017). "Florida's Orange Trees Are Dying, But a Weaponized Virus Could Save Them". Wired. Retrieved 17 April 2017.
  130. ^ a b Jelley J (7 August 2002). "GM virus curbs rabbits". Retrieved 16 December 2018.
  131. ^ O'Riordan B (26 February 2005). "Virus planned to counter cane toad". The Guardian. ISSN 0261-3077. Retrieved 16 December 2018.
  132. ^ Mildura GO. "Virus could sterilise Australia's rabbits". New Scientist. Retrieved 16 December 2018.
  133. ^ Angulo E, Cooke B (December 2002). "First synthesize new viruses then regulate their release? The case of the wild rabbit". Molecular Ecology. 11 (12): 2703–9. doi:10.1046/j.1365-294X.2002.01635.x. hdl:10261/45541. PMID 12453252. S2CID 23916432.
  134. ^ Pires DP, Cleto S, Sillankorva S, Azeredo J, Lu TK (September 2016). "Genetically Engineered Phages: a Review of Advances over the Last Decade". Microbiology and Molecular Biology Reviews. 80 (3): 523–43. doi:10.1128/MMBR.00069-15. PMC 4981678. PMID 27250768.
  135. ^ Lee YJ, Yi H, Kim WJ, Kang K, Yun DS, Strano MS, Ceder G, Belcher AM (May 2009). "Fabricating genetically engineered high-power lithium-ion batteries using multiple virus genes". Science. 324 (5930): 1051–5. Bibcode:2009Sci...324.1051L. doi:10.1126/science.1171541. PMID 19342549. S2CID 32017913.
  136. ^ Branduardi P, Smeraldi C, Porro D (2008). "Metabolically engineered yeasts: 'potential' industrial applications". Journal of Molecular Microbiology and Biotechnology. 15 (1): 31–40. doi:10.1159/000111990. PMID 18349548.
  137. ^ "GM fungi: New way to produce cheap biofuel". The Times of India. 4 June 2013. Retrieved 17 December 2018.
  138. ^ Fang W, Vega-Rodríguez J, Ghosh AK, Jacobs-Lorena M, Kang A, St Leger RJ (February 2011). "Development of transgenic fungi that kill human malaria parasites in mosquitoes". Science. 331 (6020): 1074–7. Bibcode:2011Sci...331.1074F. doi:10.1126/science.1199115. PMC 4153607. PMID 21350178.
    • Francie Diep (28 February 2011). "Genetically altered fungus designed to attack malaria in mosquitoes". Scientific American.
  139. ^ Hokanson KE, Dawson WO, Handler AM, Schetelig MF, St Leger RJ (December 2014). "Not all GMOs are crop plants: non-plant GMO applications in agriculture". Transgenic Research. 23 (6): 1057–68. doi:10.1007/s11248-013-9769-5. PMID 24242193. S2CID 255108053.
  140. ^ a b Zhao H, Lovett B, Fang W (1 January 2016). "Genetically Engineering Entomopathogenic Fungi". Advances in Genetics. 94: 137–63. doi:10.1016/bs.adgen.2015.11.001. ISBN 9780128046944. PMID 27131325.
  141. ^ Koenraadt CJ, Takken W (April 2011). "Viability of GM fungi crucial to malaria control". Science. 332 (6026): 175. Bibcode:2011Sci...332..175K. doi:10.1126/science.332.6026.175. PMID 21474739.
  142. ^ Waltz E (14 April 2016). "Gene-edited CRISPR mushroom escapes US regulation". Nature. 532 (7599): 293. Bibcode:2016Natur.532..293W. doi:10.1038/nature.2016.19754. PMID 27111611.
  143. ^ Charles D (15 April 2016). "Will Genetically 'Edited' Food Be Regulated? The Case of the Mushroom". All Things Considered. National Public Radio. Retrieved 17 December 2018.
  144. ^ Zimmer C (27 July 2018). "What Is a Genetically Modified Crop? A European Ruling Sows Confusion". The New York Times. Archived from the original on 2 January 2022. Retrieved 17 December 2018.
  145. ^ a b c Walter P, Roberts K, Raff M, Lewis J, Johnson A, Alberts B (2002). "Studying Gene Expression and Function". Molecular Biology of the Cell (4th ed.). Garland Science.
  146. ^ Ganapathi TR, Suprasanna P, Rao PS, Bapat VA (2004). "Tobacco (Nicotiana tabacum L.) — A Model System for Tissue Culture Interventions and Genetic Engineering". Indian Journal of Biotechnology. 3: 171–184.
  147. ^ Koszowski B, Goniewicz ML, Czogała J, Sobczak A (2007). [Genetically modified tobacco--chance or threat for smokers?] (PDF). Przeglad Lekarski (in Polish). 64 (10): 908–12. PMID 18409340. Archived from the original (PDF) on 23 January 2013.
  148. ^ Mou B, Scorza R (15 June 2011). Transgenic Horticultural Crops: Challenges and Opportunities. CRC Press. p. 104. ISBN 978-1-4200-9379-7.
  149. ^ Gepstein S, Horwitz BA (1995). "The impact of Arabidopsis research on plant biotechnology". Biotechnology Advances. 13 (3): 403–14. doi:10.1016/0734-9750(95)02003-l. PMID 14536094.
  150. ^ Holland CK, Jez JM (October 2018). "Arabidopsis: the original plant chassis organism". Plant Cell Reports. 37 (10): 1359–1366. doi:10.1007/s00299-018-2286-5. PMID 29663032. S2CID 253806270.
  151. ^ Jefferson RA, Kavanagh TA, Bevan MW (December 1987). "GUS fusions: beta-glucuronidase as a sensitive and versatile gene fusion marker in higher plants". The EMBO Journal. 6 (13): 3901–7. doi:10.1002/j.1460-2075.1987.tb02730.x. PMC 553867. PMID 3327686.
  152. ^ a b "Biotechnology in Ornamental Plants – Pocket K". www.isaaa.org. Retrieved 17 December 2018.
  153. ^ Chandler SF, Sanchez C (October 2012). "Genetic modification; the development of transgenic ornamental plant varieties". Plant Biotechnology Journal. 10 (8): 891–903. doi:10.1111/j.1467-7652.2012.00693.x. PMID 22537268.
  154. ^ Nosowitz D (15 September 2011). "Suntory Creates Mythical Blue (Or, Um, Lavender-ish) Rose". Popular Science. Retrieved 30 August 2012.
  155. ^ . The Japan Times. 11 September 2011. Archived from the original on 22 November 2012. Retrieved 30 August 2012.
  156. ^ "World's First 'Blue' Rose Soon Available in U.S". Wired. 14 September 2011.
  157. ^ Boehm (27 October 2009). . Bioeconomy in Baden-Württemberg. Archived from the original on 3 April 2019. Retrieved 17 December 2018.
  158. ^ a b Adams JM, Piovesan G, Strauss S, Brown S (1 August 2002). "The Case for Genetic Engineering of Native and Landscape Trees against Introduced Pests and Diseases". Conservation Biology. 16 (4): 874–79. doi:10.1046/j.1523-1739.2002.00523.x. S2CID 86697592.
  159. ^ Tripathi S, Suzuki J, Gonsalves D (2007). "Development of Genetically Engineered Resistant Papaya for papaya ringspot virus in a Timely Manner: A Comprehensive and Successful Approach". Development of genetically engineered resistant papaya for papaya ringspot virus in a timely manner: a comprehensive and successful approach. Methods in Molecular Biology. Vol. 354. pp. 197–240. doi:10.1385/1-59259-966-4:197. ISBN 978-1-59259-966-0. PMID 17172756.
  160. ^ a b c Qaim M (29 April 2016). "Introduction". Genetically Modified Crops and Agricultural Development. Springer. pp. 1–10. ISBN 978-1-137-40572-2.
  161. ^ a b "Global Status of Commercialized Biotech/GM Crops: 2014 – ISAAA Brief 49-2014". ISAAA.org. Retrieved 15 September 2016.
  162. ^ a b ISAAA 2013 Annual Report Executive Summary, Global Status of Commercialized Biotech/GM Crops: 2013 ISAAA Brief 46-2013, Retrieved 6 August 2014
  163. ^ Hakim D (29 October 2016). "Doubts About the Promised Bounty of Genetically Modified Crops". The New York Times. ISSN 0362-4331. Archived from the original on 2 January 2022. Retrieved 5 May 2017.
  164. ^ Areal FJ, Riesgo L, Rodríguez-Cerezo E (February 2013). "Economic and agronomic impact of commercialized GM crops: a meta-analysis". The Journal of Agricultural Science. 151 (1): 7–33. doi:10.1017/S0021859612000111. ISSN 0021-8596. S2CID 85891950.
  165. ^ Finger R, El Benni N, Kaphengst T, Evans C, Herbert S, Lehmann B, et al. (10 May 2011). "A Meta Analysis on Farm-Level Costs and Benefits of GM Crops". Sustainability. 3 (5): 743–762. doi:10.3390/su3050743. hdl:20.500.11850/42242.
  166. ^ Klümper W, Qaim M (3 November 2014). "A meta-analysis of the impacts of genetically modified crops". PLOS ONE. 9 (11): e111629. Bibcode:2014PLoSO...9k1629K. doi:10.1371/journal.pone.0111629. PMC 4218791. PMID 25365303.
  167. ^ Darmency H (August 2013). "Pleiotropic effects of herbicide-resistance genes on crop yield: a review". Pest Management Science. 69 (8): 897–904. doi:10.1002/ps.3522. PMID 23457026.
  168. ^ Green JM (September 2014). "Current state of herbicides in herbicide-resistant crops". Pest Management Science. 70 (9): 1351–7. doi:10.1002/ps.3727. PMID 24446395.
  169. ^ Fleischer SJ, Hutchison WD, Naranjo SE (2014). "Sustainable Management of Insect-Resistant Crops". Plant Biotechnology. pp. 115–127. doi:10.1007/978-3-319-06892-3_10. ISBN 978-3-319-06891-6.
  170. ^ "SGK321". GM Approval Database. ISAAA.org. Retrieved 27 April 2017.
  171. ^ Qiu J (October 2008). "Is China ready for GM rice?". Nature. 455 (7215): 850–2. doi:10.1038/455850a. PMID 18923484.
  172. ^ Frist B (21 November 2006). "'Green revolution' hero". The Washington Times. One existing crop, genetically engineered 'golden rice' that produces vitamin A, already holds enormous promise for reducing blindness and dwarfism that result from a vitamin-A deficient diet.
  173. ^ Black RE, Allen LH, Bhutta ZA, Caulfield LE, de Onis M, Ezzati M, Mathers C, Rivera J (January 2008). "Maternal and child undernutrition: global and regional exposures and health consequences". Lancet. 371 (9608): 243–60. doi:10.1016/S0140-6736(07)61690-0. PMID 18207566. S2CID 3910132.
  174. ^ Humphrey JH, West KP, Sommer A (1992). "Vitamin A deficiency and attributable mortality among under-5-year-olds". Bulletin of the World Health Organization. 70 (2): 225–32. PMC 2393289. PMID 1600583.
  175. ^ Paine JA, Shipton CA, Chaggar S, Howells RM, Kennedy MJ, Vernon G, Wright SY, Hinchliffe E, Adams JL, Silverstone AL, Drake R (April 2005). "Improving the nutritional value of Golden Rice through increased pro-vitamin A content". Nature Biotechnology. 23 (4): 482–7. doi:10.1038/nbt1082. PMID 15793573. S2CID 632005.
  176. ^ "US FDA approves GMO Golden Rice as safe to eat". Genetic Literacy Project. 29 May 2018. Retrieved 30 May 2018.
  177. ^ Gasdaska JR, Spencer D, Dickey L (March 2003). "Advantages of Therapeutic Protein Production in the Aquatic Plant Lemna". BioProcessing Journal. 2 (2): 49–56. doi:10.12665/J22.Gasdaska.[permanent dead link]
  178. ^ (10 December 2012) "Engineering algae to make complex anti-cancer 'designer' drug". PhysOrg, Retrieved 15 April 2013
  179. ^ Büttner-Mainik A, Parsons J, Jérôme H, Hartmann A, Lamer S, Schaaf A, Schlosser A, Zipfel PF, Reski R, Decker EL (April 2011). "Production of biologically active recombinant human factor H in Physcomitrella". Plant Biotechnology Journal. 9 (3): 373–83. doi:10.1111/j.1467-7652.2010.00552.x. PMID 20723134.
  180. ^ Baur A, Reski R, Gorr G (May 2005). "Enhanced recovery of a secreted recombinant human growth factor using stabilizing additives and by co-expression of human serum albumin in the moss Physcomitrella patens". Plant Biotechnology Journal. 3 (3): 331–40. doi:10.1111/j.1467-7652.2005.00127.x. PMID 17129315.
  181. ^ a b Hammond J, McGarvey P, Yusibov V (6 December 2012). Plant Biotechnology: New Products and Applications. Springer Science & Business Media. pp. 7–8. ISBN 978-3-642-60234-4.
  182. ^ Börnke F, Broer I (June 2010). "Tailoring plant metabolism for the production of novel polymers and platform chemicals". Current Opinion in Plant Biology. 13 (3): 354–62. doi:10.1016/j.pbi.2010.01.005. PMID 20171137.
  183. ^ Lehr F, Posten C (June 2009). "Closed photo-bioreactors as tools for biofuel production". Current Opinion in Biotechnology. 20 (3): 280–5. doi:10.1016/j.copbio.2009.04.004. PMID 19501503.
  184. ^ "UNL's AgBiosafety for Educators". agbiosafety.unl.edu. Retrieved 18 December 2018.
  185. ^ . Protalix Biotherapeutics. Archived from the original on 27 October 2012.
  186. ^ Gali Weinreb and Koby Yeshayahou for Globes 2 May 2012. "FDA approves Protalix Gaucher treatment". 29 May 2013 at the Wayback Machine
  187. ^ Concha C, Cañas R, Macuer J, Torres MJ, Herrada AA, Jamett F, Ibáñez C (May 2017). "Disease Prevention: An Opportunity to Expand Edible Plant-Based Vaccines?". Vaccines. 5 (2): 14. doi:10.3390/vaccines5020014. PMC 5492011. PMID 28556800.
  188. ^ Kovak, Emma; Qaim, Matin; Blaustein-Rejto, Dan (10 February 2021). "The climate benefits of yield increases in genetically engineered crops". bioRxiv 10.1101/2021.02.10.430488.
  189. ^ a b Forabosco F, Löhmus M, Rydhmer L, Sundström LF (May 2013). "Genetically modified farm animals and fish in agriculture: A review". Livestock Science. 153 (1–3): 1–9. doi:10.1016/j.livsci.2013.01.002.
  190. ^ "The Superpowers of Genetically Modified Pigs". The Scientist. Retrieved 5 February 2019.
  191. ^ Rudinko, Larisa (20). Guidance for industry. USA: Center for veterinary medicine Link.
  192. ^ Murray, Joo (20). Genetically modified animals 13 October 2019 at the Wayback Machine. Canada: Brainwaving
  193. ^ "How CRISPR is Spreading Through the Animal Kingdom". www.pbs.org. 23 May 2018. Retrieved 20 December 2018.
  194. ^ a b Perleberg C, Kind A, Schnieke A (January 2018). "Genetically engineered pigs as models for human disease". Disease Models & Mechanisms. 11 (1). doi:10.1242/dmm.030783. PMC 5818075. PMID 29419487.
  195. ^ Sato K, Sasaki E (February 2018). "Genetic engineering in nonhuman primates for human disease modeling". Journal of Human Genetics. 63 (2): 125–131. doi:10.1038/s10038-017-0351-5. PMC 8075926. PMID 29203824.
  196. ^ Sasaki E, Suemizu H, Shimada A, Hanazawa K, Oiwa R, Kamioka M, Tomioka I, Sotomaru Y, Hirakawa R, Eto T, Shiozawa S, Maeda T, Ito M, Ito R, Kito C, Yagihashi C, Kawai K, Miyoshi H, Tanioka Y, Tamaoki N, Habu S, Okano H, Nomura T (May 2009). "Generation of transgenic non-human primates with germline transmission". Nature. 459 (7246): 523–7. Bibcode:2009Natur.459..523S. doi:10.1038/nature08090. PMID 19478777. S2CID 4404433.
  197. ^ Schatten G, Mitalipov S (May 2009). "Developmental biology: Transgenic primate offspring". Nature. 459 (7246): 515–6. Bibcode:2009Natur.459..515S. doi:10.1038/459515a. PMC 2777739. PMID 19478771.
  198. ^ Cyranoski D (May 2009). "Marmoset model takes centre stage". Nature. 459 (7246): 492. doi:10.1038/459492a. PMID 19478751.
  199. ^ Britt Erickson, 10 February 2009, for Chemical & Engineering News. FDA Approves Drug From Transgenic Goat Milk Accessed 6 October 2012
  200. ^ Spencer LT, Humphries JE, Brantly ML (May 2005). "Antibody response to aerosolized transgenic human alpha1-antitrypsin". The New England Journal of Medicine. 352 (19): 2030–1. doi:10.1056/nejm200505123521923. PMID 15888711.
  201. ^ Zimmer C (15 October 2015). "Editing of Pig DNA May Lead to More Organs for People l". The New York Times. Archived from the original on 2 January 2022.
  202. ^ Zeyland J, Gawrońska B, Juzwa W, Jura J, Nowak A, Słomski R, Smorąg Z, Szalata M, Woźniak A, Lipiński D (August 2013). "Transgenic pigs designed to express human α-galactosidase to avoid humoral xenograft rejection". Journal of Applied Genetics. 54 (3): 293–303. doi:10.1007/s13353-013-0156-y. PMC 3720986. PMID 23780397.
  203. ^ "Pig Heart Transplants For Humans Could Be on Their Way". IFLScience. 30 April 2014.
  204. ^ Reardon S (November 2015). "New life for pig-to-human transplants". Nature. 527 (7577): 152–4. Bibcode:2015Natur.527..152R. doi:10.1038/527152a. PMID 26560282.
  205. ^ "Genetically modified pig lungs or lab-grown lungs: Which is the future of our organ supply?". Genetic Literacy Project. 6 May 2014.
  206. ^ Wu J, Platero-Luengo A, Sakurai M, Sugawara A, Gil MA, Yamauchi T, Suzuki K, Bogliotti YS, Cuello C, Morales Valencia M, Okumura D, Luo J, Vilariño M, Parrilla I, Soto DA, Martinez CA, Hishida T, Sánchez-Bautista S, Martinez-Martinez ML, Wang H, Nohalez A, Aizawa E, Martinez-Redondo P, Ocampo A, Reddy P, Roca J, Maga EA, Esteban CR, Berggren WT, Nuñez Delicado E, Lajara J, Guillen I, Guillen P, Campistol JM, Martinez EA, Ross PJ, Izpisua Belmonte JC (January 2017). "Interspecies Chimerism with Mammalian Pluripotent Stem Cells". Cell. 168 (3): 473–486.e15. doi:10.1016/j.cell.2016.12.036. PMC 5679265. PMID 28129541.
  207. ^ Lai L, Kang JX, Li R, Wang J, Witt WT, Yong HY, Hao Y, Wax DM, Murphy CN, Rieke A, Samuel M, Linville ML, Korte SW, Evans RW, Starzl TE, Prather RS, Dai Y (April 2006). "Generation of cloned transgenic pigs rich in omega-3 fatty acids". Nature Biotechnology. 24 (4): 435–6. doi:10.1038/nbt1198. PMC 2976610. PMID 16565727.
  208. ^ Tucker I (24 June 2018). "Genetically modified animals". The Guardian. ISSN 0261-3077. Retrieved 21 December 2018.
  209. ^ Zyga L (2010). . Phys.org. Archived from the original on 30 April 2015.
  210. ^ a b . Canada: University of Guelph. 2010. Archived from the original on 30 January 2016.
  211. ^ Schimdt S (22 June 2012). "Genetically engineered pigs killed after funding ends". Postmedia News. Retrieved 31 July 2012.
  212. ^ . Canada: University of Guelph. Archived from the original on 27 February 2010. Retrieved 8 March 2010.
  213. ^ Gray R (2011). . Archived from the original on 4 April 2011.
  214. ^ . Classical Medicine Journal. 14 April 2010. Archived from the original on 6 November 2014.
  215. ^ Yapp R (11 June 2011). "Scientists create cow that produces 'human' milk". The Daily Telegraph. London. Retrieved 15 June 2012.
  216. ^ Jabed A, Wagner S, McCracken J, Wells DN, Laible G (October 2012). "Targeted microRNA expression in dairy cattle directs production of β-lactoglobulin-free, high-casein milk". Proceedings of the National Academy of Sciences of the United States of America. 109 (42): 16811–6. Bibcode:2012PNAS..10916811J. doi:10.1073/pnas.1210057109. PMC 3479461. PMID 23027958.
  217. ^ "Green fluorescent protein takes Nobel prize". Lewis Brindley. Retrieved 31 May 2015.
  218. ^ Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2002). "Studying Gene Expression and Function". Molecular Biology of the Cell (4th ed.). Garland Science.
  219. ^ Randall S (2008). (PDF). Biotechnology and Genetic Engineering Reviews. 25: 245–66. doi:10.7313/upo9781904761679.011. PMID 21412358. Archived from the original (PDF) on 26 March 2014.
  220. ^ Wongsrikeao P, Saenz D, Rinkoski T, Otoi T, Poeschla E (September 2011). "Antiviral restriction factor transgenesis in the domestic cat". Nature Methods. 8 (10): 853–9. doi:10.1038/nmeth.1703. PMC 4006694. PMID 21909101.
  221. ^ Staff (3 April 2012). . National Institute of Allergy and Infectious Diseases. Archived from the original on 11 April 2014.
  222. ^ Biello D. "Ancient DNA Could Return Passenger Pigeons to the Sky". Scientific American. Retrieved 23 December 2018.
  223. ^ Sarchet P. "Can we grow woolly mammoths in the lab? George Church hopes so". New Scientist. Retrieved 23 December 2018.
  224. ^ Hawks J (19 February 2017). "How mammoth cloning became fake news". John Hawks. Retrieved 20 January 2019.
  225. ^ Shapiro B (November 2015). "Mammoth 2.0: will genome engineering resurrect extinct species?". Genome Biology. 16 (1): 228. doi:10.1186/s13059-015-0800-4. PMC 4632474. PMID 26530525.
  226. ^ Selkirk SM (October 2004). "Gene therapy in clinical medicine". Postgraduate Medical Journal. 80 (948): 560–70. doi:10.1136/pgmj.2003.017764. PMC 1743106. PMID 15466989.
  227. ^ Cavazzana-Calvo M, Fischer A (June 2007). "Gene therapy for severe combined immunodeficiency: are we there yet?". The Journal of Clinical Investigation. 117 (6): 1456–65. doi:10.1172/JCI30953. PMC 1878528. PMID 17549248.
  228. ^ Richards S (6 November 2012). "Gene therapy arrives in Europe". The Scientist.
  229. ^ Rosenecker J, Huth S, Rudolph C (October 2006). "Gene therapy for cystic fibrosis lung disease: current status and future perspectives". Current Opinion in Molecular Therapeutics. 8 (5): 439–45. PMID 17078386.
  230. ^ Persons DA, Nienhuis AW (July 2003). "Gene therapy for the hemoglobin disorders". Current Hematology Reports. 2 (4): 348–55. PMID 12901333.
  231. ^ LeWitt PA, Rezai AR, Leehey MA, Ojemann SG, Flaherty AW, Eskandar EN, et al. (April 2011). "AAV2-GAD gene therapy for advanced Parkinson's disease: a double-blind, sham-surgery controlled, randomized trial". The Lancet. Neurology. 10 (4): 309–19. doi:10.1016/S1474-4422(11)70039-4. PMID 21419704. S2CID 37154043.
  232. ^ Gallaher, James (17 March 2011). "Gene therapy 'treats' Parkinson's disease". BBC News Health. Retrieved 24 April 2011
  233. ^ Urbina Z (12 February 2013). . United Academics. Archived from the original on 16 February 2013. Retrieved 15 February 2013.
  234. ^ "Treatment for Leukemia Is Showing Early Promise". The New York Times. Associated Press. 11 August 2011. p. A15. Retrieved 21 January 2013.
  235. ^ Coghlan A (26 March 2013). "Gene therapy cures leukaemia in eight days". New Scientist. Retrieved 15 April 2013.
  236. ^ "Gene therapy cures diabetic dogs". New Scientist. 13 February 2013. Retrieved 15 February 2013.
  237. ^ "New gene therapy trial gives hope to people with heart failure". British Heart Foundation. 30 April 2013. Retrieved 5 May 2013.
  238. ^ Foster K, Foster H, Dickson JG (December 2006). "Gene therapy progress and prospects: Duchenne muscular dystrophy". Gene Therapy. 13 (24): 1677–85. doi:10.1038/sj.gt.3302877. PMID 17066097.
  239. ^ . 5 August 2001. Archived from the original on 5 August 2001.
  240. ^ Smith KR, Chan S, Harris J (October 2012). "Human germline genetic modification: scientific and bioethical perspectives". Archives of Medical Research. 43 (7): 491–513. doi:10.1016/j.arcmed.2012.09.003. PMID 23072719.
  241. ^ Kolata G (23 April 2015). "Chinese Scientists Edit Genes of Human Embryos, Raising Concerns". The New York Times. Archived from the original on 2 January 2022. Retrieved 24 April 2015.
  242. ^ Liang P, Xu Y, Zhang X, Ding C, Huang R, Zhang Z, Lv J, Xie X, Chen Y, Li Y, Sun Y, Bai Y, Songyang Z, Ma W, Zhou C, Huang J (May 2015). "CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes". Protein & Cell. 6 (5): 363–372. doi:10.1007/s13238-015-0153-5. PMC 4417674. PMID 25894090.
  243. ^ Begley S (28 November 2018). "Amid uproar, Chinese scientist defends creating gene-edited babies – STAT". STAT.
  244. ^ Wang Q, Tan X, Jiao S, You F, Zhang PJ (24 July 2014). "Analyzing cold tolerance mechanism in transgenic zebrafish (Danio rerio)". PLOS ONE. 9 (7): e102492. Bibcode:2014PLoSO...9j2492W. doi:10.1371/journal.pone.0102492. PMC 4109919. PMID 25058652.
  245. ^ "Half of Fish Consumed Globally Is Now Raised on Farms, Study Finds". ScienceDaily. Retrieved 21 December 2018.
  246. ^ Tonelli FM, Lacerda SM, Tonelli FC, Costa GM, de França LR, Resende RR (November 2017). "Progress and biotechnological prospects in fish transgenesis". Biotechnology Advances. 35 (6): 832–844. doi:10.1016/j.biotechadv.2017.06.002. PMID 28602961.
  247. ^ Nebert DW, Stuart GW, Solis WA, Carvan MJ (January 2002). "Use of reporter genes and vertebrate DNA motifs in transgenic zebrafish as sentinels for assessing aquatic pollution". Environmental Health Perspectives. 110 (1): A15. doi:10.1289/ehp.110-1240712. PMC 1240712. PMID 11813700.
  248. ^ Mattingly CJ, McLachlan JA, Toscano WA (August 2001). "Green fluorescent protein (GFP) as a marker of aryl hydrocarbon receptor (AhR) function in developing zebrafish (Danio rerio)". Environmental Health Perspectives. 109 (8): 845–849. doi:10.1289/ehp.01109845. PMC 1240414. PMID 11564622.
  249. ^ Hallerman E (June 2004). "Glofish, the first GM animal commercialized: profits amid controversy". ISB News Report.
  250. ^ Hackett PB, Ekker SE, Essner JJ (2004). "Chapter 16: Applications of transposable elements in fish for transgenesis and functional genomics". In Gong Z, Korzh V (eds.). Fish Development and Genetics. World Scientific, Inc. pp. 532–80.
  251. ^ Meyers JR (2018). "Zebrafish: Development of a Vertebrate Model Organism". Current Protocols in Essential Laboratory Techniques. 16 (1): e19. doi:10.1002/cpet.19.
  252. ^ Lu JW, Ho YJ, Ciou SC, Gong Z (September 2017). "Innovative Disease Model: Zebrafish as an In Vivo Platform for Intestinal Disorder and Tumors". Biomedicines. 5 (4): 58. doi:10.3390/biomedicines5040058. PMC 5744082. PMID 28961226.
  253. ^ Barriuso J, Nagaraju R, Hurlstone A (March 2015). "Zebrafish: a new companion for translational research in oncology". Clinical Cancer Research. 21 (5): 969–75. doi:10.1158/1078-0432.CCR-14-2921. PMC 5034890. PMID 25573382.
  254. ^ Burket CT, Montgomery JE, Thummel R, Kassen SC, LaFave MC, Langenau DM, et al. (April 2008). "Generation and characterization of transgenic zebrafish lines using different ubiquitous promoters". Transgenic Research. 17 (2): 265–79. doi:10.1007/s11248-007-9152-5. PMC 3660017. PMID 17968670.
  255. ^ Du SJ, Gong Z, Fletcher GL, Shears MA, King MJ, Idler DR, Hew CL (1992). "Growth Enhancement in Transgenic Atlantic Salmon by the Use of an 'All Fish' Chimeric Growth Hormone Gene Construct". Nature Biotechnology. 10 (2): 176–181. doi:10.1038/nbt0292-176. PMID 1368229. S2CID 27048646.
  256. ^ Devlin RH, Biagi CA, Yesaki TY, Smailus DE, Byatt JC (February 2001). "Growth of domesticated transgenic fish". Nature. 409 (6822): 781–782. Bibcode:2001Natur.409..781D. doi:10.1038/35057314. PMID 11236982. S2CID 5293883.
  257. ^ Rahman MA, et al. (2001). "Growth and nutritional trials on transgenic Nile tilapia containing an exogenous fish growth hormone gene". Journal of Fish Biology. 59 (1): 62–78. doi:10.1111/j.1095-8649.2001.tb02338.x.
  258. ^ Pollack A (21 December 2012). "Engineered Fish Moves a Step Closer to Approval". The New York Times. Archived from the original on 2 January 2022.
genetically, modified, organism, redirects, here, other, uses, disambiguation, genetically, modified, organism, organism, whose, genetic, material, been, altered, using, genetic, engineering, techniques, exact, definition, genetically, modified, organism, what. GMO redirects here For other uses see GMO disambiguation A genetically modified organism GMO is any organism whose genetic material has been altered using genetic engineering techniques The exact definition of a genetically modified organism and what constitutes genetic engineering varies with the most common being an organism altered in a way that does not occur naturally by mating and or natural recombination 1 A wide variety of organisms have been genetically modified GM including animals plants and microorganisms Genetic modification can include the introduction of new genes or enhancing altering or knocking out endogenous genes In some genetic modifications genes are transferred within the same species across species creating transgenic organisms and even across kingdoms Creating a genetically modified organism is a multi step process Genetic engineers must isolate the gene they wish to insert into the host organism and combine it with other genetic elements including a promoter and terminator region and often a selectable marker A number of techniques are available for inserting the isolated gene into the host genome Recent advancements using genome editing techniques notably CRISPR have made the production of GMOs much simpler Herbert Boyer and Stanley Cohen made the first genetically modified organism in 1973 a bacterium resistant to the antibiotic kanamycin The first genetically modified animal a mouse was created in 1974 by Rudolf Jaenisch and the first plant was produced in 1983 In 1994 the Flavr Savr tomato was released the first commercialized genetically modified food The first genetically modified animal to be commercialized was the GloFish 2003 and the first genetically modified animal to be approved for food use was the AquAdvantage salmon in 2015 Bacteria are the easiest organisms to engineer and have been used for research food production industrial protein purification including drugs agriculture and art There is potential to use them for environmental purposes or as medicine Fungi have been engineered with much the same goals Viruses play an important role as vectors for inserting genetic information into other organisms This use is especially relevant to human gene therapy There are proposals to remove the virulent genes from viruses to create vaccines Plants have been engineered for scientific research to create new colors in plants deliver vaccines and to create enhanced crops Genetically modified crops are publicly the most controversial GMOs in spite of having the most human health and environmental benefits 2 The majority are engineered for herbicide tolerance or insect resistance Golden rice has been engineered with three genes that increase its nutritional value Other prospects for GM crops are as bioreactors for the production of biopharmaceuticals biofuels or medicines Animals are generally much harder to transform and the vast majority are still at the research stage Mammals are the best model organisms for humans making ones genetically engineered to resemble serious human diseases important to the discovery and development of treatments Human proteins expressed in mammals are more likely to be similar to their natural counterparts than those expressed in plants or microorganisms Livestock is modified with the intention of improving economically important traits such as growth rate quality of meat milk composition disease resistance and survival Genetically modified fish are used for scientific research as pets and as a food source Genetic engineering has been proposed as a way to control mosquitos a vector for many deadly diseases Although human gene therapy is still relatively new it has been used to treat genetic disorders such as severe combined immunodeficiency and Leber s congenital amaurosis Many objections have been raised over the development of GMOs particularly their commercialization Many of these involve GM crops and whether food produced from them is safe and what impact growing them will have on the environment Other concerns are the objectivity and rigor of regulatory authorities contamination of non genetically modified food control of the food supply patenting of life and the use of intellectual property rights Although there is a scientific consensus that currently available food derived from GM crops poses no greater risk to human health than conventional food GM food safety is a leading issue with critics Gene flow impact on non target organisms and escape are the major environmental concerns Countries have adopted regulatory measures to deal with these concerns There are differences in the regulation for the release of GMOs between countries with some of the most marked differences occurring between the US and Europe Key issues concerning regulators include whether GM food should be labeled and the status of gene edited organisms Contents 1 Definition 2 Production 3 History 4 Bacteria 5 Viruses 6 Fungi 7 Plants 7 1 Crops 8 Animals 8 1 Mammals 8 1 1 Humans 8 2 Fish 8 3 Insects 8 4 Other 9 Regulation 10 Controversy 11 References 12 External linksDefinitionThe definition of a genetically modified organism GMO is not clear and varies widely between countries international bodies and other communities At its broadest the definition of a GMO can include anything that has had its genes altered including by nature 3 4 Taking a less broad view it can encompass every organism that has had its genes altered by humans which would include all crops and livestock In 1993 the Encyclopedia Britannica defined genetic engineering as any of a wide range of techniques among them artificial insemination in vitro fertilization e g test tube babies sperm banks cloning and gene manipulation 5 The European Union EU included a similarly broad definition in early reviews specifically mentioning GMOs being produced by selective breeding and other means of artificial selection 6 These definitions were promptly adjusted with a number of exceptions added as the result of pressure from scientific and farming communities as well as developments in science The EU definition later excluded traditional breeding in vitro fertilization induction of polyploidy mutation breeding and cell fusion techniques that do not use recombinant nucleic acids or a genetically modified organism in the process 7 8 9 Another approach was the definition provided by the Food and Agriculture Organization the World Health Organization and the European Commission stating that the organisms must be altered in a way that does not occur naturally by mating and or natural recombination 10 11 12 Progress in science such as the discovery of horizontal gene transfer being a relatively common natural phenomenon further added to the confusion on what occurs naturally which led to further adjustments and exceptions 13 There are examples of crops that fit this definition but are not normally considered GMOs 14 For example the grain crop triticale was fully developed in a laboratory in 1930 using various techniques to alter its genome 15 Genetically engineered organism GEO can be considered a more precise term compared to GMO when describing organisms genomes that have been directly manipulated with biotechnology 16 8 The Cartagena Protocol on Biosafety used the synonym living modified organism LMO in 2000 and defined it as any living organism that possesses a novel combination of genetic material obtained through the use of modern biotechnology 17 Modern biotechnology is further defined as In vitro nucleic acid techniques including recombinant deoxyribonucleic acid DNA and direct injection of nucleic acid into cells or organelles or fusion of cells beyond the taxonomic family 18 Originally the term GMO was not commonly used by scientists to describe genetically engineered organisms until after usage of GMO became common in popular media 19 The United States Department of Agriculture USDA considers GMOs to be plants or animals with heritable changes introduced by genetic engineering or traditional methods while GEO specifically refers to organisms with genes introduced eliminated or rearranged using molecular biology particularly recombinant DNA techniques such as transgenesis 20 The definitions focus on the process more than the product which means there could be GMOS and non GMOs with very similar genotypes and phenotypes 21 22 This has led scientists to label it as a scientifically meaningless category 23 saying that it is impossible to group all the different types of GMOs under one common definition 24 It has also caused issues for organic institutions and groups looking to ban GMOs 25 26 It also poses problems as new processes are developed The current definitions came in before genome editing became popular and there is some confusion as to whether they are GMOs The EU has adjudged that they are 27 changing their GMO definition to include organisms obtained by mutagenesis but has excluded them from regulation based on their long safety record and that they have been conventionally been used in a number of applications 9 In contrast the USDA has ruled that gene edited organisms are not considered GMOs 28 Even greater inconsistency and confusion is associated with various Non GMO or GMO free labeling schemes in food marketing where even products such as water or salt which do not contain any organic substances and genetic material and thus cannot be genetically modified by definition are being labeled to create an impression of being more healthy 29 30 31 ProductionMain article Genetic engineering techniques nbsp A gene gun uses biolistics to insert DNA into plant tissue Creating a genetically modified organism GMO is a multi step process Genetic engineers must isolate the gene they wish to insert into the host organism This gene can be taken from a cell 32 or artificially synthesized 33 If the chosen gene or the donor organism s genome has been well studied it may already be accessible from a genetic library The gene is then combined with other genetic elements including a promoter and terminator region and a selectable marker 34 A number of techniques are available for inserting the isolated gene into the host genome Bacteria can be induced to take up foreign DNA usually by exposed heat shock or electroporation 35 DNA is generally inserted into animal cells using microinjection where it can be injected through the cell s nuclear envelope directly into the nucleus or through the use of viral vectors 36 In plants the DNA is often inserted using Agrobacterium mediated recombination 37 38 biolistics 39 or electroporation As only a single cell is transformed with genetic material the organism must be regenerated from that single cell In plants this is accomplished through tissue culture 40 41 In animals it is necessary to ensure that the inserted DNA is present in the embryonic stem cells 37 Further testing using PCR Southern hybridization and DNA sequencing is conducted to confirm that an organism contains the new gene 42 Traditionally the new genetic material was inserted randomly within the host genome Gene targeting techniques which creates double stranded breaks and takes advantage on the cells natural homologous recombination repair systems have been developed to target insertion to exact locations Genome editing uses artificially engineered nucleases that create breaks at specific points There are four families of engineered nucleases meganucleases 43 44 zinc finger nucleases 45 46 transcription activator like effector nucleases TALENs 47 48 and the Cas9 guideRNA system adapted from CRISPR 49 50 TALEN and CRISPR are the two most commonly used and each has its own advantages 51 TALENs have greater target specificity while CRISPR is easier to design and more efficient 51 HistoryMain article History of genetic engineering nbsp Herbert Boyer pictured and Stanley Cohen created the first genetically modified organism in 1973 Humans have domesticated plants and animals since around 12 000 BCE using selective breeding or artificial selection as contrasted with natural selection 52 25 The process of selective breeding in which organisms with desired traits and thus with the desired genes are used to breed the next generation and organisms lacking the trait are not bred is a precursor to the modern concept of genetic modification 53 1 54 1 Various advancements in genetics allowed humans to directly alter the DNA and therefore genes of organisms In 1972 Paul Berg created the first recombinant DNA molecule when he combined DNA from a monkey virus with that of the lambda virus 55 56 Herbert Boyer and Stanley Cohen made the first genetically modified organism in 1973 57 They took a gene from a bacterium that provided resistance to the antibiotic kanamycin inserted it into a plasmid and then induced other bacteria to incorporate the plasmid The bacteria that had successfully incorporated the plasmid was then able to survive in the presence of kanamycin 58 Boyer and Cohen expressed other genes in bacteria This included genes from the toad Xenopus laevis in 1974 creating the first GMO expressing a gene from an organism of a different kingdom 59 nbsp In 1974 Rudolf Jaenisch created the first genetically modified animal In 1974 Rudolf Jaenisch created a transgenic mouse by introducing foreign DNA into its embryo making it the world s first transgenic animal 60 61 However it took another eight years before transgenic mice were developed that passed the transgene to their offspring 62 63 Genetically modified mice were created in 1984 that carried cloned oncogenes predisposing them to developing cancer 64 Mice with genes removed termed a knockout mouse were created in 1989 The first transgenic livestock were produced in 1985 65 and the first animal to synthesize transgenic proteins in their milk were mice in 1987 66 The mice were engineered to produce human tissue plasminogen activator a protein involved in breaking down blood clots 67 In 1983 the first genetically engineered plant was developed by Michael W Bevan Richard B Flavell and Mary Dell Chilton They infected tobacco with Agrobacterium transformed with an antibiotic resistance gene and through tissue culture techniques were able to grow a new plant containing the resistance gene 68 The gene gun was invented in 1987 allowing transformation of plants not susceptible to Agrobacterium infection 69 In 2000 Vitamin A enriched golden rice was the first plant developed with increased nutrient value 70 In 1976 Genentech the first genetic engineering company was founded by Herbert Boyer and Robert Swanson a year later the company produced a human protein somatostatin in E coli Genentech announced the production of genetically engineered human insulin in 1978 71 The insulin produced by bacteria branded Humulin was approved for release by the Food and Drug Administration in 1982 72 In 1988 the first human antibodies were produced in plants 73 In 1987 a strain of Pseudomonas syringae became the first genetically modified organism to be released into the environment 74 when a strawberry and potato field in California were sprayed with it 75 The first genetically modified crop an antibiotic resistant tobacco plant was produced in 1982 76 China was the first country to commercialize transgenic plants introducing a virus resistant tobacco in 1992 77 In 1994 Calgene attained approval to commercially release the Flavr Savr tomato the first genetically modified food 78 Also in 1994 the European Union approved tobacco engineered to be resistant to the herbicide bromoxynil making it the first genetically engineered crop commercialized in Europe 79 An insect resistant Potato was approved for release in the US in 1995 80 and by 1996 approval had been granted to commercially grow 8 transgenic crops and one flower crop carnation in 6 countries plus the EU 81 In 2010 scientists at the J Craig Venter Institute announced that they had created the first synthetic bacterial genome They named it Synthia and it was the world s first synthetic life form 82 83 The first genetically modified animal to be commercialized was the GloFish a Zebra fish with a fluorescent gene added that allows it to glow in the dark under ultraviolet light 84 It was released to the US market in 2003 85 In 2015 AquAdvantage salmon became the first genetically modified animal to be approved for food use 86 Approval is for fish raised in Panama and sold in the US 86 The salmon were transformed with a growth hormone regulating gene from a Pacific Chinook salmon and a promoter from an ocean pout enabling it to grow year round instead of only during spring and summer 87 BacteriaMain article Genetically modified bacteria nbsp nbsp Left Bacteria transformed with pGLO under ambient lightRight Bacteria transformed with pGLO visualized under ultraviolet light Bacteria were the first organisms to be genetically modified in the laboratory due to the relative ease of modifying their chromosomes 88 This ease made them important tools for the creation of other GMOs Genes and other genetic information from a wide range of organisms can be added to a plasmid and inserted into bacteria for storage and modification Bacteria are cheap easy to grow clonal multiply quickly and can be stored at 80 C almost indefinitely Once a gene is isolated it can be stored inside the bacteria providing an unlimited supply for research 89 A large number of custom plasmids make manipulating DNA extracted from bacteria relatively easy 90 Their ease of use has made them great tools for scientists looking to study gene function and evolution The simplest model organisms come from bacteria with most of our early understanding of molecular biology coming from studying Escherichia coli 91 Scientists can easily manipulate and combine genes within the bacteria to create novel or disrupted proteins and observe the effect this has on various molecular systems Researchers have combined the genes from bacteria and archaea leading to insights on how these two diverged in the past 92 In the field of synthetic biology they have been used to test various synthetic approaches from synthesizing genomes to creating novel nucleotides 93 94 95 Bacteria have been used in the production of food for a long time and specific strains have been developed and selected for that work on an industrial scale They can be used to produce enzymes amino acids flavorings and other compounds used in food production With the advent of genetic engineering new genetic changes can easily be introduced into these bacteria Most food producing bacteria are lactic acid bacteria and this is where the majority of research into genetically engineering food producing bacteria has gone The bacteria can be modified to operate more efficiently reduce toxic byproduct production increase output create improved compounds and remove unnecessary pathways 96 Food products from genetically modified bacteria include alpha amylase which converts starch to simple sugars chymosin which clots milk protein for cheese making and pectinesterase which improves fruit juice clarity 97 The majority are produced in the US and even though regulations are in place to allow production in Europe as of 2015 no food products derived from bacteria are currently available there 98 Genetically modified bacteria are used to produce large amounts of proteins for industrial use The bacteria are generally grown to a large volume before the gene encoding the protein is activated The bacteria are then harvested and the desired protein purified from them 99 The high cost of extraction and purification has meant that only high value products have been produced at an industrial scale 100 The majority of these products are human proteins for use in medicine 101 Many of these proteins are impossible or difficult to obtain via natural methods and they are less likely to be contaminated with pathogens making them safer 99 The first medicinal use of GM bacteria was to produce the protein insulin to treat diabetes 102 Other medicines produced include clotting factors to treat hemophilia 103 human growth hormone to treat various forms of dwarfism 104 105 interferon to treat some cancers erythropoietin for anemic patients and tissue plasminogen activator which dissolves blood clots 99 Outside of medicine they have been used to produce biofuels 106 There is interest in developing an extracellular expression system within the bacteria to reduce costs and make the production of more products economical 100 With a greater understanding of the role that the microbiome plays in human health there is a potential to treat diseases by genetically altering the bacteria to themselves be therapeutic agents Ideas include altering gut bacteria so they destroy harmful bacteria or using bacteria to replace or increase deficient enzymes or proteins One research focus is to modify Lactobacillus bacteria that naturally provide some protection against HIV with genes that will further enhance this protection If the bacteria do not form colonies inside the patient the person must repeatedly ingest the modified bacteria in order to get the required doses Enabling the bacteria to form a colony could provide a more long term solution but could also raise safety concerns as interactions between bacteria and the human body are less well understood than with traditional drugs There are concerns that horizontal gene transfer to other bacteria could have unknown effects As of 2018 there are clinical trials underway testing the efficacy and safety of these treatments 107 For over a century bacteria have been used in agriculture Crops have been inoculated with Rhizobia and more recently Azospirillum to increase their production or to allow them to be grown outside their original habitat Application of Bacillus thuringiensis Bt and other bacteria can help protect crops from insect infestation and plant diseases With advances in genetic engineering these bacteria have been manipulated for increased efficiency and expanded host range Markers have also been added to aid in tracing the spread of the bacteria The bacteria that naturally colonize certain crops have also been modified in some cases to express the Bt genes responsible for pest resistance Pseudomonas strains of bacteria cause frost damage by nucleating water into ice crystals around themselves This led to the development of ice minus bacteria which have the ice forming genes removed When applied to crops they can compete with the non modified bacteria and confer some frost resistance 108 nbsp This artwork is made with bacteria modified to express 8 different colors of fluorescent proteins Other uses for genetically modified bacteria include bioremediation where the bacteria are used to convert pollutants into a less toxic form Genetic engineering can increase the levels of the enzymes used to degrade a toxin or to make the bacteria more stable under environmental conditions 109 Bioart has also been created using genetically modified bacteria In the 1980s artist Jon Davis and geneticist Dana Boyd converted the Germanic symbol for femininity ᛉ into binary code and then into a DNA sequence which was then expressed in Escherichia coli 110 This was taken a step further in 2012 when a whole book was encoded onto DNA 111 Paintings have also been produced using bacteria transformed with fluorescent proteins 110 VirusesMain article Genetically modified virus Viruses are often modified so they can be used as vectors for inserting genetic information into other organisms This process is called transduction and if successful the recipient of the introduced DNA becomes a GMO Different viruses have different efficiencies and capabilities Researchers can use this to control for various factors including the target location insert size and duration of gene expression Any dangerous sequences inherent in the virus must be removed while those that allow the gene to be delivered effectively are retained 112 While viral vectors can be used to insert DNA into almost any organism it is especially relevant for its potential in treating human disease Although primarily still at trial stages 113 there has been some successes using gene therapy to replace defective genes This is most evident in curing patients with severe combined immunodeficiency rising from adenosine deaminase deficiency ADA SCID 114 although the development of leukemia in some ADA SCID patients 115 along with the death of Jesse Gelsinger in a 1999 trial set back the development of this approach for many years 116 In 2009 another breakthrough was achieved when an eight year old boy with Leber s congenital amaurosis regained normal eyesight 116 and in 2016 GlaxoSmithKline gained approval to commercialize a gene therapy treatment for ADA SCID 114 As of 2018 there are a substantial number of clinical trials underway including treatments for hemophilia glioblastoma chronic granulomatous disease cystic fibrosis and various cancers 115 The most common virus used for gene delivery comes from adenoviruses as they can carry up to 7 5 kb of foreign DNA and infect a relatively broad range of host cells although they have been known to elicit immune responses in the host and only provide short term expression Other common vectors are adeno associated viruses which have lower toxicity and longer term expression but can only carry about 4kb of DNA 115 Herpes simplex viruses make promising vectors having a carrying capacity of over 30kb and providing long term expression although they are less efficient at gene delivery than other vectors 117 The best vectors for long term integration of the gene into the host genome are retroviruses but their propensity for random integration is problematic Lentiviruses are a part of the same family as retroviruses with the advantage of infecting both dividing and non dividing cells whereas retroviruses only target dividing cells Other viruses that have been used as vectors include alphaviruses flaviviruses measles viruses rhabdoviruses Newcastle disease virus poxviruses and picornaviruses 115 Most vaccines consist of viruses that have been attenuated disabled weakened or killed in some way so that their virulent properties are no longer effective Genetic engineering could theoretically be used to create viruses with the virulent genes removed This does not affect the viruses infectivity invokes a natural immune response and there is no chance that they will regain their virulence function which can occur with some other vaccines As such they are generally considered safer and more efficient than conventional vaccines although concerns remain over non target infection potential side effects and horizontal gene transfer to other viruses 118 Another potential approach is to use vectors to create novel vaccines for diseases that have no vaccines available or the vaccines that do not work effectively such as AIDS malaria and tuberculosis 119 The most effective vaccine against Tuberculosis the Bacillus Calmette Guerin BCG vaccine only provides partial protection A modified vaccine expressing a M tuberculosis antigen is able to enhance BCG protection 120 It has been shown to be safe to use at phase II trials although not as effective as initially hoped 121 Other vector based vaccines have already been approved and many more are being developed 119 Another potential use of genetically modified viruses is to alter them so they can directly treat diseases This can be through expression of protective proteins or by directly targeting infected cells In 2004 researchers reported that a genetically modified virus that exploits the selfish behavior of cancer cells might offer an alternative way of killing tumours 122 123 Since then several researchers have developed genetically modified oncolytic viruses that show promise as treatments for various types of cancer 124 125 126 127 128 In 2017 researchers genetically modified a virus to express spinach defensin proteins The virus was injected into orange trees to combat citrus greening disease that had reduced orange production by 70 since 2005 129 Natural viral diseases such as myxomatosis and rabbit hemorrhagic disease have been used to help control pest populations Over time the surviving pests become resistant leading researchers to look at alternative methods Genetically modified viruses that make the target animals infertile through immunocontraception have been created in the laboratory 130 as well as others that target the developmental stage of the animal 131 There are concerns with using this approach regarding virus containment 130 and cross species infection 132 Sometimes the same virus can be modified for contrasting purposes Genetic modification of the myxoma virus has been proposed to conserve European wild rabbits in the Iberian peninsula and to help regulate them in Australia To protect the Iberian species from viral diseases the myxoma virus was genetically modified to immunize the rabbits while in Australia the same myxoma virus was genetically modified to lower fertility in the Australian rabbit population 133 Outside of biology scientists have used a genetically modified virus to construct a lithium ion battery and other nanostructured materials It is possible to engineer bacteriophages to express modified proteins on their surface and join them up in specific patterns a technique called phage display These structures have potential uses for energy storage and generation biosensing and tissue regeneration with some new materials currently produced including quantum dots liquid crystals nanorings and nanofibres 134 The battery was made by engineering M13 bacteriaophages so they would coat themselves in iron phosphate and then assemble themselves along a carbon nanotube This created a highly conductive medium for use in a cathode allowing energy to be transferred quickly They could be constructed at lower temperatures with non toxic chemicals making them more environmentally friendly 135 FungiFungi can be used for many of the same processes as bacteria For industrial applications yeasts combine the bacterial advantages of being a single celled organism that is easy to manipulate and grow with the advanced protein modifications found in eukaryotes They can be used to produce large complex molecules for use in food pharmaceuticals hormones and steroids 136 Yeast is important for wine production and as of 2016 two genetically modified yeasts involved in the fermentation of wine have been commercialized in the United States and Canada One has increased malolactic fermentation efficiency while the other prevents the production of dangerous ethyl carbamate compounds during fermentation 96 There have also been advances in the production of biofuel from genetically modified fungi 137 Fungi being the most common pathogens of insects make attractive biopesticides Unlike bacteria and viruses they have the advantage of infecting the insects by contact alone although they are out competed in efficiency by chemical pesticides Genetic engineering can improve virulence usually by adding more virulent proteins 138 increasing infection rate or enhancing spore persistence 139 Many of the disease carrying vectors are susceptible to entomopathogenic fungi An attractive target for biological control are mosquitos vectors for a range of deadly diseases including malaria yellow fever and dengue fever Mosquitos can evolve quickly so it becomes a balancing act of killing them before the Plasmodium they carry becomes the infectious disease but not so fast that they become resistant to the fungi By genetically engineering fungi like Metarhizium anisopliae and Beauveria bassiana to delay the development of mosquito infectiousness the selection pressure to evolve resistance is reduced 140 Another strategy is to add proteins to the fungi that block transmission of malaria 140 or remove the Plasmodium altogether 141 Agaricus bisporus the common white button mushroom has been gene edited to resist browning giving it a longer shelf life The process used CRISPR to knock out a gene that encodes polyphenol oxidase As it didn t introduce any foreign DNA into the organism it was not deemed to be regulated under existing GMO frameworks and as such is the first CRISPR edited organism to be approved for release 142 This has intensified debates as to whether gene edited organisms should be considered genetically modified organisms 143 and how they should be regulated 144 PlantsMain article Genetically modified plant nbsp Tissue culture used to regenerate Arabidopsis thalianaPlants have been engineered for scientific research to display new flower colors deliver vaccines and to create enhanced crops Many plants are pluripotent meaning that a single cell from a mature plant can be harvested and under the right conditions can develop into a new plant This ability can be taken advantage of by genetic engineers by selecting for cells that have been successfully transformed in an adult plant a new plant can then be grown that contains the transgene in every cell through a process known as tissue culture 145 Much of the advances in the field of genetic engineering has come from experimentation with tobacco Major advances in tissue culture and plant cellular mechanisms for a wide range of plants has originated from systems developed in tobacco 146 It was the first plant to be altered using genetic engineering and is considered a model organism for not only genetic engineering but a range of other fields 147 As such the transgenic tools and procedures are well established making tobacco one of the easiest plants to transform 148 Another major model organism relevant to genetic engineering is Arabidopsis thaliana Its small genome and short life cycle makes it easy to manipulate and it contains many homologs to important crop species 149 It was the first plant sequenced has a host of online resources available and can be transformed by simply dipping a flower in a transformed Agrobacterium solution 150 In research plants are engineered to help discover the functions of certain genes The simplest way to do this is to remove the gene and see what phenotype develops compared to the wild type form Any differences are possibly the result of the missing gene Unlike mutagenisis genetic engineering allows targeted removal without disrupting other genes in the organism 145 Some genes are only expressed in certain tissues so reporter genes like GUS can be attached to the gene of interest allowing visualization of the location 151 Other ways to test a gene is to alter it slightly and then return it to the plant and see if it still has the same effect on phenotype Other strategies include attaching the gene to a strong promoter and see what happens when it is overexpressed forcing a gene to be expressed in a different location or at different developmental stages 145 nbsp Suntory blue roseSome genetically modified plants are purely ornamental They are modified for flower color fragrance flower shape and plant architecture 152 The first genetically modified ornamentals commercialized altered color 153 Carnations were released in 1997 with the most popular genetically modified organism a blue rose actually lavender or mauve created in 2004 154 The roses are sold in Japan the United States and Canada 155 156 Other genetically modified ornamentals include Chrysanthemum and Petunia 152 As well as increasing aesthetic value there are plans to develop ornamentals that use less water or are resistant to the cold which would allow them to be grown outside their natural environments 157 It has been proposed to genetically modify some plant species threatened by extinction to be resistant to invasive plants and diseases such as the emerald ash borer in North American and the fungal disease Ceratocystis platani in European plane trees 158 The papaya ringspot virus devastated papaya trees in Hawaii in the twentieth century until transgenic papaya plants were given pathogen derived resistance 159 However genetic modification for conservation in plants remains mainly speculative A unique concern is that a transgenic species may no longer bear enough resemblance to the original species to truly claim that the original species is being conserved Instead the transgenic species may be genetically different enough to be considered a new species thus diminishing the conservation worth of genetic modification 158 Crops Main article Genetically modified cropsSee also Genetically modified food nbsp Wild type peanut top and transgenic peanut with Bacillus thuringiensis gene added bottom exposed to cornstalk borer larvaGenetically modified crops are genetically modified plants that are used in agriculture The first crops developed were used for animal or human food and provide resistance to certain pests diseases environmental conditions spoilage or chemical treatments e g resistance to a herbicide The second generation of crops aimed to improve the quality often by altering the nutrient profile Third generation genetically modified crops could be used for non food purposes including the production of pharmaceutical agents biofuels and other industrially useful goods as well as for bioremediation 160 nbsp Kenyans examining insect resistant transgenic Bacillus thuringiensis Bt cornThere are three main aims to agricultural advancement increased production improved conditions for agricultural workers and sustainability GM crops contribute by improving harvests through reducing insect pressure increasing nutrient value and tolerating different abiotic stresses Despite this potential as of 2018 the commercialized crops are limited mostly to cash crops like cotton soybean maize and canola and the vast majority of the introduced traits provide either herbicide tolerance or insect resistance 160 Soybeans accounted for half of all genetically modified crops planted in 2014 161 Adoption by farmers has been rapid between 1996 and 2013 the total surface area of land cultivated with GM crops increased by a factor of 100 162 Geographically though the spread has been uneven with strong growth in the Americas and parts of Asia and little in Europe and Africa 160 Its socioeconomic spread has been more even with approximately 54 of worldwide GM crops grown in developing countries in 2013 162 Although doubts have been raised 163 most studies have found growing GM crops to be beneficial to farmers through decreased pesticide use as well as increased crop yield and farm profit 164 165 166 The majority of GM crops have been modified to be resistant to selected herbicides usually a glyphosate or glufosinate based one Genetically modified crops engineered to resist herbicides are now more available than conventionally bred resistant varieties 167 in the USA 93 of soybeans and most of the GM maize grown is glyphosate tolerant 168 Most currently available genes used to engineer insect resistance come from the Bacillus thuringiensis bacterium and code for delta endotoxins A few use the genes that encode for vegetative insecticidal proteins 169 The only gene commercially used to provide insect protection that does not originate from B thuringiensis is the Cowpea trypsin inhibitor CpTI CpTI was first approved for use cotton in 1999 and is currently undergoing trials in rice 170 171 Less than one percent of GM crops contained other traits which include providing virus resistance delaying senescence and altering the plants composition 161 nbsp Golden rice compared to white riceGolden rice is the most well known GM crop that is aimed at increasing nutrient value It has been engineered with three genes that biosynthesise beta carotene a precursor of vitamin A in the edible parts of rice 70 It is intended to produce a fortified food to be grown and consumed in areas with a shortage of dietary vitamin A 172 a deficiency which each year is estimated to kill 670 000 children under the age of 5 173 and cause an additional 500 000 cases of irreversible childhood blindness 174 The original golden rice produced 1 6mg g of the carotenoids with further development increasing this 23 times 175 It gained its first approvals for use as food in 2018 176 Plants and plant cells have been genetically engineered for production of biopharmaceuticals in bioreactors a process known as pharming Work has been done with duckweed Lemna minor 177 the algae Chlamydomonas reinhardtii 178 and the moss Physcomitrella patens 179 180 Biopharmaceuticals produced include cytokines hormones antibodies enzymes and vaccines most of which are accumulated in the plant seeds Many drugs also contain natural plant ingredients and the pathways that lead to their production have been genetically altered or transferred to other plant species to produce greater volume 181 Other options for bioreactors are biopolymers 182 and biofuels 183 Unlike bacteria plants can modify the proteins post translationally allowing them to make more complex molecules They also pose less risk of being contaminated 184 Therapeutics have been cultured in transgenic carrot and tobacco cells 185 including a drug treatment for Gaucher s disease 186 Vaccine production and storage has great potential in transgenic plants Vaccines are expensive to produce transport and administer so having a system that could produce them locally would allow greater access to poorer and developing areas 181 As well as purifying vaccines expressed in plants it is also possible to produce edible vaccines in plants Edible vaccines stimulate the immune system when ingested to protect against certain diseases Being stored in plants reduces the long term cost as they can be disseminated without the need for cold storage don t need to be purified and have long term stability Also being housed within plant cells provides some protection from the gut acids upon digestion However the cost of developing regulating and containing transgenic plants is high leading to most current plant based vaccine development being applied to veterinary medicine where the controls are not as strict 187 Genetically modified crops have been proposed as one of the ways to reduce farming related CO2 emissions due to higher yield reduced use of pesticides reduced use of tractor fuel and no tillage According to a 2021 study in EU alone widespread adoption of GE crops would reduce greenhouse gas emissions by 33 million tons of CO2 equivalent or 7 5 of total farming related emissions 188 AnimalsMain article Genetically modified animal The vast majority of genetically modified animals are at the research stage with the number close to entering the market remaining small 189 As of 2018 only three genetically modified animals have been approved all in the USA A goat and a chicken have been engineered to produce medicines and a salmon has increased its own growth 190 Despite the differences and difficulties in modifying them the end aims are much the same as for plants GM animals are created for research purposes production of industrial or therapeutic products agricultural uses or improving their health There is also a market for creating genetically modified pets 191 Mammals Main article Genetically modified mammals nbsp Some chimeras like the blotched mouse shown are created through genetic modification techniques like gene targeting The process of genetically engineering mammals is slow tedious and expensive However new technologies are making genetic modifications easier and more precise 192 The first transgenic mammals were produced by injecting viral DNA into embryos and then implanting the embryos in females 60 The embryo would develop and it would be hoped that some of the genetic material would be incorporated into the reproductive cells Then researchers would have to wait until the animal reached breeding age and then offspring would be screened for the presence of the gene in every cell The development of the CRISPR Cas9 gene editing system as a cheap and fast way of directly modifying germ cells effectively halving the amount of time needed to develop genetically modified mammals 193 source source source source A porcine model of hemophilia AMammals are the best models for human disease making genetic engineered ones vital to the discovery and development of cures and treatments for many serious diseases Knocking out genes responsible for human genetic disorders allows researchers to study the mechanism of the disease and to test possible cures Genetically modified mice have been the most common mammals used in biomedical research as they are cheap and easy to manipulate Pigs are also a good target as they have a similar body size and anatomical features physiology pathophysiological response and diet 194 Nonhuman primates are the most similar model organisms to humans but there is less public acceptance towards using them as research animals 195 In 2009 scientists announced that they had successfully transferred a gene into a primate species marmosets for the first time 196 197 Their first research target for these marmosets was Parkinson s disease but they were also considering amyotrophic lateral sclerosis and Huntington s disease 198 Human proteins expressed in mammals are more likely to be similar to their natural counterparts than those expressed in plants or microorganisms Stable expression has been accomplished in sheep pigs rats and other animals In 2009 the first human biological drug produced from such an animal a goat was approved The drug ATryn is an anticoagulant which reduces the probability of blood clots during surgery or childbirth and is extracted from the goat s milk 199 Human alpha 1 antitrypsin is another protein that has been produced from goats and is used in treating humans with this deficiency 200 Another medicinal area is in creating pigs with greater capacity for human organ transplants xenotransplantation Pigs have been genetically modified so that their organs can no longer carry retroviruses 201 or have modifications to reduce the chance of rejection 202 203 Pig lungs from genetically modified pigs are being considered for transplantation into humans 204 205 There is even potential to create chimeric pigs that can carry human organs 194 206 Livestock are modified with the intention of improving economically important traits such as growth rate quality of meat milk composition disease resistance and survival Animals have been engineered to grow faster be healthier 207 and resist diseases 208 Modifications have also improved the wool production of sheep and udder health of cows 189 Goats have been genetically engineered to produce milk with strong spiderweb like silk proteins in their milk 209 A GM pig called Enviropig was created with the capability of digesting plant phosphorus more efficiently than conventional pigs 210 211 They could reduce water pollution since they excrete 30 to 70 less phosphorus in manure 210 212 Dairy cows have been genetically engineered to produce milk that would be the same as human breast milk 213 This could potentially benefit mothers who cannot produce breast milk but want their children to have breast milk rather than formula 214 215 Researchers have also developed a genetically engineered cow that produces allergy free milk 216 nbsp Mice expressing the green fluorescent proteinScientists have genetically engineered several organisms including some mammals to include green fluorescent protein GFP for research purposes 217 GFP and other similar reporting genes allow easy visualization and localization of the products of the genetic modification 218 Fluorescent pigs have been bred to study human organ transplants regenerating ocular photoreceptor cells and other topics 219 In 2011 green fluorescent cats were created to help find therapies for HIV AIDS and other diseases 220 as feline immunodeficiency virus is related to HIV 221 There have been suggestions that genetic engineering could be used to bring animals back from extinction It involves changing the genome of a close living relative to resemble the extinct one and is currently being attempted with the passenger pigeon 222 Genes associated with the woolly mammoth have been added to the genome of an African Elephant although the lead researcher says he has no intention of creating live elephants and transferring all the genes and reversing years of genetic evolution is a long way from being feasible 223 224 It is more likely that scientists could use this technology to conserve endangered animals by bringing back lost diversity or transferring evolved genetic advantages from adapted organisms to those that are struggling 225 Humans Gene therapy 226 uses genetically modified viruses to deliver genes which can cure disease in humans Although gene therapy is still relatively new it has had some successes It has been used to treat genetic disorders such as severe combined immunodeficiency 227 and Leber s congenital amaurosis 228 Treatments are also being developed for a range of other currently incurable diseases such as cystic fibrosis 229 sickle cell anemia 230 Parkinson s disease 231 232 cancer 233 234 235 diabetes 236 heart disease 237 and muscular dystrophy 238 These treatments only effect somatic cells meaning any changes would not be inheritable Germline gene therapy results in any change being inheritable which has raised concerns within the scientific community 239 240 In 2015 CRISPR was used to edit the DNA of non viable human embryos 241 242 In November 2018 He Jiankui announced that he had edited the genomes of two human embryos in an attempt to disable the CCR5 gene which codes for a receptor that HIV uses to enter cells He said that twin girls Lulu and Nana had been born a few weeks earlier and that they carried functional copies of CCR5 along with disabled CCR5 mosaicism and were still vulnerable to HIV The work was widely condemned as unethical dangerous and premature 243 Fish Main article Genetically modified fish source source source source source source source source When exposed to 13 C water the zebrafish modified to express a carp creatine kinase right maintained swimming behavior while wild type zebrafish left could not 244 Genetically modified fish are used for scientific research as pets and as a food source Aquaculture is a growing industry currently providing over half the consumed fish worldwide 245 Through genetic engineering it is possible to increase growth rates reduce food intake remove allergenic properties increase cold tolerance and provide disease resistance Fish can also be used to detect aquatic pollution or function as bioreactors 246 Several groups have been developing zebrafish to detect pollution by attaching fluorescent proteins to genes activated by the presence of pollutants The fish will then glow and can be used as environmental sensors 247 248 The GloFish is a brand of genetically modified fluorescent zebrafish with bright red green and orange fluorescent color It was originally developed by one of the groups to detect pollution but is now part of the ornamental fish trade becoming the first genetically modified animal to become publicly available as a pet when in 2003 it was introduced for sale in the USA 249 GM fish are widely used in basic research in genetics and development Two species of fish zebrafish and medaka are most commonly modified because they have optically clear chorions membranes in the egg rapidly develop and the one cell embryo is easy to see and microinject with transgenic DNA 250 Zebrafish are model organisms for developmental processes regeneration genetics behavior disease mechanisms and toxicity testing 251 Their transparency allows researchers to observe developmental stages intestinal functions and tumour growth 252 253 The generation of transgenic protocols whole organism cell or tissue specific tagged with reporter genes has increased the level of information gained by studying these fish 254 GM fish have been developed with promoters driving an over production of growth hormone for use in the aquaculture industry to increase the speed of development and potentially reduce fishing pressure on wild stocks This has resulted in dramatic growth enhancement in several species including salmon 255 trout 256 and tilapia 257 AquaBounty Technologies a biotechnology company have produced a salmon called AquAdvantage salmon that can mature in half the time as wild salmon 258 It obtained regulatory approval in 2015 the first non plant GMO food to be commercialized 259 As of August 2017 GMO salmon is being sold in Canada 260 Sales in the US started in May 2021 261 Insects See also Genetically modified insect source source source Overexpression of methyl CpG binding protein 2 in Drosophila impairs climbing ability right compared to the control group left 262 In biological research transgenic fruit flies Drosophila melanogaster are model organisms used to study the effects of genetic changes on development 263 Fruit flies are often preferred over other animals due to their short life cycle and low maintenance requirements They also have a relatively simple genome compared to many vertebrates with typically only one copy of each gene making phenotypic analysis easy 264 Drosophila have been used to study genetics and inheritance embryonic development learning behavior and aging 265 The discovery of transposons in particular the p element in Drosophila provided an early method to add transgenes to their genome although this has been taken over by more modern gene editing techniques 266 Due to their significance to human health scientists are looking at ways to control mosquitoes through genetic engineering Malaria resistant mosquitoes have been developed in the laboratory by inserting a gene that reduces the development of the malaria parasite 267 and then use homing endonucleases to rapidly spread that gene throughout the male population known as a gene drive 268 269 This approach has been taken further by using the gene drive to spread a lethal gene 270 271 In trials the populations of Aedes aegypti mosquitoes the single most important carrier of dengue fever and Zika virus were reduced by between 80 and by 90 272 273 271 Another approach is to use a sterile insect technique whereby males genetically engineered to be sterile out compete viable males to reduce population numbers 274 Other insect pests that make attractive targets are moths Diamondback moths cause US 4 to 5 billion of damage each year worldwide 275 The approach is similar to the sterile technique tested on mosquitoes where males are transformed with a gene that prevents any females born from reaching maturity 276 They underwent field trials in 2017 275 Genetically modified moths have previously been released in field trials 277 In this case a strain of pink bollworm that were sterilized with radiation were genetically engineered to express a red fluorescent protein making it easier for researchers to monitor them 278 Silkworm the larvae stage of Bombyx mori is an economically important insect in sericulture Scientists are developing strategies to enhance silk quality and quantity There is also potential to use the silk producing machinery to make other valuable proteins 279 Proteins currently developed to be expressed by silkworms include human serum albumin human collagen a chain mouse monoclonal antibody and N glycanase 280 Silkworms have been created that produce spider silk a stronger but extremely difficult to harvest silk 281 and even novel silks 282 Other nbsp Frog expressing green fluorescent proteinSystems have been developed to create transgenic organisms in a wide variety of other animals Chickens have been genetically modified for a variety of purposes This includes studying embryo development 283 preventing the transmission of bird flu 284 and providing evolutionary insights using reverse engineering to recreate dinosaur like phenotypes 285 A GM chicken that produces the drug Kanuma an enzyme that treats a rare condition in its egg passed US regulatory approval in 2015 286 Genetically modified frogs in particular Xenopus laevis and Xenopus tropicalis are used in developmental biology research GM frogs can also be used as pollution sensors especially for endocrine disrupting chemicals 287 There are proposals to use genetic engineering to control cane toads in Australia 288 289 The nematode Caenorhabditis elegans is one of the major model organisms for researching molecular biology 290 RNA interference RNAi was discovered in C elegans 291 and could be induced by simply feeding them bacteria modified to express double stranded RNA 292 It is also relatively easy to produce stable transgenic nematodes and this along with RNAi are the major tools used in studying their genes 293 The most common use of transgenic nematodes has been studying gene expression and localization by attaching reporter genes Transgenes can also be combined with RNAi techniques to rescue phenotypes study gene function image cell development in real time or control expression for different tissues or developmental stages 293 Transgenic nematodes have been used to study viruses 294 toxicology 295 diseases 296 297 and to detect environmental pollutants 298 nbsp Transgenic Hydra expressing green fluorescent proteinThe gene responsible for albinism in sea cucumbers has been found and used to engineer white sea cucumbers a rare delicacy The technology also opens the way to investigate the genes responsible for some of the cucumbers more unusual traits including hibernating in summer eviscerating their intestines and dissolving their bodies upon death 299 Flatworms have the ability to regenerate themselves from a single cell 300 Until 2017 there was no effective way to transform them which hampered research By using microinjection and radiation scientists have now created the first genetically modified flatworms 301 The bristle worm a marine annelid has been modified It is of interest due to its reproductive cycle being synchronized with lunar phases regeneration capacity and slow evolution rate 302 Cnidaria such as Hydra and the sea anemone Nematostella vectensis are attractive model organisms to study the evolution of immunity and certain developmental processes 303 Other animals that have been genetically modified include snails 304 geckos turtles 305 crayfish oysters shrimp clams abalone 306 and sponges 307 RegulationMain article Regulation of genetic engineering Genetically modified organisms are regulated by government agencies This applies to research as well as the release of genetically modified organisms including crops and food The development of a regulatory framework concerning genetic engineering began in 1975 at Asilomar California The Asilomar meeting recommended a set of guidelines regarding the cautious use of recombinant technology and any products resulting from that technology 308 The Cartagena Protocol on Biosafety was adopted on 29 January 2000 and entered into force on 11 September 2003 309 It is an international treaty that governs the transfer handling and use of genetically modified organisms 310 One hundred and fifty seven countries are members of the Protocol and many use it as a reference point for their own regulations 311 Universities and research institutes generally have a special committee that is responsible for approving any experiments that involve genetic engineering Many experiments also need permission from a national regulatory group or legislation All staff must be trained in the use of GMOs and all laboratories must gain approval from their regulatory agency to work with GMOs 312 The legislation covering GMOs are often derived from regulations and guidelines in place for the non GMO version of the organism although they are more severe 313 There is a near universal system for assessing the relative risks associated with GMOs and other agents to laboratory staff and the community They are assigned to one of four risk categories based on their virulence the severity of the disease the mode of transmission and the availability of preventive measures or treatments There are four biosafety levels that a laboratory can fall into ranging from level 1 which is suitable for working with agents not associated with disease to level 4 working with life threatening agents Different countries use different nomenclature to describe the levels and can have different requirements for what can be done at each level 313 nbsp A label marking this peanut butter as being non GMO nbsp Detail of a French cheese box declaring GMO free production i e below 0 9 There are differences in the regulation for the release of GMOs between countries with some of the most marked differences occurring between the US and Europe 314 Regulation varies in a given country depending on the intended use of the products of the genetic engineering For example a crop not intended for food use is generally not reviewed by authorities responsible for food safety 315 Some nations have banned the release of GMOs or restricted their use and others permit them with widely differing degrees of regulation 316 317 318 319 In 2016 thirty eight countries officially ban or prohibit the cultivation of GMOs and nine Algeria Bhutan Kenya Kyrgyzstan Madagascar Peru Russia Venezuela and Zimbabwe ban their importation 320 Most countries that do not allow GMO cultivation do permit research using GMOs 321 Despite regulation illegal releases have sometimes occurred due to weakness of enforcement 8 The European Union EU differentiates between approval for cultivation within the EU and approval for import and processing 322 While only a few GMOs have been approved for cultivation in the EU a number of GMOs have been approved for import and processing 323 The cultivation of GMOs has triggered a debate about the market for GMOs in Europe 324 Depending on the coexistence regulations incentives for cultivation of GM crops differ 325 The US policy does not focus on the process as much as other countries looks at verifiable scientific risks and uses the concept of substantial equivalence 326 Whether gene edited organisms should be regulated the same as genetically modified organism is debated USA regulations sees them as separate and does not regulate them under the same conditions while in Europe a GMO is any organism created using genetic engineering techniques 28 One of the key issues concerning regulators is whether GM products should be labeled The European Commission says that mandatory labeling and traceability are needed to allow for informed choice avoid potential false advertising 327 and facilitate the withdrawal of products if adverse effects on health or the environment are discovered 328 The American Medical Association 329 and the American Association for the Advancement of Science 330 say that absent scientific evidence of harm even voluntary labeling is misleading and will falsely alarm consumers Labeling of GMO products in the marketplace is required in 64 countries 331 Labeling can be mandatory up to a threshold GM content level which varies between countries or voluntary In Canada and the US labeling of GM food is voluntary 332 while in Europe all food including processed food or feed which contains greater than 0 9 of approved GMOs must be labeled 333 In 2014 sales of products that had been labeled as non GMO grew 30 percent to 1 1 billion 334 ControversySee also Genetically modified food controversies There is controversy over GMOs especially with regard to their release outside laboratory environments The dispute involves consumers producers biotechnology companies governmental regulators non governmental organizations and scientists Many of these concerns involve GM crops and whether food produced from them is safe and what impact growing them will have on the environment These controversies have led to litigation international trade disputes and protests and to restrictive regulation of commercial products in some countries 335 Most concerns are around the health and environmental effects of GMOs These include whether they may provoke an allergic reaction whether the transgenes could transfer to human cells and whether genes not approved for human consumption could outcross into the food supply 336 nbsp A protester advocating for the labeling of GMOsThere is a scientific consensus 337 338 339 340 that currently available food derived from GM crops poses no greater risk to human health than conventional food 341 342 343 344 345 but that each GM food needs to be tested on a case by case basis before introduction 346 347 348 Nonetheless members of the public are much less likely than scientists to perceive GM foods as safe 349 350 351 352 The legal and regulatory status of GM foods varies by country with some nations banning or restricting them and others permitting them with widely differing degrees of regulation 353 354 355 356 As late as the 1990s gene flow into wild populations was thought to be unlikely and rare and if it were to occur easily eradicated It was thought that this would add no additional environmental costs or risks no effects were expected other than those already caused by pesticide applications 357 However in the decades since several such examples have been observed Gene flow between GM crops and compatible plants along with increased use of broad spectrum herbicides 358 can increase the risk of herbicide resistant weed populations 359 Debate over the extent and consequences of gene flow intensified in 2001 when a paper was published showing transgenes had been found in landrace maize in Mexico the crop s center of diversity 360 361 Gene flow from GM crops to other organisms has been found to generally be lower than what would occur naturally 362 In order to address some of these concerns some GMOs have been developed with traits to help control their spread To prevent the genetically modified salmon inadvertently breeding with wild salmon all the fish raised for food are females triploid 99 are reproductively sterile and raised in areas where escaped salmon could not survive 363 364 Bacteria have also been modified to depend on nutrients that cannot be found in nature 365 and genetic use restriction technology has been developed though not yet marketed that causes the second generation of GM plants to be sterile 366 Other environmental and agronomic concerns include a decrease in biodiversity an increase in secondary pests non targeted pests and evolution of resistant insect pests 367 368 369 In the areas of China and the US with Bt crops the overall biodiversity of insects has increased and the impact of secondary pests has been minimal 370 Resistance was found to be slow to evolve when best practice strategies were followed 370 The impact of Bt crops on beneficial non target organisms became a public issue after a 1999 paper suggested they could be toxic to monarch butterflies Follow up studies have since shown that the toxicity levels encountered in the field were not high enough to harm the larvae 371 Accusations that scientists are playing God and other religious issues have been ascribed to the technology from the beginning 372 With the ability to genetically engineer humans now possible there are ethical concerns over how far this technology should go or if it should be used at all 373 Much debate revolves around where the line between treatment and enhancement is and whether the modifications should be inheritable 374 Other concerns include contamination of the non genetically modified food supply 375 376 the rigor of the regulatory process 377 378 consolidation of control of the food supply in companies that make and sell GMOs 379 exaggeration of the benefits of genetic modification 380 or concerns over the use of herbicides with glyphosate 381 Other issues raised include the patenting of life 382 and the use of intellectual property rights 383 There are large differences in consumer acceptance of GMOs with Europeans more likely to view GM food negatively than North Americans 384 GMOs arrived on the scene as the public confidence in food safety attributed to recent food scares such as Bovine spongiform encephalopathy and other scandals involving government regulation of products in Europe was low 385 This along with campaigns run by various non governmental organizations NGO have been very successful in blocking or limiting the use of GM crops 386 NGOs like the Organic Consumers Association the Union of Concerned Scientists 387 388 389 Greenpeace and other groups have said that risks have not been adequately identified and managed 390 and that there are unanswered questions regarding the potential long term impact on human health from food derived from GMOs They propose mandatory labeling 391 392 or a moratorium on such products 379 377 393 References Food genetically modified www who int Retrieved 15 August 2023 Smyth SJ April 2020 The human health benefits from GM crops Plant Biotechnology Journal 18 4 887 888 doi 10 1111 pbi 13261 PMC 7061863 PMID 31544299 Chilton MD 4 October 2016 Nature The First Creator of GMOs Forbes Retrieved 4 January 2019 Blakemore E The First GMO Is 8 000 Years Old Smithsonian Retrieved 5 January 2019 The new encyclopaedia Britannica 15th ed Chicago Encyclopaedia Britannica 1993 pp 178 ISBN 0 85229 571 5 OCLC 27665641 Staff Economic Impacts of Genetically Modified Crops on the Agri Food Sector p 42 Glossary Term and Definitions Archived 14 May 2013 at the Wayback Machine The European Commission Directorate General for Agriculture Genetic engineering The manipulation of an organism s genetic endowment by introducing or eliminating specific genes through modern molecular biology techniques A broad definition of genetic engineering also includes selective breeding and other means of artificial selection Retrieved 5 November 2012 The European Parliament and the council of the European Union 12 March 2001 Directive on the release of genetically modified organisms GMOs Directive 2001 18 EC ANNEX I A Official Journal of the European Communities a b c Freedman W 27 August 2018 6 Evolution Environmental Science a Canadian perspective 6 ed Dalhousie University a b Organisms obtained by mutagenesis are GMOs and are in principle subject to the obligations laid down by the GMO Directive PDF curia europa eu Archived PDF from the original on 25 July 2018 Retrieved 5 January 2019 Section 2 Description and Definitions www fao org Retrieved 3 January 2019 Frequently asked questions on genetically modified foods WHO Retrieved 3 January 2019 The EU Legislation on GMOs An Overview EU Science Hub European Commission 29 June 2010 Retrieved 3 January 2019 GMOs and Horizontal Gene Transfer NeuroLogica Blog 13 October 2016 Retrieved 9 July 2021 Zhang C Wohlhueter R Zhang H September 2016 Genetically modified foods A critical review of their promise and problems Food Science and Human Wellness 5 3 116 123 doi 10 1016 j fshw 2016 04 002 Oliver MJ 2014 Why we need GMO crops in agriculture Missouri Medicine 111 6 492 507 PMC 6173531 PMID 25665234 Center for Food Safety and Applied Nutrition Food from Genetically Engineered Plants Consumer Info About Food from Genetically Engineered Plants www fda gov Retrieved 8 January 2019 Secretariat of the Convention on Biological Diversity Montreal 2000 The Cartagena Protocol on Biosafety to the Convention on Biological Diversity Frequently Asked Questions FAQs on the Cartagena Protocol The Biosafety Clearing House BCH 29 February 2012 Retrieved 3 January 2019 What Is the Difference Between Genetically Modified Organisms and Genetically Engineered Organisms agbiotech ces ncsu edu Retrieved 8 January 2019 Agricultural Biotechnology Glossary USDA www usda gov Retrieved 8 January 2019 Colombo L 2007 The semantics of the term genetically modified organism Genetic impact of aquaculture activities on native populations Genimpact Final Scientific Report E U Contract N RICA CT 2005 022802 123 125 Chassy BM 2007 The History and Future of GMOs in Food and Agriculture Cereal Foods World doi 10 1094 cfw 52 4 0169 ISSN 0146 6283 Why the term GMO is scientifically meaningless Public Radio International Retrieved 5 January 2019 Tagliabue G September 2015 The nonsensical GMO pseudo category and a precautionary rabbit hole Nature Biotechnology 33 9 907 908 doi 10 1038 nbt 3333 PMID 26348954 S2CID 205281930 National Organic Standards Board Materials GMO Subcommittee Second Discussion Document on Excluded Methods Terminology PDF United States Department of Agriculture 22 August 2014 Archived PDF from the original on 2 October 2015 Retrieved 4 January 2019 Here s Why You Should Vote Against Measure P Even If You Hate GMOs Lost Coast Outpost Retrieved 4 January 2019 Neslen A 25 July 2018 Gene edited plants and animals are GM foods EU court rules The Guardian ISSN 0261 3077 Retrieved 5 January 2019 a b A CRISPR definition of genetic modification Nature Plants 4 5 233 May 2018 doi 10 1038 s41477 018 0158 1 PMID 29725105 Viewpoint Non GMO salt exploits Americans scientific illiteracy Genetic Literacy Project 1 June 2018 Retrieved 9 July 2021 Knutson J 28 May 2018 A sad day for our society when salt is labeled non GMO Agweek Retrieved 9 July 2021 Non GMO salt Water Food companies exploit GMO free labels misleading customers promoting misinformation Genetic Literacy Project 24 August 2015 Retrieved 9 July 2021 Nicholl DS 29 May 2008 An Introduction to Genetic Engineering Cambridge University Press p 34 ISBN 978 1 139 47178 7 Liang J Luo Y Zhao H 2011 Synthetic biology putting synthesis into biology Wiley Interdisciplinary Reviews Systems Biology and Medicine 3 1 7 20 doi 10 1002 wsbm 104 PMC 3057768 PMID 21064036 Berg P Mertz JE January 2010 Personal reflections on the origins and emergence of recombinant DNA technology Genetics 184 1 9 17 doi 10 1534 genetics 109 112144 PMC 2815933 PMID 20061565 Rahimzadeh M Sadeghizadeh M Najafi F Arab S Mobasheri H December 2016 Impact of heat shock step on bacterial transformation efficiency Molecular Biology Research Communications 5 4 257 261 PMC 5326489 PMID 28261629 Chen I Dubnau D March 2004 DNA uptake during bacterial transformation Nature Reviews Microbiology 2 3 241 9 doi 10 1038 nrmicro844 PMID 15083159 S2CID 205499369 a b National Research Council US Committee on Identifying and Assessing Unintended Effects of Genetically Engineered Foods on Human Health 1 January 2004 Methods and Mechanisms for Genetic Manipulation of Plants Animals and Microorganisms National Academies Press US Gelvin SB March 2003 Agrobacterium mediated plant transformation the biology behind the gene jockeying tool Microbiology and Molecular Biology Reviews 67 1 16 37 table of contents doi 10 1128 MMBR 67 1 16 37 2003 PMC 150518 PMID 12626681 Head G Hull RH Tzotzos GT 2009 Genetically Modified Plants Assessing Safety and Managing Risk London Academic Press p 244 ISBN 978 0 12 374106 6 Tuomela M Stanescu I Krohn K October 2005 Validation overview of bio analytical methods Gene Therapy 12 S1 S131 8 doi 10 1038 sj gt 3302627 PMID 16231045 S2CID 23000818 Narayanaswamy S 1994 Plant Cell and Tissue Culture Tata McGraw Hill Education pp vi ISBN 978 0 07 460277 5 Setlow JK 31 October 2002 Genetic Engineering Principles and Methods Springer Science amp Business Media p 109 ISBN 978 0 306 47280 0 Grizot S Smith J Daboussi F Prieto J Redondo P Merino N Villate M Thomas S Lemaire L Montoya G Blanco FJ Paques F Duchateau P September 2009 Efficient targeting of a SCID gene by an engineered single chain homing endonuclease Nucleic Acids Research 37 16 5405 19 doi 10 1093 nar gkp548 PMC 2760784 PMID 19584299 Gao H Smith J Yang M Jones S Djukanovic V Nicholson MG West A Bidney D Falco SC Jantz D Lyznik LA January 2010 Heritable targeted mutagenesis in maize using a designed endonuclease The Plant Journal 61 1 176 87 doi 10 1111 j 1365 313X 2009 04041 x PMID 19811621 Townsend JA Wright DA Winfrey RJ Fu F Maeder ML Joung JK Voytas DF May 2009 High frequency modification of plant genes using engineered zinc finger nucleases Nature 459 7245 442 5 Bibcode 2009Natur 459 442T doi 10 1038 nature07845 PMC 2743854 PMID 19404258 Shukla VK Doyon Y Miller JC DeKelver RC Moehle EA Worden SE Mitchell JC Arnold NL Gopalan S Meng X Choi VM Rock JM Wu YY Katibah GE Zhifang G McCaskill D Simpson MA Blakeslee B Greenwalt SA Butler HJ Hinkley SJ Zhang L Rebar EJ Gregory PD Urnov FD May 2009 Precise genome modification in the crop species Zea mays using zinc finger nucleases Nature 459 7245 437 41 Bibcode 2009Natur 459 437S doi 10 1038 nature07992 PMID 19404259 S2CID 4323298 Christian M Cermak T Doyle EL Schmidt C Zhang F Hummel A Bogdanove AJ Voytas DF October 2010 Targeting DNA double strand breaks with TAL effector nucleases Genetics 186 2 757 61 doi 10 1534 genetics 110 120717 PMC 2942870 PMID 20660643 Li T Huang S Jiang WZ Wright D Spalding MH Weeks DP Yang B January 2011 TAL nucleases TALNs hybrid proteins composed of TAL effectors and FokI DNA cleavage domain Nucleic Acids Research 39 1 359 72 doi 10 1093 nar gkq704 PMC 3017587 PMID 20699274 Esvelt KM Wang HH 2013 Genome scale engineering for systems and synthetic biology Molecular Systems Biology 9 641 doi 10 1038 msb 2012 66 PMC 3564264 PMID 23340847 Tan WS Carlson DF Walton MW Fahrenkrug SC Hackett PB 2012 Precision editing of large animal genomes Advances in Genetics Volume 80 Vol 80 pp 37 97 doi 10 1016 B978 0 12 404742 6 00002 8 ISBN 978 0 12 404742 6 PMC 3683964 PMID 23084873 a b Malzahn A Lowder L Qi Y 24 April 2017 Plant genome editing with TALEN and CRISPR Cell amp Bioscience 7 21 doi 10 1186 s13578 017 0148 4 PMC 5404292 PMID 28451378 Kingsbury N 2009 Hybrid The History and Science of Plant Breeding University of Chicago Press ISBN 978 0 226 43705 7 Clive Root 2007 Domestication Greenwood Publishing Groups Zohary D Hopf M Weiss E 2012 Domestication of Plants in the Old World The Origin and Spread of Plants in the Old World Oxford University Press Jackson DA Symons RH Berg P October 1972 Biochemical method for inserting new genetic information into DNA of Simian Virus 40 circular SV40 DNA molecules containing lambda phage genes and the galactose operon of Escherichia coli Proceedings of the National Academy of Sciences of the United States of America 69 10 2904 9 Bibcode 1972PNAS 69 2904J doi 10 1073 pnas 69 10 2904 PMC 389671 PMID 4342968 Sateesh MK 25 August 2008 Bioethics And Biosafety I K International Pvt Ltd pp 456 ISBN 978 81 906757 0 3 Retrieved 27 March 2013 Zhang C Wohlhueter R Zhang H 2016 Genetically modified foods A critical review of their promise and problems Food Science and Human Wellness 5 3 116 123 doi 10 1016 j fshw 2016 04 002 Russo E January 2003 The birth of biotechnology Nature 421 6921 456 7 Bibcode 2003Natur 421 456R doi 10 1038 nj6921 456a PMID 12540923 Morrow JF Cohen SN Chang AC Boyer HW Goodman HM Helling RB May 1974 Replication and transcription of eukaryotic DNA in Escherichia coli Proceedings of the National Academy of Sciences of the United States of America 71 5 1743 7 Bibcode 1974PNAS 71 1743M doi 10 1073 pnas 71 5 1743 PMC 388315 PMID 4600264 a b Jaenisch R Mintz B April 1974 Simian virus 40 DNA sequences in DNA of healthy adult mice derived from preimplantation blastocysts injected with viral DNA Proceedings of the National Academy of Sciences of the United States of America 71 4 1250 4 Bibcode 1974PNAS 71 1250J doi 10 1073 pnas 71 4 1250 PMC 388203 PMID 4364530 Any idiot can do it Genome editor CRISPR could put mutant mice in everyone s reach Science AAAS 2 November 2016 Retrieved 2 December 2016 Gordon JW Ruddle FH December 1981 Integration and stable germ line transmission of genes injected into mouse pronuclei Science 214 4526 1244 6 Bibcode 1981Sci 214 1244G doi 10 1126 science 6272397 PMID 6272397 Costantini F Lacy E November 1981 Introduction of a rabbit beta globin gene into the mouse germ line Nature 294 5836 92 4 Bibcode 1981Natur 294 92C doi 10 1038 294092a0 PMID 6945481 S2CID 4371351 Hanahan D Wagner EF Palmiter RD September 2007 The origins of oncomice a history of the first transgenic mice genetically engineered to develop cancer Genes amp Development 21 18 2258 70 doi 10 1101 gad 1583307 PMID 17875663 Brophy B Smolenski G Wheeler T Wells D L Huillier P Laible G February 2003 Cloned transgenic cattle produce milk with higher levels of beta casein and kappa casein Nature Biotechnology 21 2 157 62 doi 10 1038 nbt783 PMID 12548290 S2CID 45925486 Clark AJ July 1998 The mammary gland as a bioreactor expression processing and production of recombinant proteins Journal of Mammary Gland Biology and Neoplasia 3 3 337 50 doi 10 1023 a 1018723712996 PMID 10819519 Gordon K Lee E Vitale JA Smith AE Westphal H Hennighausen L 1987 Production of human tissue plasminogen activator in transgenic mouse milk 1987 Biotechnology 24 11 425 8 doi 10 1038 nbt1187 1183 PMID 1422049 S2CID 3261903 Bevan MW Flavell RB Chilton MD 1983 A chimaeric antibiotic resistance gene as a selectable marker for plant cell transformation 1983 Nature 304 5922 184 Bibcode 1983Natur 304 184B doi 10 1038 304184a0 S2CID 28713537 Jinturkar KA Rathi MN Misra A 2011 Gene Delivery Using Physical Methods Challenges in Delivery of Therapeutic Genomics and Proteomics pp 83 126 doi 10 1016 b978 0 12 384964 9 00003 7 ISBN 978 0 12 384964 9 a b Ye X Al Babili S Kloti A Zhang J Lucca P Beyer P Potrykus I January 2000 Engineering the provitamin A beta carotene biosynthetic pathway into carotenoid free rice endosperm Science 287 5451 303 5 Bibcode 2000Sci 287 303Y doi 10 1126 science 287 5451 303 PMID 10634784 S2CID 40258379 Goeddel DV Kleid DG Bolivar F Heyneker HL Yansura DG Crea R Hirose T Kraszewski A Itakura K Riggs AD January 1979 Expression in Escherichia coli of chemically synthesized genes for human insulin Proceedings of the National Academy of Sciences of the United States of America 76 1 106 10 Bibcode 1979PNAS 76 106G doi 10 1073 pnas 76 1 106 PMC 382885 PMID 85300 Artificial Genes Time 15 November 1982 Archived from the original on 27 October 2011 Retrieved 17 July 2010 Horn ME Woodard SL Howard JA May 2004 Plant molecular farming systems and products Plant Cell Reports 22 10 711 20 doi 10 1007 s00299 004 0767 1 PMC 7079917 PMID 14997337 BBC News 14 June 2002 GM crops A bitter harvest Maugh Thomas H II 9 June 1987 Altered Bacterium Does Its Job Frost Failed to Damage Sprayed Test Crop Company Says Los Angeles Times Fraley RT Rogers SG Horsch RB Sanders PR Flick JS Adams SP Bittner ML Brand LA Fink CL Fry JS Galluppi GR Goldberg SB Hoffmann NL Woo SC August 1983 Expression of bacterial genes in plant cells Proceedings of the National Academy of Sciences of the United States of America 80 15 4803 7 Bibcode 1983PNAS 80 4803F doi 10 1073 pnas 80 15 4803 PMC 384133 PMID 6308651 James Clive 1997 Global Status of Transgenic Crops in 1997 PDF ISAAA Briefs No 5 31 Archived PDF from the original on 16 January 2009 Bruening G Lyons JM 2000 The case of the FLAVR SAVR tomato California Agriculture 54 4 6 7 doi 10 3733 ca v054n04p6 Debora MacKenzie 18 June 1994 Transgenic tobacco is European first New Scientist Genetically Altered Potato Ok d For Crops Lawrence Journal World 6 May 1995 James C 1996 Global Review of the Field Testing and Commercialization of Transgenic Plants 1986 to 1995 PDF The International Service for the Acquisition of Agri biotech Applications Archived PDF from the original on 16 June 2010 Retrieved 17 July 2010 Gibson DG Glass JI Lartigue C Noskov VN Chuang RY Algire MA Benders GA Montague MG Ma L Moodie MM Merryman C Vashee S Krishnakumar R Assad Garcia N Andrews Pfannkoch C Denisova EA Young L Qi ZQ Segall Shapiro TH Calvey CH Parmar PP Hutchison CA Smith HO Venter JC July 2010 Creation of a bacterial cell controlled by a chemically synthesized genome Science 329 5987 52 6 Bibcode 2010Sci 329 52G doi 10 1126 science 1190719 PMID 20488990 S2CID 7320517 Sample I 20 May 2010 Craig Venter creates synthetic life form guardian co uk London Vazquez Salat N Salter B Smets G Houdebine LM 1 November 2012 The current state of GMO governance are we ready for GM animals Biotechnology Advances Special issue on ACB 2011 30 6 1336 43 doi 10 1016 j biotechadv 2012 02 006 PMID 22361646 Glowing fish to be first genetically changed pet CNN 21 November 2003 Retrieved 25 December 2018 a b Pollack A 19 November 2015 Genetically Engineered Salmon Approved for Consumption The New York Times ISSN 0362 4331 Archived from the original on 2 January 2022 Retrieved 27 January 2019 Bodnar A October 2010 Risk Assessment and Mitigation of AquAdvantage Salmon PDF ISB News Report Archived from the original PDF on 8 March 2021 Retrieved 22 January 2016 Melo EO Canavessi AM Franco MM Rumpf R March 2007 Animal transgenesis state of the art and applications PDF Journal of Applied Genetics 48 1 47 61 doi 10 1007 BF03194657 PMID 17272861 S2CID 24578435 Rediscovering Biology Online Textbook Unit 13 Genetically Modified Organisms www learner org Archived from the original on 3 December 2019 Retrieved 18 August 2017 Fan M Tsai J Chen B Fan K LaBaer J March 2005 A central repository for published plasmids Science 307 5717 1877 doi 10 1126 science 307 5717 1877a PMID 15790830 S2CID 27404861 Cooper GM 2000 Cells As Experimental Models The Cell A Molecular Approach 2nd ed Patel P June 2018 Microbe Mystery Scientific American 319 1 18 Bibcode 2018SciAm 319a 18P doi 10 1038 scientificamerican0718 18a PMID 29924081 S2CID 49310760 Arpino JA Hancock EJ Anderson J Barahona M Stan GB Papachristodoulou A Polizzi K July 2013 Tuning the dials of Synthetic Biology Microbiology 159 Pt 7 1236 53 doi 10 1099 mic 0 067975 0 PMC 3749727 PMID 23704788 Pollack A 7 May 2014 Researchers Report Breakthrough in Creating Artificial Genetic Code The New York Times Archived from the original on 2 January 2022 Retrieved 7 May 2014 Malyshev DA Dhami K Lavergne T Chen T Dai N Foster JM Correa IR Romesberg FE May 2014 A semi synthetic organism with an expanded genetic alphabet Nature 509 7500 385 8 Bibcode 2014Natur 509 385M doi 10 1038 nature13314 PMC 4058825 PMID 24805238 a b Karenlampi SO von Wright AJ 1 January 2016 Genetically Modified Microorganisms Encyclopedia of Food and Health Encyclopedia of Food and Health pp 211 216 doi 10 1016 B978 0 12 384947 2 00356 1 ISBN 978 0 12 384953 3 Panesar Pamit et al 2010 Enzymes in Food Processing Fundamentals and Potential Applications Chapter 10 I K International Publishing House ISBN 978 93 80026 33 6 Blair R Regenstein JM 3 August 2015 Genetic Modification and Food Quality A Down to Earth Analysis John Wiley amp Sons pp 20 24 ISBN 978 1 118 75641 6 a b c Jumba M 2009 Genetically Modified Organisms the Mystery Unraveled Durham Eloquent Books pp 51 54 ISBN 978 1 60911 081 9 a b Zhou Y Lu Z Wang X Selvaraj JN Zhang G February 2018 Genetic engineering modification and fermentation optimization for extracellular production of recombinant proteins using Escherichia coli Applied Microbiology and Biotechnology 102 4 1545 1556 doi 10 1007 s00253 017 8700 z PMID 29270732 S2CID 253769838 Leader B Baca QJ Golan DE January 2008 Protein therapeutics a summary and pharmacological classification Nature Reviews Drug Discovery A guide to drug discovery 7 1 21 39 doi 10 1038 nrd2399 PMID 18097458 S2CID 3358528 Walsh G April 2005 Therapeutic insulins and their large scale manufacture Applied Microbiology and Biotechnology 67 2 151 9 doi 10 1007 s00253 004 1809 x PMID 15580495 S2CID 5986035 Pipe SW May 2008 Recombinant clotting factors Thrombosis and Haemostasis 99 5 840 50 doi 10 1160 TH07 10 0593 PMID 18449413 S2CID 2701961 Bryant J Baxter L Cave CB Milne R July 2007 Bryant J ed Recombinant growth hormone for idiopathic short stature in children and adolescents PDF The Cochrane Database of Systematic Reviews 3 CD004440 doi 10 1002 14651858 CD004440 pub2 PMID 17636758 Baxter L Bryant J Cave CB Milne R January 2007 Bryant J ed Recombinant growth hormone for children and adolescents with Turner syndrome PDF The Cochrane Database of Systematic Reviews 1 CD003887 doi 10 1002 14651858 CD003887 pub2 PMID 17253498 Summers Rebecca 24 April 2013 Bacteria churn out first ever petrol like biofuel New Scientist Retrieved 27 April 2013 Reardon S June 2018 Genetically modified bacteria enlisted in fight against disease Nature 558 7711 497 498 Bibcode 2018Natur 558 497R doi 10 1038 d41586 018 05476 4 PMID 29946090 Amarger N November 2002 Genetically modified bacteria in agriculture Biochimie 84 11 1061 72 doi 10 1016 s0300 9084 02 00035 4 PMID 12595134 Sharma B Dangi AK Shukla P March 2018 Contemporary enzyme based technologies for bioremediation A review Journal of Environmental Management 210 10 22 doi 10 1016 j jenvman 2017 12 075 PMID 29329004 a b Yetisen AK Davis J Coskun AF Church GM Yun SH December 2015 Bioart Trends in Biotechnology 33 12 724 734 doi 10 1016 j tibtech 2015 09 011 PMID 26617334 S2CID 259584956 Church GM Gao Y Kosuri S September 2012 Next generation digital information storage in DNA Science 337 6102 1628 Bibcode 2012Sci 337 1628C doi 10 1126 science 1226355 PMID 22903519 Baldo A van den Akker E Bergmans HE Lim F Pauwels K December 2013 General considerations on the biosafety of virus derived vectors used in gene therapy and vaccination Current Gene Therapy 13 6 385 94 doi 10 2174 15665232113136660005 PMC 3905712 PMID 24195604 Is gene therapy available to treat my disorder Genetics Home Reference Retrieved 14 December 2018 a b Aiuti A Roncarolo MG Naldini L June 2017 ex vivo gene therapy in Europe paving the road for the next generation of advanced therapy medicinal products EMBO Molecular Medicine 9 6 737 740 doi 10 15252 emmm 201707573 PMC 5452047 PMID 28396566 a b c d Lundstrom K May 2018 Viral Vectors in Gene Therapy Diseases 6 2 42 doi 10 3390 diseases6020042 PMC 6023384 PMID 29883422 a b Sheridan C February 2011 Gene therapy finds its niche Nature Biotechnology 29 2 121 8 doi 10 1038 nbt 1769 PMID 21301435 S2CID 5063701 Manservigi R Epstein AL Argnani R Marconi P 2013 HSV as a Vector in Vaccine Development and Gene Therapy Landes Bioscience Chan VS November 2006 Use of genetically modified viruses and genetically engineered virus vector vaccines environmental effects Journal of Toxicology and Environmental Health Part A 69 21 1971 7 doi 10 1080 15287390600751405 PMID 16982535 S2CID 41198650 a b Ramezanpour B Haan I Osterhaus A Claassen E December 2016 Vector based genetically modified vaccines Exploiting Jenner s legacy Vaccine 34 50 6436 6448 doi 10 1016 j vaccine 2016 06 059 PMC 7115478 PMID 28029542 Tameris MD Hatherill M Landry BS Scriba TJ Snowden MA Lockhart S Shea JE McClain JB Hussey GD Hanekom WA Mahomed H McShane H March 2013 Safety and efficacy of MVA85A a new tuberculosis vaccine in infants previously vaccinated with BCG a randomized placebo controlled phase 2b trial Lancet 381 9871 1021 8 doi 10 1016 S0140 6736 13 60177 4 PMC 5424647 PMID 23391465 Delany I Rappuoli R De Gregorio E June 2014 Vaccines for the 21st century EMBO Molecular Medicine 6 6 708 20 doi 10 1002 emmm 201403876 PMC 4203350 PMID 24803000 Bhattacharya S Genetically modified virus explodes cancer cells New Scientist Khamsi R GM virus shrinks cancer tumours in humans New Scientist Leja J Yu D Nilsson B Gedda L Zieba A Hakkarainen T Akerstrom G Oberg K Giandomenico V Essand M November 2011 Oncolytic adenovirus modified with somatostatin motifs for selective infection of neuroendocrine tumor cells Gene Therapy 18 11 1052 62 doi 10 1038 gt 2011 54 PMID 21490682 S2CID 22520171 Perett Linda 30 June 2011 Measles viruses genetically modified to treat ovarian cancer National Cancer Institute Benchmarks Retrieved 5 September 2012 Breitbach CJ Thorne SH Bell JC Kirn DH July 2012 Targeted and armed oncolytic poxviruses for cancer the lead example of JX 594 Current Pharmaceutical Biotechnology 13 9 1768 72 doi 10 2174 138920112800958922 PMID 21740365 Beasley Deena 31 August 2011 Cancer fighting virus shown to target tumors alone Reuters Science Retrieved 5 September 2012 Garber K March 2006 China approves world s first oncolytic virus therapy for cancer treatment Journal of the National Cancer Institute 98 5 298 300 doi 10 1093 jnci djj111 PMID 16507823 Molteni M 12 April 2017 Florida s Orange Trees Are Dying But a Weaponized Virus Could Save Them Wired Retrieved 17 April 2017 a b Jelley J 7 August 2002 GM virus curbs rabbits Retrieved 16 December 2018 O Riordan B 26 February 2005 Virus planned to counter cane toad The Guardian ISSN 0261 3077 Retrieved 16 December 2018 Mildura GO Virus could sterilise Australia s rabbits New Scientist Retrieved 16 December 2018 Angulo E Cooke B December 2002 First synthesize new viruses then regulate their release The case of the wild rabbit Molecular Ecology 11 12 2703 9 doi 10 1046 j 1365 294X 2002 01635 x hdl 10261 45541 PMID 12453252 S2CID 23916432 Pires DP Cleto S Sillankorva S Azeredo J Lu TK September 2016 Genetically Engineered Phages a Review of Advances over the Last Decade Microbiology and Molecular Biology Reviews 80 3 523 43 doi 10 1128 MMBR 00069 15 PMC 4981678 PMID 27250768 Lee YJ Yi H Kim WJ Kang K Yun DS Strano MS Ceder G Belcher AM May 2009 Fabricating genetically engineered high power lithium ion batteries using multiple virus genes Science 324 5930 1051 5 Bibcode 2009Sci 324 1051L doi 10 1126 science 1171541 PMID 19342549 S2CID 32017913 Branduardi P Smeraldi C Porro D 2008 Metabolically engineered yeasts potential industrial applications Journal of Molecular Microbiology and Biotechnology 15 1 31 40 doi 10 1159 000111990 PMID 18349548 GM fungi New way to produce cheap biofuel The Times of India 4 June 2013 Retrieved 17 December 2018 Fang W Vega Rodriguez J Ghosh AK Jacobs Lorena M Kang A St Leger RJ February 2011 Development of transgenic fungi that kill human malaria parasites in mosquitoes Science 331 6020 1074 7 Bibcode 2011Sci 331 1074F doi 10 1126 science 1199115 PMC 4153607 PMID 21350178 Francie Diep 28 February 2011 Genetically altered fungus designed to attack malaria in mosquitoes Scientific American Hokanson KE Dawson WO Handler AM Schetelig MF St Leger RJ December 2014 Not all GMOs are crop plants non plant GMO applications in agriculture Transgenic Research 23 6 1057 68 doi 10 1007 s11248 013 9769 5 PMID 24242193 S2CID 255108053 a b Zhao H Lovett B Fang W 1 January 2016 Genetically Engineering Entomopathogenic Fungi Advances in Genetics 94 137 63 doi 10 1016 bs adgen 2015 11 001 ISBN 9780128046944 PMID 27131325 Koenraadt CJ Takken W April 2011 Viability of GM fungi crucial to malaria control Science 332 6026 175 Bibcode 2011Sci 332 175K doi 10 1126 science 332 6026 175 PMID 21474739 Waltz E 14 April 2016 Gene edited CRISPR mushroom escapes US regulation Nature 532 7599 293 Bibcode 2016Natur 532 293W doi 10 1038 nature 2016 19754 PMID 27111611 Charles D 15 April 2016 Will Genetically Edited Food Be Regulated The Case of the Mushroom All Things Considered National Public Radio Retrieved 17 December 2018 Zimmer C 27 July 2018 What Is a Genetically Modified Crop A European Ruling Sows Confusion The New York Times Archived from the original on 2 January 2022 Retrieved 17 December 2018 a b c Walter P Roberts K Raff M Lewis J Johnson A Alberts B 2002 Studying Gene Expression and Function Molecular Biology of the Cell 4th ed Garland Science Ganapathi TR Suprasanna P Rao PS Bapat VA 2004 Tobacco Nicotiana tabacum L A Model System for Tissue Culture Interventions and Genetic Engineering Indian Journal of Biotechnology 3 171 184 Koszowski B Goniewicz ML Czogala J Sobczak A 2007 Genetycznie modyfikowany tyton szansa czy zagrozenie dla palaczy Genetically modified tobacco chance or threat for smokers PDF Przeglad Lekarski in Polish 64 10 908 12 PMID 18409340 Archived from the original PDF on 23 January 2013 Mou B Scorza R 15 June 2011 Transgenic Horticultural Crops Challenges and Opportunities CRC Press p 104 ISBN 978 1 4200 9379 7 Gepstein S Horwitz BA 1995 The impact of Arabidopsis research on plant biotechnology Biotechnology Advances 13 3 403 14 doi 10 1016 0734 9750 95 02003 l PMID 14536094 Holland CK Jez JM October 2018 Arabidopsis the original plant chassis organism Plant Cell Reports 37 10 1359 1366 doi 10 1007 s00299 018 2286 5 PMID 29663032 S2CID 253806270 Jefferson RA Kavanagh TA Bevan MW December 1987 GUS fusions beta glucuronidase as a sensitive and versatile gene fusion marker in higher plants The EMBO Journal 6 13 3901 7 doi 10 1002 j 1460 2075 1987 tb02730 x PMC 553867 PMID 3327686 a b Biotechnology in Ornamental Plants Pocket K www isaaa org Retrieved 17 December 2018 Chandler SF Sanchez C October 2012 Genetic modification the development of transgenic ornamental plant varieties Plant Biotechnology Journal 10 8 891 903 doi 10 1111 j 1467 7652 2012 00693 x PMID 22537268 Nosowitz D 15 September 2011 Suntory Creates Mythical Blue Or Um Lavender ish Rose Popular Science Retrieved 30 August 2012 Suntory to sell blue roses overseas The Japan Times 11 September 2011 Archived from the original on 22 November 2012 Retrieved 30 August 2012 World s First Blue Rose Soon Available in U S Wired 14 September 2011 Boehm 27 October 2009 Green genetic engineering now conquers the ornamental plant market as well Bioeconomy in Baden Wurttemberg Archived from the original on 3 April 2019 Retrieved 17 December 2018 a b Adams JM Piovesan G Strauss S Brown S 1 August 2002 The Case for Genetic Engineering of Native and Landscape Trees against Introduced Pests and Diseases Conservation Biology 16 4 874 79 doi 10 1046 j 1523 1739 2002 00523 x S2CID 86697592 Tripathi S Suzuki J Gonsalves D 2007 Development of Genetically Engineered Resistant Papaya for papaya ringspot virus in a Timely Manner A Comprehensive and Successful Approach Development of genetically engineered resistant papaya for papaya ringspot virus in a timely manner a comprehensive and successful approach Methods in Molecular Biology Vol 354 pp 197 240 doi 10 1385 1 59259 966 4 197 ISBN 978 1 59259 966 0 PMID 17172756 a b c Qaim M 29 April 2016 Introduction Genetically Modified Crops and Agricultural Development Springer pp 1 10 ISBN 978 1 137 40572 2 a b Global Status of Commercialized Biotech GM Crops 2014 ISAAA Brief 49 2014 ISAAA org Retrieved 15 September 2016 a b ISAAA 2013 Annual Report Executive Summary Global Status of Commercialized Biotech GM Crops 2013 ISAAA Brief 46 2013 Retrieved 6 August 2014 Hakim D 29 October 2016 Doubts About the Promised Bounty of Genetically Modified Crops The New York Times ISSN 0362 4331 Archived from the original on 2 January 2022 Retrieved 5 May 2017 Areal FJ Riesgo L Rodriguez Cerezo E February 2013 Economic and agronomic impact of commercialized GM crops a meta analysis The Journal of Agricultural Science 151 1 7 33 doi 10 1017 S0021859612000111 ISSN 0021 8596 S2CID 85891950 Finger R El Benni N Kaphengst T Evans C Herbert S Lehmann B et al 10 May 2011 A Meta Analysis on Farm Level Costs and Benefits of GM Crops Sustainability 3 5 743 762 doi 10 3390 su3050743 hdl 20 500 11850 42242 Klumper W Qaim M 3 November 2014 A meta analysis of the impacts of genetically modified crops PLOS ONE 9 11 e111629 Bibcode 2014PLoSO 9k1629K doi 10 1371 journal pone 0111629 PMC 4218791 PMID 25365303 Darmency H August 2013 Pleiotropic effects of herbicide resistance genes on crop yield a review Pest Management Science 69 8 897 904 doi 10 1002 ps 3522 PMID 23457026 Green JM September 2014 Current state of herbicides in herbicide resistant crops Pest Management Science 70 9 1351 7 doi 10 1002 ps 3727 PMID 24446395 Fleischer SJ Hutchison WD Naranjo SE 2014 Sustainable Management of Insect Resistant Crops Plant Biotechnology pp 115 127 doi 10 1007 978 3 319 06892 3 10 ISBN 978 3 319 06891 6 SGK321 GM Approval Database ISAAA org Retrieved 27 April 2017 Qiu J October 2008 Is China ready for GM rice Nature 455 7215 850 2 doi 10 1038 455850a PMID 18923484 Frist B 21 November 2006 Green revolution hero The Washington Times One existing crop genetically engineered golden rice that produces vitamin A already holds enormous promise for reducing blindness and dwarfism that result from a vitamin A deficient diet Black RE Allen LH Bhutta ZA Caulfield LE de Onis M Ezzati M Mathers C Rivera J January 2008 Maternal and child undernutrition global and regional exposures and health consequences Lancet 371 9608 243 60 doi 10 1016 S0140 6736 07 61690 0 PMID 18207566 S2CID 3910132 Humphrey JH West KP Sommer A 1992 Vitamin A deficiency and attributable mortality among under 5 year olds Bulletin of the World Health Organization 70 2 225 32 PMC 2393289 PMID 1600583 Paine JA Shipton CA Chaggar S Howells RM Kennedy MJ Vernon G Wright SY Hinchliffe E Adams JL Silverstone AL Drake R April 2005 Improving the nutritional value of Golden Rice through increased pro vitamin A content Nature Biotechnology 23 4 482 7 doi 10 1038 nbt1082 PMID 15793573 S2CID 632005 US FDA approves GMO Golden Rice as safe to eat Genetic Literacy Project 29 May 2018 Retrieved 30 May 2018 Gasdaska JR Spencer D Dickey L March 2003 Advantages of Therapeutic Protein Production in the Aquatic Plant Lemna BioProcessing Journal 2 2 49 56 doi 10 12665 J22 Gasdaska permanent dead link 10 December 2012 Engineering algae to make complex anti cancer designer drug PhysOrg Retrieved 15 April 2013 Buttner Mainik A Parsons J Jerome H Hartmann A Lamer S Schaaf A Schlosser A Zipfel PF Reski R Decker EL April 2011 Production of biologically active recombinant human factor H in Physcomitrella Plant Biotechnology Journal 9 3 373 83 doi 10 1111 j 1467 7652 2010 00552 x PMID 20723134 Baur A Reski R Gorr G May 2005 Enhanced recovery of a secreted recombinant human growth factor using stabilizing additives and by co expression of human serum albumin in the moss Physcomitrella patens Plant Biotechnology Journal 3 3 331 40 doi 10 1111 j 1467 7652 2005 00127 x PMID 17129315 a b Hammond J McGarvey P Yusibov V 6 December 2012 Plant Biotechnology New Products and Applications Springer Science amp Business Media pp 7 8 ISBN 978 3 642 60234 4 Bornke F Broer I June 2010 Tailoring plant metabolism for the production of novel polymers and platform chemicals Current Opinion in Plant Biology 13 3 354 62 doi 10 1016 j pbi 2010 01 005 PMID 20171137 Lehr F Posten C June 2009 Closed photo bioreactors as tools for biofuel production Current Opinion in Biotechnology 20 3 280 5 doi 10 1016 j copbio 2009 04 004 PMID 19501503 UNL s AgBiosafety for Educators agbiosafety unl edu Retrieved 18 December 2018 ProCellEx Platform Protalix Biotherapeutics Archived from the original on 27 October 2012 Gali Weinreb and Koby Yeshayahou for Globes 2 May 2012 FDA approves Protalix Gaucher treatment Archived 29 May 2013 at the Wayback Machine Concha C Canas R Macuer J Torres MJ Herrada AA Jamett F Ibanez C May 2017 Disease Prevention An Opportunity to Expand Edible Plant Based Vaccines Vaccines 5 2 14 doi 10 3390 vaccines5020014 PMC 5492011 PMID 28556800 Kovak Emma Qaim Matin Blaustein Rejto Dan 10 February 2021 The climate benefits of yield increases in genetically engineered crops bioRxiv 10 1101 2021 02 10 430488 a b Forabosco F Lohmus M Rydhmer L Sundstrom LF May 2013 Genetically modified farm animals and fish in agriculture A review Livestock Science 153 1 3 1 9 doi 10 1016 j livsci 2013 01 002 The Superpowers of Genetically Modified Pigs The Scientist Retrieved 5 February 2019 Rudinko Larisa 20 Guidance for industry USA Center for veterinary medicine Link Murray Joo 20 Genetically modified animals Archived 13 October 2019 at the Wayback Machine Canada Brainwaving How CRISPR is Spreading Through the Animal Kingdom www pbs org 23 May 2018 Retrieved 20 December 2018 a b Perleberg C Kind A Schnieke A January 2018 Genetically engineered pigs as models for human disease Disease Models amp Mechanisms 11 1 doi 10 1242 dmm 030783 PMC 5818075 PMID 29419487 Sato K Sasaki E February 2018 Genetic engineering in nonhuman primates for human disease modeling Journal of Human Genetics 63 2 125 131 doi 10 1038 s10038 017 0351 5 PMC 8075926 PMID 29203824 Sasaki E Suemizu H Shimada A Hanazawa K Oiwa R Kamioka M Tomioka I Sotomaru Y Hirakawa R Eto T Shiozawa S Maeda T Ito M Ito R Kito C Yagihashi C Kawai K Miyoshi H Tanioka Y Tamaoki N Habu S Okano H Nomura T May 2009 Generation of transgenic non human primates with germline transmission Nature 459 7246 523 7 Bibcode 2009Natur 459 523S doi 10 1038 nature08090 PMID 19478777 S2CID 4404433 Schatten G Mitalipov S May 2009 Developmental biology Transgenic primate offspring Nature 459 7246 515 6 Bibcode 2009Natur 459 515S doi 10 1038 459515a PMC 2777739 PMID 19478771 Cyranoski D May 2009 Marmoset model takes centre stage Nature 459 7246 492 doi 10 1038 459492a PMID 19478751 Britt Erickson 10 February 2009 for Chemical amp Engineering News FDA Approves Drug From Transgenic Goat Milk Accessed 6 October 2012 Spencer LT Humphries JE Brantly ML May 2005 Antibody response to aerosolized transgenic human alpha1 antitrypsin The New England Journal of Medicine 352 19 2030 1 doi 10 1056 nejm200505123521923 PMID 15888711 Zimmer C 15 October 2015 Editing of Pig DNA May Lead to More Organs for People l The New York Times Archived from the original on 2 January 2022 Zeyland J Gawronska B Juzwa W Jura J Nowak A Slomski R Smorag Z Szalata M Wozniak A Lipinski D August 2013 Transgenic pigs designed to express human a galactosidase to avoid humoral xenograft rejection Journal of Applied Genetics 54 3 293 303 doi 10 1007 s13353 013 0156 y PMC 3720986 PMID 23780397 Pig Heart Transplants For Humans Could Be on Their Way IFLScience 30 April 2014 Reardon S November 2015 New life for pig to human transplants Nature 527 7577 152 4 Bibcode 2015Natur 527 152R doi 10 1038 527152a PMID 26560282 Genetically modified pig lungs or lab grown lungs Which is the future of our organ supply Genetic Literacy Project 6 May 2014 Wu J Platero Luengo A Sakurai M Sugawara A Gil MA Yamauchi T Suzuki K Bogliotti YS Cuello C Morales Valencia M Okumura D Luo J Vilarino M Parrilla I Soto DA Martinez CA Hishida T Sanchez Bautista S Martinez Martinez ML Wang H Nohalez A Aizawa E Martinez Redondo P Ocampo A Reddy P Roca J Maga EA Esteban CR Berggren WT Nunez Delicado E Lajara J Guillen I Guillen P Campistol JM Martinez EA Ross PJ Izpisua Belmonte JC January 2017 Interspecies Chimerism with Mammalian Pluripotent Stem Cells Cell 168 3 473 486 e15 doi 10 1016 j cell 2016 12 036 PMC 5679265 PMID 28129541 Lai L Kang JX Li R Wang J Witt WT Yong HY Hao Y Wax DM Murphy CN Rieke A Samuel M Linville ML Korte SW Evans RW Starzl TE Prather RS Dai Y April 2006 Generation of cloned transgenic pigs rich in omega 3 fatty acids Nature Biotechnology 24 4 435 6 doi 10 1038 nbt1198 PMC 2976610 PMID 16565727 Tucker I 24 June 2018 Genetically modified animals The Guardian ISSN 0261 3077 Retrieved 21 December 2018 Zyga L 2010 Scientist bred goats that produce spider silk Phys org Archived from the original on 30 April 2015 a b Enviropig Canada University of Guelph 2010 Archived from the original on 30 January 2016 Schimdt S 22 June 2012 Genetically engineered pigs killed after funding ends Postmedia News Retrieved 31 July 2012 Enviropig Environmental Benefits Canada University of Guelph Archived from the original on 27 February 2010 Retrieved 8 March 2010 Gray R 2011 Genetically modified cows produce human milk Archived from the original on 4 April 2011 Genetically modified cows producing human milk Classical Medicine Journal 14 April 2010 Archived from the original on 6 November 2014 Yapp R 11 June 2011 Scientists create cow that produces human milk The Daily Telegraph London Retrieved 15 June 2012 Jabed A Wagner S McCracken J Wells DN Laible G October 2012 Targeted microRNA expression in dairy cattle directs production of b lactoglobulin free high casein milk Proceedings of the National Academy of Sciences of the United States of America 109 42 16811 6 Bibcode 2012PNAS 10916811J doi 10 1073 pnas 1210057109 PMC 3479461 PMID 23027958 Green fluorescent protein takes Nobel prize Lewis Brindley Retrieved 31 May 2015 Alberts B Johnson A Lewis J Raff M Roberts K Walter P 2002 Studying Gene Expression and Function Molecular Biology of the Cell 4th ed Garland Science Randall S 2008 Genetically Modified Pigs for Medicine and Agriculture PDF Biotechnology and Genetic Engineering Reviews 25 245 66 doi 10 7313 upo9781904761679 011 PMID 21412358 Archived from the original PDF on 26 March 2014 Wongsrikeao P Saenz D Rinkoski T Otoi T Poeschla E September 2011 Antiviral restriction factor transgenesis in the domestic cat Nature Methods 8 10 853 9 doi 10 1038 nmeth 1703 PMC 4006694 PMID 21909101 Staff 3 April 2012 Biology of HIV National Institute of Allergy and Infectious Diseases Archived from the original on 11 April 2014 Biello D Ancient DNA Could Return Passenger Pigeons to the Sky Scientific American Retrieved 23 December 2018 Sarchet P Can we grow woolly mammoths in the lab George Church hopes so New Scientist Retrieved 23 December 2018 Hawks J 19 February 2017 How mammoth cloning became fake news John Hawks Retrieved 20 January 2019 Shapiro B November 2015 Mammoth 2 0 will genome engineering resurrect extinct species Genome Biology 16 1 228 doi 10 1186 s13059 015 0800 4 PMC 4632474 PMID 26530525 Selkirk SM October 2004 Gene therapy in clinical medicine Postgraduate Medical Journal 80 948 560 70 doi 10 1136 pgmj 2003 017764 PMC 1743106 PMID 15466989 Cavazzana Calvo M Fischer A June 2007 Gene therapy for severe combined immunodeficiency are we there yet The Journal of Clinical Investigation 117 6 1456 65 doi 10 1172 JCI30953 PMC 1878528 PMID 17549248 Richards S 6 November 2012 Gene therapy arrives in Europe The Scientist Rosenecker J Huth S Rudolph C October 2006 Gene therapy for cystic fibrosis lung disease current status and future perspectives Current Opinion in Molecular Therapeutics 8 5 439 45 PMID 17078386 Persons DA Nienhuis AW July 2003 Gene therapy for the hemoglobin disorders Current Hematology Reports 2 4 348 55 PMID 12901333 LeWitt PA Rezai AR Leehey MA Ojemann SG Flaherty AW Eskandar EN et al April 2011 AAV2 GAD gene therapy for advanced Parkinson s disease a double blind sham surgery controlled randomized trial The Lancet Neurology 10 4 309 19 doi 10 1016 S1474 4422 11 70039 4 PMID 21419704 S2CID 37154043 Gallaher James 17 March 2011 Gene therapy treats Parkinson s disease BBC News Health Retrieved 24 April 2011 Urbina Z 12 February 2013 Genetically Engineered Virus Fights Liver Cancer United Academics Archived from the original on 16 February 2013 Retrieved 15 February 2013 Treatment for Leukemia Is Showing Early Promise The New York Times Associated Press 11 August 2011 p A15 Retrieved 21 January 2013 Coghlan A 26 March 2013 Gene therapy cures leukaemia in eight days New Scientist Retrieved 15 April 2013 Gene therapy cures diabetic dogs New Scientist 13 February 2013 Retrieved 15 February 2013 New gene therapy trial gives hope to people with heart failure British Heart Foundation 30 April 2013 Retrieved 5 May 2013 Foster K Foster H Dickson JG December 2006 Gene therapy progress and prospects Duchenne muscular dystrophy Gene Therapy 13 24 1677 85 doi 10 1038 sj gt 3302877 PMID 17066097 1990 The Declaration of Inuyama 5 August 2001 Archived from the original on 5 August 2001 Smith KR Chan S Harris J October 2012 Human germline genetic modification scientific and bioethical perspectives Archives of Medical Research 43 7 491 513 doi 10 1016 j arcmed 2012 09 003 PMID 23072719 Kolata G 23 April 2015 Chinese Scientists Edit Genes of Human Embryos Raising Concerns The New York Times Archived from the original on 2 January 2022 Retrieved 24 April 2015 Liang P Xu Y Zhang X Ding C Huang R Zhang Z Lv J Xie X Chen Y Li Y Sun Y Bai Y Songyang Z Ma W Zhou C Huang J May 2015 CRISPR Cas9 mediated gene editing in human tripronuclear zygotes Protein amp Cell 6 5 363 372 doi 10 1007 s13238 015 0153 5 PMC 4417674 PMID 25894090 Begley S 28 November 2018 Amid uproar Chinese scientist defends creating gene edited babies STAT STAT Wang Q Tan X Jiao S You F Zhang PJ 24 July 2014 Analyzing cold tolerance mechanism in transgenic zebrafish Danio rerio PLOS ONE 9 7 e102492 Bibcode 2014PLoSO 9j2492W doi 10 1371 journal pone 0102492 PMC 4109919 PMID 25058652 Half of Fish Consumed Globally Is Now Raised on Farms Study Finds ScienceDaily Retrieved 21 December 2018 Tonelli FM Lacerda SM Tonelli FC Costa GM de Franca LR Resende RR November 2017 Progress and biotechnological prospects in fish transgenesis Biotechnology Advances 35 6 832 844 doi 10 1016 j biotechadv 2017 06 002 PMID 28602961 Nebert DW Stuart GW Solis WA Carvan MJ January 2002 Use of reporter genes and vertebrate DNA motifs in transgenic zebrafish as sentinels for assessing aquatic pollution Environmental Health Perspectives 110 1 A15 doi 10 1289 ehp 110 1240712 PMC 1240712 PMID 11813700 Mattingly CJ McLachlan JA Toscano WA August 2001 Green fluorescent protein GFP as a marker of aryl hydrocarbon receptor AhR function in developing zebrafish Danio rerio Environmental Health Perspectives 109 8 845 849 doi 10 1289 ehp 01109845 PMC 1240414 PMID 11564622 Hallerman E June 2004 Glofish the first GM animal commercialized profits amid controversy ISB News Report Hackett PB Ekker SE Essner JJ 2004 Chapter 16 Applications of transposable elements in fish for transgenesis and functional genomics In Gong Z Korzh V eds Fish Development and Genetics World Scientific Inc pp 532 80 Meyers JR 2018 Zebrafish Development of a Vertebrate Model Organism Current Protocols in Essential Laboratory Techniques 16 1 e19 doi 10 1002 cpet 19 Lu JW Ho YJ Ciou SC Gong Z September 2017 Innovative Disease Model Zebrafish as an In Vivo Platform for Intestinal Disorder and Tumors Biomedicines 5 4 58 doi 10 3390 biomedicines5040058 PMC 5744082 PMID 28961226 Barriuso J Nagaraju R Hurlstone A March 2015 Zebrafish a new companion for translational research in oncology Clinical Cancer Research 21 5 969 75 doi 10 1158 1078 0432 CCR 14 2921 PMC 5034890 PMID 25573382 Burket CT Montgomery JE Thummel R Kassen SC LaFave MC Langenau DM et al April 2008 Generation and characterization of transgenic zebrafish lines using different ubiquitous promoters Transgenic Research 17 2 265 79 doi 10 1007 s11248 007 9152 5 PMC 3660017 PMID 17968670 Du SJ Gong Z Fletcher GL Shears MA King MJ Idler DR Hew CL 1992 Growth Enhancement in Transgenic Atlantic Salmon by the Use of an All Fish Chimeric Growth Hormone Gene Construct Nature Biotechnology 10 2 176 181 doi 10 1038 nbt0292 176 PMID 1368229 S2CID 27048646 Devlin RH Biagi CA Yesaki TY Smailus DE Byatt JC February 2001 Growth of domesticated transgenic fish Nature 409 6822 781 782 Bibcode 2001Natur 409 781D doi 10 1038 35057314 PMID 11236982 S2CID 5293883 Rahman MA et al 2001 Growth and nutritional trials on transgenic Nile tilapia containing an exogenous fish growth hormone gene Journal of Fish Biology 59 1 62 78 doi 10 1111 j 1095 8649 2001 tb02338 x Pollack A 21 December 2012 Engineered Fish Moves a Step Closer to Approval The New York Times Archived from the original on 2 January 2022 span, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.