Encyclopedia Britannica

  • History & Society
  • Science & Tech
  • Biographies
  • Animals & Nature
  • Geography & Travel
  • Arts & Culture
  • Games & Quizzes
  • On This Day
  • One Good Fact
  • New Articles
  • Lifestyles & Social Issues
  • Philosophy & Religion
  • Politics, Law & Government
  • World History
  • Health & Medicine
  • Browse Biographies
  • Birds, Reptiles & Other Vertebrates
  • Bugs, Mollusks & Other Invertebrates
  • Environment
  • Fossils & Geologic Time
  • Entertainment & Pop Culture
  • Sports & Recreation
  • Visual Arts
  • Demystified
  • Image Galleries
  • Infographics
  • Top Questions
  • Britannica Kids
  • Saving Earth
  • Space Next 50
  • Student Center
  • Introduction

Historical developments

  • Process and techniques
  • Applications
  • Controversy and ethical issues

genetically engineered salmon

  • Should genetically modified crops be grown?
  • Are genetically modified organisms safe for the environment?
  • What is recombinant DNA technology?
  • When was recombinant DNA technology invented?
  • How is recombinant DNA technology useful?

Rows of tassled corn in a Nebraska field. (agriculture)

genetic engineering

Our editors will review what you’ve submitted and determine whether to revise the article.

  • National Center for Biotechnology Information - PubMed Central - Genetic engineering of animals: Ethical issues, including welfare concerns
  • LiveScience - What's Genetic Engineering?
  • Iowa State University Digital Press - Genetic Engineering
  • Biology LibreTexts - Genetic Engineering
  • University of Marland - Department of English - Interpolations - Genetic Engineering: A Serious Threat to Human Society
  • BMC - Agriculture and Food Security - Genetic engineering for improving quality and productivity of crops
  • Khan Academy - Introduction to genetic engineering
  • OER at MSU Libraries - An Interactive Introduction to Organismal and Molecular Biology, 2nd ed. - Genetic Engineering
  • genetic engineering - Children's Encyclopedia (Ages 8-11)
  • genetic engineering - Student Encyclopedia (Ages 11 and up)
  • Table Of Contents

genetically engineered salmon

Recent News

genetic engineering , the artificial manipulation, modification, and recombination of DNA or other nucleic acid molecules in order to modify an organism or population of organisms. The term genetic engineering is generally used to refer to methods of recombinant DNA technology , which emerged from basic research in microbial genetics . The techniques employed in genetic engineering have led to the production of medically important products, including human insulin , human growth hormone , and hepatitis B vaccine , as well as to the development of genetically modified organisms such as disease-resistant plants.

genetically modified humans

The term genetic engineering initially referred to various techniques used for the modification or manipulation of organisms through the processes of heredity and reproduction . As such, the term embraced both artificial selection and all the interventions of biomedical techniques, among them artificial insemination , in vitro fertilization (e.g., “test-tube” babies), cloning , and gene manipulation. In the latter part of the 20th century, however, the term came to refer more specifically to methods of recombinant DNA technology (or gene cloning ), in which DNA molecules from two or more sources are combined either within cells or in vitro and are then inserted into host organisms in which they are able to propagate .

The possibility for recombinant DNA technology emerged with the discovery of restriction enzymes in 1968 by Swiss microbiologist Werner Arber . The following year American microbiologist Hamilton O. Smith purified so-called type II restriction enzymes , which were found to be essential to genetic engineering for their ability to cleave a specific site within the DNA (as opposed to type I restriction enzymes, which cleave DNA at random sites). Drawing on Smith’s work, American molecular biologist Daniel Nathans helped advance the technique of DNA recombination in 1970–71 and demonstrated that type II enzymes could be useful in genetic studies. Genetic engineering based on recombination was pioneered in 1973 by American biochemists Stanley N. Cohen and Herbert W. Boyer, who were among the first to cut DNA into fragments, rejoin different fragments, and insert the new genes into E. coli bacteria , which then reproduced.

FREE K-12 standards-aligned STEM

curriculum for educators everywhere!

Find more at TeachEngineering.org .

  • TeachEngineering
  • Introduction to Genetic Engineering and Its Applications

Lesson Introduction to Genetic Engineering and Its Applications

Grade Level: 9 (9-12)

(Consider adding 30 minutes for a thorough ethics discussion.)

Lesson Dependency: None

Subject Areas: Biology

NGSS Performance Expectations:

NGSS Three Dimensional Triangle

  • Print lesson and its associated curriculum

Activities Associated with this Lesson Units serve as guides to a particular content or subject area. Nested under units are lessons (in purple) and hands-on activities (in blue). Note that not all lessons and activities will exist under a unit, and instead may exist as "standalone" curriculum.

  • Bacteria Transformation
Lesson Activity

TE Newsletter

Engineering connection, learning objectives, worksheets and attachments, more curriculum like this, pre-req knowledge, introduction/motivation, associated activities, lesson closure, vocabulary/definitions, additional multimedia support, user comments & tips.

Engineers help design and create healthier tomorrows

Genetic engineers have developed genetic recombination techniques to manipulate gene sequences in plants, animals and other organisms to express specific traits. Applications for genetic engineering are increasing as engineers and scientists work together to identify the locations and functions of specific genes in the DNA sequence of various organisms. Once each gene is classified, engineers develop ways to alter them to create organisms that provide benefits such as cows that produce larger volumes of meat, fuel- and plastics-generating bacteria, and pest-resistant crops.

After this lesson, students should be able to:

  • List several present day applications of genetic engineering.
  • Describe general techniques used by genetic engineers to modify DNA.
  • Analyze the benefits and drawbacks of manipulating an organism's DNA.

Educational Standards Each TeachEngineering lesson or activity is correlated to one or more K-12 science, technology, engineering or math (STEM) educational standards. All 100,000+ K-12 STEM standards covered in TeachEngineering are collected, maintained and packaged by the Achievement Standards Network (ASN) , a project of D2L (www.achievementstandards.org). In the ASN, standards are hierarchically structured: first by source; e.g. , by state; within source by type; e.g. , science or mathematics; within type by subtype, then by grade, etc .

Ngss: next generation science standards - science.

NGSS Performance Expectation

HS-LS1-1. Construct an explanation based on evidence for how the structure of DNA determines the structure of proteins which carry out the essential functions of life through systems of specialized cells. (Grades 9 - 12)

Do you agree with this alignment? Thanks for your feedback!

This lesson focuses on the following aspects of NGSS:
Science & Engineering Practices Disciplinary Core Ideas Crosscutting Concepts
Construct an explanation based on valid and reliable evidence obtained from a variety of sources (including students' own investigations, models, theories, simulations, peer review) and the assumption that theories and laws that describe the natural world operate today as they did in the past and will continue to do so in the future.

Alignment agreement: Thanks for your feedback!

Systems of specialized cells within organisms help them perform the essential functions of life.

Alignment agreement: Thanks for your feedback!

All cells contain genetic information in the form of DNA molecules. Genes are regions in the DNA that contain the instructions that code for the formation of proteins, which carry out most of the work of cells.

Alignment agreement: Thanks for your feedback!

Investigating or designing new systems or structures requires a detailed examination of the properties of different materials, the structures of different components, and connections of components to reveal its function and/or solve a problem.

Alignment agreement: Thanks for your feedback!

International Technology and Engineering Educators Association - Technology

View aligned curriculum

Do you agree with this alignment? Thanks for your feedback!

State Standards

Texas - science.

A basic understanding of protein synthesis and DNA's role in the cell/body is helpful so students can follow how changes in DNA result in major changes in the characteristics of organisms.

(Make copies of the Genetic Engineering Flow Chart , one per student. Hand out the blank flow charts for students to fill in during the presentation and lecture. Then show the class the 16-slide Genetic Engineering Presentation , a PowerPoint® file. Open with two images of the same organism: one that has been genetically engineered and one that has not. Examples: two ears of corn in which the non-modified one is diseased; two cows in which the modified one is larger; or, since students really respond to bioluminescent organisms, show two mice in which one has been modified to glow green. Slide 2 shows two examples of modified versus non-modified mice. Another idea is to show two organisms that look the same even though one has been modified as an example of how most modifications are not visible.)

What is the difference between these two organisms? (Answers will vary, depending on the image shown.) Even though they are the same organism, why are they are different? (Answer: Genetic engineering. Some students may not come to this answer on their own. Expect some to suggest mutations.) The difference is due to genetic engineering. The animal (or plant) that has been changed is called a genetically modified organism, or GMO.

How do engineers change the traits of organisms? (Listen to student ideas.) DNA contains all of the genetic information to determine an organism's traits or characteristics. By modifying the DNA, engineers are able to determine which traits an organism will possess.

(Continue through the presentation: What is genetic engineering? History of GMO Development, What is the GMO process? Then starting with slide 6 , go through the provided examples of GMO bacteria, plants and animals. Emphasize the reasons for modifying each organism [ slide 10 ].)

(Show the slide 14 picture of a man and spider.) Can anyone guess what would happen if we combined the DNA from these two creatures? (Expect students to enthusiastically answer "spiderman.") Could engineers create a "spiderman" in the lab today? (Expect some yes responses, while most students answer no.) Not quite. However, in 2000, engineers created the first goat able to produce spider silk proteins (an amazingly strong and elastic fiber with futuristic benefits in construction [bridge suspension cables, airbags that are gentler for passengers], medicine [artificial skin to heal burns, artificial ligaments, thread for stitching wounds] and the military [body armor] if sufficient quantities could be generated), so maybe it is not too far away.

(Show slide 15 .) Genetic engineering is so new and astonishing that people are still trying to figure out the pros and cons. We saw some examples of the benefits from genetically modified organisms, what about the disadvantages and harm caused by genetic engineering? (After listening to student ideas, go through the concerns listed on the slide. Alternatively, go through the contents of this slide and background information as a class discussion during the Lesson Closure, extending the lesson time as necessary.)

(Continue on to present students with the content in the Lesson Background section, and then a class review of the completed flow charts.)

Lesson Background and Concepts for Teachers

A drawing shows a double helix DNA molecule that looks like a "twisted ladder" with color and letter identifiers composing each structural sub-component.

What is DNA?

Deoxyribonucleic acid (DNA) is a large biomolecule that contains the complete genetic information for an organism. Every cell of living organisms and many viruses, contains DNA. The basic building block of a DNA molecule is called a nucleotide , and a single strand of DNA may contain billions of nucleotides. (Refer to Figure 1 to see the DNA structure with labeled parts.) Although each DNA molecule contains many of these building blocks, only four unique nucleotides are used to create the entire DNA sequence; these are written as A, G, C and T. Analogous to how the 26 letters of the alphabet can be arranged to create words with different meanings, these four nucleotides can be arranged in sequences to "spell" the genetic instructions to create all of the different proteins organisms need to live.

A line drawing shows a long DNA molecule (double helix-shaped) with a small segment selected as a gene.

Why are proteins important?

Proteins perform all of the work in organisms. Some functions of proteins include:

  • Serving as catalysts for reactions
  • Performing cell signaling
  • Transporting molecules across membranes
  • Creating structures

When a protein is created by its gene, it is said that the gene is "expressed," or used. Most gene expressions do not produce results visible to the unaided eye. However some genes, such as those that code for proteins responsible for pigment, do have visual expression. The expression of a gene in an observable manner is called a phenotypic trait ; one example is an organism's hair color. In fact, everything you can see in an organism is a result of proteins or protein actions.

How is DNA used in genetic engineering?

A diagram titled "Genetically Engineered Animals," shows a six-step process to create a modified goat capable of producing a therapeutic protein.

By definition, genetic engineering is the direct altering of an organism's genome. This is achieved through manipulation of the DNA. Doing this is possible because DNA is like a universal language; all DNA for all organisms is made up of the same nucleotide building blocks. Thus, it is possible for genes from one organism to be read by another organism. In the cookbook analogy, this equates to taking a recipe from one organism's cookbook and putting into another cookbook. Now imagine that all cookbooks are written in the same language; thus, any recipe can be inserted and used in any other cookbook.

In practice, since DNA contains the genes to build certain proteins, by changing the DNA sequence, engineers are able to provide a new gene for a cell/organism to create a different protein. The new instructions may supplement the old instructions such that an extra trait is exhibited, or they may completely replace the old instructions such that a trait is changed.

Genetic Engineering Technique

The process for genetic engineering begins the same for any organism being modified (see Figure 3 for an example of this procedure).

  • Identify an organism that contains a desirable gene.
  • Extract the entire DNA from the organism.
  • Remove this gene from the rest of the DNA. One way to do this is by using a restriction enzyme . These enzymes search for specific nucleotide sequences where they will "cut" the DNA by breaking the bonds at this location.
  • Insert the new gene to an existing organism's DNA. This may be achieved through a number of different processes.

A drawing shows a selected gene is used to build a recombinant plasmid, which is inserted into a bacteria.

Once the recombinant DNA has been built, it can be passed to the organism to be modified. If modifying bacteria, this process is quite simple. The plasmid can be easily inserted into the bacteria where the bacteria treat it as their own DNA. For plant modification, certain bacteria such as Agrobacterium tumefaciens may be used because these bacteria permit their plasmids to be passed to the plant's DNA.

Applications and Economics

The number of applications for genetic engineering are increasing as more and more is learned about the genomes of different organisms. A few interesting or notable application areas are described below.

How many of today's crops are genetically modified? As of 2010, in the U.S., 86% of corn produced was genetically modified. Bt -corn is a common GMO that combines a gene from the Bt bacteria with corn DNA to produce a crop that is insect-resistant. The bacteria gene used contains a recipe for a protein that is toxic when consumed by insects, but safe when consumed by humans.

A number of other genes can be combined with crops to produce desirable properties such as:

  • Herbicide-, drought-, freeze- or disease-resistance
  • Higher yield
  • Faster growth
  • Improved nutrition
  • Longer shelf life

The creation of genetically modified crops provides many incentives for farmers and businesses. When farmers are able to plant a crop that has a higher yield per acre, they can significantly increase production, and thus sales, with minimal cost. Disease, pest and other resistances reduce crop loss, which also helps to increase profits. Besides farmers, other benefactors from modified crops include seed, agrochemical and agriculture equipment companies as well as distributors and universities that are involved in GMO research. In 2011, the value of genetically modified seed was $13.2 billion in the U.S. alone. The value of the end products produced from these seeds topped $160 billion.

Due to their simple structures, the most commonly modified organisms are bacteria. The first modified bacteria were created in 1973. Bacteria can be modified to produce desirable proteins that can be harvested and used. One example is insulin or spider silk, which is difficult to gather naturally. Other modifications to bacteria include making changes to the cellular respiration process to alter the byproducts; typically CO 2 is produced, however engineers have made modifications so that hydrocarbon byproducts such as diesel and polyethylene (a fuel and a plastic) are produced.

(The 30-minute lesson time leaves a fair amount of time for discussion, but since class participation will vary, you may want to extend the lesson another 30-minutes to allow for a thorough discussion of the ethical implications of genetic engineering. This makes a good student research and debate topic, too.)

The main reason genetically modified organisms are not more widely used is due to ethical concerns. Nearly 50 countries around the world, including Australia, Japan and all of the countries in the European Union, have enacted significant restrictions or full bans on the production and sale of genetically modified organism food products, and 64 countries have GMO labeling requirements. Some issues to consider when deciding whether to create and/or use GMOs include:

Safety: This generally arises in the case of GMO foods. Are the foods safe for human consumption? Is GMO feed healthy for animals? Many opponents of GMO foods say not enough independent testing is done before the food is approved for sale to consumers. In general, research has shown that GMO foods are safe for humans. Another safety consideration is the health of farmers and their families, animals and communities who are put at risk with exposure to chemicals used in tandem with GMO seeds.

Environmental Impact: Consider that genetic engineers have the ability to create trees that grow faster than their unmodified counterparts. This seems like a great deal for the lumber industry, but might some unintended consequences result? Being outdoors and grown in large quantities, the modified trees may cross-pollinate with unmodified trees to form hybrids outside of designated growing areas. This in return could create trees that could disrupt the ecosystem. For example, they could overpopulate the area or grow so large that they smother other plant life. This same scenario has unintended and undesirable consequences when the pollen from GMO crops drifts into non-GMO fields.

Humans: Should humans be genetically engineered? Doing so could have medical applications that reduce or prevent genetic disorders such as Down's syndrome. However, the bigger question is where should engineering humans stop? Should parents be allowed to decide their children's eye colors, heights or even genders before birth?

Watch this activity on YouTube

What part of an organism contains all of the information needed for it to function? (Answer: DNA) When genes are expressed, what is the final product made? (Answer: Proteins) Does anyone know why bacteria are modified more than other organisms? (Answer: With their very simple structures and ability to use plasmids, bacteria are much easier and less costly to modify.)

What are some ethical and moral concerns that genetic engineers must consider? Does anyone think it is a good idea to genetically modify people? Some researchers say this could be an approach to cure diseases such as Down's syndrome and other genetic defects. Superficial changes could also be made, such as determining a person's height, eye color or gender, by making changes to embryos in the mothers' wombs. But just because something can be done, does that make it a good idea? (Answer: No. This is a good topic for an extended discussion.)

DNA: Acronym for deoxyribonucleic acid, which is a molecule that contains an organism's complete genetic information.

gene: The molecular unit of an organism that contains information for a specific trait (specific DNA sequence).

genome: An entire set of genes for an organism.

GMO: Acronym for genetically modified organism.

nucleotide: The building block of DNA.

plasmid: The circular DNA structure used by bacteria.

protein: Large biomolecules used by an organism for a number of purposes; in this context, to express a desired trait.

recombinant DNA: DNA to which a section has been removed and replaced (recombined) with a new sequence.

restriction enzyme: An enzyme that "cuts" DNA when specific base pair sequences are present.

trait: A distinguishing characteristic.

Pre-Lesson Assessment

Discussion Questions: Initiate a brief discussion to gauge whether students have heard of or know anything about genetics. Ask questions such as:

  • Why are your eyes the color that they are?
  • Would anyone like to be taller (or shorter)?
  • Is there any way to make these changes?

Post-Introduction Assessment

Flow Chart: Have students complete the Genetic Engineering Flow Chart during the course of the lesson. After delivering the presentation and lecture, go through the flow chart as a class, so that students can complete anything they missed and check their flow charts for accuracy. Answers are provided on the Genetic Engineering Flow Chart Answer Key .

Lesson Summary Assessment

Recombinant Creature Design : Have students in pairs (or individually) create their own recombinant organisms. Direct students to pick any organism and decide what gene they would like to add. If desired, provide a list of genes from which they can choose (such as genes that makes an organism smarter, bigger, faster, grow extra limbs, etc.). To encourage critical thinking, require students to write down a potential use for the resulting creatures. Finally, have students sketch what their recombinant creatures would look like.

View some genetic engineering examples (with photographs) at: http://www.mnn.com/green-tech/research-innovations/photos/12-bizarre-examples-of-genetic-engineering/

Show students some applications of spider silk at Popular Mechanics' "6 Spider-Silk Superpowers" slide show at http://www.popularmechanics.com/science/health/med-tech/6-spider-silk-superpowers#slide-1

introduction of genetic engineering essay

As a class, students work through an example showing how DNA provides the "recipe" for making human body proteins. They see how the pattern of nucleotide bases (adenine, thymine, guanine, cytosine) forms the double helix ladder shape of DNA, and serves as the code for the steps required to make gene...

preview of 'DNA: The Human Body Recipe' Lesson

Students learn about mutations to both DNA and chromosomes, and uncontrolled changes to the genetic code. They are introduced to small-scale mutations (substitutions, deletions and insertions) and large-scale mutations (deletion duplications, inversions, insertions, translocations and nondisjunction...

preview of 'All Sorts of Mutations: Changes in the Genetic Code' Lesson

Students reinforce their knowledge that DNA is the genetic material for all living things by modeling it using toothpicks and gumdrops that represent the four biochemicals (adenine, thiamine, guanine, and cytosine) that pair with each other in a specific pattern, making a double helix. Student teams...

preview of 'DNA Build' Activity

Students construct paper recombinant plasmids to simulate the methods genetic engineers use to create modified bacteria. They learn what role enzymes, DNA and genes play in the modification of organisms.

preview of 'Bacteria Transformation' Activity

12 Bizarre Examples of Genetic Engineering. Posted October 27, 2010. MNN Holdings, Mother Nature Network. Accessed December 8, 2013. http://www.mnn.com/green-tech/research-innovations/photos/12-bizarre-examples-of-genetic-engineering

Biello, David. Turning Bacteria into Plastic Factories. Posted September 16, 2008. Scientific American. Accessed December 11, 2013. http://www.scientificamerican.com/article.cfm?id=turning-bacteria-into-plastic-factories-replacing-fossil-fuels

DNA. Updated June 7, 2014. Wikipedia, The Free Encyclopedia. Accessed June 16, 2014. http://en.wikipedia.org/wiki/DNA

Emspak, Jesse. Gut Bacteria Make Diesel Fuel. Posted April 23, 2013. Discovery Communications. Accessed December 11, 2013. http://news.discovery.com/tech/biotechnology/gut-bacteria-make-diesel-fuel-130423.htm

Genetic engineering. Updated December 7, 2013. Wikipedia, The Free Encyclopedia. Accessed December 9, 2013. http://en.wikipedia.org/wiki/Genetic_engineering

Genetically modified crops. Updated June 12, 2014. Wikipedia, The Free Encyclopedia. Accessed June 16, 2014. http://en.wikipedia.org/wiki/Genetically_modified_crops

Straley, Regan. GMO Food Concerns. Posted August 29, 2014. Lancaster Online, Lancaster, PA. Accessed August 31, 2014. http://lancasteronline.com/opinion/gmo-food-concerns/article_3c5092ba-2ed0-11e4-ab00-001a4bcf6878.html

Vierra, Craig, et al. The Future of Biomaterial Manufacturing: Spider Silk Production from Bacteria. Posted July 17, 2012. Journal of Visualized Experiments (JoVE). Accessed December 11, 2013. http://www.jove.com/about/press-releases/39/the-future-biomaterial-manufacturing-spider-silk-production-from

What is genetic engineering and how does it work? Updated 2005. University of Nebraska. Accessed December 10, 2013. http://agbiosafety.unl.edu/basic_genetics.shtml

Other Related Information

(optional: Show students the What Is Engineering? video)

Contributors

Supporting program, acknowledgements.

This digital library content was developed by the University of Houston's College of Engineering under National Science Foundation GK-12 grant number DGE 0840889. However, these contents do not necessarily represent the policies of the NSF and you should not assume endorsement by the federal government.

Last modified: May 12, 2021

  • DOI: 10.1017/CBO9780511800986
  • Corpus ID: 31472912

An Introduction to Genetic Engineering

  • Published 10 February 2023

Figures and Tables from this paper

figure 1.1

103 Citations

Genetic engineering and its applications: a way to engineer and manipulate genes, chapter six gene cloning, genetic engineering and cloning: focus on animal biotechnology, an overview of the importance of bacterial elements for plant genetic engineering.

  • Highly Influenced
  • 10 Excerpts

Comparative Genomics in Eukaryotes

Genetic engineering in animal production: applications and prospects, molecular cloning and recombinant dna technology, designing genetic engineering technologies for human values, discussion on the implications upon the environment of some technologies from the second green revolution, unveiling the molecular symphony: exploring mechanisms, diversity and applications of restriction enzymes in biology, related papers.

Showing 1 through 3 of 0 Related Papers

This page has been archived and is no longer updated

Genetically Modified Organisms (GMOs): Transgenic Crops and Recombinant DNA Technology

introduction of genetic engineering essay

People have been altering the genomes of plants and animals for many years using traditional breeding techniques. Artificial selection for specific, desired traits has resulted in a variety of different organisms, ranging from sweet corn to hairless cats. But this artificial selection , in which organisms that exhibit specific traits are chosen to breed subsequent generations, has been limited to naturally occurring variations. In recent decades, however, advances in the field of genetic engineering have allowed for precise control over the genetic changes introduced into an organism . Today, we can incorporate new genes from one species into a completely unrelated species through genetic engineering, optimizing agricultural performance or facilitating the production of valuable pharmaceutical substances. Crop plants, farm animals, and soil bacteria are some of the more prominent examples of organisms that have been subject to genetic engineering.

Current Use of Genetically Modified Organisms

Table 1: Examples of GMOs Resulting from Agricultural Biotechnology

Herbicide tolerance Soybean Glyphosate herbicide (Roundup) tolerance conferred by expression of a glyphosate-tolerant form of the plant enzyme 5-enolpyruvylshikimate-3-phosphate synthase (EPSPS) isolated from the soil bacterium , strain CP4 Insect resistance Corn Resistance to insect pests, specifically the European corn borer, through expression of the insecticidal protein Cry1Ab from Altered fatty acid composition Canola High laurate levels achieved by inserting the gene for ACP thioesterase from the California bay tree Virus resistance Plum Resistance to plum pox virus conferred by insertion of a coat protein (CP) gene from the virus Vitamin enrichment Rice Three genes for the manufacture of beta-carotene, a precursor to vitamin A, in the endosperm of the rice prevent its removal (from husks) during milling Vaccines Tobacco Hepatitis B virus surface antigen (HBsAg) produced in transgenic tobacco induces immune response when injected into mice Oral vaccines Maize Fusion protein (F) from Newcastle disease virus (NDV) expressed in corn seeds induces an immune response when fed to chickens Faster maturation Coho salmon A type 1 growth hormone gene injected into fertilized fish eggs results in 6.2% retention of the vector at one year of age, as well as significantly increased growth rates

The pharmaceutical industry is another frontier for the use of GMOs. In 1986, human growth hormone was the first protein pharmaceutical made in plants (Barta et al ., 1986), and in 1989, the first antibody was produced (Hiatt et al ., 1989). Both research groups used tobacco, which has since dominated the industry as the most intensively studied and utilized plant species for the expression of foreign genes (Ma et al ., 2003). As of 2003, several types of antibodies produced in plants had made it to clinical trials. The use of genetically modified animals has also been indispensible in medical research. Transgenic animals are routinely bred to carry human genes, or mutations in specific genes, thus allowing the study of the progression and genetic determinants of various diseases.

Potential GMO Applications

Many industries stand to benefit from additional GMO research. For instance, a number of microorganisms are being considered as future clean fuel producers and biodegraders. In addition, genetically modified plants may someday be used to produce recombinant vaccines. In fact, the concept of an oral vaccine expressed in plants (fruits and vegetables) for direct consumption by individuals is being examined as a possible solution to the spread of disease in underdeveloped countries, one that would greatly reduce the costs associated with conducting large-scale vaccination campaigns. Work is currently underway to develop plant-derived vaccine candidates in potatoes and lettuce for hepatitis B virus (HBV), enterotoxigenic Escherichia coli (ETEC), and Norwalk virus. Scientists are also looking into the production of other commercially valuable proteins in plants, such as spider silk protein and polymers that are used in surgery or tissue replacement (Ma et al ., 2003). Genetically modified animals have even been used to grow transplant tissues and human transplant organs, a concept called xenotransplantation. The rich variety of uses for GMOs provides a number of valuable benefits to humans, but many people also worry about potential risks.

Risks and Controversies Surrounding the Use of GMOs

Despite the fact that the genes being transferred occur naturally in other species, there are unknown consequences to altering the natural state of an organism through foreign gene expression . After all, such alterations can change the organism's metabolism , growth rate, and/or response to external environmental factors. These consequences influence not only the GMO itself, but also the natural environment in which that organism is allowed to proliferate. Potential health risks to humans include the possibility of exposure to new allergens in genetically modified foods, as well as the transfer of antibiotic-resistant genes to gut flora.

Horizontal gene transfer of pesticide, herbicide, or antibiotic resistance to other organisms would not only put humans at risk , but it would also cause ecological imbalances, allowing previously innocuous plants to grow uncontrolled, thus promoting the spread of disease among both plants and animals. Although the possibility of horizontal gene transfer between GMOs and other organisms cannot be denied, in reality, this risk is considered to be quite low. Horizontal gene transfer occurs naturally at a very low rate and, in most cases, cannot be simulated in an optimized laboratory environment without active modification of the target genome to increase susceptibility (Ma et al ., 2003).

In contrast, the alarming consequences of vertical gene transfer between GMOs and their wild-type counterparts have been highlighted by studying transgenic fish released into wild populations of the same species (Muir & Howard, 1999). The enhanced mating advantages of the genetically modified fish led to a reduction in the viability of their offspring . Thus, when a new transgene is introduced into a wild fish population, it propagates and may eventually threaten the viability of both the wild-type and the genetically modified organisms.

Unintended Impacts on Other Species: The Bt Corn Controversy

One example of public debate over the use of a genetically modified plant involves the case of Bt corn. Bt corn expresses a protein from the bacterium Bacillus thuringiensis . Prior to construction of the recombinant corn, the protein had long been known to be toxic to a number of pestiferous insects, including the monarch caterpillar, and it had been successfully used as an environmentally friendly insecticide for several years. The benefit of the expression of this protein by corn plants is a reduction in the amount of insecticide that farmers must apply to their crops. Unfortunately, seeds containing genes for recombinant proteins can cause unintentional spread of recombinant genes or exposure of non-target organisms to new toxic compounds in the environment.

The now-famous Bt corn controversy started with a laboratory study by Losey et al . (1999) in which the mortality of monarch larvae was reportedly higher when fed with milkweed (their natural food supply) covered in pollen from transgenic corn than when fed milkweed covered with pollen from regular corn. The report by Losey et al . was followed by another publication (Jesse & Obrycki, 2000) suggesting that natural levels of Bt corn pollen in the field were harmful to monarchs.

Debate ensued when scientists from other laboratories disputed the study, citing the extremely high concentration of pollen used in the laboratory study as unrealistic, and concluding that migratory patterns of monarchs do not place them in the vicinity of corn during the time it sheds pollen. For the next two years, six teams of researchers from government, academia, and industry investigated the issue and concluded that the risk of Bt corn to monarchs was "very low" (Sears et al ., 2001), providing the basis for the U.S. Environmental Protection Agency to approve Bt corn for an additional seven years.

Unintended Economic Consequences

Another concern associated with GMOs is that private companies will claim ownership of the organisms they create and not share them at a reasonable cost with the public. If these claims are correct, it is argued that use of genetically modified crops will hurt the economy and environment, because monoculture practices by large-scale farm production centers (who can afford the costly seeds) will dominate over the diversity contributed by small farmers who can't afford the technology. However, a recent meta-analysis of 15 studies reveals that, on average, two-thirds of the benefits of first-generation genetically modified crops are shared downstream, whereas only one-third accrues upstream (Demont et al ., 2007). These benefit shares are exhibited in both industrial and developing countries. Therefore, the argument that private companies will not share ownership of GMOs is not supported by evidence from first-generation genetically modified crops.

GMOs and the General Public: Philosophical and Religious Concerns

In a 2007 survey of 1,000 American adults conducted by the International Food Information Council (IFIC), 33% of respondents believed that biotech food products would benefit them or their families, but 23% of respondents did not know biotech foods had already reached the market. In addition, only 5% of those polled said they would take action by altering their purchasing habits as a result of concerns associated with using biotech products.

According to the Food and Agriculture Organization of the United Nations, public acceptance trends in Europe and Asia are mixed depending on the country and current mood at the time of the survey (Hoban, 2004). Attitudes toward cloning, biotechnology, and genetically modified products differ depending upon people's level of education and interpretations of what each of these terms mean. Support varies for different types of biotechnology; however, it is consistently lower when animals are mentioned.

Furthermore, even if the technologies are shared fairly, there are people who would still resist consumable GMOs, even with thorough testing for safety, because of personal or religious beliefs. The ethical issues surrounding GMOs include debate over our right to "play God," as well as the introduction of foreign material into foods that are abstained from for religious reasons. Some people believe that tampering with nature is intrinsically wrong, and others maintain that inserting plant genes in animals, or vice versa, is immoral. When it comes to genetically modified foods, those who feel strongly that the development of GMOs is against nature or religion have called for clear labeling rules so they can make informed selections when choosing which items to purchase. Respect for consumer choice and assumed risk is as important as having safeguards to prevent mixing of genetically modified products with non-genetically modified foods. In order to determine the requirements for such safeguards, there must be a definitive assessment of what constitutes a GMO and universal agreement on how products should be labeled.

These issues are increasingly important to consider as the number of GMOs continues to increase due to improved laboratory techniques and tools for sequencing whole genomes, better processes for cloning and transferring genes, and improved understanding of gene expression systems. Thus, legislative practices that regulate this research have to keep pace. Prior to permitting commercial use of GMOs, governments perform risk assessments to determine the possible consequences of their use, but difficulties in estimating the impact of commercial GMO use makes regulation of these organisms a challenge.

History of International Regulations for GMO Research and Development

In 1971, the first debate over the risks to humans of exposure to GMOs began when a common intestinal microorganism, E. coli , was infected with DNA from a tumor-inducing virus (Devos et al ., 2007). Initially, safety issues were a concern to individuals working in laboratories with GMOs, as well as nearby residents. However, later debate arose over concerns that recombinant organisms might be used as weapons. The growing debate, initially restricted to scientists, eventually spread to the public, and in 1974, the National Institutes of Health (NIH) established the Recombinant DNA Advisory Committee to begin to address some of these issues.

In the 1980s, when deliberate releases of GMOs to the environment were beginning to occur, the U.S. had very few regulations in place. Adherence to the guidelines provided by the NIH was voluntary for industry. Also during the 1980s, the use of transgenic plants was becoming a valuable endeavor for production of new pharmaceuticals, and individual companies, institutions, and whole countries were beginning to view biotechnology as a lucrative means of making money (Devos et al ., 2007). Worldwide commercialization of biotech products sparked new debate over the patentability of living organisms, the adverse effects of exposure to recombinant proteins, confidentiality issues, the morality and credibility of scientists, the role of government in regulating science, and other issues. In the U.S., the Congressional Office of Technology Assessment initiatives were developed, and they were eventually adopted worldwide as a top-down approach to advising policymakers by forecasting the societal impacts of GMOs.

Then, in 1986, a publication by the Organization for Economic Cooperation and Development (OECD), called "Recombinant DNA Safety Considerations," became the first intergovernmental document to address issues surrounding the use of GMOs. This document recommended that risk assessments be performed on a case-by-case basis. Since then, the case-by-case approach to risk assessment for genetically modified products has been widely accepted; however, the U.S. has generally taken a product-based approach to assessment, whereas the European approach is more process based (Devos et al ., 2007). Although in the past, thorough regulation was lacking in many countries, governments worldwide are now meeting the demands of the public and implementing stricter testing and labeling requirements for genetically modified crops.

Increased Research and Improved Safety Go Hand in Hand

Proponents of the use of GMOs believe that, with adequate research, these organisms can be safely commercialized. There are many experimental variations for expression and control of engineered genes that can be applied to minimize potential risks. Some of these practices are already necessary as a result of new legislation, such as avoiding superfluous DNA transfer (vector sequences) and replacing selectable marker genes commonly used in the lab (antibiotic resistance) with innocuous plant-derived markers (Ma et al ., 2003). Issues such as the risk of vaccine-expressing plants being mixed in with normal foodstuffs might be overcome by having built-in identification factors, such as pigmentation, that facilitate monitoring and separation of genetically modified products from non-GMOs. Other built-in control techniques include having inducible promoters (e.g., induced by stress, chemicals, etc.), geographic isolation, using male-sterile plants, and separate growing seasons.

GMOs benefit mankind when used for purposes such as increasing the availability and quality of food and medical care, and contributing to a cleaner environment. If used wisely, they could result in an improved economy without doing more harm than good, and they could also make the most of their potential to alleviate hunger and disease worldwide. However, the full potential of GMOs cannot be realized without due diligence and thorough attention to the risks associated with each new GMO on a case-by-case basis.

References and Recommended Reading

Barta, A., et al . The expression of a nopaline synthase-human growth hormone chimaeric gene in transformed tobacco and sunflower callus tissue. Plant Molecular Biology 6 , 347–357 (1986)

Beyer, P., et al . Golden rice: Introducing the β-carotene biosynthesis pathway into rice endosperm by genetic engineering to defeat vitamin A deficiency. Journal of Nutrition 132 , 506S–510S (2002)

Demont, M., et al . GM crops in Europe: How much value and for whom? EuroChoices 6 , 46–53 (2007)

Devlin, R., et al . Extraordinary salmon growth. Nature 371 , 209–210 (1994) ( link to article )

Devos, Y., et al . Ethics in the societal debate on genetically modified organisms: A (re)quest for sense and sensibility. Journal of Agricultural and Environmental Ethics 21 , 29–61 (2007) doi:10.1007/s10806-007-9057-6

Guerrero-Andrade, O., et al . Expression of the Newcastle disease virus fusion protein in transgenic maize and immunological studies. Transgenic Research 15 , 455–463(2006) doi:10.1007/s11248-006-0017-0

Hiatt, A., et al . Production of antibodies in transgenic plants. Nature 342 , 76–79 (1989) ( link to article )

Hoban, T. Public attitudes towards agricultural biotechnology. ESA working papers nos. 4-9. Agricultural and Development Economics Division, Food and Agricultural Organization of the United Nations (2004)

Jesse, H., & Obrycki, J. Field deposition of Bt transgenic corn pollen: Lethal effects on the monarch butterfly. Oecologia 125 , 241–248 (2000)

Losey, J., et al . Transgenic pollen harms monarch larvae. Nature 399 , 214 (1999) doi:10.1038/20338 ( link to article )

Ma, J., et al . The production of recombinant pharmaceutical proteins in plants. Nature Reviews Genetics 4 , 794–805 (2003) doi:10.1038/nrg1177 ( link to article )

Muir, W., & Howard, R. Possible ecological risks of transgenic organism release when transgenes affect mating success: Sexual selection and the Trojan gene hypothesis. Proceedings of the National Academy of Sciences 96 , 13853–13856 (1999)

Sears, M., et al . Impact of Bt corn on monarch butterfly populations: A risk assessment. Proceedings of the National Academy of Sciences 98 , 11937–11942 (2001)

Spurgeon, D. Call for tighter controls on transgenic foods. Nature 409 , 749 (2001) ( link to article )

Takeda, S., & Matsuoka, M. Genetic approaches to crop improvement: Responding to environmental and population changes. Nature Reviews Genetics 9 , 444–457 (2008) doi:10.1038/nrg2342 ( link to article )

United States Department of Energy, Office of Biological and Environmental Research, Human Genome Program. Human Genome Project information: Genetically modified foods and organisms, (2007)

  • Add Content to Group

Article History

Flag inappropriate.

Google Plus+

StumbleUpon

Email your Friend

introduction of genetic engineering essay

  •  |  Lead Editor:  Bob Moss

Topic Rooms

Within this Subject (34)

  • Applications in Biotechnology (4)
  • Discovery of Genetic Material (4)
  • DNA Replication (6)
  • Gene Copies (5)
  • Jumping Genes (4)
  • RNA (7)
  • Transcription & Translation (4)

Other Topic Rooms

  • Gene Inheritance and Transmission
  • Gene Expression and Regulation
  • Nucleic Acid Structure and Function
  • Chromosomes and Cytogenetics
  • Evolutionary Genetics
  • Population and Quantitative Genetics
  • Genes and Disease
  • Genetics and Society
  • Cell Origins and Metabolism
  • Proteins and Gene Expression
  • Subcellular Compartments
  • Cell Communication
  • Cell Cycle and Cell Division

ScholarCast

© 2014 Nature Education

  • Press Room |
  • Terms of Use |
  • Privacy Notice |

Send

Visual Browse

  • IAS Preparation
  • UPSC Preparation Strategy
  • Genetic Engineering

Genetic Engineering [UPSC Notes]

Genetic engineering, also called genetic modification, is the direct manipulation of an organism’s genome using biotechnology. It is a set of technologies used to change the genetic makeup of cells, including the transfer of genes within and across species boundaries to produce improved or novel organisms. Read important facts about Genetic Engineering in this article for the IAS Exam .

by following the below-mentioned links:

What is Genetic Engineering

  • In simple words, genetic engineering can be described as the manual addition of a new DNA into an organism.
  •  It aids the addition of such traits that are not originally found in the organisms.
  • Recombinant DNA is required to create Genetically Modified Organisms (GMO.)
  • An area of chromosome (gene) is spliced.
  • Genetic disorders in humans can be corrected using genetic engineering.
  • Selective breeding has been in the world since ancient times.
  • Jack Williamson used the word ‘Genetic Engineering’ in his science fiction novel Dragon’s Island which was published in 1951.
  • First recombinant DNA molecules were created by an American Biochemist, Paul Berg.

New DNA may be inserted in the host genome by first isolating and copying the genetic material of interest using molecular cloning methods to generate a DNA sequence, or by synthesizing the DNA and then inserting this construct into the host organism. Genes may be removed, or “knocked out”, using a nuclease. Gene targeting is a different technique that uses homologous recombination to change an endogenous gene and can be used to delete a gene, remove exons, add a gene, or introduce point mutations.

Aspirants reading, ‘GEAC’ can also refer to topics lined below:

Applications of Genetic Engineering

Medicine, research, industry and agriculture are a few sectors where genetic engineering applies. It can be used on various plants, animals and microorganisms. The first microorganism to be genetically modified is bacteria.

  • Manufacturing of drugs
  • Creation of model animals that mimic human conditions and,
  • Gene therapy
  • Human growth hormones
  • Follicle-stimulating hormones
  • Human albumin
  • Monoclonal antibodies
  • Antihemophilic factors
  • In Research: Genes and other genetic information from a wide range of organisms can be inserted into bacteria for storage and modification, creating genetically modified bacteria in the process.
  • Transformation of cells in organisms with a gene coding to get a useful protein.
  • Medicines like insulin, human growth hormone, and vaccines, supplements such as tryptophan, aid in the production of food (chymosin in cheese making) and fuels are produced using such techniques.
  • Genetically modified crops are produced using genetic engineering in agriculture.
  • Such crops are produced that provide protection from insect pests.
  • It is used or can be used in the creation of fungal and virus-resistant crops.
  • Conservation
  • Natural area management
  • Microbial art

Benefits of Genetic Engineering

  • The production of genetically modified crops is a boon to agriculture.
  • The crops that are drought-resistant, disease-resistant can be grown with it.
  • As described earlier, genetic disorders can be treated.
  • The diseases such as malaria, dengue can be eliminated by sterilising the mosquitoes using genetic engineering.
  • Therapeutic cloning

Challenges of Genetic Engineering

  • The production of genetically-engineered entities may result in an adverse manner and produce undesired results which are unforeseen.
  • With the introduction of a genetically-engineered entity into one ecosystem for a desirable result, may lead to distortion of the existing biodiversity.
  • Genetically-engineered crops can also produce adverse health effects.
  • The concept of genetic-engineering is debated for its bioethics where community against it argue over the right of distorting or moulding the nature as per our needs.

Regulations in India

Genetic Engineering Appraisal Committee (GEAC) is the biotech regulator in India. It is created under the Ministry of Environment and Forests. Read more about GEAC in the linked article.

There are five bodies that are authorized to handle rules noted under Environment Protection Act 1986 “Rules for Manufacture, Use, Import, Export and Storage of Hazardous Microorganisms/Genetically Engineered Organisms or Cells 1989”. These are:

  • Institutional Biosafety Committees (IBSC)
  • Review Committee of Genetic Manipulation (RCGM)
  • Genetic Engineering Approval Committee (GEAC)
  • State Biotechnology Coordination Committee (SBCC) and
  • District Level Committee (DLC)

Which are the genetically modified crops in India?

  • Bt Cotton is the genetically modified crop that is under cultivation in India.
  • Bt Brinjal was initially approved but later was blocked from production.
  • GM Mustard is yet to be allowed for cultivated. It will be the first genetically modified food crop in the country.

FAQ about Genetic Engineering

What are some examples of genetic engineering, what are the pros and cons of genetic engineering.

UPSC Preparation:

IAS General Studies Notes Links

Leave a Comment Cancel reply

Your Mobile number and Email id will not be published. Required fields are marked *

Request OTP on Voice Call

Post My Comment

introduction of genetic engineering essay

IAS 2024 - Your dream can come true!

Download the ultimate guide to upsc cse preparation, register with byju's & download free pdfs, register with byju's & watch live videos.

EDUCBA

Essay on Genetic Engineering

Narayan Bista

Introduction to Genetic Engineering

Genetic engineering, the deliberate modification of an organism’s genome, has revolutionized fields from agriculture to medicine, offering unprecedented possibilities and raising profound ethical questions. For example, the creation of genetically modified (GM) crops has significantly increased agricultural yields and reduced reliance on pesticides. In medicine, genetic engineering has paved the way for groundbreaking treatments like gene therapy, offering hope for previously incurable genetic diseases. However, these advancements come with challenges, including ethical concerns about altering the fundamental building blocks of life. As we delve into the world of genetic engineering, we must carefully navigate the complex interplay between science, ethics, and society.

Essay on Genetic Engineering

Historical Background

  • Early Understanding of Heredity : Genetic engineering’s roots trace back to ancient times when people observed hereditary traits in plants and animals. Early farmers selected seeds from plants with desirable traits, unknowingly practicing a rudimentary form of selective breeding.
  • Mendel’s Laws : In the mid-19th century, Gregor Mendel conducted experiments with pea plants that laid the groundwork for modern genetics. He discovered the laws of inheritance, showing that traits are passed down through discrete units (now known as genes) and follow predictable patterns.
  • Discovery of DNA : During the 1950s, James Watson, Francis Crick, and Rosalind Franklin unveiled the double helix structure of DNA, the molecule pivotal for conveying genetic information. This discovery revolutionized biology and set the stage for genetic engineering.
  • Recombinant DNA Technology : In the 1970s, scientists developed recombinant DNA technology to combine DNA from different sources. This breakthrough paved the way for genetic engineering by enabling the transfer of genes between organisms.
  • Development of Transgenic Organisms : In the 1980s, researchers created the first transgenic organisms, such as bacteria that produce insulin. This marked the beginning of practical applications of genetic engineering in medicine and industry.
  • Advancements in Genome Sequencing : Completing the Human Genome Project in 2003 provided a comprehensive map of the human genome, further advancing genetic engineering research and applications.
  • CRISPR-Cas9 Revolution : In recent years, the development of the CRISPR-Cas9 gene-editing technology has revolutionized genetic engineering, allowing for precise and efficient editing of genes in various organisms.
  • Current Landscape : Today, various fields utilize genetic engineering, including agriculture, medicine, and environmental conservation, with ongoing advancements and debates surrounding its ethical and societal implications.

Principles of Genetic Engineering

Several key principles guide genetic engineering, underpinning its techniques and applications:

Watch our Demo Courses and Videos

Valuation, Hadoop, Excel, Mobile Apps, Web Development & many more.

  • DNA as the Genetic Material : At the core of genetic engineering is the understanding that DNA contains an organism’s genetic information, determining its traits. Manipulating DNA allows scientists to modify these traits.
  • Gene Isolation and Cloning : The first step in genetic engineering is isolating the gene of interest. Researchers then clone this gene to produce multiple copies for further manipulation. Cloning involves inserting the gene into a vector, such as a plasmid, which can replicate inside a host cell.
  • Vector-Mediated Gene Transfer : Once cloned, a vector transfers the gene into the target organism’s genome. Vectors are vehicles that can carry the gene into the target cell. Plasmids, viruses, and artificial chromosomes are some of the most common vectors.
  • Gene Expression : After inserting the gene into the target organism, researchers must express it to produce the desired trait. Gene expression involves transcribing the gene into messenger RNA (mRNA) and translating it into a functional protein.
  • CRISPR-Cas9 Technology : CRISPR-Cas9 is a groundbreaking gene-editing tool, enabling meticulous alterations to DNA sequences with unprecedented precision. It works by using a guide RNA to target specific DNA sequences, and the Cas9 enzyme cuts the DNA at that location. This enables highly precise insertion, deletion, or modification of genes.
  • Applications in Agriculture, Medicine, and Industry : Genetic engineering has diverse applications, including creating genetically modified crops with improved traits, developing gene therapies for genetic disorders, and producing recombinant proteins for industrial use.
  • Ethical and Safety Considerations : Genetic engineering raises ethical concerns regarding manipulating living organisms and potential unintended consequences. Safety measures, regulatory frameworks, and public engagement are essential to ensure the responsible use of genetic engineering technologies.

Applications of Genetic Engineering

Genetic engineering has many applications across different fields, including agriculture, medicine, industry, and research. Some key applications include:

  • Agriculture : GM crops with better characteristics, like increased nutrition, extended shelf life, disease and herbicide resistance, and pest and disease resistance, are produced by genetic engineering. GM crops can help increase crop yields, reduce the use of pesticides, and improve food security.
  • Medicine : In medicine, researchers utilize genetic engineering to develop therapies for genetic disorders, such as gene therapy, involving the replacement of faulty genes with functional ones. Researchers utilize genetic engineering to manufacture pharmaceuticals, including insulin and growth hormones, by leveraging genetically engineered bacteria or yeast.
  • Bioremediation : Genetic engineering is used in bioremediation to clean up environmental pollutants. Researchers can genetically engineer microorganisms to degrade pollutants like oil spills or toxic chemicals into less harmful substances.
  • Industrial Applications : Genetic engineering is used in industry to produce enzymes, proteins, and chemicals. For example, industries use enzymes produced by genetically engineered microorganisms to produce biofuels, detergents, and textiles.
  • Research : Genetic engineering is used in research to study gene function and regulation. Techniques like gene knockout, which deactivates specific genes, and gene editing, using CRISPR-Cas9, enable researchers to comprehend the functions of genes in health and disease.
  • Forensics and DNA Profiling : Genetic engineering is used in forensics to analyze DNA evidence and identify individuals. DNA profiling techniques, such as polymerase chain reaction (PCR) and gel electrophoresis, are based on genetic engineering principles.
  • Livestock Improvement : Genetic engineering enhances livestock by introducing favorable traits like disease resistance, enhanced growth rates, and increased milk or meat production.

Ethical and Social Considerations

Genetic engineering raises several ethical and social considerations that require careful addressing:

  • Genetic Privacy : As the utilization of genetic information grows in healthcare and other sectors, concerns regarding the privacy and security of genetic data are on the rise. Unauthorized access to genetic information could lead to discrimination or other harmful consequences.
  • Genetic Discrimination : Concerns exist that individuals could face discrimination in areas such as employment, insurance, and education due to the use of genetic information. Laws and regulations are needed to protect against such discrimination.
  • Informed Consent : In genetic research and medical treatment, informed consent is essential. Individuals must receive full information about the risks and benefits of genetic testing or treatment and must freely consent to participate.
  • Human Enhancement : Genetic engineering raises questions about the ethics of enhancing human traits beyond what is considered normal or natural. This includes concerns about creating “designer babies” with desired traits.
  • Environmental Concerns : Introducing genetically modified organisms (GMOs) into the environment raises concerns regarding potential ecological impacts, including unintended effects on non-target species or ecosystems.
  • Equitable Access : There are concerns about equitable access to genetic technologies and treatments. Factors such as income, geography, or ethnicity should not limit access to genetic testing or treatment.
  • Long-Term Effects : The enduring repercussions of genetic engineering on individuals, populations , and ecosystems necessitate thorough understanding and investigation. More research is needed to assess potential risks and benefits.
  • Cultural and Religious Beliefs : Genetic engineering raises ethical questions that intersect with cultural and religious beliefs. Some groups may object to certain genetic technology applications based on their values and beliefs.
  • Regulation and Oversight : Robust regulation and oversight of genetic engineering are necessary to ensure responsible and ethical use. This includes monitoring the safety of GMOs and ensuring the appropriate use of genetic information.

Realizing the full potential of genetic engineering and ensuring responsible use requires addressing several challenges:

  • Safety Concerns : Concerns persist regarding the safety of genetically modified organisms (GMOs) and products derived from genetic modification. Ensuring the safety of GMOs for human health and the environment requires rigorous testing and evaluation.
  • Ethical Dilemmas : Genetic engineering raises complex ethical questions, such as the morality of altering the genetic makeup of organisms, the implications of creating genetically modified humans, and the potential for genetic discrimination.
  • Regulatory Framework : The regulation of genetic engineering varies widely across countries and regions. Establishing consistent and effective regulatory frameworks is crucial to ensuring genetic engineering technologies’ safe and ethical application.
  • Environmental Impact : The release of GMOs into the environment can have unintended consequences, such as spreading modified genes to wild populations or disrupting ecosystems. Assessing and mitigating these risks is a significant challenge.
  • Public Perception and Acceptance : The public often meets genetic engineering with skepticism and fear, driven by concerns about safety, ethics, and the potential for negative impacts. Building public trust and acceptance is crucial for the responsible development and use of genetic engineering technologies.
  • Intellectual Property Rights : Patenting genetically engineered organisms and technologies raises questions about access to and control over genetic resources. Balancing the need for innovation and investment with equitable access to genetic technologies is a complex challenge.
  • Unintended Consequences : Genetic engineering can have unintended consequences, such as developing resistance to pests or losing genetic diversity in crops. Anticipating and mitigating these unintended consequences is essential.
  • Socioeconomic Impacts : The adoption of genetic engineering technologies can have socioeconomic impacts, such as changes in agricultural practices, access to food, and distribution of benefits. Ensuring that these technologies benefit all stakeholders is a challenge.

Future Prospects

The future of genetic engineering holds exciting possibilities and challenges:

  • Precision Medicine : Scientists will crucially advance personalized medicine and tailor treatments to an individual’s genetic makeup through genetic engineering. This may result in more focused and efficient treatments for various diseases.
  • Gene Editing : Continued advancements in gene-editing technologies, such as CRISPR-Cas9, will enable precise modifications to the genome, offering new opportunities for treating genetic disorders and improving crop traits.
  • Synthetic Biology : Genetic engineering is driving the field of synthetic biology, where researchers design and construct new biological parts, devices, and systems. This could lead to novel biofuels, materials, and pharmaceuticals.
  • Environmental Applications : Genetic engineering will continue to find applications in environmental efforts, such as bioremediation and conservation. Engineered organisms could help clean up pollution or restore ecosystems.
  • Ethical and Regulatory Challenges : As genetic engineering technologies advance, there will be ongoing ethical and regulatory challenges to address, such as ensuring the responsible use of gene editing in humans and the environment.
  • Global Collaboration : International collaboration will be essential for addressing the global challenges facing genetic engineering, such as ensuring equitable access to genetic technologies and sharing benefits and risks across countries.
  • Public Engagement : Engaging the public in discussions about the benefits, risks, and ethical implications of genetic engineering will be critical for building trust and ensuring responsible development and use of these technologies.

Genetic engineering is a powerful tool with diverse applications and immense potential to improve human health, agriculture, industry, and the environment. It has revolutionized medicine by enabling the development of therapies for genetic disorders and the production of pharmaceuticals. It has resulted in the development of genetically engineered crops with enhanced features and higher yields in agriculture. However, genetic engineering also raises ethical, social, and environmental concerns that must be carefully considered and addressed. With responsible use and thoughtful regulation, genetic engineering can continue to drive innovation and benefit society while minimizing risks and upholding ethical standards.

EDUCBA

*Please provide your correct email id. Login details for this Free course will be emailed to you

By signing up, you agree to our Terms of Use and Privacy Policy .

Valuation, Hadoop, Excel, Web Development & many more.

Forgot Password?

This website or its third-party tools use cookies, which are necessary to its functioning and required to achieve the purposes illustrated in the cookie policy. By closing this banner, scrolling this page, clicking a link or continuing to browse otherwise, you agree to our Privacy Policy

Quiz

Explore 1000+ varieties of Mock tests View more

Submit Next Question

🚀 Limited Time Offer! - 🎁 ENROLL NOW

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Front Genome Ed
  • PMC10773783

Logo of fge

What do people think about genetic engineering? A systematic review of questionnaire surveys before and after the introduction of CRISPR

Pedro dias ramos.

1 i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal

2 ICBAS–Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal

Maria Strecht Almeida

Ingrid anna sofia olsson.

Jinxue Ruan , Huazhong Agricultural University, China

Associated Data

The original contributions presented in the study are included in the article/ Supplementary Material ; further inquiries can be directed to the corresponding authors.

The advent of CRISPR-Cas9 in 2012 started revolutionizing the field of genetics by broadening the access to a method for precise modification of the human genome. It also brought renewed attention to the ethical issues of genetic modification and the societal acceptance of technology for this purpose. So far, many surveys assessing public attitudes toward genetic modification have been conducted worldwide. Here, we present the results of a systematic review of primary publications of surveys addressing public attitudes toward genetic modification as well as the awareness and knowledge about the technology required for genetic modification. A total of 53 primary publications (1987–2020) focusing on applications in humans and non-human animals were identified, covering countries in four continents. Of the 53 studies, 30 studies from until and including 2012 (pre-CRISPR) address gene therapy in humans and genetic modification of animals for food production and biomedical research. The remaining 23 studies from after 2013 (CRISPR) address gene editing in humans and animals. Across countries, respondents see gene therapy for disease treatment or prevention in humans as desirable and highly acceptable, whereas enhancement is generally met with opposition. When the study distinguishes between somatic and germline applications, somatic gene editing is generally accepted, whereas germline applications are met with ambivalence. The purpose of the application is also important for assessing attitudes toward genetically modified animals: modification in food production is much less accepted than for biomedical application in pre-CRISPR studies. A relationship between knowledge/awareness and attitude toward genetic modification is often present. A critical appraisal of methodology quality in the primary publications with regards to sampling and questionnaire design, development, and administration shows that there is considerable scope for improvement in the reporting of methodological detail. Lack of information is more common in earlier studies, which probably reflects the changing practice in the field.

Introduction

The advent of CRISPR-Cas9 in 2012 started revolutionizing the field of genetics by democratizing the access to a method for precise modification of the mammalian genome ( Camporesi and Cavaliere, 2016 ; Barrangou and Horvath, 2017 ). The finding that the technique is straightforward and of low cost—while being precise and efficient—underlies the wide uptake of CRISPR-Cas9 by research groups and industries ( Camporesi and Cavaliere, 2016 ; Nordberg et al., 2018 ). This has resulted in an explosion of laboratories engaging in research using genetic modification of organisms, including applications in clinical practice, biomedical research, food production, and for environmental purposes ( Nordberg et al., 2018 ; Brokowski and Adli, 2019 ). The possibility of CRISPR-Cas9 application to human embryos has nonetheless raised concern among scientists and in society and led to revisit previous regulations on human genetic manipulation, such as Article 13 of the Oviedo Convention, the Universal Declaration on the Human Genome and Human Rights, and the EU Charter of Fundamental Rights ( Nordberg et al., 2018 ). The first years of CRISPR-Cas9 were marked by uncertainty, and an international moratorium on human germline manipulation was adopted by a range of countries ( Isasi et al., 2016 ; Boggio et al., 2019 ; Brokowski and Adli, 2019 ). However, in 2018, media announced the first case of human embryo manipulation that resulted in the birth of the first gene-edited twin babies and the expected arrival of another gene-edited baby in the summer of 2019 ( Hirsch et al., 2019 ; Meagher et al., 2020 ). This story initiated a frenzy of media articles, generally characterized by strong and general disapproval, conveying concern that scientists were “crossing the line” and almost unanimous rejection by members of the scientific community ( Nordberg et al., 2018 ; Morrison and de Saille, 2019 ). The discussion around CRISPR-Cas9 has also reignited concerns about gene editing of animals, including those used for food, and their potential release into the environment and the food supply chain ( Caplan et al., 2015 ).

By the time the CRISPR-Cas9 technique became available, the question of genetic modification of living organisms had already been discussed for more than 3 decades. Following the first study by Thomas and Capecchi in 1987, where recombinant DNA could be transferred as a tool to mammalian cells, the first international conference in 1975 led to the creation of the Recombinant Advisory Committee (RAC) to discuss ethical and societal issues related to the application of this new biotechnology tool ( Hurlbut et al., 2015 ; Rufo and Ficorilli, 2019 ). Subsequent landmark events where genetic engineering was applied to humans, such as the first clinical introduction of retrovirus in gene-modified cells by Rosenberg in 1989 ( Hanna et al., 2017 ), the death of Jesse Gelsinger in 1999 after gene therapy intervention to treat a metabolic disorder ( Caplan, 2019 ), and the death of X-SCID patients in a gene therapy trial in 2002 ( Couzin and Kaiser, 2005 ), were reflected in public distrust and a delay in the development of gene therapy over the first decade of the 21st century. Other major scientific milestones include the first genome-edited embryos ( Liang et al., 2015 ), human clinical trials with genome editing therapies ( ClinicalTrials.gov, 2016a ; ClinicalTrials.gov, 2016b ; ClinicalTrials.gov, 2018 ), the genome-edited human babies referred to above, and the attribution of the 2020 Nobel Prize in Chemistry to Jennifer Doudna and Emmanuelle Charpentier for their work leading to the CRISPR technology ( Royal Swedish Academy of Science, 2020 ).

As it makes gene editing much easier and more widely applicable, CRISPR-Cas9 comes across as a technology perceived as both promising and threatening and, as such, is particularly interesting in the context of initiatives such as RRI (Responsible Research and Innovation), which aim to open up research to society ( Shelley-Egan et al., 2020 ). The underlying objective is to align the research and development of new technologies with societal values and priorities. Understanding public knowledge and awareness of a new technology is an important part of the process, as is the measurement of citizens’ attitudes toward such development, for two main reasons. First, in representative democracies, questionnaires are important sources of information about how citizens position themselves in specific issues. Second, it is important to understand how receptive citizens are to adopting new technologies in their daily lives.

Opinion surveys measure the views of society within a given context in relation to a certain topic, often with a cross-sectional approach that measures opinions at a specific time-point and allows for comparison, such as between countries or regions but not over time ( Stockemer, 2019a ; Stockemer, 2019b ). When used as research instruments, surveys of public opinion are designed to provide quantitative information that allows researchers to answer underlying research questions by assessing the attitudes of surveyed people ( Haddock and Maio, 2008 ). A critical appraisal of the study methodology is an important complement to a systematic review of study outcomes. Despite being most common in reviews of randomized clinical trials, critical appraisal is relevant for many types of studies, including quantitative, qualitative, mixed-methods, and surveys ( National Health and Medical Research Council, 2000a ; National Health and Medical Research Council, 2000b ; Moher et al., 2009 ; Crowe and Sheppard, 2011 ; Nolan et al., 2012 ; Pace et al., 2012 ; Oluka et al., 2014 ). An important aspect of methodological quality is the survey instrument, that is, the set of questions and the accompanying measurement scales such as Likert and semantic differential scales, which are constructs that need to be evaluated in terms of validity and reliability before the survey is administered ( Haddock and Maio, 2008 ; Boateng et al., 2018 ; Hair et al., 2019 ). In systematic reviews of quantitative questionnaire studies, critical appraisal also includes the validity and how representative the sample is of the population under study, how the variables have been defined, whether potential biases are considered, and other factors that may interfere with result interpretation ( COGEM, 2018 ).

The aim of the present systematic review is to map the existing body of evidence concerning public attitudes toward genetic modification since the first survey on the topic was applied nearly 35 years ago. The review includes 53 primary publications covering countries in Asia, Europe, North America, South America, and Oceania, integrating public attitudes and awareness and knowledge about genetic modification. Our approach is comprehensive as it includes cross-sectional surveys measuring public opinions on matters of biotechnology and genetic engineering when applied to humans and other animals and introduces critical appraisal as a means to assess the methodology quality surrounding questionnaire design, development, and administration together with population sampling and the main limitations and successes drawn from studies in this type of analysis. This systematic review will complement existing narrative reviews and perspective papers on the topic, such as Lassen et al. (2006) ; Condit (2010) ; Howell et al. (2020) .

Methodology

Web of Science (WOS) was selected as the primary source for scholarly publications, focusing the search to identify surveys done with citizens on three different themes: gene therapy , genetically modified animals (GM animals) , and genome editing . The search was conducted between July and November 2019 and reviewed again in February 2020 and August 2022. This database search was complemented with Google search engine to look specifically for the gray literature that could not be found through WOS, namely, governmental reports and other studies not published in academic journals. Although not peer-reviewed by academic scholars, their relevance for policy advising means this type of literature is worth considering ( Haddaway et al., 2015 ; Piasecki et al., 2018 ). For the WOS search, the themes gene therapy and GM animals included only publications until 2012 since this was the year of the advent of CRISPR-Cas9 biotechnology, which changed the terminology of scientific articles from “genetic modification” to “genome editing.” Conversely and likewise, for the genome editing theme, only studies from 2013 onward were included. All WOS databases were investigated: WOS Core Collection, Current Contents Connect, Derwent Innovations Index, KCI—Korean Journal Database, MEDLINE ® , Russian Science Citation Index, and SciELO Citation Index. Pilot studies were searched using different combinations of keywords until the identification of the final Boolean strings to be used for the searching process was completed (see Supplementary Material ). For this, the numbers of publications retrieved from WOS for a specific combination of strings were analyzed, and only the ones with the highest numbers were considered. For GM animals , the different combinations of strings yielded the highest number of all, while for gene therapy and genome editing themes, it was irrelevant to add more string terms since it would always yield equal or lower numbers of publications. These allowed us to conduct the search in a broadened way, finding the most publications possible for each theme and discarding unintended ones. As for the Google search, the terms used included the theme name, adding “public” plus “attitude” terms, and the search results were screened thoroughly until the titles of the links showed redundancy in the upcoming search pages. After the identification of websites conveying multiple surveys, these were also used as a source to search for additional gray literature studies.

The screening process is described in the PRISMA flowchart presented in Figure 1 . All WOS publications that featured surveys with the general public regarding genetic modification of humans or animals were included in an Endnote library. All publications only addressing genetic modification of plants or crops were excluded from the library, and so were publications in the format of reviews and meeting or conference abstracts. All publications without access to its full-text or PDF document or not written in English were equally excluded. From the initial set of 2,981 publications, following duplicate removal and implementation of the exclusion criteria, 60 publications were left. After a careful reading of these, 33 publications reporting qualitative rather than quantitative studies and/or with low sample sizes (lower than 100 respondents) were excluded. To the WOS final list of 27 publications, 26 from the gray literature not meeting the exclusion criteria were added, equaling a total of 53 primary publications eligible for the systematic review.

An external file that holds a picture, illustration, etc.
Object name is fgeed-05-1284547-g001.jpg

PRISMA flowchart and exclusion criteria used for the search and selection of primary publications in the systematic review.

Survey parameters

The systematic review followed the PICOS guidelines (population, intervention, comparator, outcome, and study design) for the evaluation of studies, resulting from the initial search, except for the intervention index since we were not performing any statistical or meta-analysis ( Centre for Reviews and Dissemination, 2009a ; Centre for Reviews and Dissemination, 2009b ). Population concerned the number of participants featured in the surveys and the country where the surveys took place. Comparison concerned the differences and similarities of public attitudes toward genetic modification procedures among citizens of different countries, comparison between years, and comparison of the type of questions and terminology used by surveyors. Outcomes analyzed were as follows: percentage of agreement with genetic modification in broad terms and for specific applications in humans and animals, the reasoning behind those attitudes, and respondents’ level of knowledge and/or the level of familiarity with biotechnology and/or genetic engineering topics. For more details, please see Supplementary Table S1 .

Critical appraisal of primary publications

All included primary publications were evaluated with regard to the methodological quality of the studies they reported. This was done by assessing if certain indicators were present or absent and by evaluating how well-described and appropriate they were for the studies in question ( Supplementary Table S2 ).

The critical appraisal addressed the following: content of questionnaires —whether authors generated their own items or adapted them from previous surveys; validity— cross-checking between authors and/or external advisers and testing with the target population for both clarity and efficacy of measuring concepts; reliability —trustworthiness of the same items and constructs used within the surveys; sampling —representativeness and randomness; risk of bias —potential response, non-response, and selection bias; and ethical practices —details on informed consent obtained, if there were incentives given to respondents, and disclosure of any ethical statements by authors either related to ethical approval of studies or the potential conflict of interests experienced.

The search, selection, and first analysis were performed by the first author. Feedback was obtained by the other two authors. The critical appraisal was performed by PDR and IASO, while MSA performed the co-authorship network analysis (see Supplementary Material ).

Of the 53 primary publications identified in this review, the 30 studies conducted prior to the advent of CRISPR-Cas9 technology in 2012 represent the pre-CRISPR period ( Supplementary Tables S3, S4 ), whereas the 23 studies conducted from 2013 onward represent the CRISPR period ( Supplementary Table S5 ). Pre-CRISPR studies were conducted between 1987 and 2010 and comprised 25 surveys with questions assessing attitudes toward the genetic modification of animals (GM animals) and 14 surveys assessing attitudes toward the genetic modification of humans. In the CRISPR period, eight survey studies addressed the genetic modification of animals, and 15 addressed the genetic modification of humans.

Generally speaking, the surveys conducted in the pre-CRISPR period focused on the opinion of the general public toward the genetic modification of animals for use in medical applications, food products derived from such animals (meat and milk), and the genetic modification of humans as gene therapy applications for the cure, prevention, and reduction of the risk of diseases. Some of these surveys also included additional aspects of human genetic modification, such as adults and children, prevention and therapy, and modification to change characteristics not related to diseases.

Table 1 summarizes the number of approvers of the genome editing technology in both periods in a proportion of 10 citizens, considering the previously mentioned applications and the region where the surveys took place.

Number of approvers of the genetic modification of humans and animals in pre-CRISPR (1987–2012) and CRISPR (2013–2022) periods. The number of approvers in both periods is given for a total of 10 respondents for each primary publication included in the systematic review. Studies are listed according to their year of publication and include information about authors, country(ies) of survey administration, and the genetic modification of animals and humans’ features. For the pre-CRISPR period, studies with approvers of GM animals for transplants, meat, and milk in a total of 10 respondents and approvers of the genetic modification of humans for somatic and germline applications, and disease and enhancement settings are both represented. For the CRISPR period, studies with approvers of GE animals for transplants/medicines, milk, and welfare purposes in a total of 10 respondents and with approvers of GE humans for somatic and germline applications, and disease and enhancement settings are both represented.

Approvers (in a total of 10 respondents)
Genetic modification of animalsGenetic modification of humans
Authors (year)Country(ies)Transplants and/or medicinesMeat (pork, sheep, and cow)Milk (cow and sheep)Somatic (disease)Somatic (enhancement)Germline (disease)Germline (enhancement)
US7 (farm animals)8 ***Prevent–85***Prevent–84 (intelligence) and 4 (physical)
9–children8 (non-fatal)
Japan---5---
7–children
Asia/Oceania10 (TH), 9 (NZ, AU, and IS), 8 (J and RU), and 7 (IN) ***PreventMore ethical10 (TH), 9 (AU and IN), and 8 (NZ, J, RU, and IS)Physical
8 (NZ, AU, IN, TH, RU, and IS) and 7 (J)8 (TH), 6 (IN), 3 (NZ, AU, RU, and IS), and 2 (J)Non-fatal8 (TH) and 6 (IN)
Children9 (TH), 8 (AU and NZ), 7 (RU and IS), and 6 (J and IN)4 (RU), 3 (AU and J), and 2 (NZ and IS)
8 (AU, TH, IN, NZ, and IS), 7 (J), and 6 (RU)Intelligence
7 (TH and IN), 3 (AU, J, and RU), and 2 (NZ and IS)
EC129447---
EC12-447---
EU154 - mice and pigs------
EU154------
EU23-3-5---
Japan---Opinion formation increased greatly after the first gene therapy success (only 56% formed an opinion)
United Kingdom---Trust in the government and people in charge reveal favoring to allow gene therapy
Belief in public involvement, awareness, interest, and levels of education do not favor to allow gene therapy
EU276--654-
Japan5 (mice)547 ***Prevent - 6-75
3 (pigs)3 (physical)
2 (intelligence)
Asia/Oceania-8 (TH), 7 (IN), 6 (AU and J), 5 (NZ), 4 (IS), and 3 (RU)8 (TH), 7 (IN), 4 (NZ, AU, J, and IS), and 2 (RU)10 (TH), 9 (NZ, AU, and IS), 8 (J and RU), and 7 (IN)More ethical10 (TH), 9 (AU and IN), and 8 (NZ, J, RU, and IS)Physical
***Prevent8 (TH), 6 (IN), 3 (NZ, AU, RU, and IS), and 2 (J)Non-fatal8 (TH), 6 (IN), 4 (RU), 3 (AU and J), 2 (NZ and IS)
8 (NZ, AU, IN, TH, RU, and IS) and 7 (J)9 (TH), 8 (AU and NZ), 7 (RU and IS), and 6 (J and IN)Intelligence
7 (TH and IN), 3 (RU, AU, and J), and 2 (NZ and IS)
Australia-----4 (serious defect)1 - cosmetic
3 (minor defect)
2 (aggression and violence)
New Zealand5--4---
United Kingdom---9---
8–children
United Kingdom---9–cystic fibrosis, 8–heart disease, and 6–baldness4–average height6–8—cystic fibrosis and 5–6—heart disease1–sex of the unborn baby
7–schizophrenia2–height, intelligence, and sexual option2–4—baldness
6–less aggressive *** Prevent
7–heart disease
2–baldness
United Kingdom---2–aggressive behavior and alcoholism1–adults (intelligence/specific skills)--
1–children (appearance/behavior)
Germany4–laboratory: medical2–farm: agricultural-7---
Australia-3–sheep and pork-----
Sweden-2–pork and salmon-----
Asia/Oceania-8 (TH), 7 (J and IN), 5 (AU and NZ), 4 (IS), and 3 (RU)8 (TH and IN), 4 (J, AU, NZ, and IS), and 2 (RU)----
Japan354----
Japan5–mosquitoes54----
New Zealand2-2----
US and South Korea-3 (US)-----
2 (South Korea)
South Korea-2-----
US8–sheep38–sheep----
US-3-----
US-3-----
Japan and New ZealandPigs-4 (J and NZ)----
5 (J)
3 (NZ)
Mice
6 (J)
5 (NZ)
Japan3------
New Zealand11621--
Global6 (life-threatening and debilitating)-6 (life-threatening and debilitating)3
US--31 (intelligence of physical)
US--5 ***Prevent: 1–4-
Australia53-7 (general)
China6Intelligence: 26-
Disease: 7–8Skin color: 1
Non-disease (high cholesterol): 3
Europe8260
US---6463
US---Risk−7; no risk–8
China---8–adults and children464
United Kingdom7 (mosquitoes and organs)5–efficiency of food7–resistant to disease8–(in)curable5–prolonged life8-
3–profit6–invasive species7–non-life-threatening2–cosmetic
5–control pest and hornless cows6–disorder not inherited3–intelligence
Netherlands---9-7–Neuromuscular2
3–HIV
Japan---The higher the awareness, the higher the support
Europe-3–6-8–9 (disease) ***Prevent--3–5
8–9 (disease)
7–9 (disabilities)
Pew Research Center (2018)US--7 (treatment of serious illnesses) ***Prevent - 62 (intelligence)
US6 (transplants)4-
7 (mosquitoes)
Global6 (life-threatening and debilitating)-6 (life-threatening and debilitating)3
Australia76-***Prevent - 85***Prevent - 84
US-69
Brazil-43
USWildlife - 1

Genetically modified animals in pre-CRISPR and CRISPR periods

  • A) Pre-CRISPR: GM animals for food purposes are mostly rejected, and medical applications are seen ambivalently worldwide.

Overall, 25 of the 30 surveys from the pre-CRISPR period covered the genetic modification of animals (GM animals). In a quick overview of Table 1 , we can see that transplants and medicines face a higher approval from respondents than food products derived from GM animals. For all cases of food derived from GM animals, either to obtain “leaner meat,” “meat less fatty,” or simply “meat from these animals,” the approval rate is very low among respondents in almost all countries analyzed, and this trend is consistent from 1987 to 2006, although there are some studies where approval for meat consumption of GM animals reaches more than half of the respondents (the US in 1987, Japan in 1997, Thailand and India in 1997 and 2000, and Australia in 2000; Figure 2A ). Approval of GM animals for organ transplantation and medicines dropped considerably between 1991 and 2010 in Europe. The lowest approval reached 4 in every 10 European citizens in 1996 and 2002 and only 3 in every 10 citizens in 2005, according to Eurobarometer ( Figure 2A ). Conversely, medicines derived from GM cows gained approval among Europeans between 2002 and 2010, according to Eurobarometer ( Figure 3A ). Australians and New Zealanders are among the lowest approvers of GM animals worldwide for both medical and food purposes, and their approval has been decreasing in surveys after the 2000s ( Figures 2A , ​ ,3A). 3A ). A similar trend is seen for citizens from the US who rejected meat derived from GM pigs in all surveys conducted after 2000 ( Figure 2A ). Japanese citizens were the most surveyed public in the pre-CRISPR period, regarding attitudes toward GM animals, which they approved slightly more for food—meat and milk—than for medical purposes (organs for transplantation in pigs ( Figure 2A ) and mice for cancer research ( Supplementary Figure S5 ), going against the general trend. A note of remark is their decrease in approval for the meat of GM pigs from 1997 to 2003, as well as for transplants and medicines ( Figure 2A ). The use of GM mice for cancer research is seen as “to be encouraged” more than GM pigs for transplants among Japanese citizens ( Table 1 –pre-CRISPR). In two studies of single European countries, in Germany, less than half of the citizens supported GM laboratory animals for cancer research, and Swedish citizens categorically rejected GM salmon for food consumption ( Supplementary Figure S5 ), similar to their choice regarding GM pigs ( Figure 2A ).

An external file that holds a picture, illustration, etc.
Object name is fgeed-05-1284547-g002.jpg

Public support for gene modification in pigs worldwide for a proportion of 10 citizens upon survey inquiry in pre-CRISPR (A) and CRISPR (B) periods. (B) CRISPR: Animal welfare in focus and genome-edited animals for food applications continue to be less approved than for medical applications.

An external file that holds a picture, illustration, etc.
Object name is fgeed-05-1284547-g003.jpg

Public support for gene modification in cows worldwide for a proportion of 10 citizens upon survey inquiry in pre-CRISPR (A) and CRISPR (B) periods.

The CRISPR period surveys on attitudes toward GM animals represent a total of eight surveys worldwide over a 10-year period, with the highest number conducted in the US ( Funk and Heferon, 2018a ; Kohl et al., 2019 ; Lull et al., 2019 ; McConnachie et al., 2019 ). Table 1 (CRISPR period) shows that US citizens approve of the genetic modification of animals for human health purposes, in this case, genome-edited pigs for transplants of organs to humans (6 in 10) and genome-edited mosquitoes to eradicate the spreading of diseases into humans (7 in 10). Upon examining Oceanic countries, Australian citizens are more supportive of GE cows for medicines than for meat- and milk-derived products, while New Zealanders are profound rejecters of GE animals for both applications ( Figure 3B ). Regarding the approval for genome-edited pigs for food consumption, Brazilian citizens are mostly rejecters (only 4 in 10) in contrast to US citizens, where more than half support gene editing either for derived products such as meat from GE pigs or meat and milk from GE cows ( Figures 2B , ​ ,3B). 3B ). A new type of question present in surveys from the CRISPR era deals with the genetic engineering of animals for improved animal welfare. Here, we can see that US citizens frankly approve of “GE cows to become hornless” as a way to avoid invasive and painful dehorning ( Figure 3B ). The majority of citizens in New Zealand approve of GE pigs for better animal health and safety, whereas among Brazilian citizens, the approval for GE pigs to “reduce boar taint in pigs” (as an alternative to invasive and painful castration) is below half of the respondents ( Figure 2B ). The only study covering genome editing in wildlife reported a profound rejection among US citizens ( Table 1 ; Supplementary Figure S5 –CRISPR period) because this was perceived as a risk for both humans and nature.

Genetic modification of humans in pre-CRISPR and CRISPR periods

  • A) CRISPR: Somatic genetic modification for therapy is a yes, while enhancement is a no.

Overall, the genetic modification of humans for gene therapy purposes receives medium to high acceptance worldwide ( Table 1 ; Figure 4A ). Only three exceptions can be identified: two related to disease prevention, where 4 in every 10 New Zealand respondents agree with it for “preventing stomach cancer by modifying a person’s genetic code,” and 2 in every 10 United Kingdom citizens approve it to prevent baldness ( Table 1 ). The same low proportion of United Kingdom citizens approved of gene therapy to treat aggressive behavior and alcoholism identified as diseases ( Figure 4A ). The overall greatest support for gene therapy is found among Thai citizens, followed by Australians, New Zealanders, and Israeli and Japanese citizens in the 1990s to cure fatal diseases and United Kingdom citizens in the 2000s for genetic diseases like cystic fibrosis and heart diseases ( Table 1 ; Figure 4A ). On the other side of the genetic modification of humans, enhancement is mostly rejected by all citizens surveyed during the pre-CRISPR period, with the only exceptions being in 1995 and 2000 studies, where Thai and Indian citizens show high approval to “make people more ethical” and the ambivalence demonstrated by US citizens in 1987 toward “changing the genetic makeup of human cells” as well as European Union respondents in 2010 regarding human enhancement ( Table 1 ; Figure 4A ).

An external file that holds a picture, illustration, etc.
Object name is fgeed-05-1284547-g004.jpg

Public support for gene modification in human adults worldwide for a proportion of 10 citizens upon survey inquiry in pre-CRISPR (A) and CRISPR (B) periods.

Germline genetic modification for therapy purposes gained high approval, similar to somatic genetic modification. Once again, there are exceptions, and these involve citizens from New Zealand in 2005 and Europeans in 2010. For New Zealanders, this represents a drop from much higher levels in the second half of the 1990s (almost 8 in every 10 citizens supporting it to cure fatal disease ( Figure 5A ); then, 10 years later, the number decreased to 4 in 10 citizens for approving GE for serious defects and further decreased to 2 for minor defects and to 1 in every 10 citizens for preventing aggression and violence ( Table 1 ; Figure 6A )). Among the most approving respondents of the germline genome modification for therapy are Thai respondents, followed closely by Australian and Indian citizens ( Figure 5A ).

An external file that holds a picture, illustration, etc.
Object name is fgeed-05-1284547-g005.jpg

Public support for gene modification in human germline cells worldwide for a proportion of 10 citizens upon survey inquiry in pre-CRISPR (A) and CRISPR (B) periods.

An external file that holds a picture, illustration, etc.
Object name is fgeed-05-1284547-g006.jpg

Public support for gene modification in human adults and human germline cells for preventing disease worldwide for a proportion of 10 citizens upon survey inquiry in pre-CRISPR (A) and CRISPR (B) periods.

Germline genetic modification for enhancement purposes is approved largely by Thai and Indian citizens to improve the physical characteristics and intelligence level “that children would inherit” ( Table 1 ; Figure 5A ). All the other countries surveyed about this rejection of those applications, particularly for the improvement of intelligence, cosmetic modifications in children, and determination of sex in an unborn baby ( Table 1 ).

  • B) CRISPR: Genome editing of humans for therapy is considered more acceptable in somatic than in germline modifications, but enhancement is opposed.

Surveys in the CRISPR period inquired citizens about genome editing of humans for therapy, similar to that in the pre-CRISPR period, with results showing strong approval worldwide. At this point, Europeans are the most approving of GE to cure diseases, although by a low margin when compared with Chinese and US citizens and with New Zealand citizens following closely. For the prevention of diseases, all citizens surveyed demonstrate an equally high approval rate of 8 in every 10 citizens ( Figure 6B ). In children, the approval rate of gene therapy was only assessed in China and showed to be similarly high among citizens ( Table 1 ).

Similar to the pre-CRISPR period, genetic enhancement of human beings was generally rejected by citizens worldwide ( Figures 4A, B ). Intelligence and the change in skin color were purposes profoundly rejected by Chinese citizens ( Table 1 ). The only case with less than a majority rejecting enhancement (genome editing of “human body cells to change one’s appearance”) was among Australians and to “prolong life” among United Kingdom citizens ( Figure 4B ).

Overall, GE in the human germline, as in the cases of unborn babies and embryos to cure serious diseases, gained approval among citizens ( Figure 5B ). US citizens were the most surveyed public in the CRISPR period, and multiple surveys conducted consecutively from 2016 to 2018 demonstrate a growth in approval of this type of genetic intervention for disease during this period, increasing from 3 to 5 in every 10 citizens in two surveys conducted in 2016 to 6 and 7 in every 10 citizens in surveys conducted in 2017 and 2018, respectively. The remaining studies include European and Chinese publics, and approval rates fall between the highest and the lowest of the US studies ( Figure 5B ; Table 1 ). In fact, 7 in every 10 citizens from the Netherlands approve GGE to avoid hereditary neuromuscular disease, while only 3 in every 10 citizens agree with it for HIV resistance ( Table 1 ). Australian citizens are the most approving of GE in germ cells and embryos, whereas US citizens have lower approval rates, between 3 in every 10 citizens in 2016 and 5 in every 10 citizens in 2018 that approve genetic interventions in unborn babies ( Figure 5B ). Likewise, this is similar for “prevention of disease” scenarios ( Figure 6B ).

Finally, the idea of genetic enhancement of unborn babies is not approved by members of the public anywhere in the world. It was even completely rejected among Europeans and US citizens in a survey conducted in 2017 ( Figure 5B ), and although the surveys conducted demonstrate a higher approval rate among Europeans 1 year later, still less than half of the participants agree with germline genetic enhancement, which is similar to the responses of Australian and Chinese citizens ( Figure 5B ; Table 1 ).

Awareness and knowledge correlation with public attitudes toward the genetic modification of humans and animals

Overall, the more aware or knowledgeable inquired publics are about topics of science and technology, in general, biotechnology, genetics, genetic modification, and gene editing, the most approving they are of genetic modification in humans and animals. In total, 44 surveys assessed the awareness and knowledge of participants about genetic modification topics, and from these, 17 surveys assessed only awareness (level of familiarity) and eight surveys assessed only knowledge (the level of education of respondents) about these topics.

From the total of surveys administered during the pre-CRISPR period, almost none showed a significant correlation between the awareness and knowledge of citizens about topics of science and their approval of genetic modification of humans or animals ( OTA, 1987 ; Macer, 1992 ; Ng et al., 2000 ; Hallman et al., 2002 ; Nayga et al., 2006 ). There was, however, in the United Kingdom in 2007, a survey that demonstrated a significant correlation between higher awareness of citizens to genes and genetics and lower approval of gene therapy in humans ( Barnett et al., 2007 ). Surveys measuring awareness and knowledge of genetics demonstrated a tendency or association between approval of gene therapy in humans and higher awareness or knowledge of these topics ( Macer et al., 1995 ; Sturgis et al., 2005 ; Sato et al., 2006 ). Curiously, a tendency for citizens to reject GM animals when they are more aware of the technology can be observed ( Macer et al., 1997 ; Ng et al., 2000 ; Inaba and Macer, 2003a ; Inaba and Macer, 2003b ). In terms of knowledge, the attitude of citizens showed a tendency to approve when the knowledge was higher ( OTA, 1987 ; Commission of the European Communities, 1991 ; Commission of the European Community, 1993 ; European Commission Directorate-General ScienceResearch and Development XII, 1996 ; Hallman et al., 2002 ; Hallman et al., 2003 ; Puduri et al., 2005 ), except for one study with German respondents ( Hampel et al., 2000 ).

In the CRISPR period, no studies demonstrated a significant correlation between the approval of GE animals or GE humans and awareness or knowledge about the topics among citizens. Nevertheless, there was a tendency for citizens who were more aware of scientific topics to show increased acceptance of gene therapy in humans and GE animals ( Funk et al., 2016 ; STAT and Harvard, 2016 ; Scheufele et al., 2017 ; Funk and Heferon, 2018b ; Funk and Heferon, 2018a ; Lakomý et al., 2018 ; Uchiyama et al., 2018 ; Critchley et al., 2019 ; Lull et al., 2019 ; Chikhazhe 2015; McConnachie et al., 2019 ), except for one study finding no correlation ( Yunes et al., 2019 ).

Methodological quality: reporting of critical issues

This section presents the results of critical appraisal of the methodology, as reported in the primary publications selected for analysis, to provide an indication of the methodological quality ( Petticrew and Roberts, 2005 ). All data are summarized in Supplementary Table S2 .

Questionnaire development

Surveys may be the result of original item generation or adaptation of items used in previous surveys. For pre-CRISPR surveys, there was an approximately even distribution between the eight studies originally generating their own items and the 10 that adapted existing studies. For studies in the CRISPR period, generating own items was much more common (11 versus 4). As for the remaining 18 surveys that have information available both in the pre-CRISPR and CRISPR periods, a hybrid approach was followed.

The validity of a survey instrument is reflected by how well it measures what it is supposed to measure. Face validity (whether it appears to measure what it should) and content validity (if it is understandable to respondents) were the most reported types in 29 and 37 studies, respectively ( Boateng et al., 2018 ; Hair et al., 2019 ). Construct validity, to check if the construct used is suitable, appears mostly in the form of hypothesis testing in 16 pre-CRISPR and 14 CRISPR surveys.

Reliability is about how reproducible survey instrument data are across different applications of the survey. Most papers (32 of the 53) included in the review did not report this parameter. Among the papers that did, the most used was Cronbach’s alpha index to measure internal consistency and split-half reliability, where samples are divided into halves or thirds to ensure that there is not a significant difference between groups of individuals studied.

Sampling: method, response rate, and weighing

The methodology of sampling participants for surveys is very diverse across the different surveys analyzed. CRISPR surveys were conducted mostly online, and pre-CRISPR surveys overlap between telephone, face-to-face, and mail responses. Quota sampling from databases (rather than random sampling) was more common in CRISPR surveys than in pre-CRISPR surveys (10 vs. 3). Weighing of the sample was used to overcome potential sampling bias but was reported in less than half of the studies. In the studies where weighing was reported, the correction tool mostly used was based on demographics for surveys in both time periods. The majority of the studies report a medium response rate (25%–75% of invited participants responded). CRISPR studies show a medium to high response rate compared with pre-CRISPR studies, for which response rates were generally low to medium. Multinational surveys such as Eurobarometer and intercontinental surveys demonstrate a different response rate per country, and therefore, sample weighing was used. Furthermore, an equal number of pre-CRISPR and CRISPR surveys did not report on the response rate (7 each).

Methodology accountability and reporting

Only half of the studies provide information on bias, and this is transversal to both pre-CRISPR and CRISPR studies. The most commonly referred by authors in the studies from the systematic review is recruitment bias, with under- or over-representation of certain demographic groups for education, age, gender, race, and socio-economic status. Some studies report techniques to avoid bias, namely, the use of random digit dialing to avoid inadequate telephone surveys ( OTA, 1987 ), demographic comparisons to the census to avoid sample distortions ( OTA, 1987 ), standardization of questionnaires and their delivery ( Macer et al., 1995 ), use of open responses ( Macer et al., 1995 ; Macer et al., 2000 ), background campaigning ( McCaughey et al., 2019 ), online survey to have a more robust sample ( Weisberg et al., 2017 ), online tools to avoid age bias ( Wang et al., 2017 ), and not mentioning the survey nature to avoid self-selection bias ( McConnachie et al., 2019 ; Yunes et al., 2019 ). In CRISPR studies, authors report about ethical practices taken during survey conduction, whereas this is mostly not reported in pre-CRISPR studies. Such practices involve obtaining informed consent from participants, voluntary participation invitation, obtaining a privacy statement, or even the chance of withdrawal from the study. Formal ethics approval for the study was only reported for 10 studies from the total of publications in the systematic review. Finally, incentives to participants in order to increase their willingness to participate were disclosed in nine studies.

This systematic review of 53 primary publications on attitudes toward genetic modification in humans and non-human animals provides a comprehensive picture of studies in Europe, North America, Asia, and Oceania over 35 years. The review shows some variation between countries but a clear pattern in how different applications are viewed, which does not change substantially over time.

There is an overall positive attitude to gene therapy for medical purposes in humans, both for adults and children, and both as treatment for a fatal genetic disease and as prevention from developing a disease that would otherwise be likely to occur. This is transversal from the early studies before the 1990s to the most recent studies, with little variation among the public and regardless of their origin. This is in agreement with international and national policies ( Walters, 1991 ; Horst, 2007 ; DH-Bio, Committee on Bioethics, 2015 ; Polcz and Lewis, 2016 ; Nicol et al., 2017 ), and indeed, several clinical trials of somatic gene therapy are underway ( EASAC, 2017 ; Karagyaur et al., 2019 ). Key challenges in the use of these therapies in the clinic raised by scholars regard their definition and regulation ( Nicol et al., 2017 ; Sherkow et al., 2018 ) and were partly recognized in some of the public opinion surveys, including the “need for strict regulation” in somatic therapy (Eurobarometer, 2010) and the need for FDA approval to proceed ( STAT and Harvard, 2016 ).

The differentiation between germline and somatic cells becomes important over time. Surveys administered pre-CRISPR hardly ever distinguish between the correction of genes carrying disease for the individual and those that can be passed onto future generations. In contrast, post-CRISPR surveys address this directly not just by questioning explicitly about germline and unborn babies but also when asking both about germline versus somatic therapy and adult versus prenatal therapy. Overall, somatic gene therapy is widely accepted in most surveys, whereas there is much ambivalence about germline gene therapy, with higher support to prevent future health issues in unborn babies and lower support if the purposes are non-health-related issues like physical and psychological characteristics. The ethics of germline gene editing experienced a spike of interest with the advent of the CRISPR-Cas9 technology ( National Academy of Sciences, 2017 ; Nordberg et al., 2018 ; Brokowski and Adli, 2019 ; Morrison and de Saille, 2019 ), and the ethical issues are discussed by the general public and the scientific community in distinct ways. The surveyed public often mentions unnaturalness, messing with nature, and humans playing God in the creation of designer babies as main arguments to reject germline gene editing and health benefits as a reason to accept it. Researchers, on the other hand, primarily refer to technical hurdles and uncertainties, such as off-target effects and mosaicism, as the background of ethical questions related to unintended consequences and safety and also the problem of introducing irreversible changes to the genome of future individuals whose consent cannot be obtained ( Bosley et al., 2015 ; Gyngell et al., 2017 ; National Academy of Sciences, 2017 ; Brokowski and Adli, 2019 ; Morrison and de Saille, 2019 ). Many scholars defend that, while germline gene editing will eventually be inevitable, the technology should not be pursued in the clinic except when no other alternative exists to prevent a severe or deadly genetically transmitted disease and only after the technology has proven to be safe to proceed to clinical trials ( Bosley et al., 2015 ; Gyngell et al., 2017 ; Brokowski, 2018 ; Browkoski and Adli, 2019 ; Morrison and de Saille, 2019 ). Others argue that research on gene editing could improve the understanding of genetic diseases and should be used for single-gene disorders and other disorders arising from polygenic traits ( Gyngell et al., 2017 ). Scholars have defended the adoption of a moratorium on germline gene editing more than once: following the first edit on human cells and after the birth of the first gene-edited babies in late November 2018, respectively ( Baltimore et al., 2015 ; EASAC, 2017 ; Brokowski and Adli, 2019 ; Lander, 2019 ), often justified by the precautionary principle and taking into account the unpredictability of an emerging new form of technology ( Nordberg et al., 2018 ).

A third relevant point is the differentiation between therapy and enhancement. Across countries, citizens are generally opposed to genetic modification for the purpose of enhancement. When asked to distinguish between different types of enhancement, intelligence or psychological features were favored over physical abilities and appearance in US and British studies. Across the countries where there is some support for non-therapeutic gene editing, the most supported purpose is improved human health. This is in line with the establishment of a purpose for genome editing beforehand and the clear distinction between what is a disease and what is a deviation from a societal norm ( Brokowski and Adli, 2019 ). As for current guidelines, the US National Academies of Sciences, Engineering, and Medicine exclude the use of genome modification for any type of enhancement under any circumstance ( National Academy of Sciences, 2017 ; Brokowski and Adli, 2019 ). The reasons for this are also aligned with the slippery-slope argument that gene editing will ultimately lead to social harm by the creation of new genetically modified humans that may lead to “new forms of inequality, discrimination, and societal conflict” if regulation fails to limit germline gene editing to therapeutic uses ( Gyngell et al., 2017 ).

With regard to GM animals, the aspect that stands out as a continued trend is the way acceptance differs between different purposes. Overall, GM animals appear as generally not acceptable for food purposes, be it for leaner or healthier meat, as in the case of GM pigs, or to produce more milk, as in the case of GM cows. In 2007, Novoselova et al. highlighted the important role of consumers in the potential integration of GM products derived from animals into the food chain, pointing out the perception of healthy and safe food, as well as understanding of environmental and ethical concerns as key issues ( Novoselova et al., 2007 ). This perception is based on arguments that “genetic modification is intrinsically wrong” for food applications ( Frewer, 2003 ), with many people even questioning the usefulness of such applications ( Macnaghten, 2004 ). Risk and benefit perceptions regarding food are affected by many factors which interact in complex ways; specifically, with regard to animals, this is further complicated by the duality of the animal as a friend and food ( Ueland et al., 2012 ). As for GM pigs or GM sheep, for medical purposes such as organs for transplantation and derived products to help with diseases, the acceptance is higher. Furthermore, among professionals who are involved with animal research, support for GM pigs in medical applications like xenotransplantation was greater than that for food applications ( Schuppli and Weary, 2010 ). Although this would overcome the shortage of human organs for transplantation, this discussion is again reflecting current and older moral reservations regarding the mixing of tissues from human and non-human species, as well as the unnaturalness and invasiveness of the process and ultimately the risk for human health ( Einsieddel, 2005 ; EASAC, 2017 ; Luna, 2017 ; de Graeff et al., 2019 ). Similarly, it has been found over the years of public opinion surveys that public perceptions of risk are higher when they concern GM animals rather than GM crops/microorganisms and are also perceived as riskier and having more ethical concerns if the context is food applications rather than medical applications as the latter tend to be evaluated on a more specific or case-by-case basis ( Frewer, 2003 ; Frewer et al., 2011 ). The two differences that appear when comparing surveys from before and after the introduction of CRISPR-Cas9 technology are largely associated with the type of questions that were asked. In pre-CRISPR surveys, most respondents see laboratory research in animal models like GM mice as useful but not morally acceptable. This reflects an ambivalence between what is perceived to be a valuable objective (the study of human disease) and the concerns over animals’ welfare ( Spencer, 1999 ; EASAC, 2017 ; de Graeff et al., 2019 ). In the CRISPR surveys that include animal applications, the questions are about applications where genetic modification is done to avoid animal welfare problems, and while people mention some concerns, in particular about potential suffering, overall, they see it as something good. However, they also reveal an unwillingness to consume products derived from these animals, similar to respondents in pre-CRISPR surveys. This follows the usual perception of risks and ethical concerns where the public has also been found to be willing to pay less for GM foods than conventional ones ( Frewer et al., 2011 ). Impacts on human health by the introduction of genetically modified species in the food chain, unnaturalness, and potential ecosystem disturbance are also recognized as moral issues of these interventions ( Nuffield Council of Bioethics, 2016 ; EASAC, 2017 ; Nordberg et al., 2018 ; de Graeff et al., 2019 ). Impacts on biodiversity and sustainability are repeatedly identified ethical concerns about the genetic modification of animals, together with animal welfare, tampering with nature, and unnaturalness ( Frewer et al., 2004 ; MacNaghten, 2004 ; Schuppli and Weary, 2010 ; Frewer et al., 2011 ). Furthermore, GM animals are also seen more negatively than GM plants, and the perception that the technology is unnatural has increased over the years ( Frewer, 2017 ).

Across many surveys, there is a correlation with support for gene technology: the higher the awareness and knowledge levels, the higher the support as well. This lends some support for the deficit model, according to which education and an improved public understanding of science would lead to a higher acceptance of food that is genetically engineered and gene therapy as a clinical treatment approach ( Uzogara, 2000 ; Gottweis, 2002 ). However, in most cases, this relationship is weak, and awareness and knowledge levels toward genetic engineering or modification and biotechnology are generally not considered predictive of public attitude ( Priest, 2000 ; Gottweis, 2002 ; Chen and Chern, 2004 ; Saher et al., 2006 ; Wheeler, 2008 ; Frewer et al., 2011 ). In this context, it is relevant to consider the role of social media. Huber et al. (2019) found that the use of social media news and trust in science was positively correlated across data from 20 countries. They also found that trust in science was more strongly related to social media news use than traditional media news. However, an important caveat highlighted by the authors is that their analysis did not consider the quality of the information. The social media discussion of COVID-19 has made the question of whether what is disseminated is verified scientific information or misinformation/fake news increasingly critical. Radrizzani et al. (2023) surveyed a sample of the United Kingdom public about how their trust in science had been affected by the introduction of the first COVID-19 vaccines. They found that it was much more common for people to report that not only their trust had increased than that it had decreased but also that trust decreased among those who had little trust in science to begin with. In the US, Xiao et al. (2021) found that individuals who get most news from social media had greater beliefs in conspiracies in general and in COVID-19-related conspiracies in particular. Social media may also play a different role in survey research, as illustrated by studies covered by our systematic review, such as McCaughey et al., 2016 , Wang et al., 2017 ; McCaughey et al., 2019 , that included online social media as a method for participant recruitment and response to surveys.

The critical appraisal of methodological quality shows that most studies provide low- to medium-quality information. Only two publications ( Magnusson and Hursti, 2002 ; Kohl et al., 2019 ) fulfill all the criteria recommended for questionnaire surveys ( Petticrew and Roberts, 2005 ; Malhotra, 2006 ; Stockemer, 2019a ; Stockemer, 2019b ). Most studies report or demonstrate the consideration of two to three of the criteria but typically not on the aspects considered more relevant for ensuring the methodological quality, such as the item generation method and response rate. Characteristics of greater relevance, such as validity, reliability, risk of bias, and sampling, are reported at a much lower frequency than what is desired. Poor methodological quality may justify the exclusion of studies from a systematic review. We nevertheless included all surveys in this systematic review because, first, our priority was comprehensiveness and, second, in order to be able to highlight the issue of study quality, which is not yet receiving as much attention in reviews of social science research as it does in biomedical research. Although not reporting does not necessarily mean that the practice was absent, it does, at least, suggest limited attention to the methodology. Lack of information is more common in earlier studies, which probably reflects the changing practice in the field. One also needs to distinguish between survey reports in the gray literature, which focus on reporting the results, from articles in scholarly journals with peer review, where a discussion of methods and issues such as the risk of bias are expected to be an integral part of reporting. Finally, the lack of information regarding formal ethics approval might simply mean that the context in which the study was implemented was considered exempt from formal approval, even though mentioning the exemption would be expected.

To the best of our knowledge, our study is unique in comprehensiveness. First, it includes publications covering almost 35 years and addressing attitudes to human and non-human genetic modifications. Although the 2020 systematic review by Delhove et al. undertook a similar approach in terms of timespan and definition of primary publications, it covers only attitudes to human genetic modification ( Delhove et al., 2020 ). The limitations to our study include the choice of databases, studies, and information to include. We used WOS as the source database and Google web search for publication retrieval. It is possible that other databases would have generated a somewhat different outcome in terms of selected publications. We chose only to include studies of the general public, excluding studies of only specific publics ( Frewer et al., 1997 ; Chen and Chern, 2004 ; Napier et al., 2004 ). Additionally, we must admit some delay regarding the change in terminology from “genetic modification” to “genome editing” that occurred with the advent of CRISPR in 2012 and which was considered in our literature search (see Methodology ). In terms of analysis of results, we opted to only assess the influence of awareness and knowledge in public attitudes and did not include other parameters that could have had an influence here, like trust in organizations, demographics (e.g., socio–economic status), and religious index. The reason to only include awareness and knowledge is because these variables have been continuously assessed, and therefore, we could have a parallel view of how they would have influenced public opinions toward genetic modification over time. Finally, the present paper includes only a qualitative analysis of quantitative results, and we did not perform a meta-analysis.

Future perspectives

Public consultation is critical in controversial matters in relation to genetics and biotechnology, especially when applications will potentially directly influence citizens’ lives and, therefore, have to ensure accurate representation ( Halpern et al., 2019 ). Although cross-sectional surveys such as those we analyzed are important because they provide an overview of how public opinion evolved during the last 35 years, real comprehensive initiatives of public engagement and societal debate on genome modification beforehand are indispensable ( Tait et al., 2017 ; Jasanoff and Hurlbut, 2018 ; Wirz et al., 2020 ). This could include a citizen policy approach, such as that described for climate action policy ( Wintle et al., 2017 ; O’Grady, 2020 ). This would be particularly important in the context of policy-making for CRISPR-Cas9 technology implementation. The design of citizen engagement initiatives with multiple stakeholders in the discussion of genome editing driven by the intervention of some associations already in place like the Association for Responsible Research and Innovation in Genome Editing (ARRIGE) may elevate the dialog and contribute to the adoption of a participatory governance framework that may resemble such reflections ( Montoliu et al., 2018 ; Hirsch et al., 2019 ; Pereira and Völker, 2020 ). This path would also entail the best opportunity for scientists and policymakers to consolidate RRI practices in an era where the speed of technology implementation is key but responsibility for its adoption is mandatory ( Tait et al., 2017 ; Shelley-Egan et al., 2020 ).

The surveys we analyzed varied widely in methodology, and more standardized approaches across countries and over time would be important for such future studies. Good examples to follow are Eurobarometer surveys and international surveys that demand a higher collaboration between teams and offer a consistent overview that may transform a cross-sectional view into a more longitudinal one, allowing for more robust hypothesized theories over time ( Stockemer, 2019a ; Stockemer, 2019b ). Co-authorship analysis for the studies included in the present review ( Supplementary Figure S2 ) enabled addressing the connectedness of the authors involved. Although some extensive networks can be seen, most studies seem authored by independent groups of researchers. More collaborations may benefit methodological consistency in future studies.

Additionally, the bioethics literature on biotechnology recognizes a wider range of issues than those that have been covered in the public attitude surveys, such as eugenics, access to technology, funding of genome technologies, and social justice. These are subjects that impact the public and which they often care about, and should be included in future studies as well ( Isasi et al., 2016 ; Nuffield Council of Bioethics, 2016 ; Brokowski and Adli, 2019 ). In policy-making, principles such as solidarity, social justice, and the welfare of future generations are worth considering in the case of GE ( Halpern et al., 2019 ; Mulvihill et al., 2017 ). Finally, it is important to include an assessment of technology awareness and knowledge as part of the survey. Many surveys indicate low levels of knowledge and awareness, and these factors seem to be related to opinion, at least to some extent.

Acknowledgments

The authors would like to thank the librarian at i3S, Anabela Costa, for her guidance on the methodology for the search and selection of primary publications to feature in this systematic review. The authors also wish to thank Cord Brakebusch for the coordination of IMGENE as part of the European Union’s Horizon 2020 Research and Innovation Program under the Marie Sklodowska-Curie Grant, agreement no. 765269, which enabled the funding of this project.

Funding Statement

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This project has received funding from the European Union’s Horizon 2020 Research and Innovation Program under the Marie Skłodowska-Curie Grant, agreement no. 765269.

Data availability statement

Author contributions.

PR: conceptualization, data curation, formal analysis, investigation, methodology, resources, software, validation, visualization, writing–original draft, and writing–review and editing. MA: conceptualization, data curation, formal analysis, methodology, supervision, validation, writing–original draft, writing–review and editing, software, and visualization. IO: conceptualization, data curation, formal analysis, methodology, supervision, validation, writing–original draft, writing–review and editing, funding acquisition, project administration, and resources.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors, and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fgeed.2023.1284547/full#supplementary-material

  • Baltimore D., Baylis F., Berg P., Daley G. Q., Doudna J. A., Lander E. S., et al. (2015). Human gene editing: international summit statement . Available at: https://www.nationalacademies.org/news/2015/12/on-human-gene-editing-international-summit-statement .
  • Barnett J., Cooper H., Senior V. (2007). Belief in public efficacy, trust, and attitudes toward modern genetic science . Risk Anal. 27 ( 4 ), 921–933. 10.1111/j.1539-6924.2007.00932.x [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Barrangou R., Horvath P. (2017). A decade of discovery: CRISPR functions and applications . Nat. Microbiol. 2 , 17092. 10.1038/nmicrobiol.2017.92 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Boateng G. O., Neilands T. B., Frongillo E. A., Melgar-Quinonez H. R., Young S. L. (2018). Best practices for developing and validating scales for health, social, and behavioral research: a primer . Front. Public Health 6 , 149. 10.3389/fpubh.2018.00149 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Boggio A., Knoppers B. M., Almqvist J., Romano C. P. R. (2019). The human right to science and the regulation of human germline engineering . CRISPR J. 2 , 134–142. 10.1089/crispr.2018.0053 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Bosley K., Botchan M., Bredenoord A., Carrol D., Alta Charo R., Chapentier E., et al. (2015). CRISPR germline engineering – the community speaks . Nat. Biotechnol. 33 ( 5 ), 478–486. 10.1038/nbt.3227 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Brokowski C. (2018). Do CRISPR germline ethics statements cut it? CRISPR J. 1 , 115–125. 10.1089/crispr.2017.0024 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Brokowski C., Adli M. (2019). CRISPR ethics: moral considerations for applications of a powerful tool . J. Mol. Biol. 431 ( 1 ), 88–101. 10.1016/j.jmb.2018.05.044 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Camporesi S., Cavaliere G. (2016). Emerging ethical perspectives in the clustered regularly interspaced short palindromic repeats genome-editing debate . Per Med. 13 ( 6 ), 575–586. 10.2217/pme-2016-0047 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Caplan A. (2019). Getting serious about the challenge of regulating germline gene therapy . PLoS Biol. 17 ( 4 ), e3000223. 10.1371/journal.pbio.3000223 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Caplan A. L., Parent B., Shen M., Plunkett C. (2015). No time to waste--the ethical challenges created by CRISPR: CRISPR/Cas, being an efficient, simple, and cheap technology to edit the genome of any organism, raises many ethical and regulatory issues beyond the use to manipulate human germ line cells . EMBO Rep. 16 ( 11 ), 1421–1426. 10.15252/embr.201541337 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Centre for Reviews and Dissemination (2009a). “ Core principles and methods for conducting a systematic review of health interventions ,” in Systematic reviews . Editor Centre of Reviews and Dissemination (York: University of York; ), 1–108. [ Google Scholar ]
  • Centre for Reviews and Dissemination (2009b). “ Incorporating qualitative evidence in or alongside effectiveness reviews ,” in Systematic reviews . Editor Centre of Reviews and Dissemination (York: University of York; ), 219–237. [ Google Scholar ]
  • Chen C., Liang Z. (2018). Chinese public attitudes on gene editing . Sun-Yat Sen University. [ Google Scholar ]
  • Chen H. Y., Chern W. S. (2004). “ Willingness to pay for GM foods: results from a public survey in the USA ,” in 6th international conference of the international-consortium-on-agricultural-biotechnology-research . Editors Evenson R. E., Santaniello V. (CABI Digital Library; ), 117–129. [ Google Scholar ]
  • Chikhazhe T. L. (2015). New Zealand public attitudes towards genetically modified food . M.Sc. Thesis. Lincoln: Lincoln University. Available at: https://researcharchive.lincoln.ac.nz/handle/10182/6825 . [ Google Scholar ]
  • ClinicalTrials.gov (2016a). Identifier NCT02793856, PD-1 knockout 150 engineered T cells for metastatic non-small cell lung cancer . Chengdu Medgencell (MD): Sichuan University; National Library of Medicine. [about 6 screens]. Available at: https://www.clinicaltrials.gov/ct2/show/ {"type":"clinical-trial","attrs":{"text":"NCT02793856","term_id":"NCT02793856"}} NCT02793856 ?term=CRISPR&cond=cancer&cntry=CN&rank=5 (Accessed August 3, 2021). [ Google Scholar ]
  • ClinicalTrials.gov (2016b). Identifier NCT03872479, single ascending dose study in participants with LCA10 . Editas Medicine (MD): National Library of Medicine. [about 5 screens]. Available at: https://www.clinicaltrials.gov/ct2/show/ {"type":"clinical-trial","attrs":{"text":"NCT03872479","term_id":"NCT03872479"}} NCT03872479 ?term=NCT%2303872479&rank=1 (Accessed August 3, 2021). [ Google Scholar ]
  • ClinicalTrials.gov (2018). Identifier NCT03545815, study of CRISPR-cas9 mediated PD-1 and TCR gene-knocked out mesothelin-directed CAR-T cells in patients with mesothelin positive multiple solid tumors . Chinese PLA General Hospital (MD): National Library of Medicine. [about 4 screens]. Available from: https://www.clinicaltrials.gov/ct2/show/ {"type":"clinical-trial","attrs":{"text":"NCT03545815","term_id":"NCT03545815"}} NCT03545815 ?term=CRISPR&cond=cancec&cntry=CN&rank=1 (Accessed August 3, 2021). [ Google Scholar ]
  • COGEM, The Netherlands Commission on Genetic Modification (2018). CRISPR and Animals: implications of genome editing for policy and society . Policy Report CGM/180501-01. [ Google Scholar ]
  • Commission of the European Communities (1991). Opinions of Europeans on biotechnology in 1991 . Eurobarometer 35.1. [ Google Scholar ]
  • Commission of the European Community (1993). Biotechnology and genetic engineering: what Europeans think about it in 1993 . Eurobarometer 39.1. [ Google Scholar ]
  • Condit C. M. (2010). Public attitudes and beliefs about genetics . Annu. Rev. Genomics Hum. Genet. 11 , 339–359. 10.1146/annurev-genom-082509-141740 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Cook A. J., Fairweather J. R., Satterfield T., Hunt L. M. (2004). New Zealand public acceptance of biotechnology . Research Report n.269. AERU: Lincoln University. [ Google Scholar ]
  • Cormick C., Mercer R. (2017). Community attitudes to gene technology . Australia: The Office of the Gene Technology Regulator, Instinct and reason, J2537. [ Google Scholar ]
  • Couzin J., Kaiser J. (2005). Gene therapy. As Gelsinger case ends, gene therapy suffers another blow . Science 307 , 1028. 10.1126/science.307.5712.1028b [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Critchley C., Nicol D., Bruce G., Walshe J., Treleaven T., Tuc h B. (2019). Predicting public attitudes toward gene editing of germlines: the impact of moral and hereditary concern in human and animal applications . Front. Genet. 9 , 704. 10.3389/fgene.2018.00704 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Crowe M., Sheppard L. (2011). A review of critical appraisal tools show they lack rigor: alternative tool structure is proposed . J. Clin. Epidemiol. 64 ( 1 ), 79–89. 10.1016/j.jclinepi.2010.02.008 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • de Graeff N., Jongsma K. R., Johnston J., Hartley S., Bredenoord A. L. (2019). The ethics of genome editing in non-human animals: a systematic review of reasons reported in the academic literature . Philos. Trans. R. Soc. Lond B Biol. Sci. 374 ( 1772 ), 20180106. 10.1098/rstb.2018.0106 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Delhove J., Osenk I., Prichard I., Donnelley M. (2020). Public acceptability of gene therapy and gene editing for human use: a systematic review . Hum. Gene Ther. 31 ( 1-2 ), 20–46. 10.1089/hum.2019.197 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • DH-Bio, Committee on Bioethics (2015). Statement on genome editing technologies . Statement of Council of Europe DH-BIO/INF 13 Final. [ Google Scholar ]
  • Einsieddel E. F. (2005). Public perceptions of transgenic animals . Rev. Sci. Tech. 24 ( 1 ), 149–157. [ PubMed ] [ Google Scholar ]
  • European Academies Science Advisory Council (2017). Genome editing: scientific opportunities, public interests and policy options in the European Union . EASAC policy report 31. [ Google Scholar ]
  • European Commission Directorate General for Research (2002). Europeans and biotechnology in 2002 . Eurobarometer 58.0. [ Google Scholar ]
  • European Commission Directorate-General for Research (2006). Europeans and biotechnology in 2005: patterns and trends . Eurobarometer 63.1. [ Google Scholar ]
  • European Commission Directorate-General for Research (2010). Europeans and biotechnology in 2010: winds of change? Eurobarometer 73.1. [ Google Scholar ]
  • European Commission Directorate-General Science, Research and Development XII (1996). European opinions on modern biotechnology . Eurobarometer 46.1. [ Google Scholar ]
  • Evans M. D. R., Kelley J., Zanjani E. D. (2005). The ethics of gene therapy and abortion: public opinion . Fetal Diagn Ther. 20 ( 3 ), 223–234. 10.1159/000083910 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Frewer L. (2003). 10. Societal issues and public attitudes towards genetically modified foods . Trends Food Sci. Technol. 14 ( 5-8 ), 319–332. 10.1016/S0924-2244(03)00064-5 [ CrossRef ] [ Google Scholar ]
  • Frewer L., Lassen J., Kettlitz B., Scholderer J., Beekman V., Berdal K. G. (2004). Societal aspects of genetically modified foods . Food Chem. Toxicol. 42 ( 7 ), 1181–1193. 10.1016/j.fct.2004.02.002 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Frewer L. J. (2017). Consumer acceptance and rejection of emerging agrifood technologies and their applications . Eur. Rev. Agric. Econ. 44 ( 4 ), 683–704. 10.1093/erae/jbx007 [ CrossRef ] [ Google Scholar ]
  • Frewer L. J., Bergmann K., Brennan M., Lion R., Meertens R., Rowe G., et al. (2011). Consumer response to novel agri-food technologies: implications for predicting consumer acceptance of emerging food technologies . Trends Food Sci. Technol. 22 ( 8 ), 442–456. 10.1016/j.tifs.2011.05.005 [ CrossRef ] [ Google Scholar ]
  • Frewer L. J., Howard C., Shepherd R. (1997). Public concerns in the United Kingdom about general and specific applications of genetic engineering: risk, benefit, and ethics . Sci. Technol. Hum. Values 22 ( 1 ), 98–124. 10.1177/016224399702200105 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Funk C., Heferon M. (2018a). Most Americans accept genetic engineering of animals that benefits human health, but many oppose other uses . Pew Research Center. [ Google Scholar ]
  • Funk C., Heferon M. (2018b). Public views of gene editing for babies depend on how it would Be used . Pew Research Center. [ Google Scholar ]
  • Funk C., Kennedy B., Sciupac E. U. S. (2016). U.S. Public wary of biomedical technologies to “Enhance” human abilities . Pew Research Center. [ Google Scholar ]
  • Gaskell G., Bard I., Allansdottir A., da Cunha R. V., Eduard P., Hampel J., et al. (2017). Public views on gene editing and its uses . Nat. Biotechnol. 35 ( 11 ), 1021–1023. 10.1038/nbt.3958 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Gottweis H. (2002). Gene therapy and the public: a matter of trust . Gene Ther. 9 , 667–669. 10.1038/sj.gt.3301752 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Govindasamy R., Onyango B., Hallman W. K., Jang H.-M., Puduri V. (2008). Public approval of plant and animal biotechnology in South Korea: an ordered probit analysis . Agribusiness 24 ( 1 ), 102–118. 10.1002/agr.20149 [ CrossRef ] [ Google Scholar ]
  • Gyngell C., Douglas T., Savulescu J. (2017). The ethics of germline gene editing . J. Appl. Philos. 34 ( 4 ), 498–513. 10.1111/japp.12249 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Haddaway N. R., Collins A. M., Coughlin D., Kirk S. (2015). The role of Google scholar in evidence reviews and its applicability to grey literature searching . PLoS One 17 ( 9 ), e0138237. 10.1371/journal.pone.0138237 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Haddock G., Maio G. R. (2008). “ Attitudes: content, structure and functions ,” in Introduction to social psychology: a European perspective. BPS textbooks in psychology . Editors Hewstone M., Stroebe W., Jonas K. (Oxford: Blackwell; ), 112–133. [ Google Scholar ]
  • Hair J. F., Gabriel L. D. S., da Silva D., Junior S. B. (2019). Development and validation of attitudes measurement scales: fundamental and practical aspects . RAUSP Manag. J. 54 ( 4 ), 490–507. 10.1108/RAUSP-05-2019-0098 [ CrossRef ] [ Google Scholar ]
  • Hallman W. K., Adelaja A. O., Schilling B. J., Lang J. T. (2002). Public perceptions of genetically modified foods: Americans know not what they eat . Washington, D.C., USA: Food Policy Institute. 10.13140/RG.2.2.25011.63521 [ CrossRef ] [ Google Scholar ]
  • Hallman W. K., Hebden W. C., Aquino H. L., Cuite C. L., Lang J. T. (2003). Public perceptions of genetically modified foods: a national study of American knowledge and opinion . Washington, D.C., USA: Food Policy Institute. 10.7282/T37M0B7R [ CrossRef ] [ Google Scholar ]
  • Halpern J., O'Hara S. E., Doxzen K. W., Witkowsky L. B., Owen A. L. (2019). Societal and ethical impacts of germline genome editing: how can we secure human Rights? CRISPR J. 2 ( 5 ), 293–298. 10.1089/crispr.2019.0042 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hampel J., Pfenning U., Peters H. P. (2000). Attitudes towards genetic engineering . New Genet. Soc. 19 ( 3 ), 233–249. 10.1080/713687604 [ CrossRef ] [ Google Scholar ]
  • Hanna E., Rémuzat C., Auquier P., Toumi M. (2017). Gene therapies development: slow progress and promising prospect . J. Mark. Access and Health Policy 5 , 1265293. 10.1080/20016689.2017.1265293 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hendriks S., Giesbertz N. A. A., Bredenoord A. L., Repping S. (2018). Reasons for being in favour of or against genome modification: a survey of the Dutch general public . Hum. Reprod. Open 2018 ( 3 ), hoy008. 10.1093/hropen/hoy008 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hirsch F., Lemaitre C., Chneiweiss H., Montoliu L. (2019). Genome editing: promoting responsible research . Pharm. Med. 33 ( 3 ), 187–191. 10.1007/s40290-019-00276-1 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Horst M. (2007). Public expectations in gene therapy: scientific futures and their performative effects on scientific citizenship . Sci. Technol. Hum. Values 32 ( 2 ), 150–171. 10.1177/0162243906296852 [ CrossRef ] [ Google Scholar ]
  • Howell E. L., Yang S., Beets B., Brossard D., Scheufele D. A., Xenos M. A. (2020). What do we (not) know about global views of human gene editing? Insights and blind spots in the CRISPR era . CRISPR J. 3 ( 3 ), 148–155. 10.1089/crispr.2020.0004 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Huber B., Barnidge M., Gil de Zúñiga H., Liu J. (2019). Fostering public trust in science: the role of social media . Public Underst. Sci. 28 ( 7 ), 759–777. 10.1177/0963662519869097 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Human Genetics Commission (2001). Public attitudes to human genetic information . London, UK: Human Genetics Commission. [ Google Scholar ]
  • Hurlbut J. B., Saha K., Jasanoff S. (2015). CRISPR democracy: gene editing and the need for inclusive deliberation . Issues Sci. Technol. 32 ( 1 ), 1–14. [ Google Scholar ]
  • Inaba M., Macer D. R. J. (2003a). Attitudes to biotechnology in Japan in 2003 . Eubios J. Asian Int. Biotech. 13 , 78–89. Available from: https://www.eubios.info/EJ133/ej133b.htm . [ Google Scholar ]
  • Inaba M., Macer D. R. J. (2003b). “ Japanese views of medical biotechnology ,” in Asian bioethics in the 21st century . Editors Sang-youg S., Young-Mo K., Macer D. R. J. (Tsukuba: Eubios Ethics Institute; ), 178–196. [ Google Scholar ]
  • Isasi R., Kleiderman E., Knoppers B. (2016). Editing policy to fit the genome? Science 351 ( 6271 ), 337–339. 10.1126/science.aad6778 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Jasanoff S., Hurlbut J. B. (2018). A global observatory for gene editing . Nature 555 , 435–437. 10.1038/d41586-018-03270-w [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Karagyaur M. N., Efimenko A. Y., Makarevich P. I., Vasiluev P. A., Akopyan Z. A., Bryzgalina E. V., et al. (2019). Ethical and legal aspects of using genome editing technologies in medicine (review) . Sovrem. Tekhnologii Med. 11 ( 3 ), 117. 10.17691/stm2019.11.3.16 [ CrossRef ] [ Google Scholar ]
  • Kohl P. A., Brossard D., Scheufele D. A., Xenos M. A. (2019). Public views about editing genes in wildlife for conservation . Conserv. Biol. 33 ( 6 ), 1286–1295. 10.1111/cobi.13310 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Lakomý M., Bohlin G., Hlavová R., Macháčková H., Bergman M., Lindholm M. (2018). Public attitudes to life sciences research in six European countries . ORION DELIVERABLE 2.3. [ Google Scholar ]
  • Lander S., Baylis F., Zhang F., Charpentier E., Berg P., Bourgain C., et al. (2019). Adopt a moratorium on heritable genome editing . Nature 567 , 165–168. 10.1038/d41586-019-00726-5 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Lassen J., Gjerris M., Sandøe P. (2006). After Dolly--ethical limits to the use of biotechnology on farm animals . Theriogenology 65 ( 5 ), 992–1004. 10.1016/j.theriogenology.2005.09.012 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Liang P., Xu Y., Zhang X., Ding C., Huang R., Zhang Z., et al. (2015). CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes . Protein Cell 6 ( 5 ), 363–372. 10.1007/s13238-015-0153-5 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Lull R. B., Akin H., Hallman W. K., Brossard D., Jamieson K. H. (2019). Modeling risk perceptions, benefit perceptions, and approval of releasing genetically engineered mosquitoes as a response to zika virus . Environ. Commun. 14 , 933–953. 10.1080/17524032.2019.1685558 [ CrossRef ] [ Google Scholar ]
  • Luna A. (2017). The unknown of human animal chimera research . Intersect 10 ( 3 ), 1–8. [ Google Scholar ]
  • Macer D., Bezar H., Harman N., Kamada H., Macer N. (1997). Attitudes to biotechnology in Japan and New Zealand in 1997, with international comparisons . Eubios J. Asian Inst. Biotech. 7 , 137–151. Available from: https://www.eubios.info/EJ75/ej75h.htm . [ Google Scholar ]
  • Macer D., Ng M. A. C. (2000). Changing attitudes to biotechnology in Japan . Nat. Biotechnol. 18 ( 9 ), 945–947. 10.1038/79425 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Macer D. R. J. (1992). Public acceptance of human gene-therapy and perceptions of human genetic manipulation . Hum. Gene Ther. 3 ( 5 ), 511–518. 10.1089/hum.1992.3.5-511 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Macer D. R. J. (1997). “ Ethics and public acceptance of agricultural biotechnology ,” in Biotechnology in agriculture . Editor Altman A. (New York: Marcel Dekker; ), 661–690. [ Google Scholar ]
  • Macer D. R. J., Akiyama S., Alora A. T., Asada Y., Azariah J., Azariah H., et al. (1995). International perceptions and approval of gene-therapy . Hum. Gene Ther. 6 ( 6 ), 791–803. 10.1089/hum.1995.6.6-791 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Macer D. R. J., Azariah J., Srinives P. (2000). Attitudes to biotechnology in Asia . Int. J. Biotechnol. 2 , 313–332. 10.1504/IJBT.2000.000142 [ CrossRef ] [ Google Scholar ]
  • Macnaghten P. (2004). Animals in their nature . Sociology 38 ( 3 ), 533–551. 10.1177/0038038504043217 [ CrossRef ] [ Google Scholar ]
  • Magnusson M. K., Hursti U. K. K. (2002). Consumer attitudes towards genetically modified foods . Appetite 39 ( 1 ), 9–24. 10.1006/appe.2002.0486 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Malhotra N. (2006). “ Questionnaire design and development ,” in The handbook of marketing research: uses, misuses, and future advances . Editors Grover R., Vriens M. (California: Sage Publications; ), 176–202. [ Google Scholar ]
  • Marteau T., Michie S., Drake H., Bobrow M. (1995). Public-attitudes towards the selection of desirable characteristics in children . J. Med. Genet. 32 ( 10 ), 796–798. 10.1136/jmg.32.10.796 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • McCaughey T., Budden D. M., Sanfilippo P. G., Gooden G. E. C., Fan L., Fenwick E., et al. (2016). A need for better understanding is the major determinant for public perceptions of human gene editing . Hum. Gene Ther. 30 ( 1 ), 36–43. 10.1089/hum.2018.033 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • McCaughey T., Sanfilippo P. G., Gooden G. E. C., Budden D. M., Fan L., Fenwick E., et al. (2019). A global social media survey of attitudes to human genome editing . Cell Stem Cell 18 ( 5 ), 569–572. 10.1016/j.stem.2016.04.011 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • McConnachie E., Hotzel M. J., Robbins J. A., Shriver A., Weary D. M., von Keyserlingk M. A. G. (2019). Public attitudes towards genetically modified polled cattle . PLoS One 14 ( 5 ), e0216542. 10.1371/journal.pone.0216542 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Meagher K. M., Allyse M. A., Master Z., Sharp R. R. (2020). Reexamining the ethics of human germline editing in the wake of scandal . Mayo Clin. Proc. 95 ( 2 ), 330–338. 10.1016/j.mayocp.2019.11.018 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Moher D., Liberati A., Tetzlaff J., Altman D. G., Group P. (2009). Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement . PLoS Med. 6 ( 7 ), e1000097. 10.1371/journal.pmed.1000097 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Montoliu L., Merchant J., Hirsch F., Abecassis M., Jouannet P., Baertschi B., et al. (2018). ARRIGE arrives: toward the responsible use of genome editing . CRISPR J. 1 ( 2 ), 128–129. 10.1089/crispr.2018.29012.mon [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Morrison M., de Saille S. (2019). CRISPR in context: towards a socially responsible debate on embryo editing . Palgrave Commun. 5 ( 1 ), 110. 10.1057/s41599-019-0319-5 [ CrossRef ] [ Google Scholar ]
  • Mulvihill J. J., Capps B., Joly Y., Lysaght T., Zwart H. A. E., Chadwick R., et al. (2017). Ethical issues of CRISPR technology and gene editing through the lens of solidarity . Br. Med. Bull. 122 ( 1 ), 17–29. 10.1093/bmb/ldx002 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Napier T. L., Tucker M. A., Henry C., Yang X. (2004). Ethical orientations of Ohio residents toward genetically engineered plants and animals: an urban/rural comparison . J. Food Agric. Environ. 2 ( 2 ), 400–411. [ Google Scholar ]
  • National Academy of Sciences, Engineering and Medicine (2017). Human genome editing: science, ethics, and governance . Washington (DC): National Academies Press. [ PubMed ] [ Google Scholar ]
  • National Health and Medical Research Council (2000a). “ Aetiology and risk factors ,” in How to review the evidence: systematic identification and review of the scientific literature . Editor National Health and Medical Research Council (Canberra: Commonwealth of Australia; ), 71–77. [ Google Scholar ]
  • National Health and Medical Research Council (2000b). “ Appraising and selecting studies ,” in How to review the evidence: systematic identification and review of the scientific literature . Editor National Health and Medical Research Council (Canberra: Commonwealth of Australia; ), 23–25. [ Google Scholar ]
  • Nayga R. M., Fishera M. G., Onyango B. (2006). Acceptance of genetically modified food: comparing consumer perspectives in the United States and South Korea . Agr. Econ. 34 ( 3 ), 331–341. 10.1111/j.1574-0864.2006.00129.x [ CrossRef ] [ Google Scholar ]
  • Ng M. A. C., Takeda C., Watanabe T., Macer D. (2000). Attitudes of the public and scientists to biotechnology in Japan at the start of 2000 . Eubios J. Asian Int. Bioeth. 10 ( 4 ), 106–113. [ PubMed ] [ Google Scholar ]
  • Nicol D., Eckstein L., Morrison M., Sherkow J. S., Otlowski M., Whitton T., et al. (2017). Key challenges in bringing CRISPR-mediated somatic cell therapy into the clinic . Genome Med. 9 ( 1 ), 85. 10.1186/s13073-017-0475-4 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Nolan M., Beran T., Hecker K. G. (2012). Surveys assessing students’ attitudes toward statistics: a systematic review of validity and reliability . Stat. Ed. Res. J. 11 ( 2 ), 103–123. 10.52041/serj.v11i2.333 [ CrossRef ] [ Google Scholar ]
  • Nordberg A., Minssen T., Holm S., Horst M., Mortensen K., Moller B. L. (2018). Cutting edges and weaving threads in the gene editing (Я)evolution: reconciling scientific progress with legal, ethical, and social concerns . J. Law Biosci. 5 ( 1 ), 35–83. 10.1093/jlb/lsx043 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Norton J., Lawrence G., Wood G. (1998). The Australian public's perception of genetically-engineered foods . Austral Biotech. 8 ( 3 ), 172–181. [ Google Scholar ]
  • Novoselova T. A., Meuwissen M. P. M., Huirne R. B. M. (2007). Adoption of GM technology in livestock production chains: an integrating framework . Trends Food Sci. Tech. 18 ( 4 ), 175–188. 10.1016/j.tifs.2006.12.005 [ CrossRef ] [ Google Scholar ]
  • Nuffield Council on Bioethics (2016). Genome Editing: an ethical review . [ Google Scholar ]
  • Office of Technology Assessment (1987). New developments in biotechnology—background paper: public perceptions of biotechnology . Washington, DC: U.S. Government Printing Office. OTA-BP-BA-45. [ Google Scholar ]
  • O’Grady C. (2020). Jury duty for global warming: citizen help solve the puzzle for climate action. Science . Available at: https://www.sciencemag.org/news/2020/10/jury-duty-global-warming-citizen-groups-help-solve-puzzle-climate-action (Assessed January 16, 2021). [ Google Scholar ]
  • Oluka O. C., Nie S., Sun Y. (2014). Quality assessment of TPB-based questionnaires: a systematic review . PLoS One 9 ( 4 ), e94419. 10.1371/journal.pone.0094419 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Pace R., Pluye P., Bartlett G., Macaulay A. C., Salsberg J., Jagosh J., et al. (2012). Testing the reliability and efficiency of the pilot Mixed Methods Appraisal Tool (MMAT) for systematic mixed studies review . Int. J. Nurs. Stud. 49 ( 1 ), 47–53. 10.1016/j.ijnurstu.2011.07.002 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Pereira Â. G., Völker T. (2020). “ Engaging with citizens ,” in Science for policy handbook . Editors Sucha V., Sinkiewicz M. (European Union: Elsevier; ), 78–95. [ Google Scholar ]
  • Petticrew M., Roberts H. (2005). Systematic reviews in the social sciences – a practical guide . 1st ed. Oxford: Blackwell Publishing. [ Google Scholar ]
  • Piasecki J., Waligora M., Dranseika V. (2018). Google search as an additional source in systematic reviews . Sci. Eng. Ethics 24 ( 2 ), 809–810. 10.1007/s11948-017-0010-4 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Polcz S., Lewis A. (2016). CRISPR-Cas9 and the non-germline non-controversy . J. Law Biosci. 3 ( 2 ), 413–425. 10.2139/ssrn.2697333 [ CrossRef ] [ Google Scholar ]
  • Priest S. (2000). US public opinion divided over biotechnology? Nat. Biotechnol. 18 , 939–942. 10.1038/79412 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Puduri V., Govindasamy R., Lang J. T., Onyango B. (2005). I will not eat it with a fox; I will not eat it in a box: what determines acceptance of GM food for American consumers? Choices 20 ( 4 ), 257. [ Google Scholar ]
  • Radrizzani S., Fonseca C., Woollard A., Pettitt J., Hurst L. D. (2023). Both trust in, and polarization of trust in, relevant sciences have increased through the COVID-19 pandemic . PLoS ONE 18 ( 3 ), e0278169. 10.1371/journal.pone.0278169 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Royal Swedish Academy of Sciences (2020). The Nobel prize in Chemistry 2020 . [ Google Scholar ]
  • Rufo F., Ficorilli A. (2019). From asilomar to genome editing: research ethics and models of decision . NanoEthics 13 ( 3 ), 223–232. 10.1007/s11569-019-00356-1 [ CrossRef ] [ Google Scholar ]
  • Saher M., Lindeman M., Hursti U. K. (2006). Attitudes towards genetically modified and organic foods . Appetite 46 ( 3 ), 324–331. 10.1016/j.appet.2006.01.015 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Sato H., Akabayashi A., Kai I. (2006). Public, experts, and acceptance of advanced medical technologies: the case of organ transplant and gene therapy in Japan . Health Care Anal. 14 ( 4 ), 203–214. 10.1007/s10728-006-0026-4 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Scheufele D. A., Xenos M. A., Howell E. L., Rose K. M., Brossard D., Hardy B. W. (2017). U.S. attitudes on human genome editing . Science 357 ( 6351 ), 553–554. 10.1126/science.aan3708 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Schuppli C. A., Weary D. M. (2010). Attitudes towards the use of genetically modified animals in research . Public Underst. Sci. 19 ( 6 ), 686–697. 10.1177/0963662510362834 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Shelley-Egan C., Gjefsen M. D., Nydal R. (2020). Consolidating RRI and Open Science: understanding the potential for transformative change . Life Sci. Soc. Policy 16 ( 1 ), 7. 10.1186/s40504-020-00103-5 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Sherkow J. S., Zettler P. J., Greely H. T. (2018). Is it 'gene therapy . J. Law Biosci. 5 ( 3 ), 786–793. 10.1093/jlb/lsy020 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Small B. H., Parminter T. G., Fisher M. W. (2005). Understanding public responses to genetic engineering through exploring intentions to purchase a hypothetical functional food derived from genetically modified dairy cattle . New Zeal J. Agr. Res. 48 ( 4 ), 391–400. 10.1080/00288233.2005.9513672 [ CrossRef ] [ Google Scholar ]
  • Spencer S. (1999). The animal scientist in a changing society . Domest. Anim. Endocrin 17 , 95–100. 10.1016/s0739-7240(99)00027-2 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • STAT and Harvard, T.H. Chan School of Public Health (2016). The public and genetic editing, testing, and therapy . Harvard T.H. Chan School of Public Health. [ Google Scholar ]
  • Stockemer D. (2019a). “ A short introduction to survey research ,” in Quantitative methods for the social sciences: a practical introduction with examples in SPSS and Stata . Editor Stockerner D. (Ottawa: Springer; ), 23–35. [ Google Scholar ]
  • Stockemer D. (2019b). “ Conducting a survey ,” in Quantitative methods for the social sciences: a practical introduction with examples in SPSS and Stata . Editor Stockerner D. (Ottawa: Springer; ), 57–72. [ Google Scholar ]
  • Sturgis P., Cooper H., Fife-Schaw C. (2005). Attitudes to biotechnology: estimating the opinions of a better-informed public . New Genet. Soc. 24 ( 1 ), 31–56. 10.1080/14636770500037693 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Tait J., Banda G., Watkins A. (2017). “ Proportionate and adaptive governance of innovative technologies (PAGIT) ,” in Case study: responsible governance of innovative technologies (Innogen Institute: Univeristy of Edinburgh; ). Innogen Summary Report. [ Google Scholar ]
  • Uchiyama M., Nagai A., Muto K. (2018). Survey on the perception of germline genome editing among the general public in Japan . J. Hum. Genet. 63 ( 6 ), 745–748. 10.1038/s10038-018-0430-2 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Ueland O., Gunnlaugsdottir H., Holm F., Kalogeras N., Leino O., Luteijn J. M., et al. (2012). State of the art in benefit-risk analysis: consumer perception . Food Chem. Toxicol. 50 ( 1 ), 67–76. 10.1016/j.fct.2011.06.006 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Uzogara S. (2000). The impact of genetic modification of human foods in the 21st century: a review . Biotechnol. Adv. 18 , 179–206. 10.1016/s0734-9750(00)00033-1 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • van Mill A., Hopkins H., Kinsella S. (2017). Potential uses for genetic technologies: dialogue and engagement research conducted on behalf of the Royal Society , Hopkins van Mill: Creating connections, Ltd. Findings report. [ Google Scholar ]
  • Walters L. (1991). Human gene therapy: ethics and public policy . Hum. Gene Ther. 2 , 115–122. 10.1089/hum.1991.2.2-115 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Wang J.-H., Wang R., Lee J. H., Iao T. W. U., Hu X., Wang Y.-M., et al. (2017). Public attitudes toward gene therapy in China . Mol. Ther. Methods Clin. Dev. 6 , 40–42. 10.1016/j.omtm.2017.05.008 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Weisberg S. M., Badgio D., Chatterjee A. (2017). A CRISPR new world: attitudes in the public toward innovations in human genetic modification . Front. Public Health 5 ( 117 ), 117–119. 10.3389/fpubh.2017.00117 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Wheeler S. A. (2008). The barriers to further adoption of organic farming and genetic engineering in Australia: views of agricultural professionals and their information sources . Renew. Agr. Food Syst. 23 ( 2 ), 161–170. 10.1017/S1742170507002128 [ CrossRef ] [ Google Scholar ]
  • Wintle B. C., Boehm C. R., Rhodes C., Molloy J. C., Millett P., Adam L., et al. (2017). A transatlantic perspective on 20 emerging issues in biological engineering . Elife 6 , e30247. 10.7554/eLife.30247 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Wirz C. D., Scheufele D. A., Brossard D. (2020). Societal debates about emerging genetic technologies: toward a science of public engagement . Environ. Commun. 14 ( 7 ), 859–864. 10.1080/17524032.2020.1811478 [ CrossRef ] [ Google Scholar ]
  • Xiao X., Borah P., Su Y. (2021). The dangers of blind trust: examining the interplay among social media news use, misinformation identification, and news trust on conspiracy beliefs . Public Underst. Sci. 30 ( 8 ), 977–992. 10.1177/0963662521998025 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Yunes M. C., Teixeira D. L., von Keyserlingk M. A. G., Hotzel M. J. (2019). Is gene editing an acceptable alternative to castration in pigs? PLoS One 14 ( 6 ), e0218176. 10.1371/journal.pone.0218176 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Search Menu
  • Sign in through your institution
  • Volume 12, Issue 1, 2024 (In Progress)
  • Volume 11, Issue 1, 2023
  • Advance articles
  • Editor's Choice
  • Virtual Issues
  • Clinical Briefs
  • ISEMPH Prizes
  • Why Publish
  • Author Guidelines
  • Submission Site
  • Open Access
  • Calls for Papers
  • About Evolution, Medicine, and Public Health
  • About the International Society for Evolution, Medicine and Public Health
  • Editorial Board
  • Advertising and Corporate Services
  • Journals Career Network
  • Self-Archiving Policy
  • For Reviewers
  • Journals on Oxford Academic
  • Books on Oxford Academic

International Society for Evolution, Medicine & Public Health

Article Contents

Introduction, human enhancement, genetic engineering, conclusions.

  • < Previous

Human enhancement: Genetic engineering and evolution

  • Article contents
  • Figures & tables
  • Supplementary Data

Mara Almeida, Rui Diogo, Human enhancement: Genetic engineering and evolution, Evolution, Medicine, and Public Health , Volume 2019, Issue 1, 2019, Pages 183–189, https://doi.org/10.1093/emph/eoz026

  • Permissions Icon Permissions

Genetic engineering opens new possibilities for biomedical enhancement requiring ethical, societal and practical considerations to evaluate its implications for human biology, human evolution and our natural environment. In this Commentary, we consider human enhancement, and in particular, we explore genetic enhancement in an evolutionary context. In summarizing key open questions, we highlight the importance of acknowledging multiple effects (pleiotropy) and complex epigenetic interactions among genotype, phenotype and ecology, and the need to consider the unit of impact not only to the human body but also to human populations and their natural environment (systems biology). We also propose that a practicable distinction between ‘therapy’ and ‘enhancement’ may need to be drawn and effectively implemented in future regulations. Overall, we suggest that it is essential for ethical, philosophical and policy discussions on human enhancement to consider the empirical evidence provided by evolutionary biology, developmental biology and other disciplines.

Lay Summary: This Commentary explores genetic enhancement in an evolutionary context. We highlight the multiple effects associated with germline heritable genetic intervention, the need to consider the unit of impact to human populations and their natural environment, and propose that a practicable distinction between ‘therapy’ and ‘enhancement’ is needed.

There are countless examples where technology has contributed to ameliorate the lives of people by improving their inherent or acquired capabilities. For example, over time, there have been biomedical interventions attempting to restore functions that are deficient, such as vision, hearing or mobility. If we consider human vision, substantial advances started from the time spectacles were developed (possibly in the 13th century), continuing in the last few years, with researchers implanting artificial retinas to give blind patients partial sight [ 1–3 ]. Recently, scientists have also successfully linked the brain of a paralysed man to a computer chip, which helped restore partial movement of limbs previously non-responsive [ 4 , 5 ]. In addition, synthetic blood substitutes have been created, which could be used in human patients in the future [ 6–8 ].

The progress being made by technology in a restorative and therapeutic context could in theory be applied in other contexts to treat non-pathological conditions. Many of the technologies and pharmaceutical products developed in a medical context to treat patients are already being used by humans to ‘enhance’ some aspect of their bodies, for example drugs to boost brain power, nutritional supplements, brain stimulating technologies to control mood or growth hormones for children of short stature. Assistive technology for disabled people, reproductive medicine and pharmacology, beside their therapeutic and restorative use, have a greater potential for human ‘enhancement’ than currently thought. There are also dual outcomes as some therapies can have effects that amount to an enhancement as for example, the artificial legs used by the South African sprinter Oscar Pistorius providing him with a competitive advantage.

This commentary will provide general ethical considerations on human enhancement, and within the several forms of so-called human biomedical enhancement, it will focus on genetic engineering, particularly on germline (heritable) genetic interventions and on the insights evolutionary biology can provide in rationalizing its likely impact. These insights are a subject often limited in discussions on genetic engineering and human enhancement in general, and its links to ethical, philosophical and policy discussions, in particular [ 9 ]. The rapid advances in genetic technology make this debate very topical. Moreover, genes are thought to play a very substantial role in biological evolution and development of the human species, thus making this a topic requiring due consideration. With this commentary, we explore how concepts based in evolutionary biology could contribute to better assess the implications of human germline modifications, assuming they were widely employed. We conclude our brief analysis by summarizing key issues requiring resolution and potential approaches to progress them. Overall, the aim is to contribute to the debate on human genetic enhancement by looking not only at the future, as it is so often done, but also at our evolutionary past.

The noun ‘enhancement’ comes from the verb ‘enhance’, meaning ‘to increase or improve’. The verb enhance can be traced back to the vulgar Latin inaltiare and late Latin inaltare (‘raise, exalt’), from ‘ altare ’ (‘make high’) and altus (‘high’), literally ‘grown tall’. For centuries human enhancement has populated our imagination outlined by stories ranging from the myths of supernormal strengths and eternal life to the superpowers illustrated by the 20th century comic books superheroes. The desire of overcoming normal human capacities and the transformation to an almost ‘perfect’ form has been part of the history of civilization, extending from arts and religion to philosophy. The goal of improving the human condition and health has always been a driver for innovation and biomedical developments.

In the broadest sense, the process of human enhancement can be considered as an improvement of the ‘limitations’ of a ‘natural version’ of the human species with respect to a specific reference in time, and to different environments, which can vary depending on factors such as, for example, climate change. The limitations of the human condition can be physical and/or mental/cognitive (e.g. vision, strength or memory). This poses relevant questions of what a real or perceived human limitation is in the environment and times in which we are living and how it can be shifted over time considering social norms and cultural values of modern societies. Besides, the impact that overcoming these limitations will have on us humans, and the environment, should also be considered. For example, if we boost the immune system of specific people, this may contribute to the development/evolution of more resistant viruses and bacteria or/and lead to new viruses and bacteria to emerge. In environmental terms, enhancing the longevity of humans could contribute to a massive increase in global population, creating additional pressures on ecosystems already under human pressure.

Two decades ago, the practices of human enhancement have been described as ‘biomedical interventions that are used to improve human form or functioning beyond what is necessary to restore or sustain health’ [ 10 ]. The range of these practices has now increased with technological development, and they are ‘any kind of genetic, biomedical, or pharmaceutical intervention aimed at improving human dispositions, capacities, or well-being, even if there is no pathology to be treated’ [ 11 ]. Practices of human enhancement could be visualized as upgrading a ‘system’, where interventions take place for a better performance of the original system. This is far from being a hypothetical situation. The rapid progress within the fields of nanotechnology, biotechnology, information technology and cognitive science has brought back discussions about the evolutionary trajectory of the human species by the promise of new applications which could provide abilities beyond current ones [ 12 , 13 ]. If such a possibility was consciously embraced and actively pursued, technology could be expected to have a revolutionary interference with human life, not just helping humans in achieving general health and capabilities commensurate with our current ones but helping to overcome human limitations far beyond of what is currently possible for human beings. The emergence of new technologies has provided a broader range of potential human interventions and the possibility of transitioning from external changes to our bodies (e.g. external prosthesis) to internal ones, especially when considering genetic manipulation, whose changes can be permanent and transmissible.

The advocates of a far-reaching human enhancement have been referred to as ‘transhumanists’. In their vision, so far, humans have largely worked to control and shape their exterior environments (niche construction) but with new technologies (e.g. biotechnology, information technology and nanotechnology) they will soon be able to control and fundamentally change their own bodies. Supporters of these technologies agree with the possibility of a more radical interference in human life by using technology to overcome human limitations [ 14–16 ], that could allow us to live longer, healthier and even happier lives [ 17 ]. On the other side, and against this position, are the so-called ‘bioconservatives’, arguing for the conservation and protection of some kind of ‘human essence’, with the argument that it exists something intrinsically valuable in human life that should be preserved [ 18 , 19 ].

There is an ongoing debate between transhumanists [ 20–22 ] and bioconservatives [ 18 , 19 , 23 ] on the ethical issues regarding the use of technologies in humans. The focus of this commentary is not centred on this debate, particularly because the discussion of these extreme, divergent positions is already very prominent in the public debate. In fact, it is interesting to notice that the ‘moderate’ discourses around this topic are much less known. In a more moderate view, perhaps one of the crucial questions to consider, independently of the moral views on human enhancement, is whether human enhancement (especially if considering germline heritable genetic interventions) is a necessary development, and represents an appropriate use of time, funding and resources compared to other pressing societal issues. It is crucial to build space for these more moderate, and perhaps less polarized voices, allowing the consideration of other positions and visions beyond those being more strongly projected so far.

Ethical and societal discussions on what constitutes human enhancement will be fundamental to support the development of policy frameworks and regulations on new technological developments. When considering the ethical implications of human enhancement that technology will be available to offer now and in the future, it could be useful to group the different kinds of human enhancements in the phenotypic and genetic categories: (i) strictly phenotypic intervention (e.g. ranging from infrared vision spectacles to exoskeletons and bionic limbs); (ii) somatic, non-heritable genetic intervention (e.g. editing of muscle cells for stronger muscles) and (iii) germline, heritable genetic intervention (e.g. editing of the C–C chemokine receptor type 5 (CCR5) gene in the Chinese baby twins, discussed later on). These categories of enhancement raise different considerations and concerns and currently present different levels of acceptance by our society. The degree of ethical, societal and environmental impacts is likely to be more limited for phenotypic interventions (i) but higher for genetic interventions (ii and iii), especially for the ones which are transmissible to future generations (iii).

The rapid advances in technology seen in the last decades, have raised the possibility of ‘radical enhancement’, defined by Nicholas Agar, ‘as the improvement of human attributes and abilities to levels that greatly exceed what is currently possible for human beings’ [ 24 ]. Genetic engineering offers the possibility of such an enhancement by providing humans a profound control over their own biology. Among other technologies, genetic engineering comprises genome editing (also called gene editing), a group of technologies with the ability to directly modify an organism’s DNA through a targeted intervention in the genome (e.g. insertion, deletion or replacement of specific genetic material) [ 25 ]. Genome editing is considered to achieve much greater precision than pre-existing forms of genetic engineering. It has been argued to be a revolutionary tool due to its efficiency, reducing cost and time. This technology is considered to have many applications for human health, in both preventing and tackling disease. Much of the ethical debate associated with this technology concerns the possible application of genome editing in the human germline, i.e. the genome that can be transmitted to following generations, be it from gametes, a fertilized egg or from first embryo divisions [ 26–28 ]. There has been concern as well as enthusiasm on the potential of the technology to modify human germline genome to provide us with traits considered positive or useful (e.g. muscle strength, memory and intelligence) in the current and future environments.

Genetic engineering: therapy or enhancement and predictability of outcomes

To explore some of the possible implications of heritable interventions we will take as an example the editing (more specifically ‘deletion’ using CRISPR genome editing technology) of several base pairs of the CCR5 gene. Such intervention was practised in 2018 in two non-identical twin girls born in China. Loss of function mutations of the CCR5 had been previously shown to provide resistance to HIV. Therefore, the gene deletion would be expected to protect the twin baby girls from risk of transmission of HIV which could have occurred from their father (HIV-positive). However, the father had the infection kept under control and the titre of HIV virus was undetectable, which means that risk of transmission of HIV infection to the babies was negligible [ 29 ].

From an ethical ground, based on current acceptable practices, this case has been widely criticized by the scientific community beside being considered by many a case of human enhancement intervention rather than therapy [ 29 , 30 ]. One of the questions this example helps illustrate is that the ethical boundary between a therapy that ‘corrects’ a disorder by restoring performance to a ‘normal’ scope, and an intervention that ‘enhances’ human ability outside the accepted ‘normal’ scope, is not always easy to draw. For the sake of argument, it could be assumed that therapy involves attempts to restore a certain condition of health, normality or sanity of the ‘natural’ condition of a specific individual. If we take this approach, the question is how health, normality and sanity, as well as natural per se, are defined, as the meaning of these concepts shift over time to accommodate social norms and cultural values of modern societies. It could be said that the difficulty of developing a conceptual distinction between therapy and enhancement has always been present. However, the potential significance of such distinction is only now, with the acceleration and impact of technological developments, becoming more evident.

Beyond ethical questions, a major problem of this intervention is that we do not (yet?) know exactly the totality of the effects that the artificial mutation of the CCR5 may have, at both the genetic and phenotypic levels. This is because we now know that, contrary to the idea of ‘one gene-one trait’ accepted some decades ago, a gene—or its absence—can affect numerous traits, many of them being apparently unrelated (a phenomenon also known as pleiotropy). That is, due to constrained developmental interactions, mechanisms and genetic networks, a change in a single gene can result in a cascade of multiple effects [ 31 ]. In the case of CCR5, we currently know that the mutation offers protection against HIV infection, and also seems to increase the risk of severe or fatal reactions to some infectious diseases, such as the influenza virus [ 32 ]. It has also been observed that among people with multiple sclerosis, the ones with CCR5 mutation are twice as likely to die early than are people without the mutation [ 33 ]. Some studies have also shown that defective CCR5 can have a positive effect in cognition to enhance learning and memory in mice [ 34 ]. However, it’s not clear if this effect would be translated into humans. The example serves to illustrate that, even if human enhancement with gene editing methods was considered ethically sound, assessing the totality of its implications on solid grounds may be difficult to achieve.

Genetic engineering and human evolution: large-scale impacts

Beyond providing the opportunity of enhancing human capabilities in specific individuals, intervening in the germline is likely to have an impact on the evolutionary processes of the human species raising questions on the scale and type of impacts. In fact, the use of large-scale genetic engineering might exponentially increase the force of ‘niche construction’ in human evolution, and therefore raise ethical and practical questions never faced by our species before. It has been argued that natural selection is a mechanism of lesser importance in the case of current human evolution, as compared to other organisms, because of advances in medicine and healthcare [ 35 ]. According to such a view, among many others advances, natural selection has been conditioned by our ‘niche-construction’ ability to improve healthcare and access to clean water and food, thus changing the landscape of pressures that humans have been facing for survival. An underlying assumption or position of the current debate is that, within our human species, the force of natural selection became minimized and that we are somehow at the ‘end-point’ of our evolution [ 36 ]. If this premise holds true, one could argue that evolution is no longer a force in human history and hence that any human enhancement would not be substituting itself to human evolution as a key driver for future changes.

However, it is useful to remember that, as defined by Darwin in his book ‘On the Origin of the Species’, natural selection is a process in which organisms that happen to be ‘better’ adapted to a certain environment tend to have higher survival and/or reproductive rates than other organisms [ 37 ]. When comparing human evolution to human genetic enhancement, an acceptable position could be to consider ethically sound those interventions that could be replicated naturally by evolution, as in the case of the CCR5 gene. Even if this approach was taken, however, it is important to bear in mind that human evolution acts on human traits sometimes increasing and sometimes decreasing our biological fitness, in a constant evolutionary trade-off and in a contingent and/or neutral—in the sense of not ‘progressive’—process. In other worlds, differently from genetic human enhancement, natural selection does not ‘ aim ’ at improving human traits [ 38 ]. Human evolution and the so-called genetic human enhancement would seem therefore to involve different underlying processes, raising several questions regarding the implications and risks of the latter.

But using genetic engineering to treat humans has been proposed far beyond the therapeutic case or to introduce genetic modifications known to already occur in nature. In particular, when looking into the views expressed on the balance between human evolution and genetic engineering, some argue that it may be appropriate to use genetic interventions to go beyond what natural selection has contributed to our species when it comes to eradicate vulnerabilities [ 17 ]. Furthermore, when considering the environmental, ecological and social issues of contemporary times, some suggest that genetic technologies could be crucial tools to contribute to human survival and well-being [ 20–22 ]. The possible need to ‘engineer’ human traits to ensure our survival could include the ability to allow our species to adapt rapidly to the rate of environmental change caused by human activity, for which Darwinian evolution may be too slow [ 39 ]. Or, for instance, to support long-distance space travel by engineering resistance to radiation and osteoporosis, along with other conditions which would be highly advantageous in space [ 40 ].

When considering the ethical and societal merits of these propositions, it is useful to consider how proto-forms of enhancement has been approached by past human societies. In particular, it can be argued that humans have already employed—as part of our domestication/‘selective breeding’ of other animals—techniques of indirect manipulation of genomes on a relatively large scale over many millennia, albeit not on humans. The large-scale selective breeding of plants and animals over prehistoric and historic periods could be claimed to have already shaped some of our natural environment. Selective breeding has been used to obtain specific characteristics considered useful at a given time in plants and animals. Therefore, their evolutionary processes have been altered with the aim to produce lineages with advantageous traits, which contributed to the evolution of different domesticated species. However, differently from genetic engineering, domestication possesses inherent limitations in its ability to produce major transformations in the created lineages, in contrast with the many open possibilities provided by genetic engineering.

When considering the impact of genetic engineering on human evolution, one of questions to be considered concerns the effects, if any, that genetic technology could have on the genetic pool of the human population and any implication on its resilience to unforeseen circumstances. This underlines a relevant question associated with the difference between ‘health’ and biological fitness. For example, a certain group of animals can be more ‘healthy’—as domesticated dogs—but be less biologically ‘fit’ according to Darwin’s definition. Specifically, if such group of animals are less genetically diverse than their ancestors, they could be less ‘adaptable’ to environmental changes. Assuming that, the human germline modification is undertaken at a global scale, this could be expected to have an effect, on the distribution of genetically heritable traits on the human population over time. Considering that gene and trait distributions have been changing under the processes of evolution for billions of years, the impact on evolution will need to be assessed by analysing which genetic alterations have been eventually associated with specific changes within the recent evolutionary history of humans. On this front, a key study has analysed the implications of genetic engineering on the evolutionary biology of human populations, including the possibility of reducing human genetic diversity, for instance creating a ‘biological monoculture’ [ 41 ]. The study argued that genetic engineering will have an insignificant impact on human diversity, while it would likely safeguard the capacity of human populations to deal with disease and new environmental challenges and therefore, ensure the health and longevity of our species [ 41 ]. If the findings of this study were considered consistent with other knowledge and encompassing, the impact of human genetic enhancements on the human genetic pool and associated impacts could be considered secondary aspects. However, data available from studies on domestication strongly suggests that domestication of both animals and plans might lead to not only decreased genetic diversity per se, but even affect patterns of variation in gene expression throughout the genome and generally decreased gene expression diversity across species [ 42–44 ]. Given that, according to recent studies within the field of biological anthropology recent human evolution has been in fact a process of ‘self-domestication’ [ 45 ], one could argue that studies on domestication could contribute to understanding the impacts of genetic engineering.

Beyond such considerations, it is useful to reflect on the fact that human genetic enhancement could occur on different geographical scales, regardless of the specific environment and geological periods in which humans are living and much more rapidly than in the case of evolution, in which changes are very slow. If this was to occur routinely and on a large scale, the implications of the resulting radical and abrupt changes may be difficult to predict and its impacts difficult to manage. This is currently highlighted by results of epigenetics studies, and also of the microbiome and of the effects of pollutants in the environment and their cumulative effect on the development of human and non-human organisms alike. Increasingly new evidence indicates a greater interdependence between humans and their environments (including other microorganisms), indicating that modifying the environment can have direct and unpredictable consequences on humans as well. This highlight the need of a ‘systems level’ approach. An approach in which the ‘bounded body’ of the individual human as a basic unit of biological or social action would need to be questioned in favour of a more encompassing and holistic unit. In fact, within biology, there is a new field, Systems Biology, which stresses the need to understand the role that pleiotropy, and thus networks at multiple levels—e.g. genetic, cellular, among individuals and among different taxa—play within biological systems and their evolution [ 46 ]. Currently, much still needs to be understood about gene function, its role in human biological systems and the interaction between genes and external factors such as environment, diet and so on. In the future if we do choose to genetically enhance human traits to levels unlikely to be achieved by human evolution, it would be crucial to consider if and how our understanding of human evolution enable us to better understand the implications of genetic interventions.

New forms of human enhancement are increasingly coming to play due to technological development. If phenotypic and somatic interventions for human enhancement pose already significant ethical and societal challenges, germline heritable genetic intervention, require much broader and complex considerations at the level of the individual, society and human species as a whole. Germline interventions associated with modern technologies are capable of much more rapid, large-scale impacts and seem capable of radically altering the balance of humans with the environment. We know now that beside the role genes play on biological evolution and development, genetic interventions can induce multiple effects (pleiotropy) and complex epigenetics interactions among genotype, phenotype and ecology of a certain environment. As a result of the rapidity and scale with which such impact could be realized, it is essential for ethical and societal debates, as well as underlying scientific studies, to consider the unit of impact not only to the human body but also to human populations and their natural environment (systems biology). An important practicable distinction between ‘therapy’ and ‘enhancement’ may need to be drawn and effectively implemented in future regulations, although a distinct line between the two may be difficult to draw.

In the future if we do choose to genetically enhance human traits to levels unlikely to be achieved by human evolution, it would be crucial to consider if and how our understanding of humans and other organisms, including domesticated ones, enable us to better understand the implications of genetic interventions. In particular, effective regulation of genetic engineering may need to be based on a deep knowledge of the exact links between phenotype and genotype, as well the interaction of the human species with the environment and vice versa .

For a broader and consistent debate, it will be essential for technological, philosophical, ethical and policy discussions on human enhancement to consider the empirical evidence provided by evolutionary biology, developmental biology and other disciplines.

This work was supported by Fundação para a Ciência e a Tecnologia (FCT) of Portugal [CFCUL/FIL/00678/2019 to M.A.].

Conflict of interest : None declared.

Pham P , Roux S , Matonti F et al.  Post-implantation impedance spectroscopy of subretinal micro-electrode arrays, OCT imaging and numerical simulation: towards a more precise neuroprosthesis monitoring tool . J Neural Eng 2013 ; 10 : 046002 .

Google Scholar

Maghami MH , Sodagar AM , Lashay A et al.  Visual prostheses: the enabling technology to give sight to the blind . J Ophthal Vis Res 2014 ; 9 : 494 – 505 .

Weitz AC , Nanduri D , Behrend MR et al.  Improving the spatial resolution of epiretinal implants by increasing stimulus pulse duration . Sci Transl Med 2015 ; 7 : 318ra203.

Bouton CE , Shaikhouni A , Annetta NV et al.  Restoring cortical control of functional movement in a human with quadriplegia . Nature 2016 ; 533 : 247 – 50 .

Geddes L. First paralysed person to be ‘reanimated’ offers neuroscience insights. Technique moves man’s arm by decoding his thoughts and electrically stimulating his own muscles . Nat News 2016 ; 533 .

Squires JE. Artificial blood . Science 2002 ; 295 : 1002 – 5 .

Lowe KC. Blood substitutes: from chemistry to clinic . J Mater Chem 2006 ; 16 : 4189 – 96 .

Moradi S , Jahanian-Najafabadi A , Roudkenar MH. Artificial blood substitutes: first steps on the long route to clinical utility . Clin Med Insights Blood Disord 2016 ; 9 : 33 – 41 .

Powell R , Kahane G , Savulescu J. Evolution, genetic engineering, and human enhancement . Philos Technol 2012 ; 25 : 439 – 58 .

Parens E (ed.). Enhancing Human Traits: Ethical and Social Implications . Washington, DC : Georgetown University Press , 1998 .

Google Preview

Giubilini A , Sanyal S. Challenging human enhancement. In: Clarke S , Savulescu J , Coady T et al.  (eds). The Ethics of Human Enhancement: Understanding the Debate . Oxford : Oxford University Press , 2016 .

Elliott C. Better Than Well: American Medicine Meets the American Dream . New York, NY : WWW Norton & Company, Inc ., 2003 .

Kramer P. Listening to Prozac . London : Fourth Estate , 1994 .

Moravec H. Mind Children: The Future of Robot and Human Intelligence . Cambridge : Harvard University Press , 1990 .

Bostrom N. Human genetic enhancements: a transhumanist perspective . J Value Inq 2003 ; 37 : 493 – 506 .

Kurzweil R. The Singularity is Near: When Humans Transcend Biology . New York, NY : Viking , 2005 .

Harris J. Enhancing Evolution: The Ethical Case for Making Better People . Princeton, NJ : Princeton University Press , 2010 .

Fukuyama F. Our Posthuman Future: Consequences of the Biotechnology Revolution . New York, NY : Picador , 2002 .

Sandel M. The Case Against Perfection: Ethics in the Age of Genetic Engineering . Cambridge : The Belknap Press of Harvard University Press , 2007 .

Savulescu J , Persson I. The perils of cognitive enhancement and the urgent imperative to enhance the moral character of humanity . J Appl Philos 2008 ; 25 : 162 – 77 .

Buchanan A. Beyond Humanity . Oxford : Oxford University Press , 2011 .

Persson I , Savulescu J. Moral enhancement, freedom, and the god machine . Monist 2012 ; 95 : 399 – 421 .

Leon K. Ageless bodies, happy souls: biotechnology and the pursuit of perfection . New Atlantis 2003 ; 1 : 9 – 28 .

Agar N. Humanity’s End: Why We Should Reject Radical Enhancement . Cambridge : MIT Press , 2010 .

Gaj T , Gersbach CA , Barbas CF III ,. ZFN, TALEN, and CRISPR/Cas based methods for genome engineering . Trends Biotechnol 2013 ; 3 : 397 – 405 .

Baltimore D , Berg P , Botchan M et al.  Biotechnology. A prudent path forward for genomic engineering and germline gene modification . Science 2015 ; 348 : 36 – 8 .

Otieno MO. CRISPR/Cas9 human genome editing: challenges, ethical concerns and implications . J Clin Res Bioeth 2015 ; 6 : 253 .

Ishii T. Germline genome-editing research and its socio-ethical implications . Trends Mol Med 2015 ; 21 : 473 – 81 .

Bionews.org.uk. First Genome-edited Babies: A Very Different Perception of Ethics , 2018 . https://www.bionews.org.uk/page_140060 (27 August 2019, date last accessed).

Cyranoski D. CRISPR-baby scientist fails to satisfy his critics . Nat News 2018 ; 564 : 13 – 4 .

Galis F , Metz JA. Evolutionary novelties: the making and breaking of pleiotropic constraints . Integr Comp Biol 2007 ; 47 : 409 – 19 .

Falcon A , Cuevas MT , Rodriguez-Frandsen A et al.  CCR5 deficiency predisposes to fatal outcome in influenza virus infection . J Gen Virol 2015 ; 96 : 2074 – 8 .

Gade-Andavolu R , Comings DE , MacMurray J et al.  Association of CCR5 Δ32 deletion with early death in multiple sclerosis . Genet Med 2004 ; 6 : 126 – 31 .

Zhou M , Greenhill S , Huang S et al.  CCR5 is a suppressor for cortical plasticity and hippocampal learning and memory . eLife 2016 ; 5 : e20985 .

Tibayrenc M , Ayala FJ (eds). On Human Nature: Biology, Psychology, Ethics, Politics, and Religion . London : Academic Press , 2017 .

Baldi P. The Shattered Self: The End of Natural Evolution . Cambridge : MIT Press , 2001 .

Darwin C. On the Origin of Species by Means of Natural Selection, or, the Preservation of Favoured Races in the Struggle for Life . London : J. Murray , 1859 .

Gould SJ. The Structure of Evolutionary Theory . Belknap, NY : Harvard University Press , 2002 .

Rees M. Our Final Century: Will the Humans Race Survive the Twenty-first Century? Eastbourne : Gardners Books , 2003 .

Nuffield Council on Bioethics. Genome Editing: An Ethical Review . London : Nuffield Council on Bioethics , 2016 .

Powell R. The evolutionary biological implications of human genetic engineering . J Med Philos 2012 ; 37 : 204 – 26 .

Liu W , Chen L , Zhang S et al.  Decrease of gene expression diversity during domestication of animals and plants . BMC Evol Biol 2019 ; 19 : 1 – 11 .

Fages A , Hanghøj K , Khan N et al.  Tracking five millennia of horse management with extensive ancient genome time series . Cell 2019 ; 177 : 1419 – 35 .

Zhang J , Wang X , Yao J et al.  Effect of domestication on the genetic diversity and structure of Saccharina japonica populations in China . Sci Rep 2017 ; 7 : 42158 .

Theofanopoulou C , Gastaldon S , O’Rourke T et al.  Self-domestication in Homo sapiens : insights from comparative genomics . PLoS One 2018 ; 13 : e0196700 .

Capra F , Luisi PL. The Systems View of Life . Cambridge : Cambridge University Press , 2014

  • genetic engineering
Month: Total Views:
September 2019 51
October 2019 571
November 2019 512
December 2019 474
January 2020 492
February 2020 712
March 2020 680
April 2020 615
May 2020 609
June 2020 599
July 2020 605
August 2020 632
September 2020 842
October 2020 1,322
November 2020 1,583
December 2020 1,688
January 2021 1,536
February 2021 2,019
March 2021 3,077
April 2021 2,893
May 2021 2,223
June 2021 1,434
July 2021 906
August 2021 911
September 2021 1,393
October 2021 2,158
November 2021 2,377
December 2021 1,736
January 2022 1,315
February 2022 1,783
March 2022 2,522
April 2022 1,946
May 2022 1,552
June 2022 789
July 2022 675
August 2022 486
September 2022 919
October 2022 1,462
November 2022 1,427
December 2022 1,216
January 2023 1,372
February 2023 1,740
March 2023 2,320
April 2023 2,195
May 2023 1,646
June 2023 1,066
July 2023 940
August 2023 1,070
September 2023 1,283
October 2023 1,953
November 2023 1,886
December 2023 1,684
January 2024 1,871
February 2024 2,312
March 2024 2,729
April 2024 2,752
May 2024 2,034
June 2024 1,117
July 2024 854
August 2024 697

Email alerts

Citing articles via, affiliations.

  • Online ISSN 2050-6201
  • Copyright © 2024 International Society for Evolution, Medicine, and Public Health
  • About Oxford Academic
  • Publish journals with us
  • University press partners
  • What we publish
  • New features  
  • Open access
  • Institutional account management
  • Rights and permissions
  • Get help with access
  • Accessibility
  • Advertising
  • Media enquiries
  • Oxford University Press
  • Oxford Languages
  • University of Oxford

Oxford University Press is a department of the University of Oxford. It furthers the University's objective of excellence in research, scholarship, and education by publishing worldwide

  • Copyright © 2024 Oxford University Press
  • Cookie settings
  • Cookie policy
  • Privacy policy
  • Legal notice

This Feature Is Available To Subscribers Only

Sign In or Create an Account

This PDF is available to Subscribers Only

For full access to this pdf, sign in to an existing account, or purchase an annual subscription.

132 Genetic Engineering Essay Topic Ideas & Examples

Welcome to our list of genetic engineering essay topics! Here, you will find everything from trending research titles to the most interesting genetic engineering topics for presentation. Get inspired with our writing ideas and bonus samples!

🔝 Top 10 Genetic Engineering Topics for 2024

🏆 best genetic engineering topic ideas & essay examples, ⭐ good genetic engineering research topics, 👍 simple & easy genetic engineering essay topics, ❓ genetic engineering discussion questions, 🔎 genetic engineering research topics, ✅ genetic engineering project ideas.

  • Ethical Issues of Synthetic Biology
  • CRISPR-Cas9 and Its Applications
  • Progress and Challenges in Gene Therapy
  • Applications of Gene Editing in Animals
  • The Process of Genetic Engineering in Plants
  • Genetic Engineering for Human Enhancement
  • Genetic Engineering for Improving Crop Yield
  • Regulatory Issues of Genetic Editing of Embryos
  • Gene Silencing in Humans through RNA Interference
  • Gene Drive Technology for Controlling Invasive Species
  • Changing the world: Genetic Engineering Effects Genes used in genetic engineering have a high impact on health and disease, therefore the inclusion of the genetic process alters the genes that influence human behavior and traits.
  • The Ethical Issues of Genetic Engineering Many people have questioned the health risks that arise from genetically modified crops, thus it is the politicians who have to ensure that the interests of the people are met and their safety is assured. […]
  • Is Genetically Engineered Food the Solution to the World’s Hunger Problems? However, the acceptance of GMO’s as the solution to the world’s food problem is not unanimously and there is still a multitude of opposition and suspicion of their use.
  • Human Genetic Engineering: Key Principles and Issues There are many options for the development of events in the field of genetic engineering, and not all of them have been studied. To conclude, human genetic engineering is one of the major medical breakthroughs, […]
  • Designer Babies Creation in Genetic Engineering The creation of designer babies is an outcome of advancements in technology hence the debate should be on the extent to which technology can be applied in changing the way human beings live and the […]
  • Future of Genetic Engineering and the Concept of “Franken-Foods” This is not limited to cows alone but extends to pigs, sheep, and poultry, the justification for the development of genetically modified food is based on the need to feed an ever growing population which […]
  • Mitochondrial Diseases Treatment Through Genetic Engineering Any disorders and abnormalities in the development of mitochondrial genetic information can lead to the dysfunction of these organelles, which in turn affects the efficiency of intracellular ATP production during the process of cellular respiration.
  • Genetic Engineering: Is It Ethical to Manipulate Life? In the case of more complex operations, genetic engineering can edit existing genes to turn on or off the synthesis of a particular protein in the organism from which the gene was taken.
  • Biotechnology and Genetic Engineering Apart from that, there are some experiments that cannot be ethically justified, at least in my opinion, for example, the cloning of human being or the attempts to find the gene for genius.
  • Genetic Engineering in the Movie “Gattaca” by Niccol This would not be right at all since a person should be responsible for their own life and not have it dictated to them as a result of a societal construct created on the basis […]
  • Religious vs Scientific Views on Genetic Engineering With the need to increase the global economy, the field of agriculture is one among the many that have been used to improve the commercial production to take care of the global needs for food […]
  • Genetic Engineering Using a Pglo Plasmid The objective of this experiment is to understand the process and importance of the genetic transformation of bacteria in real time with the aid of extrachromosomal DNA, alternatively referred to as plasmids.
  • Managing Diabetes Through Genetic Engineering Genetic engineering refers to the alteration of genetic make-up of an organism through the use of techniques to introduce a new DNA or eliminate a given hereditable material. What is the role of genetic engineering […]
  • The Role of Plant Genetic Engineering in Global Security Although it can be conveniently stated that the adequacy, abundance and reliability of the global food supply has a major role to play in the enhancement of human life, in the long run, they influence […]
  • Significance of Human Genetic Engineering The gene alteration strategy enables replacing the specific unwanted genes with the new ones, which are more resistant and freer of the particular ailment, hence an essential assurance of a healthy generation in the future.
  • Is the World Ready for Genetic Engineering? The process of manipulating genes has brought scientists to important discoveries, among which is the technology of the production of new kinds of crops and plants with selected characteristics. The problem of the advantages and […]
  • Genome: Bioethics and Genetic Engineering Additionally, towards the end of the documentary, the narrator and some of the interviewed individuals explain the problem of anonymity that is also related to genetic manipulations.
  • Is Genetic Engineering an Environmentally Sound Way to Increase Food Production? According to Thomas & Earl and Barry, genetic engineering is environmentally unsound method of increasing food production because it threatens the indigenous species.
  • Gattaca: Ethical Issues of Genetic Engineering Although the world he lives in has determined that the only measure of a man is his genetic profile, Vincent discovers another element of man that science and society have forgotten.
  • A Major Milestone in the Field of Science and Technology: Should Genetic Engineering Be Allowed? The most controversial and complicated aspect of this expertise is Human Genetic Engineering- whereby the genotype of a fetus can be altered to produce desired results.
  • Genetic Engineering Is Ethically Unacceptable However, the current application of genetic engineering is in the field of medicine particularly to treat various genetic conditions. However, this method of treatment has various consequences to the individual and the society in general.
  • Designer Genes: Different Types and Use of Genetic Engineering McKibben speaks of Somatic Gene Therapy as it is used to modify the gene and cell structure of human beings so that the cells are able to produce certain chemicals that would help the body […]
  • A Technique for Controlling Plant Characteristics: Genetic Engineering in the Agriculture A cautious investigation of genetic engineering is required to make sure it is safe for humans and the environment. The benefit credited to genetic manipulation is influenced through the utilization of herbicide-tolerant and pest-safe traits.
  • The Dangers of Genetic Engineering and the Issue of Human Genes’ Modification In this case, the ethics of human cloning and human genes’ alteration are at the center of the most heated debates. The first reason to oppose the idea of manipulation of human genes lies in […]
  • Genetically Engineered Food Against World Hunger I support the production of GMFs in large quality; I hold the opinion that they can offer a lasting solution to food problems facing the world.
  • Genetic Engineering in Food: Development and Risks Genetic engineering refers to the manipulation of the gene composition of organisms, to come up with organisms, which have different characteristics from the organic ones.
  • Genetic Engineering in the Workplace The main purpose of the paper is to evaluate and critically discuss the ethical concerns regarding the implementation of genetic testing in the workplace and to provide potential resolutions to the dilemmas.
  • Genetic Engineering and Eugenics Comparison The main idea in genetic engineering is to manipulate the genetic make-up of human beings in order to shackle their inferior traits. The concept of socially independent reproduction is replicated in both eugenics and genetic […]
  • The Film “Gattaca” and Genetic Engineering In the film, it is convincing that in the near future, science and technology at the back of genetic engineering shall be developed up to the level which makes the film a reality.
  • Ecological Effects of the Release of Genetically Engineered Organisms Beneficial soil organisms such as earthworms, mites, nematodes, woodlice among others are some of the soil living organisms that are adversely affected by introduction of genetically engineered organisms in the ecosystem since they introduce toxins […]
  • Benefits of Genetic Engineering as a Huge Part of People’s Lives Genetic Engineering is said to question whether man has the right to manipulate the course and laws of nature and thus is in constant collision with religion and the beliefs held by it regarding life.
  • Perfect Society: The Effects of Human Genetic Engineering
  • Genetic Engineering and Forensic Criminal Investigations
  • Biotechnology Assignment and Genetic Engineering
  • Genetic Engineering and Genetically Modified Organisms
  • Bio-Ethics and the Controversy of Genetic Engineering
  • Health and Environmental Risks of Genetic Engineering in Food
  • Genetic Engineering and the Risks of Enforcing Changes on Organisms
  • Genetic Engineering and How It Affects Globel Warming
  • Cloning and Genetic Engineering in the Food Animal Industry
  • Genetic Engineering and Its Impact on Society
  • Embryonic Research, Genetic Engineering, & Cloning
  • Genetic Engineering: Associated Risks and Possibilities
  • Issues Concerning Genetic Engineering in Food Production
  • Genetic Engineering, DNA Fingerprinting, Gene Therapy
  • Cloning: The Benefits and Dangers of Genetic Engineering
  • Genetic Engineering, History, and Future: Altering the Face of Science
  • Islamic and Catholic Views on Genetic Engineering
  • Gene Therapy and Genetic Engineering: Should It Be Approved in the US
  • Exploring the Real Benefits of Genetic Engineering in the Modern World
  • Genetic Engineering and Food Security: A Welfare Economics Perspective
  • Identify the Potential Impact of Genetic Engineering on the Future Course of Human Immunodeficiency Virus
  • Genetic Engineering and DNA Technology in Agricultural Productivity
  • Human Genetic Engineering: Designing the Future
  • Genetic Engineering and the Politics Behind It
  • The Potential and Consequences of Genetic Engineering
  • Genetic Engineering and Its Effect on Human Health
  • The Moral and Ethical Controversies, Benefits, and Future of Genetic Engineering
  • Gene Therapy and Genetic Engineering for Curing Disorders
  • Genetic Engineering and the Human Genome Project
  • Ethical Standards for Genetic Engineering
  • Genetic Engineering and Cryonic Freezing: A Modern Frankenstein
  • The Perfect Child: Genetic Engineering
  • Genetic Engineering and Its Effects on Future Generations
  • Agricultural Genetic Engineering: Genetically Modified Foods
  • Genetic Engineering: The Manipulation or Alteration of the Genetic Structure of a Single Cell or Organism
  • Analysing Genetic Engineering Regarding Plato Philosophy
  • The Dangers and Benefits of Human Cloning and Genetic Engineering
  • Genetic Engineering: Arguments of Both Proponents and Opponents and a Mediated Solution
  • Genetic and How Genetic Engineering Is Diffusing Individualism
  • Finding Genetic Harmony With Genetic Engineering
  • What Is Genetic Engineering?
  • Do You Think Genetically Modified Food Could Harm the Ecosystems of the Areas in Which They Grow?
  • How Agricultural Research Systems Shape a Technological Regime That Develops Genetic Engineering?
  • Can Genetic Engineering for the Poor Pay Off?
  • How Does Genetic Engineering Affect Agriculture?
  • Do You Think It’s Essential to Modify Genes to Create New Medicines?
  • How Can Genetic Engineering Stop Human Suffering?
  • Can Genetic Engineering Cure HIV/AIDS in Humans?
  • How Has Genetic Engineering Revolutionized Science and the World?
  • Do You Think Genetic Engineering Is Playing God and That We Should Leave Life as It Was Created?
  • What Are Some Advantages and Disadvantages of Genetic Engineering?
  • How Will Genetic Engineering Affect the Human Race?
  • When Does Genetic Engineering Go Bad?
  • What Are the Benefits of Human Genetic Engineering?
  • Does Genetic Engineering Affect the Entire World?
  • How Does the Christian Faith Contend With Genetic Engineering?
  • What Are the Ethical and Social Implications of Genetic Engineering?
  • How Will Genetic Engineering Impact Our Lives?
  • Why Should Genetic Engineering Be Extended?
  • Will Genetic Engineering Permanently Change Our Society?
  • What Are People Worried About Who Oppose Genetic Engineering?
  • Do You Worry About Eating GM (Genetically Modified) Food?
  • What Do You Think of the Idea of Genetically Engineering New Bodily Organs to Replace Yours When You Are Old?
  • Should Genetic Engineering Go Ahead to Eliminate Human Flaws, Such as Violence, Jealousy, Hate, Etc?
  • Does the Government Have the Right to Limit How Far We Modify Ourselves?
  • Why Is Genetic Food Not Well Accepted?
  • What Is the Best in the Genetic Modification of Plants, Plant Cell, or Chloroplasts and Why?
  • How Do You Feel About Human Gene Editing?
  • Does Climate Change Make the Genetic Engineering of Crops Inevitable?
  • What Do You Think About Plant Genetic Modification?
  • Gene Drives and Pest Control
  • The Benefits of Genetically Modified Organisms
  • Challenges of Gene Editing for Rare Genetic Diseases
  • The Use of Genetic Engineering to Treat Human Diseases
  • Ethical Considerations and Possibilities of Designer Babies
  • How Genetic Engineering Can Help Restore Ecosystems
  • Basic Techniques and Tools for Gene Manipulation
  • Latest Advancements in Genetic Engineering and Genome Editing
  • Will Engineering Resilient Organisms Help Mitigate Climate Change?
  • Creation of Renewable Resources through Genetic Engineering
  • Genetic Engineering Approach to Drought and Pest Resistance
  • Genetic Engineering Use in DNA Analysis and Identification
  • Synthetic Microorganisms and Biofactories for Sustainable Bioproduction
  • Stem Cells’ Potential for Regenerative Medicine
  • The Role of Genetic Modification in Vaccine Development
  • Can Genetic Engineering Help Eradicate Invasive Species Responsibly?
  • Genetic Engineering for Enhancing the Body’s Defense Mechanisms
  • Advancements in Transplantation Medicine and Creating Bioengineered Organs
  • Genetic Editing of Microbes for Environmental Cleanup
  • Is It Possible to Develop Living Detection Systems?
  • Infertility Essay Topics
  • Bioethics Titles
  • Genetics Research Ideas
  • Epigenetics Essay Titles
  • Morality Research Ideas
  • Stem Cell Essay Titles
  • Biochemistry Research Topics
  • Evolution Topics
  • Chicago (A-D)
  • Chicago (N-B)

IvyPanda. (2024, February 26). 132 Genetic Engineering Essay Topic Ideas & Examples. https://ivypanda.com/essays/topic/genetic-engineering-essay-topics/

"132 Genetic Engineering Essay Topic Ideas & Examples." IvyPanda , 26 Feb. 2024, ivypanda.com/essays/topic/genetic-engineering-essay-topics/.

IvyPanda . (2024) '132 Genetic Engineering Essay Topic Ideas & Examples'. 26 February.

IvyPanda . 2024. "132 Genetic Engineering Essay Topic Ideas & Examples." February 26, 2024. https://ivypanda.com/essays/topic/genetic-engineering-essay-topics/.

1. IvyPanda . "132 Genetic Engineering Essay Topic Ideas & Examples." February 26, 2024. https://ivypanda.com/essays/topic/genetic-engineering-essay-topics/.

Bibliography

IvyPanda . "132 Genetic Engineering Essay Topic Ideas & Examples." February 26, 2024. https://ivypanda.com/essays/topic/genetic-engineering-essay-topics/.

  • Genetic Engineering Essay Introduction
  • Genetic Engineering

Featured Example Essay

This featured Genetic Engineering Essay Introduction is one of many example essays available on this topic.

Sample Essay Examples

Cloning

Related Essay Topics

  • Genetic Engineering Essay
  • Essay About Genetic Engineering
  • Essay On Genetic Engineering In Food Production
  • Gattaca Essay Genetic Engineering
  • Genetic Engineering Essay Outline
  • Genetic Engineering Essay Topics
  • Genetic Engineering Opinion Essay
  • Argumentative Essay On Genetic Engineering
  • Genetic Engineering Argumentative Essay
  • Essay On Genetic Engineering
  • Gattaca Genetic Engineering Essay
  • Essays On Genetic Engineering
  • Genetic Engineering Ethics Essay
  • Genetic Engineering Pros And Cons Essay
  • Genetically Engineered Food Essay

1

Home — Essay Samples — Science — Genetic Engineering — Exploring the Pros and Cons of Genetic Engineering

test_template

Exploring The Pros and Cons of Genetic Engineering

  • Categories: Engineering Genetic Engineering

About this sample

close

Words: 571 |

Published: Feb 7, 2024

Words: 571 | Page: 1 | 3 min read

Table of contents

Introduction, pros of genetic engineering, cons of genetic engineering, regulation of genetic engineering.

Image of Alex Wood

Cite this Essay

Let us write you an essay from scratch

  • 450+ experts on 30 subjects ready to help
  • Custom essay delivered in as few as 3 hours

Get high-quality help

author

Verified writer

  • Expert in: Science

writer

+ 120 experts online

By clicking “Check Writers’ Offers”, you agree to our terms of service and privacy policy . We’ll occasionally send you promo and account related email

No need to pay just yet!

Related Essays

2 pages / 1005 words

3 pages / 1261 words

3 pages / 1487 words

3 pages / 1416 words

Remember! This is just a sample.

You can get your custom paper by one of our expert writers.

121 writers online

Still can’t find what you need?

Browse our vast selection of original essay samples, each expertly formatted and styled

Related Essays on Genetic Engineering

In Gattaca's dystopia, the dangers of a society obsessed with genetic perfection are exposed. The film challenges viewers to reflect on the implications of genetic determinism, the erosion of personal identity, and the ethical [...]

As genetic engineering continues to advance, the controversy surrounding its potential benefits and ethical considerations has become increasingly prominent. This essay aims to provide a comprehensive analysis of genetic [...]

Genetic engineering is the process of altering the genetic makeup of an organism by adding, deleting or changing its DNA. The process involves the manipulation of genes and their transfer from one organism to another, thereby [...]

Genetic engineering is the manipulation of an organism's genetic material to produce desirable traits. It has been used in various fields such as agriculture, medicine, and environmental conservation. Genetic engineering has [...]

Genetic Engineering is a powerful and potentially very dangerous too. To change the sequence of nucleotides of the DNA that code for the structure of a complex living organism, can have extremely ill effects although the [...]

Genetics is the most interesting part of science because it explains how certain traits are passed down by parents to their offspring. Gregor Mendel is considered the pioneer in explaining this theory of the genetics within [...]

Related Topics

By clicking “Send”, you agree to our Terms of service and Privacy statement . We will occasionally send you account related emails.

Where do you want us to send this sample?

By clicking “Continue”, you agree to our terms of service and privacy policy.

Be careful. This essay is not unique

This essay was donated by a student and is likely to have been used and submitted before

Download this Sample

Free samples may contain mistakes and not unique parts

Sorry, we could not paraphrase this essay. Our professional writers can rewrite it and get you a unique paper.

Please check your inbox.

We can write you a custom essay that will follow your exact instructions and meet the deadlines. Let's fix your grades together!

Get Your Personalized Essay in 3 Hours or Less!

We use cookies to personalyze your web-site experience. By continuing we’ll assume you board with our cookie policy .

  • Instructions Followed To The Letter
  • Deadlines Met At Every Stage
  • Unique And Plagiarism Free

introduction of genetic engineering essay

The Spinoff

introduction of genetic engineering essay

Politics August 14, 2024

What does lifting the ‘ban’ on genetic modification mean for new zealand.

avatar

  • Share Story

For nearly three decades, New Zealand’s strict regulations around genetic modification have more or less confined research opportunities to the lab. Yesterday, the government confirmed legislative reform was on its way.

Let’s start at square one – what exactly is GM?

Genetic modification, also known as gene technology or genetic engineering (GE), involves taking DNA and inserting it into the genome of another organism, so that organism can make new substances or perform different functions. DNA can come from microbes, animals, plants, or be synthetic (made in the laboratory). Genetically engineered organisms are known as GMOs. 

It’s worth noting that GM is not the same as gene editing, which involves making a change to the existing genome as opposed to introducing new DNA. But in New Zealand, both are regulated under the same act, the Hazardous Substances and New Organisms (HSNO) Act 1996.

What’s it used for?

GM has applications across agriculture, medicine and industry and climate change adaptation. For example, Crown research institute AgResearch is trialling inserting a gene into white clover to reduce the methane emissions from livestock that eat it. Field trials are being done in Australia, due to our tight regulations. 

In Wellington, scientists at the Malaghan Institute are engineering a patient’s own cells to fight cancer (CAR T-cell cancer therapies). They collect a patient’s immune cells, genetically modify the cells to recognise and kill their cancer and return the cells to the patient. It’s proved effective against B-cell non-Hodgkin lymphomas.

introduction of genetic engineering essay

That all sounds sensible. So why’s it controversial?

Politically, GM has been a highly contentious issue in New Zealand, culminating in a scandal that came to be known as “ Corngate ”. In a live TV interview in 2002, journalist John Campbell grilled a furious Helen Clark, then the prime minister, about a cover-up of accidentally released GM corn in New Zealand’s food supply in 2000, which was revealed by a yet-to-be-published book by investigative journalist Nicky Hager. This was a time of major debate about the safety of GM crops, which much of the environmentally minded left saw as a dangerous, unproven practice that risked our clean, green reputation. While science has essentially settled in favour of GM, it has remained a somewhat politically untouchable subject in Aotearoa.

OK, and remind me, wth is the ‘ban’?

In 1996 New Zealand introduced the Hazardous Substances and New Organisms (HSNO) Act, which put in place a very rigorous and complex approval process before GMOs could be released out of containment (laboratories or quarantine) in New Zealand.  

As the 90s progressed, the potential health risks and environmental impacts of GMOs became a major public concern in New Zealand, with Greenpeace leading campaigns to urge supermarkets not to sell genetically engineered foods. In 1999, the Green Party presented a petition of 92,000 signatures to parliament and in response, the government announced a royal commission into genetic modification, putting in place a moratorium on the commercial planting of GM crops. The commission recommended a proceed-with-caution approach, and the moratorium was lifted in 2003, amid large protests.

introduction of genetic engineering essay

So there’s not actually a ban, then?

Not technically, but the regulations are so onerous that GM research has mostly been confined to labs. It’s still regulated by the HSNO Act, which has had various amendments since 1996 but is largely based on what we knew about the risks and benefits of GMOs in the late 90s. 

OK, so what have been the effects?

As biotechnologist and geneticist Tony Conner explained last year , field tests first substantially declined after the introduction of the law in 1996, then virtually ceased after a later amendment. Amendments meant that GM plants in fields were not allowed to flower – virtually impossible to guarantee, even with manual removal of flower buds. Preventing flowering also prevents grain and fruit developing, which is usually a key purpose of field testing. Also, the procedures are cumbersome and take more effort and resource than the field experiments themselves.

As the Herald reported last year, the Environmental Protection Authority (formerly the Environmental Risk Management Authority), the body that regulates the act, had not received an application for a GM field test in more than a decade.

This has had a flow-on effect for laboratory research – there’s not much point starting something if you know you can’t finish it. Conner wrote that scientists missed out, as did consumers. We did not develop fruit and veges with higher resistance to pests and diseases, or onions that don’t make people cry. 

Since the HSNO Act came into force, only three unconditional releases of GMOs into the environment have been approved. All three were for medical uses.

Why change things?

National campaigned on updating these laws, and committed to it in both the Act Party and NZ First coalition agreements. Collins says changing the laws will “enable us to improve health outcomes, adapt to climate change, deliver massive economic gains and improve the lives of New Zealanders”. She says we’ve been lagging behind other countries like Australia, England, Canada and many European nations in allowing the use of this technology for the benefit of their people, and their economies.

Essentially, loosening the regulations will allow researchers and companies to develop and sell products (like a certain kind of crop) or healthcare treatments (like CAR T-cell therapy). The use of gene technology could help the agriculture sector mitigate emissions and increase productivity. 

What will the new legislation be like?

The full regulatory regime has not been released but the new legislation will be based on Australia’s Gene Technology Act 2000. A regulator will be a single decision maker supported by an office, a technical advisory committee and a Māori advisory committee, and will manage potential risks to human health and the environment. 

There will be tiers of risk: gene modifications indistinguishable from changes that could be achieved through selective breeding or natural mutation will be considered low-risk and fall under no regulation. The sterile pines and CAR T-cell therapies fit this tier, along with things like disease-resistant maize and non-browning mushrooms. Activities including research in labs with animals would require risk assessment and management, and licences would be needed to cover field trials, clinical trials and commercial releases.

Are we sure GM is totally safe? What are the risks?

There could be some potential health risks to eating GM food, like possible exposure to new allergens and the transfer of antibiotic-resistant genes to gut flora . However, GM food is already available in our supermarkets, mostly imported from overseas. Food Standards Australia New Zealand (FSANZ) “conducts a thorough safety assessment of all GM foods before they are allowed in the food supply” that ensures “any GM food that is approved is as safe as food already in the food supply, including in the long term”.  GM foods and ingredients (including food additives and processing aids) that contain novel DNA or novel protein must be labelled with the words “genetically modified”.  

Another concern is that private companies could claim ownership of the organisms they create and not share them at a reasonable cost – like the case with Monsanto . This could hurt small farmers who can’t afford the technology and instead favour large-scale monoculture farm production. 

More Reading

What do the experts reckon.

Scientists at Crown Research Institutes AgResearch, Scion and Plant & Food Research welcomed the changes, commenting that they would allow New Zealand to compete on an even playing field. “These new regulations will allow scientists to develop new plant varieties so the agrifood sector can adapt at the speed required to meet fast-moving challenges, such as climate change, and remain competitive on the global market,” said Richard Newcomb, chief scientist at Plant & Food Research.

Academics were a little more cautious in their praise, with Michael Bunce, genetics expert and honorary/adjunct professor at the University of Otago and Curtin University, saying that while an update to the law was needed, it was now time for New Zealand to have a “technical and nuanced conversation” that “requires us to pick up our DNA ‘game’ a little, and park debates from last century when gene technologies were still in their infancy. We may also have to accept that the country may remain divided on this topic – some New Zealanders will remain opposed to gene editing.”

University of Canterbury genetics professor Jack Heinemann, meanwhile, said that calling the current regulations a “ban” was “misleading hyperbole”. In comments via the Science Media Centre, Heinemann said, “There is no robust evidence that New Zealand’s regulation have prevented, or will prevent, truly useful products from coming to market.”

Josephine Johnson, associate professor at Otago’s Bioethics Centre, said reform made sense, but the approach would need to be adapted “in light of our country’s special features, and there I immediately think of our Treaty commitments and the high value we place on our natural environment”.

Labour’s science, innovation and technology spokesperson Deborah Russell said, “Unlike under National’s undemocratic and broken fast track legislation, Judith Collins must ensure its genetic modification and editing legislation includes the essential safeguards to protect our environment and is regulated to protect New Zealand’s interests.”

The Green Party’s response came from agriculture and food safety spokesman Steve Abel, who led Greenpeace’s GE campaign in the early 2000s. Speaking to the Herald, he said the party would continue to “ oppose the environmental release of GE crops ” though they back “ethical use of GE biotechnology in containment, including medical use”. When asked whether the Greens could be convinced to back the legislation, Abel did not rule it out, instead saying there first needed to be a “ wide-ranging and robust public discussion “. In 2019, an open letter signed by more than 150 young scientists urged the Greens to change their position in light of the climate crisis, and take the lead on GM reform.

When will this all happen?

Collins has said the legislation will be introduced to parliament by the end of the year. There will be an opportunity for the public to have their say at the select committee stage – dates haven’t been set, but will be published on the MBIE website . Collins wants to have it passed and the regulator in operation by the end of 2025. 

The Spinoff’s political coverage is powered by the generous support of our members . If you value what we do and believe in the importance of independent and freely accessible journalism – tautoko mai,  donate today .

IMAGES

  1. Genetic Engineering Essay

    introduction of genetic engineering essay

  2. Benefits of Genetic Engineering

    introduction of genetic engineering essay

  3. Genetic Engineering Essay

    introduction of genetic engineering essay

  4. Genetic Engineering Essay Example

    introduction of genetic engineering essay

  5. Impressive Genetic Engineering Essay ~ Thatsnotus

    introduction of genetic engineering essay

  6. Genetic Engineering (400 Words)

    introduction of genetic engineering essay

COMMENTS

  1. Genetic engineering

    The term genetic engineering initially referred to various techniques used for the modification or manipulation of organisms through the processes of heredity and reproduction.As such, the term embraced both artificial selection and all the interventions of biomedical techniques, among them artificial insemination, in vitro fertilization (e.g., "test-tube" babies), cloning, and gene ...

  2. Introduction to Genetic Engineering and Its Applications

    Genetic Engineering Technique. The process for genetic engineering begins the same for any organism being modified (see Figure 3 for an example of this procedure). Identify an organism that contains a desirable gene. Extract the entire DNA from the organism. Remove this gene from the rest of the DNA.

  3. Principles of Genetic Engineering

    Genetic engineering is the use of molecular biology technology to modify DNA sequence(s) in genomes, using a variety of approaches. For example, homologous recombination can be used to target specific sequences in mouse embryonic stem (ES) cell genomes or other cultured cells, but it is cumbersome, poorly efficient, and relies on drug positive/negative selection in cell culture for success.

  4. PDF An Introduction to Genetic Engineering Third Edition

    Introduction 1.1 What is genetic engineering? Progress in any scientific discipline is dependent on the availability of techniques and methods that extend the range and sophistication of experiments that may be performed. Over the past 35 years or so this has been demonstrated in a spectacular way by the emergence of genetic engineering.

  5. Genetic engineering

    Genetic engineering, also called genetic modification or genetic manipulation, is the modification and manipulation of an organism's genes using technology.It is a set of technologies used to change the genetic makeup of cells, including the transfer of genes within and across species boundaries to produce improved or novel organisms.. New DNA is obtained by either isolating and copying the ...

  6. PDF Fourth Edition An Introduction to Genetic Engineering

    An Introduction to Genetic Engineering Fourth EditionThe fourth edition of this popular textbook retains its focus on the fundamental principles of gene manipulation, providing an accessible and broad-based introductio. to the subject for beginning undergraduate students. It has been brought thoroughly up to date with new chapters on the story ...

  7. PDF An Introduction to Genetic Engineering Third Edition

    Chapter 1 Introduction 2 Chapter summary 2 1.1 What is genetic engineering? 3 1.2 Laying the foundations 5 1.3 First steps 6 1.4 What's in store? 7 Concept map 10 Part I The basis of genetic engineering Chapter 2 Introducing molecular biology 12 Chapter summary 12 2.1 The way that living systems are organised 13 2.2 The flow of genetic ...

  8. [PDF] An Introduction to Genetic Engineering

    An Introduction to Genetic Engineering. D. Nicholl. Published 10 February 2023. Biology. The fourth edition of this popular textbook retains its focus on the fundamental principles of gene manipulation, providing an accessible and broad-based introduction to the subject for beginning undergraduate students. It has been brought thoroughly up to ...

  9. Historic Overview of Genetic Engineering Technologies for Human Gene

    Introduction. Gene therapy is a therapeutic strategy using genetic engineering techniques to treat various diseases. 1,2) In the early 1960s, gene therapy first progressed with the development of recombinant DNA (rDNA) technology, 1) and was further developed using various genetic engineering tools, such as viral vectors. 3-5) More than 1900 clinical trials have been conducted with gene ...

  10. Human Genetic Engineering: Key Principles and Issues Essay

    Discussion. Genetic engineering is a recent breakthrough in humanity in the field of medicine, formulating one of the most complex processes. Genetic engineering technologies include the construction of functionally active genetic structures, their introduction into the human body, and integration into the genome (Wheale & Schomber, 2019).

  11. Genetically Modified Organisms (GMOs)

    In 1971, the first debate over the risks to humans of exposure to GMOs began when a common intestinal microorganism, E. coli, was infected with DNA from a tumor-inducing virus (Devos et al ., 2007 ...

  12. (PDF) Introduction to Genetic Engineering

    Genetic engineer ing is the direct modification of an. org anism' s genome, which is the list of specific traits. (genes) s tored in the DNA. Changing the genome. enables engineer s to giv e ...

  13. Essays on Genetic Engineering

    Genetic Engineering. 2 pages / 835 words. Genetic engineering, also known as genetic modification, is the direct manipulation of DNA to alter an organism's characteristics (phenotype) in a particular way. It is a set of technologies used to change the genetic makeup of cells to produce improved or novel organisms.

  14. PDF Genetic Engineering (3500 words)

    Soon after the publication of the short essay by Crick and Watson on DNA structure ... therapy involves the introduction of novel genes into germ cells such as egg/early . 4 ... As the use of genetic engineering expands rapidly, it's hard to generate an exhaustive list of all possible applications. However, there are at least three other ...

  15. Genetic Engineering

    What is Genetic Engineering. In simple words, genetic engineering can be described as the manual addition of a new DNA into an organism. It aids the addition of such traits that are not originally found in the organisms. Recombinant DNA is required to create Genetically Modified Organisms (GMO.) An area of chromosome (gene) is spliced.

  16. Essay on Genetic Engineering: Unlocking Human Potential

    This essay examines genetic engineering, covering its applications, ethical considerations, focusing on its impact on science and technology. ... Introduction to Genetic Engineering. Genetic engineering, the deliberate modification of an organism's genome, has revolutionized fields from agriculture to medicine, offering unprecedented ...

  17. What do people think about genetic engineering? A systematic review of

    Subsequent landmark events where genetic engineering was applied to humans, such as the first clinical introduction of retrovirus in gene-modified cells by Rosenberg in 1989 (Hanna et al., 2017), the death of Jesse Gelsinger in 1999 after gene therapy intervention to treat a metabolic disorder (Caplan, 2019), and the death of X-SCID patients in ...

  18. Human enhancement: Genetic engineering and evolution

    Genetic engineering opens new possibilities for biomedical enhancement requiring ethical, societal and practical considerations to evaluate its implications for human biology, human evolution and our natural environment. In this Commentary, we consider human enhancement, and in particular, we explore genetic enhancement in an evolutionary context.

  19. 132 Genetic Engineering Essay Topic Ideas & Examples

    Genetic Engineering and Eugenics Comparison. The main idea in genetic engineering is to manipulate the genetic make-up of human beings in order to shackle their inferior traits. The concept of socially independent reproduction is replicated in both eugenics and genetic […] The Film "Gattaca" and Genetic Engineering.

  20. Genetic Engineering Essay Introduction

    Genetic Engineering Essay Introduction; Featured Example Essay Cloning and Genetic Engineering in the Food Animal Industry INTRODUCTION When the Roslin Institute's first sheep cloning work was announced in March 1996 the papers were full of speculation about its long-term implications. Because of this discovery, the media's attention has ...

  21. Exploring The Pros and Cons of Genetic Engineering

    Introduction. Genetic engineering is the process of altering the genetic makeup of an organism by adding, deleting or changing its DNA. The process involves the manipulation of genes and their transfer from one organism to another, thereby creating new traits or characteristics. Genetic engineering has been in existence for over three decades ...

  22. What does lifting the 'ban' on genetic modification mean for New

    Genetic modification, also known as gene technology or genetic engineering (GE), involves taking DNA and inserting it into the genome of another organism, so that organism can make new substances ...

  23. The Spherical Tokamak for Energy Production: theme issue introduction

    1. The case for fusion energy. Analysis of global progress to reach net-zero greenhouse gas emissions shows that current decarbonization plans will not achieve the aim by 2050 [] and that real implementation lags even further behind.Although the proportion of energy generated from low-carbon sources is increasing, so is the absolute quantity of greenhouse gases entering the atmosphere as ...

  24. PDF An Introduction to Genetic Engineering Third Edition

    Introduction 1.1 What is genetic engineering? Progress in any scientific discipline is dependent on the availability of techniques and methods that extend the range and sophistication of experiments that may be performed. Over the past 35 years or so this has been demonstrated in a spectacular way by the emergence of genetic engineering.