We use cookies to distinguish you from other users and to provide you with a better experience on our websites. Close this message to accept cookies or find out how to manage your cookie settings.
To save content items to your account,
please confirm that you agree to abide by our usage policies.
If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account.
Find out more about saving content to .
To save content items to your Kindle, first ensure [email protected]
is added to your Approved Personal Document E-mail List under your Personal Document Settings
on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part
of your Kindle email address below.
Find out more about saving to your Kindle.
Note you can select to save to either the @free.kindle.com or @kindle.com variations.
‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi.
‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.
Genome editing has recently evolved from a theoretical concept to a powerful and versatile set of tools. The discovery and implementation of CRISPR-Cas9 technology have propelled the field further into a new era. This RNA-guided system allows for specific modification of target genes, offering high accuracy and efficiency. Encouraging results are being announced in clinical trials employed in conditions like sickle cell disease (SCD) and transfusion-dependent beta-thalassaemia (TDT). The path finally led the way to the recent FDA approval of the first gene therapy drug utilising the CRISPR/Cas9 system to edit autologous CD34+ haematopoietic stem cells in SCD patients (Casgevy). Ongoing research explores the potential of CRISPR technology for cancer therapies, HIV treatment and other complex diseases. Despite its remarkable potential, CRISPR technology faces challenges such as off-target effects, suboptimal delivery systems, long-term safety concerns, scalability, ethical dilemmas and potential repercussions of genetic alterations, particularly in the case of germline editing. Here, we examine the transformative role of CRISPR technologies, including base editing and prime editing approaches, in modifying the genetic and epigenetic codes in the human genome and provide a comprehensive focus, particularly on relevant clinical applications, to unlock the full potential and challenges of gene editing.
Since the eighteenth century, animal breeding has significantly evolved, culminating in the use of gene editing (GE) technologies like CRISPR-Cas9. These technologies offer unprecedented capabilities to modify animal genomes, potentially revolutionising breeding practices by achieving desired traits much faster compared to traditional selective breeding (SB). Because breeding programmes focussed on productivity traits have often compromised animal welfare, this article provides an analysis of the ethical issues underpinning the use of GE in animal breeding, and the current laws regulating such practices. In doing so, this article critically examines the European Union (EU)’s regulatory framework for SB and GE in farm animals, highlighting significant gaps and inconsistencies. Specifically, the use of GE animals is currently regulated under the EU’s GMO legislation, while SB is more loosely regulated, posing substantial risks to animal welfare. The authors advocate for a regulatory framework that prioritises animal welfare goals and proposes reforms to enhance animal protection objectives by ensuring a more consistent and humane approach to farm animal breeding.
Various in-vitro (induced pluripotent stem cells–derived) and in-vivo genetic models (animal models like Caenorhabditis elegans, Drosophila melanogaster, zebrafish, rodents, and non-human primates) have been used to study movement disorders such as Parkinson’s disease, hereditary ataxia, Huntington’s disease, dystonia, and essential tremor. These genetic models have provided important clues on the underlying pathophysiologic mechanisms of these diseases and serve as useful platforms to unravel potential therapeutic targets. The next generation of genetic models is promising with the advancement of gene-editing techniques, such as CRISPR-Cas9, brain organoid technology, and identification of novel genes and loci from large-scale genetic studies will facilitate development of new genetic models.
Utilizing online survey data of US consumers, this study examines the extent to which consumers' acceptance of genetically modified (GM) and gene-edited (GE) food is driven by their risk attitudes. Our results indicate that individuals with high-risk propensity are more likely to accept both GM and GE food than individuals with low- and medium-risk propensity. Our results also find differences in consumers' attitudes toward plants and animal products in the context of both GM and GE. Intriguingly, these attitudinal differences can be explained by consumers' risk propensities. Specifically, both low- and medium-risk propensity consumers differentiate between plants and animal products; the latter is less acceptable than the former, indicating a tendency to have more concerns about the application of biotechnology to animals than plants. However, individuals with high-risk propensity do not differentiate between GM and GE plants and animal products. Our results suggest that policymakers, the food industry, and researchers need to consider these attitudinal differences while studying consumer attitudes toward GM and GE food. Failing to capture these attitudinal differences in studies focusing on consumer behavior toward GM and GE food may result in either overestimating or underestimating consumer response.
New breeding methods have provided scientists with opportunities to improve traits in a wide range of crops, however, there remains resistance to foods that are produced from these crops, and mandates on labels used to describe such processes continue to be a source of policy debate. Here we focus on gene editing and examine (i) whether consumer acceptance varies when the technology is applied to different ingredients (unrefined versus highly refined ingredients) and (ii) the impact of two different claims related to gene editing (health-focused claim versus an environment-focused claim). Our results show that consumers are less likely to purchase a food product that includes gene-edited ingredients, yet the ingredient that is gene-edited is not important. We also find evidence that both of our selected claims about foods produced using gene-edited ingredients would increase consumers’ likelihood to purchase relative to the case with no claims.
Philosophers have used thought experiments to examine contentious examples of genetic modification. We hypothesised that these examples would prove useful in provoking responses from lay participants concerning technological interventions used to address welfare concerns. We asked 747 US and Canadian citizens to respond to two scenarios based on these thought experiments: genetically modifying chickens to produce blind progeny that are less likely to engage in feather-pecking (BC); and genetically modifying animals to create progeny that do not experience any subjective state (i.e. incapable of experiencing pain or fear; IA). For contrast, we assessed a third scenario that also resulted in the production of animal protein with no risk of suffering but did not involve genetically modifying animals: the development of cultured meat (CM). Participants indicated on a seven-point scale how acceptable they considered the technology (1 = very wrong to do; 7 = very right to do), and provided a text-based, open-ended explanation of their response. The creation of cultured meat was judged more acceptable than the creation of blind chickens and insentient animals. Qualitative responses indicated that some participants accepted the constraints imposed by the thought experiment, for example, by accepting perceived harms of the technology to achieve perceived benefits in reducing animal suffering. Others expressed discomfort with such trade-offs, advocating for other approaches to reducing harm. We conclude that people vary in their acceptance of interventions within existing systems, with some calling for transformational change.
What are genes? What do genes do? These questions are not simple and straightforward to answer; at the same time, simplistic answers are quite prevalent and are taken for granted. This book aims to explain the origin of the gene concept, its various meanings both within and outside science, as well as to debunk the intuitive view of the existence of 'genes for' characteristics and disease. Drawing on contemporary research in genetics and genomics, as well as on ideas from history of science, philosophy of science, psychology and science education, it explains what genes are and what they can and cannot do. By presenting complex concepts and research in a comprehensible and rigorous manner, it examines the potential impact of research in genetics and genomics and how important genes actually are for our lives. Understanding Genes is an accessible and engaging introduction to genes for any interested reader.
Regulatory agencies aim to protect the public by moderating risks associated with innovation, but a good regulatory regime should also promote justified public trust. After introducing the USDA 2020 SECURE Rule for regulation of biotech innovation as a case study, this essay develops a theory of justified public trust in regulation. On the theory advanced here, to be trustworthy, a regulatory regime must (1) fairly and effectively manage risk, must be (2) “science based” in the relevant sense, and must in addition be (3) truthful, (4) transparent, and (5) responsive to public input. Evaluated with these norms, the USDA SECURE Rule is shown to be deeply flawed, since it fails appropriately to manage risk, and similarly fails to satisfy other normative requirements for justified trust. The argument identifies ways in which the SECURE Rule itself might be improved, but more broadly provides a normative framework for the evaluation of trustworthy regulatory policy-making.
To understand what genes “do,” we have to consider what happens during development. The first and most striking evidence that the local environment matters for the outcome of development was provided by the experiments of embryologists Wilhelm Roux and Hans Driesch in the late nineteenth and early twentieth centuries. Roux had hypothesized that during the cell divisions of the embryo, hereditary particles were unevenly distributed in its cells, thus driving their differentiation. This view entailed that even the first blastomeres (the cells emerging from the first few divisions of the zygote – that is, the fertilized ovum) would each have different hereditary material and that the embryo would thus become a kind of mosaic. Roux decided to test this hypothesis. He assumed that if it were true, destroying a blastomere in the two-cell or the four-cell stage would produce a partially deformed embryo. If it were not true, then the destruction of a blastomere would have no effect. With a hot sterilized needle, Roux punctured one of the blastomeres in a two-cell frog embryo that was thus killed. The other blastomere was left to develop. The outcome was a half-developed embryo; the part occupied by the punctured blastomere was highly disorganized and undifferentiated, whereas those cells resulting from the other blastomere were well-developed and partially differentiated. This result stood as confirmation for Roux’s hypothesis.
During the 1970s, more puzzling observations were made. The first was that the genome of animals contained large amounts of DNA with unique sequences that should correspond to a larger number of genes than anticipated. It was also observed that the RNA molecules in the nuclei of cells were much longer than those found outside the nucleus, in the cytoplasm. These observations started making sense in 1977, when sequences of mRNA were compared to the corresponding DNA sequences. It was shown that certain sequences that existed in the DNA did not exist in the mRNA, and that therefore they must have been somehow removed. It was thus concluded that the genes encoding various proteins in eukaryotes included both coding sequences and ones that were not included in the mRNA that would reach the ribosomes for translation. These “removed” sequences were called introns, to contrast them with the ones that were expressed in translation, which were called exons. The procedure that removed the intron sequences from the initial mRNA and that left only the exon sequences in the mature mRNA was named “RNA splicing.”
One important, and for some the most surprising, conclusion of genome-wide association studies (GWAS) has been that in most cases numerous single nucleotide polymorphism (SNPs) in several genes were found to be associated with the development of a characteristic or the risk of developing a disease. As already mentioned, the main conclusion has been that the relationship between genes and characteristics or diseases is usually a many-to-many one, as many genes may be implicated in the same condition, and the same gene may be implicated in several different conditions. In fact, the same allele may be protective for one disease but increase the risk for another. For example, a variation in the PTPN22 (protein tyrosine phosphatase, nonreceptor type 22) gene on chromosome 1 seems to protect against Crohn’s disease but to predispose to autoimmune diseases. In other cases, certain variants are associated with more than one disease, such as the JAZF1 (JAZF1 zinc finger 1) gene on chromosome 7 that is implicated in prostate cancer and in type 2 diabetes. Therefore, we should forget the simple scheme of gene 1 → condition 1/gene 2 → condition 2, and adopt a richer – and certainly more complicated – representation of the relationship between genes and disease. Additional GWAS on more variants in larger populations might provide a better picture in the future. But insofar as we do not understand all biological processes in detail, all we are left with are probabilistic associations between genes and characteristics (or diseases). The “associated gene” may be informative, but its explanatory potential and clinical value are limited – at least for now.
This chapter is about the public image of genes. But what exactly do we mean by “public”? Here, I use the word as a noun or an adjective vaguely, in order to refer to all ordinary people who are not experts in genetics. I thus contrast them with scientists who are experts in genetics – that is, who have mastered genetics-related knowledge and skills, who practice these as their main occupation, and who have valid genetics-related credentials, confirmed experience, and affirmation by their peers. I must note that both “experts” and “the public” are complex categories that depend on the context and that change over time. There is no single group of nonexperts that we can define as “the” public, as people around the world differ in their perceptions of science, depending on their cultural contexts. We had therefore better refer to “publics.” The differences among experts nowadays might be less significant than those among nonexperts, given today’s global scientific communities, but they do exist. Finally, both the categories of experts and publics have changed across time, depending, on the one hand, on the level of experts’ knowledge and understanding of the natural world, and, on the other hand, on publics’ attitudes toward that knowledge and understanding.
If you were taught Mendelian genetics at school (see Figures 2.1 and 2.2) you should be aware that it is an oversimplified model that does not work for most cases of inherited characteristics. Human eye color is a textbook example of a monogenic characteristic. It refers to the color of the iris – the colored circle in the middle of the eye. The iris comprises two tissue layers, an inner one called the iris pigment epithelium and an outer one called the anterior iridial stroma. It is the density and cellular composition of the latter that mostly affects the color of the iris. The melanocyte cells of the anterior iridial stroma store melanin in organelles called melanosomes. White light entering the iris can absorb or reflect a spectrum of wavelengths, giving rise to the three common iris colors (blue, green–hazel, and brown) and their variations. Blue eyes contain minimal pigment levels and melanosome numbers; green–hazel eyes have moderate pigment levels and melanosome numbers; and brown eyes are the result of high melanin levels and melanosome numbers. Textbook accounts often explain that a dominant allele B is responsible for brown color, whereas a recessive allele b is responsible for blue color (Figure 4.1). According to such accounts, parents with brown eyes can have children with blue eyes, but it is not possible for parents with blue eyes to have children with brown eyes. This pattern of inheritance was first described at the beginning of the twentieth century and it is still taught in schools, although it became almost immediately evident that there were exceptions, such as that two parents with blue eyes could have offspring with brown or dark hazel eyes.
Perhaps you were taught at school that genetics began with Gregor Mendel. Because of his experiments with peas, Mendel is considered to be a pioneer of genetics and the person who discovered the laws of heredity. According to the model of “Mendelian inheritance,” things are rather simple and straightforward with inherited characteristics. Some alleles are dominant – that is, they impose their effects on other alleles that are recessive. An individual who carries two recessive alleles exhibits the respective “recessive” characteristic, whereas a single dominant allele is sufficient for the “dominant” version of the characteristic to appear. In this sense, particular genes determine particular characteristics (e.g., seed color in peas), and particular alleles of those genes determine particular versions of the respective characteristics. Mendel, the story goes, discovered that characteristics are controlled by hereditary factors, the inheritance of which follows two laws: the law of segregation and the law of independent assortment.
A large and highly heterogeneous group of individuals conducts genetic and genomic research outside of traditional corporate and academic settings. They can be an important source of innovation, but their activities largely take place beyond the purview of existing regulatory systems for promoting safe and ethical practices. Historically the gene-targeting technology available for non-traditional biology (NTB) experiments has been limited, and therefore they have attracted little regulatory attention. New techniques such as CRISPR-cas9, however, may create a need for alternate governance approaches. This chapter explores whether alternate governance approaches might be needed and, if so, what governance approaches would be most likely to enable non-traditional experiments to be conducted safely and ethically.
Gene editing technologies consist of a set of engineering tools, such as CRISPR/Cas9, that seek to deliberately target and modify specific DNA sequences of living cells. While technical and safety challenges prevail, particularly regarding germline applications, these technologies are touted as transformational for the promotion and improvement of health and well-being. Furthermore, their enhanced simplicity, efficiency, precision and affordability had spurred their development. This in turn, has brought to the fora scientific and socio-political debates concerning their wide range of actual and potential applications together with their inexorable ethical implications. So far, human gene editing (HGE) has stimulated a new wave of policy by an extensive range of national and international actors (e.g. governments, professional organizations, funding agencies, etc.). This chapter outlines some of the socio-ethical issues raised by HGE technologies, with focus on germline interventions (HGI) and addresses a variety of policy frameworks. It further analyses commonalities as well as divergences in approaches traversing a continuum of normative models.
The global food system exhibits dizzying complexity, with interaction among social, economic, biological, and technological factors. Opposition to the first generation of plants and animals transformed through rDNA-enabled gene transfer (so-called GMOs) has been a signature episode in resistance to the forces of industrialization and globalization in the food system. Yet agricultural scientists continue to tout gene technology as an essential component in meeting future global food needs. An ethical analysis of the debate over gene technologies reveals the details that matter. On the one hand, alternative regimes for institutionalizing gene technology (through regulation, trade policy, and intellectual property law) could mitigate injustices suffered by politically marginalized and economically disadvantaged actors in the food system, especially smallholding farmers in less industrialized economies. On the other hand, GMO opposition has been singularly effective in mobilizing citizens of affluent countries against policies and practices that lie at the heart of these same injustices. As part of the roundtable, “Ethics and the Future of the Global Food System,” this essay argues that charting a middle course that realizes the benefits of gene technology while blocking its use in the perpetration of unjust harms may require a more detailed grasp of intricacies in the food system than even motivated bystanders are willing to develop.
Recent advances in gene editing technology promise much for medical advances and human well-being. However, in parallel domains, there have been objections to the use of such biotechnologies. Moreover, the psychological factors that govern the willingness to use gene editing technology have been underexplored to date. In this registered report, we sought to test whether pathogen disgust sensitivity is linked with opposition to gene editing. U.K.-based adult participants (N = 347) were recruited to this study. Gene editing attitudes reflected two largely distinct latent factors concerning enhancing human traits and treating medical disorders. In contrast to prediction, pathogen disgust sensitivity was related to greater support for gene editing in both of these domains. This result suggests that gene editing, at least in the current study, is not viewed as pathogenic, or that the perceived benefits of gene editing outweigh any perceived pathogen risk.
Sweden can be considered a relatively liberal European country when it comes to research, for example, it allows creating embryos for research purposes; yet, the question of human germline genome modification has been approached with great caution. With the adoption of the Genetic Integrity Act in 2006, the Swedish legislature intended to enable some research relating to gene editing technology while simultaneously placing bans on its use in clinical trials and clinical care, and providing criminal sanctions if these bans are violated. In this way, Swedish law is also aligned with its external commitments, and in particular, the EU Clinical Trials laws. While arguably the Genetic Integrity Act could have effectively functioned prior to the advances in gene editing technology, today it may be regarded as ambiguous and outdated. Hence, risks that ethically contested practices could emerge cannot be excluded. This chapter examines the national laws and policies relating to human germline genome modification in research and in clinical care in Sweden, with due regard to Sweden’s external commitments. Importantly, in light of the ongoing regulatory discussions at the national, European and international fora, it is not obvious that, should European laws become more permissive, and enable human germline genome modification, so would Swedish national law.