Jump to content

Genetic engineering

From Wikipedia, the free encyclopedia

This is an old revision of this page, as edited by Hyarmendacil (talk | contribs) at 01:04, 18 May 2011 (→‎History: grammar). The present address (URL) is a permanent link to this revision, which may differ significantly from the current revision.

Genetic engineering, also called genetic modification, is the direct human manipulation of an organism's genome using modern DNA technology. It involves the introduction of foreign DNA or synthetic genes into the organism of interest. The introduction of new DNA does not require the use of classical genetic methods, however traditional breeding methods are typically used for the propagation of recombinant organisms.

An organism that is generated through the introduction of recombinant DNA is considered to be a genetically modified organism. The first organisms genetically engineered were bacteria in 1973 and then mice in 1974. Insulin-producing bacteria were commercialized in 1982 and genetically modified food has been sold since 1994.

The most common form of genetic engineering involves the insertion of new genetic material at an unspecified location in the host genome. This is accomplished by isolating and copying the genetic material of interest using molecular cloning methods to generate a DNA sequence containing the required genetic elements for expression, and then inserting this construct into the host organism. Other forms of genetic engineering include gene targeting and knocking out specific genes via engineered nucleases such as zinc finger nucleases or engineered homing endonucleases.

Genetic engineering techniques have been applied in numerous fields including research, biotechnology, and medicine. Medicines such as insulin and human growth hormone are now produced in bacteria, experimental mice such as the oncomouse and the knockout mouse are being used for research purposes and insect resistant and/or herbicide tolerant crops have been commercialized. Genetically engineered plants and animals capable of producing biotechnology drugs more cheaply than current methods (called pharming) are also being developed and in 2009 the FDA approved the sale of the pharmaceutical protein antithrombin produced in the milk of genetically engineered goats.

Definition

Genetic engineering alters the genetic makeup of an organism using techniques that introduce heritable material prepared outside the organism either directly into the host or into a cell that is then fused or hybridized with the host.[1] This involves using recombinant nucleic acid (DNA or RNA) techniques to form new combinations of heritable genetic material followed by the incorporation of that material either indirectly through a vector system or directly through micro-injection, macro-injection and micro-encapsulation techniques. Genetic engineering does not include traditional animal and plant breeding, in vitro fertilisation, induction of polyploidy, mutagenesis and cell fusion techniques that do not use recombinant nucleic acids or a genetically modified organism in the process.[1] Cloning and stem cell research, although not considered genetic engineering,[2] are closely related and genetic engineering can be used within them.[3] Synthetic biology is an emerging discipline that takes genetic engineering a step further by introducing artificially synthesized genetic material from raw materials into an organism.[4]

If genetic material from another species is added to the host, the resulting organism is called transgenic. If genetic material from the same species or a species that can naturally breed with the host is used the resulting organism is called cisgenic.[5] Genetic engineering can also be used to remove genetic material from the target organism, creating a knock out organism.[6] In Europe genetic modification is synonymous with genetic engineering while within the United States of America it can also refer to conventional breeding methods.[7] Within the scientific community, the term genetic engineering is not commonly used; more specific terms such as transgenic are preferred.

History

Humans have altered the genomes of species for thousands of years through artificial selection and more recently mutagenesis. Genetic engineering as the direct manipulation of DNA by humans outside breeding and mutations has only existed since the 1970s. The term "genetic engineering" was first coined by Jack Williamson in his science fiction novel Dragon's Island, published in 1951,[8] one year before DNA's role in heredity was confirmed by Alfred Hershey and Martha Chase,[9] and two years before James Watson and Francis Crick showed that the DNA molecule has a double-helix structure.

In 1972 Paul Berg created the first recombinant DNA molecules by combining DNA from the monkey virus SV40 with that of the lambda virus.[10] In 1973 Herbert Boyer and Stanley Cohen created the first transgenic organism by inserting antibiotic resistance genes into the plasmid of an E. coli bacterium.[11][12] A year later Rudolf Jaenisch created a transgenic mouse by introducing foreign DNA into its embryo, making it the world’s first transgenic animal.[13] In 1976 Genentech, the first genetic engineering company was founded by Herbert Boyer and Robert Swanson and a year later and the company produced a human protein (somatostatin) in E.coli. Genentech announced the production of genetically engineered human insulin in 1978.[14] In 1980, the U.S. Supreme Court in the Diamond v. Chakrabarty case ruled that genetically altered life could be patented.[15] The insulin produced by bacteria, branded humulin, was approved for release by the Food and Drug Administration in 1982.[16]

The first field trials of genetically engineered plants occurred in France and the USA in 1986, tobacco plants were engineered to be resistant to herbicides.[17] The People’s Republic of China was the first country to commercialize transgenic plants, introducing a virus-resistant tobacco in 1992.[18] In 1994 Calgene attained approval to commercially release the Flavr Savr tomato, a tomato engineered to have a longer shelf life.[19] 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.[20] In 1995, Bt Potato was approved safe by the Environmental Protection Agency, making it the first pesticide producing crop to be approved in the USA.[21] In 2009 11 transgenic crops were grown commercially in 25 countries, the largest of which by area grown were the USA, Brazil, Argentina, India, Canada, China, Paraguay and South Africa.[22]

In 2010, scientists at the J. Craig Venter Institute, announced that they had created the first synthetic bacterial genome, and added it to a cell containing no DNA. The resulting bacterium, named Synthia, was the world's first synthetic life form.[23][24]

Process

Isolating the Gene

Elements of genetic engineering

First, the gene to be inserted into the genetically modified organism must be chosen and isolated. Presently, most genes transferred into plants provide protection against insects or tolerance to herbicides.[25] In animals the majority of genes used are growth hormone genes.[26] Once chosen the genes must be isolated. This typically involves multiplying the gene using polymerase chain reaction (PCR). If the chosen gene or the donor organism's genome has been well studied it may be present in a genetic library. If the DNA sequence is known, but no copies of the gene are available, it can be artificially synthesized. Once isolated, the gene is inserted into a bacterial plasmid.

Constructs

The gene to be inserted into the genetically modified organism must be combined with other genetic elements in order for it to work properly. The gene can also be modified at this stage for better expression or effectiveness. As well as the gene to be inserted most constructs contain a promoter and terminator region as well as a selectable marker gene. The promoter region initiates transcription of the gene and can be used to control the location and level of gene expression, while the terminator region ends transcription. The selectable marker, which in most cases confers antibiotic resistance to the organism it is expressed in, is needed to determine which cells are transformed with the new gene. The constructs are made using recombinant DNA techniques, such as restriction digests, ligations and molecular cloning.[27]

Gene Targeting

The most common form of genetic engineering involves inserting new genetic material randomly within the host genome. Other techniques allow new genetic material to be inserted at a specific location in the host genome or generate mutations at desired genomic loci capable of knocking out endogenous genes. The technique of gene targeting uses homologous recombination to target desired changes to a specific endogenous gene. This tends to occur at a relatively low frequency in plants and animals and generally requires the use of selectable markers. The frequency of gene targeting can be greatly enhanced with the use of engineered nucleases such as zinc finger nucleases,[28] [29] engineered homing endonucleases,[30] [31] or nucleases created from TAL effectors.[32] [33] In addition to enhancing gene targeting, engineered nucleases can also be used to introduce mutations at endogenous genes that generate a gene knockout[34] .[35]

Transformation

A. tumefaciens attaching itself to a carrot cell

About 1% of bacteria are naturally able to take up foreign DNA but it can also be induced in other bacteria.[36] Stressing the bacteria for example, with a heat shock or an electric shock, can make the cell membrane permeable to DNA that may then incorporate into their genome or exist as extrachromosomal DNA. DNA is generally inserted into animal cells using microinjection, where it can be injected through the cells nuclear envelope directly into the nucleus or through the use of viral vectors. In plants the DNA is generally inserted using Agrobacterium-mediated recombination or biolistics.[37]

In Agrobacterium-mediated recombination the plasmid construct must also contain T-DNA. Agrobacterium naturally inserts DNA from a tumor inducing plasmid into any susceptible plant's genome it infects, causing crown gall disease. The T-DNA region of this plasmid is responsible for insertion of the DNA. The genes to be inserted are cloned into a binary vector, which contains T-DNA and can be grown in both E. Coli and Agrobacterium. Once the binary vector is constructed the plasmid is transformed into Agrobacterium containing no plasmids and plant cells are infected. The Agrobacterium will then naturally insert the genetic material into the plant cells.[38]

In biolistics particles of gold or tungsten are coated with DNA and then shot into young plant cells or plant embryos. Some genetic material will enter the cells and transform them. This method can be used on plants that are not susceptible to Agrobacterium infection and also allows transformation of plant plastids. Another transformation method for plant and animal cells is electroporation. Electroporation involves subjecting the plant or animal cell to an electric shock, which can make the cell membrane permeable to plasmid DNA. In some cases the electroporated cells will incorporate the DNA into their genome. Due to the damage caused to the cells and DNA the transformation efficiency of biolistics and electroporation is lower than agrobacterial mediated transformation and microinjection.[39]

Selection

Not all the organism's cells will be transformed with the new genetic material; in most cases a selectable marker is used to differentiate transformed from untransformed cells. If a cell has been successfully transformed with the DNA it will also contain the marker gene. By growing the cells in the presence of an antibiotic or chemical that selects or marks the cells expressing that gene it is possible to separate the transgenic events from the non-transgenic. Another method of screening involves using a DNA probe that will only stick to the inserted gene. A number of strategies have been developed that can remove the selectable marker from the mature transgenic plant.[40]

Regeneration

As often only a single cell is transformed with genetic material the organism must be regrown from that single cell. As bacteria consist of a single cell and reproduce clonally regeneration is not necessary. In plants this is accomplished through the use of tissue culture. Each plant species has different requirements for successful regeneration through tissue culture. If successful an adult plant is produced that contains the transgene in every cell. In animals it is necessary to ensure that the inserted DNA is present in the embryonic stem cells. When the offspring is produced they can be screened for the presence of the gene. All offspring from the first generation will be heterozygous for the inserted gene and must be mated together to produce a homozygous animal.

Confirmation

The finding that a recombinant organism contains the inserted genes is not usually sufficient to ensure that the genes will be expressed in an appropriate manner in the intended tissues of the recombinant organism. To examine the presence of the gene, further analysis frequently uses PCR, Southern hybridization, and DNA sequencing, which serve to determine the chromosomal location and copy number of the inserted gene. To examine expression of the trans-gene, an extensive analysis of transcription, RNA processing patterns, and the expression and localization of the protein product(s) is usually necessary, using methods including northern hybridization, quantitative RT-PCR, Western blot, immunofluorescence and phenotypic analysis. When appropriate, the organism's offspring are studied to confirm that the trans-gene and associated phenotype are stably inherited.

Applications

Genetic engineering has applications in medicine, research, industry and agriculture and can be used on a wide range of plants, animals and micro organism.

Medicine

In medicine genetic engineering has been used to mass-produce insulin, human growth hormones, follistim (for treating infertility), human albumin, monoclonal antibodies, antihemophilic factors, vaccines and many other drugs.[41] Vaccination generally involves injecting weak live, killed or inactivated forms of viruses or their toxins into the person being immunized.[42] Genetically engineered viruses are being developed that can still confer immunity, but lack the infectious sequences.[43] Mouse hybridomas, cells fused together to create monoclonal antibodies, have been humanised through genetic engineering to create human monoclonal antibodies.[44]

Genetic engineering is used to create animal models of human diseases. Genetically modified mice are the most common genetically engineered animal model.[45] They have been used to study and model cancer (the oncomouse), obesity, heart disease, diabetes, arthritis, substance abuse, anxiety, aging and Parkinson disease.[46] Potential cures can be tested against these mouse models. Also genetically modified pigs have been bred with the aim of increasing the success of pig to human organ transplantation.[47]

Gene therapy is the genetic engineering of humans by replacing defective human genes with functional copies. This can occur in somatic tissue or germline tissue. If the gene is inserted into the germline tissue it can be passed down to that person's descendants.[48] Gene therapy has been used to treat patients suffering from immune deficiencies (notably Severe combined immunodeficiency) and trials have been carried out on other genetic disorders.[49] The success of gene therapy so far has been limited and a patient (Jesse Gelsinger) has died during a clinical trial testing a new treatment.[50] There are also ethical concerns should the technology be used not just for treatment, but for enhancement, modification or alteration of a human beings' appearance, adaptability, intelligence, character or behavior.[51] The distinction between cure and enhancement can also be difficult to establish.[52] Transhumanists consider the enhancement of humans desirable.

Research

Knockout mice
Human cells in which some proteins are fused with green fluorescent protein to allow them to be visualised

Genetic engineering is an important tool for natural scientists. Genes and other genetic information from a wide range of organisms are transformed into bacteria for storage and modification, creating genetically modified bacteria in the process. Bacteria are cheap, easy to grow, clonal, multiply quickly, relatively easy to transform 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.

Organisms are genetically engineered to discover the functions of certain genes. This could be the effect on the phenotype of the organism, where the gene is expressed or what other genes it interacts with. These experiments generally involve loss of function, gain of function, tracking and expression.

  • Loss of function experiments, such as in a gene knockout experiment, in which an organism is engineered to lack the activity of one or more genes. A knockout experiment involves the creation and manipulation of a DNA construct in vitro, which, in a simple knockout, consists of a copy of the desired gene, which has been altered such that it is non-functional. Embryonic stem cells incorporate the altered gene, which replaces the already present functional copy. These stem cells are injected into blastocysts, which are implanted into surrogate mothers. This allows the experimenter to analyze the defects caused by this mutation and thereby determine the role of particular genes. It is used especially frequently in developmental biology. Another method, useful in organisms such as Drosophila (fruit fly), is to induce mutations in a large population and then screen the progeny for the desired mutation. A similar process can be used in both plants and prokaryotes.
  • Gain of function experiments, the logical counterpart of knockouts. These are sometimes performed in conjunction with knockout experiments to more finely establish the function of the desired gene. The process is much the same as that in knockout engineering, except that the construct is designed to increase the function of the gene, usually by providing extra copies of the gene or inducing synthesis of the protein more frequently.
  • Tracking experiments, which seek to gain information about the localization and interaction of the desired protein. One way to do this is to replace the wild-type gene with a 'fusion' gene, which is a juxtaposition of the wild-type gene with a reporting element such as green fluorescent protein (GFP) that will allow easy visualization of the products of the genetic modification. While this is a useful technique, the manipulation can destroy the function of the gene, creating secondary effects and possibly calling into question the results of the experiment. More sophisticated techniques are now in development that can track protein products without mitigating their function, such as the addition of small sequences that will serve as binding motifs to monoclonal antibodies.
  • Expression studies aim to discover where and when specific proteins are produced. In these experiments, the DNA sequence before the DNA that codes for a protein, known as a gene's promoter, is reintroduced into an organism with the protein coding region replaced by a reporter gene such as GFP or an enzyme that catalyzes the production of a dye. Thus the time and place where a particular protein is produced can be observed. Expression studies can be taken a step further by altering the promoter to find which pieces are crucial for the proper expression of the gene and are actually bound by transcription factor proteins; this process is known as promoter bashing.

Industrial

By engineering genes into bacterial plasmids it is possible to create a biological factory that can produce proteins and enzymes.[53] Some genes do not work well in bacteria, so yeast, a eukaryote, can also be used.[54] Bacteria and yeast factories have been used to produce medicines such as insulin, human growth hormone, and vaccines, supplements such as tryptophan, aid in the production of food (chymosin in cheese making) and fuels.[55] Other applications involving genetically engineered bacteria being investigated involve making the bacteria perform tasks outside their natural cycle, such as cleaning up oil spills, carbon and other toxic waste.[56]

Agriculture

Bt-toxins present in peanut leaves (bottom image) protect it from extensive damage caused by European corn borer larvae (top image).[57]

One of the best-known and controversial applications of genetic engineering is the creation of genetically modified food. There are three generations of genetically modified crops.[58] First generation crops have been commercialized and most provide protection from insects and/or resistance to herbicides. There are also fungal and virus resistant crops developed or in development.[59][60] They have been developed to make the insect and weed management of crops easier and can indirectly increase crop yield.[61]

The second generation of genetically modified crops being developed aim to directly improve yield by improving salt, cold or drought tolerance and to increase the nutritional value of the crops.[62] The third generation consists of pharmaceutical crops, crops that contain edible vaccines and other drugs.[63] Some agriculturally important animals have been genetically modified with growth hormones to increase their size[64] while others have been engineered to express drugs and other proteins in their milk.[65][66][67]

The genetic engineering of agricultural crops can increase the growth rates and resistance to different diseases caused by pathogens and parasites.[68] This is beneficial as it can greatly increase the production of food sources with the usage of fewer resources that would be required to host the world's growing populations. These modified crops would also reduce the usage of chemicals, such as fertilizers and pesticides, and therefore decrease the severity and frequency of the damages produced by these chemical pollution.[68]

Ethical and safety concerns have been raised around the use of genetically modified food.[69] A major safety concern relates to the human health implications of eating genetically modified food, in particular whether toxic or allergic reactions could occur.[70] Gene flow into related non-transgenic crops, off target effects on beneficial organisms and the impact on biodiversity are important environmental issues.[71] Ethical concerns involve religious issues, corporate control of the food supply, intellectual property rights and the level of labeling needed on genetically modified products.

Other uses

In materials science, a genetically modified virus has been used to construct a more environmentally friendly lithium-ion battery.[72][73] Some bacteria have been genetically engineered to create black and white photographs[74] while others have potential to be used as sensors by expressing a fluorescent protein under certain environmental conditions.[75] Genetic engineering is also being used to create BioArt[76] and novelty items such as blue roses,[77] and glowing fish.[78]

Opposition and criticism

A 2010 study of Canola found transgenes in 80% of wild (uncultivated or "feral") varieties in North Dakota, meaning 80% of the plants which had established themselves in the area were genetically engineered varieties. The researchers stated that "we found the highest densities of [such transgene-containing] plants near agricultural fields and along major freeways, but we were also finding plants in the middle of nowhere" adding that "over time,..the build-up of different types of herbicide resistance in feral [natural] canola and closely related weeds, like field mustard, could make it more difficult to manage these plants using herbicides."[79]

See also

References

  1. ^ a b 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" (Document). Official Journal of the European Communities. p. page 17. {{cite document}}: |page= has extra text (help); Unknown parameter |url= ignored (help)
  2. ^ Van Eenennaam, Alison. "Is Livestock Cloning Another Form of Genetic Engineering?" (PDF). agbiotech.
  3. ^ David M. Suter, Michel Dubois-Dauphin, Karl-Heinz Krause (2006). "Genetic engineering of embryonic stem cells" (PDF). Swiss Med Wkly. 136 (27–28): 413–415. PMID 16897894.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  4. ^ Ernesto Andrianantoandro, Subhayu Basu, David K Kariga & Ron Weiss (16 May 2006). "Synthetic biology: new engineering rules for an emerging discipline". Molecular Systems Biology. 2 (2006.0028): 2006.0028. doi:10.1038/msb4100073. PMC 1681505. PMID 16738572.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  5. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1007/s11540-008-9097-y, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1007/s11540-008-9097-y instead.
  6. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nm1001-1086, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nm1001-1086 instead.
  7. ^ James H. Maryanski (October 19, 1999). "Genetically Engineered Foods". Center for Food Safety and Applied Nutrition at the Food and Drug Administration.
  8. ^ Stableford, Brian M. (2004). Historical dictionary of science fiction literature. p. 133. ISBN 9780810849389.
  9. ^ Hershey A, Chase M (1952). "Independent functions of viral protein and nucleic acid in growth of bacteriophage" (PDF). J Gen Physiol. 36 (1): 39–56. doi:10.1085/jgp.36.1.39. PMC 2147348. PMID 12981234.
  10. ^ Jackson, DA; Symons, RH; Berg, P (October 1, 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". PNAS. 69 (10): 2904–2909. doi:10.1073/pnas.69.10.2904. PMC 389671. PMID 4342968.
  11. ^ Arnold, Paul (2009). "History of Genetics: Genetic Engineering Timeline".
  12. ^ Stanley N. Cohen and Annie C. Y. Chang (1973-05-01). "Recircularization and Autonomous Replication of a Sheared R-Factor DNA Segment in Escherichia coli Transformants — PNAS". Pnas.org. Retrieved 2010-07-17.
  13. ^ Jaenisch, R. and Mintz, B. (1974). "Simian virus 40 DNA sequences in DNA of healthy adult mice derived from preimplantation blastocysts injected with viral DNA". Proc. Natl. Acad. Sci. 71 (4): 1250–1254. doi:10.1073/pnas.71.4.1250. PMC 388203. PMID 4364530. {{cite journal}}: line feed character in |journal= at position 12 (help)CS1 maint: multiple names: authors list (link)
  14. ^ Goeddel, David (1979). "Expression in Escherichia coli of chemically synthesized genes for human insulin" (PDF). PNAS. 76 (1): 106–110. doi:10.1073/pnas.76.1.106. PMC 382885. PMID 85300. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help); Unknown parameter |month= ignored (help)
  15. ^ US Supreme Court Cases from Justia & Oyez (1980-06-16). "Diamond V Chakrabarty". Supreme.justia.com. Retrieved 2010-07-17. {{cite web}}: Text "volume 447" ignored (help); Text "year 1980" ignored (help)
  16. ^ "Artificial Genes". TIME. 1982-11-15. Retrieved 2010-07-17.
  17. ^ James, Clive (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. Retrieved 17 July 2010.
  18. ^ James, Clive (1997). "Global Status of Transgenic Crops in 1997" (PDF). ISAAA Briefs No. 5.: 31.
  19. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.3733/ca.v054n04p6, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.3733/ca.v054n04p6 instead.
  20. ^ Debora MacKenzie (18 June 1994). "Transgenic tobacco is European first". New Scientist. {{cite journal}}: Cite journal requires |journal= (help)
  21. ^ Genetically Altered Potato Ok'd For Crops Lawrence Journal-World - 6 May 1995
  22. ^ Global Status of Commercialized Biotech/GM Crops: 2009 ISAAA Brief 41-2009, February 23, 2010, retrieved August 10, 2010
  23. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1126/science.1190719, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1126/science.1190719 instead.
  24. ^ Ian Sample (Thursday 20 May 2010). "Craig Venter creates synthetic life form". London: guardian.co.uk. {{cite news}}: Check date values in: |date= (help)
  25. ^ James, Clive (2008). "Global Status of Commercilized Biotech/GM Crops:2008". ISSA Breif No. 39.
  26. ^ Food and Agricultural Organisation of the United Nations. "The process of genetic modification".
  27. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1534/genetics.109.112144, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1534/genetics.109.112144 instead.
  28. ^ Townsend JA, Wright DA, Winfrey RJ; et al. (2009). "High-frequency modification of plant genes using engineered zinc-finger nucleases". Nature. 459 (7245): 442–5. doi:10.1038/nature07845. PMC 2743854. PMID 19404258. {{cite journal}}: Explicit use of et al. in: |author= (help); Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  29. ^ Shukla VK, Doyon Y, Miller JC; et al. (2009). "Precise genome modification in the crop species Zea mays using zinc-finger nucleases". Nature. 459 (7245): 437–41. doi:10.1038/nature07992. PMID 19404259. {{cite journal}}: Explicit use of et al. in: |author= (help); Unknown parameter |month= ignored (help); Unknown parameter |unused_data= ignored (help)CS1 maint: multiple names: authors list (link)
  30. ^ Grizot S, Smith J, Daboussi F; et al. (2009). "Efficient targeting of a SCID gene by an engineered single-chain homing endonuclease". Nucleic Acids Res. 37 (16): 5405–19. doi:10.1093/nar/gkp548. PMC 2760784. PMID 19584299. {{cite journal}}: Explicit use of et al. in: |author= (help); Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  31. ^ Gao H, Smith J, Yang M; et al. (2010). "Heritable targeted mutagenesis in maize using a designed endonuclease". Plant J. 61 (1): 176–87. doi:10.1111/j.1365-313X.2009.04041.x. PMID 19811621. {{cite journal}}: Explicit use of et al. in: |author= (help); Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  32. ^ Christian M, Cermak T, Doyle EL; et al. (2010). "TAL Effector Nucleases Create Targeted DNA Double-strand Breaks". Genetics. 186 (2): 757–61. doi:10.1534/genetics.110.120717. PMC 2942870. PMID 20660643. {{cite journal}}: Explicit use of et al. in: |author= (help); Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  33. ^ Li T, Huang S, Jiang WZ; et al. (2010). "TAL nucleases (TALNs): hybrid proteins composed of TAL effectors and FokI DNA-cleavage domain". Nucleic Acids Res. 39 (1): 359. doi:10.1093/nar/gkq704. PMC 3017587. PMID 20699274. {{cite journal}}: Explicit use of et al. in: |author= (help); Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  34. ^ S.C. Ekker (2008). "Zinc finger-based knockout punches for zebrafish genes". Zebrafish. 5 (2): 1121–3. doi:10.1089/zeb.2008.9988. PMC 2849655. PMID 18554175.
  35. ^ Geurts AM, Cost GJ, Freyvert Y; et al. (2009). "Knockout rats via embryo microinjection of zinc-finger nucleases". Science. 325 (5939): 433. doi:10.1126/science.1172447. PMC 2831805. PMID 19628861. {{cite journal}}: Explicit use of et al. in: |author= (help); Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  36. ^ Chen I, Dubnau D (2004). "DNA uptake during bacterial transformation". Nat. Rev. Microbiol. 2 (3): 241–9. doi:10.1038/nrmicro844. PMID 15083159.
  37. ^ Graham Head; Hull, Roger H; Tzotzos, George T. (2009). Genetically Modified Plants: Assessing Safety and Managing Risk. London: Academic Pr. p. 244. ISBN 0-12-374106-8.{{cite book}}: CS1 maint: multiple names: authors list (link)
  38. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1128/MMBR.67.1.16-37.2003, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1128/MMBR.67.1.16-37.2003 instead.
  39. ^ Behrooz Darbani, Safar Farajnia, Mahmoud Toorchi, Saeed Zakerbostanabad, Shahin Noeparvar and C. Neal Stewart Jr. (2010). "DNA-Delivery Methods to Produce Transgenic Plants". Science Alert.{{cite web}}: CS1 maint: multiple names: authors list (link)
  40. ^ Barbara Hohn, Avraham A Levy and Holger Puchta (2001). "Elimination of selection markers from transgenic plants". Current Opinion in Biotechnology. 12 (2): 139–143. doi:10.1016/S0958-1669(00)00188-9. PMID 11287227.
  41. ^ John C. Avise (2004). The hope, hype & reality of genetic engineering: remarkable stories from agriculture, industry, medicine, and the environment. Oxford University Press US. p. 22. ISBN 9780195169508.
  42. ^ National Institute of Allergies and Infectious Diseases. "Vaccine Types". national Institute of Health.
  43. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.vaccine.2009.08.039, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.vaccine.2009.08.039 instead.
  44. ^ Roque AC, Lowe CR, Taipa MA. (2004). "Antibodies and genetically engineered related molecules: production and purification". Biotechnol Proress. 20 (3): 639–54. doi:10.1021/bp030070k. PMID 15176864.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  45. ^ "Background: Cloned and Genetically Modified Animals". Center for Genetics and Society. April 14, 2005.
  46. ^ "Knockout Mice". Nation Human Genome Research Institute. 2009.
  47. ^ "GM pigs best bet for organ transplant". Medical News Today. 21 september 2003. {{cite web}}: Check date values in: |date= (help)
  48. ^ "What is Genetic Engineering? A simple introduction". Physicians and scientists for responsible application of science and technology.
  49. ^ "Gene Therapy". Oak Ridge national laboratory. Thursday, June 11, 2009. Retrieved 9/7/2010. {{cite web}}: Check date values in: |accessdate= and |date= (help)
  50. ^ Sheryl Gay (Sunday, July 4, 2010). "Trials are halted on a gene therapy". The New York Times. {{cite news}}: Check date values in: |date= (help)
  51. ^ Emilie R. Bergeson (1997). "The Ethics of Gene Therapy".
  52. ^ Kathi E. Hanna. "Genetic Enhancement". National Human Genome Research Institute.
  53. ^ "Applications of Genetic Engineering". Microbiologyprocedure. Retrieved 9/7/2010. {{cite web}}: Check date values in: |accessdate= (help)
  54. ^ "Biotech: What are transgenic organisms?". Easyscience. 2002. Retrieved 9/7/2010. {{cite web}}: Check date values in: |accessdate= (help)
  55. ^ Savage, Neil (Wednesday, August 01, 2007). "Making Gasoline from Bacteria: A biotech startup wants to coax fuels from engineered microbes". Technology Review. Retrieved 9/7/2010. {{cite web}}: Check date values in: |accessdate= and |date= (help)
  56. ^ "Application of Some Genetically Engineered Bacteria". Retrieved 9/7/2010. {{cite web}}: Check date values in: |accessdate= (help)
  57. ^ Jan Suszkiw (1999.). "Tifton, Georgia: A Peanut Pest Showdown". Agricultural Research magazine. Retrieved 2008-11-23. {{cite web}}: Check date values in: |year= (help); Unknown parameter |month= ignored (help)
  58. ^ Magaña-Gómez JA, de la Barca AM (2009). "Risk assessment of genetically modified crops for nutrition and health". Nutr. Rev. 67 (1): 1–16. doi:10.1111/j.1753-4887.2008.00130.x. PMID 19146501.
  59. ^ Aparna Islam (2006). "Fungus Resistant Transgenic Plants: Strategies, Progress and Lessons Learnt". Plant Tissue Culture and Biotechnology. 16 (2).
  60. ^ "Disease resistant crops". GMO Compass.
  61. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1111/j.1744-7348.2004.tb00376.x, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1111/j.1744-7348.2004.tb00376.x instead.
  62. ^ Deborah B. Whitman (2000). "Genetically Modified Foods: Harmful or Helpful?".
  63. ^ Michelle Marvier (2008). "Pharmaceutical crops in California, benefits and risks. A review". Agron. Sustain. Dev. 28: 1–9. doi:10.1051/agro:2007050.
  64. ^ "Giant GM salmon on the way". BBC News. 2000-04-11.
  65. ^ Emma Young (2003). "GM cows to please cheese-makers". New Scientist.
  66. ^ "FDA Approves First Human Biologic Produced by GE Animals". US Food and Drug Administration.
  67. ^ Paulo Rebêlo (15 July 2004). "GM cow milk 'could provide treatment for blood disease'". SciDev.
  68. ^ a b Sustaining Life. Oxford University Press, Inc. 2008. ISBN 978-0-19-517509-7. {{cite book}}: |access-date= requires |url= (help); Unknown parameter |coauthors= ignored (|author= suggested) (help)
  69. ^ John Pickrell (4 September 2006). "Introduction: GM Organisms". New Scientist.
  70. ^ "20 questions on genetically modified foods". World Health Organization. 2010.
  71. ^ "Can GM crops harm the environment?". National Environment Research Council (NERC).
  72. ^ "New virus-built battery could power cars, electronic devices". Web.mit.edu. 2009-04-02. Retrieved 2010-07-17.
  73. ^ "Hidden Ingredient In New, Greener Battery: A Virus". Npr.org. Retrieved 2010-07-17.
  74. ^ Joab Jackson (December 6, 2005). "Genetically Modified Bacteria Produce Living Photographs". National Geographic News.
  75. ^ "Researchers Synchronize Blinking 'Genetic Clocks' -- Genetically Engineered Bacteria That Keep Track of Time". ScienceDaily. Jan. 24, 2010. {{cite web}}: Check date values in: |date= (help)
  76. ^ Jessica M. Pasko (3/4/2007). "Bio-artists bridge gap between arts, sciences: Use of living organisms is attracting attention and controversy". msnbc. {{cite web}}: Check date values in: |date= (help)
  77. ^ Yukihisa Katsumoto; et al. (October 9, 2007). "Engineering of the Rose Flavonoid Biosynthetic Pathway Successfully Generated Blue-Hued Flowers Accumulating Delphinidin". Plant and Cell Physiology. 48 (11): 1589–1600. doi:10.1093/pcp/pcm131. PMID 17925311. {{cite journal}}: Explicit use of et al. in: |author= (help)
  78. ^ C. Neal Stewart, J (April 2006). "Go with the glow: fluorescent proteins to light transgenic organisms". Trends in Biotechnology. 24 (4): 155–162. doi:10.1016/j.tibtech.2006.02.002. PMID 16488034.
  79. ^ Richard Black. "GM plants 'established in the wild'". BBC.

Further reading

External links