teknos logo.png

Welcome to the website for Teknos, Thomas Jefferson's Science Journal, showcasing student articles, papers, and editorials. Enjoy!

Ethics of Human Enhancement

Ethics of Human Enhancement

The Ethics of Human Enhancement and Genetic Modification

Anvi Padiyar, Anjali Pagidi, Jennifer Liu, Joanna Cheng, Grace Huang, and Sahana Ramesh
Edited by Teknos 2018-2019 Staff

Flying cars and civilizations on Mars may still be far away, but something much bigger is standing right at the horizon. As researchers venture into gene alterations, the human race as we know it is approaching a new biotechnological era that we look at today as “superhuman.” Human enhancements and genetic modifications may soon be able to give us superpowers such as boosted intelligence or memory (Henschke, 2017), but with these superpowers come social and ethical questions. How we answer these questions can determine the future of our society.

These technological breakthroughs are the subjects of many new innovations, such as the CRISPR-Cas9 system. First discovered in bacteria, CRISPR-Cas9 is a major component of the immune system, defending bacteria from viruses like phage infections ("What Are Genome," 2018). In the bacterial genome, there are sections of DNA containing clusters of certain palindromic repeat sequences, called Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR). When a phage attacks for the first time, the bacterium degrades the phage DNA and stores fragments of it within the CRISPR sequences in the bacterial genome ("What Are Genome," 2018). These stored viral DNA fragments are called spacers, and they allow the bacterium to easily recognize the phage in future attacks. In these subsequent attacks, the bacterium transcribes the spacers, producing CRISPR guide RNA (gRNA). The bacterium then activates the Cas-9 protein, which uses the gRNA as a template and scans the surrounding phage sequences for the protospacer: the target viral DNA complementary to the gRNA. Once the target is identified, Cas-9 cleaves the protospacer, creating a double-stranded break that deactivates the viral DNA and saves the bacteria.

So now we know that the naturally occurring CRISPR-Cas9 system helps defend prokaryotic bacteria from invading phages, but the remaining question is: what's the big deal? Since the early days of molecular biology, scientists have searched for ways to program DNA-protein interactions by localizing proteins to specific DNA sequences. Proteins do the majority of DNA modifications, so once a protein binds to a piece of DNA, the possibilities are endless: think controlling gene expression and cutting, inserting, and mutating DNA. Genome editing is one of the most promising applications of targeted DNA-protein interaction. If you can get proteins to bind to your target sequence, genes can be inserted and deleted, which opens the door to human enhancement. There are numerous obstacles, however. Because proteins must physically interact with DNA, and since protein structure is hard to predict, finding the right protein to bind to your DNA sequence of interest is difficult. Additionally, breaking DNA is a tricky, and often dangerous process. Previously carried out by chemical carcinogens that broke DNA in random locations, that often killed the cells. It's no wonder that CRISPR is the fastest, easiest, cheapest, and most precise tool in the field now. In CRISPR, the gRNA is complementary to the DNA target, not the protein. While predicting how a protein will interact with a DNA is hard, DNA and RNA interactions are simply based on complementarity! Thus, the current CRISPR-Cas9 editing system is based on choosing a gRNA complementary to your target DNA, in order to remove, insert, and edit specific genes. Due to the small size of gRNA, it can easily be expressed, modified, inserted into cells, and synthesized in labs. Thousands of gRNAs, each targeting a different sequence, are being compiled into libraries at this very moment (Kim & Colaiácovo, 2016).

Although the CRISPR method may seem to be a great improvement upon previous gene-editing methods, it is not without its drawbacks. The main issue with the CRISPR procedure is that the Cas-9 enzyme is not 100% accurate, and DNA is sometimes cut at sites other than the intended target, a phenomenon called “off-target effects.” If researchers do not find a way to make sure that the Cas-9 enzyme is precise down to micrometers, severe consequences can arise. Just one accidentally deleted base pair or the slightest tweak to an individual’s DNA sequence can cause malignancies and other health problems (Kim & Colaiácovo, 2016). Additionally, even when the DNA is cut at the correct spot, the precision of the edits may vary, leading to devastating consequences.

Although scientists are making strong headway in these gene-editing procedures, they have been a point of controversy since their conception. Genetic modifications for humans hold much promise for us in the years to come. One of the most obvious applications of this new innovation is the ability to remove inherited defective DNA segments from human embryos. Gene editing could eliminate severe hereditary diseases such as Cystic Fibrosis and Down Syndrome, bringing new hope to many families. A more controversial application of gene-editing is the creation of designer babies- engineering in vitro babies for selected traits, such as enhanced physical stamina, gender changes, and superintelligence.

While many in the medical field believe genetic modification has the potential to cure inherited diseases and other illnesses, this technology also raises many questions about the morality and ramifications of gene editing. In the future, as this technology becomes more mainstream, society may be affected in many ways (Douglas, 2015). For example, human genetic engineering will undoubtedly be an expensive procedure. Only wealthier people will be able to select and enhance their children's phenotype, and this new crop of genetically-modified people will only further widen the socioeconomic gap in society. Authors who write about utopian worlds often describe how those worlds become dystopian, how their inhabitants have manipulated and misused technology and advancements to create a state that is worse than before  (Mitchell & Kilner, 2003). In the book Red Queen, by Victoria Aveyard, there exists a world divided by the type of blood, where red-blooded commoners are ruled over by a silver-blooded race of elite superhumans. A similar broken world may become ours one day if genetic modification is not handled and used properly. We have no way of knowing what potential disputes this technology may bring, and they could be detrimental to the lives of countless individuals (Mitchell & Kilner, 2003).

People may also lose their individualities as the human race becomes more physically similar in pursuit of a certain standard of beauty (Douglas, 2015). This idea on a small scale is already intertwined with our society today, as evident in the growing plastic surgery industry. Genetic modifications, like plastic surgery and all other invasive procedures, pose the risk of side effects. This poses a question: to what extent is it ethically wrong to endanger human lives for modifications that are unnecessary for survival? Some may argue why alterations should be made to something that works properly — like the old adage says, “If it ain’t broke, don’t fix it”. However, others may argue that it is only natural for humans to evolve and their technology along with it.   

Human enhancement and genetic modification are upcoming science breakthroughs in modern medicine. Therefore, they are no longer subjects of science fiction but of real consequence. With the knowledge of current technologies, it is better to err on the side of caution, especially when considering relations to society. The short term benefits may seem groundbreaking, but we must be well informed of the potential hazards so that we are prepared for these innovations when they arrive.

References

[1] Aveyard, V. (2015). Red Queen. New York City, NYC: HarperCollins.

[2] Douglas, T. (2015). “The Harms of Enhancement and the Conclusive Reasons View.” Cambridge Quarterly of Healthcare Ethics, 24(1). https: //doi.org/10.1017/S0963180114000218

[3] Henschke, A. (2017, July 3). “‘Supersoldiers’: Ethical concerns in human enhancement technologies.” Retrieved November 5, 2018, from https://medium.com/law-and-policy/supersoldiers-ethical-concerns-in-human-enhanceme nt-technologies-fa9bf1e06889

[4] Kim, H., & Colaiácovo, M. P. (2016). CRISPR-Cas9-Guided Genome Engineering in C. elegans. Current Protocols in Molecular Biology. doi:10.1002/cpmb.7

[5] Mitchell, C. B., & Kilner, J. F. (2003, September 30). “Remaking Humans: The New Utopians Versus a Truly Human Future.” Retrieved November 5, 2018, from https://cbhd.org/content/remaking-humans-new-utopians-versus-truly-human-future

[6] Vidyasagar, A. (2018, April 20). “What is CRISPR?” Retrieved November 21, 2018, from https://www. livescience.com/58790-crispr-explained.html

[7] “What are genome editing and CRISPR-Cas9?” (2018, October 30). Retrieved November 5, 2018, from https://ghr.nlm.nih.gov/primer/genomicresearch/genomeediting

Computer-Generated Pastiche: Text Style Transfer

Computer-Generated Pastiche: Text Style Transfer

Obesity and Dementia

Obesity and Dementia