Journal articles on the topic 'Germline gene editing'

To see the other types of publications on this topic, follow the link: Germline gene editing.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Germline gene editing.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Sugarman, Jeremy. "Ethics and germline gene editing." EMBO reports 16, no. 8 (July 2, 2015): 879–80. http://dx.doi.org/10.15252/embr.201540879.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Mason, Veronica R. J. M. "Hurtling toward Germline Gene Editing." Ethics & Medics 44, no. 8 (2019): 1–4. http://dx.doi.org/10.5840/em201944811.

Full text
Abstract:
Genetic enhancement runs up against several moral issues, perhaps the chief of which is the inevitable eugenic attitude it would foster and the associated inequality it would create between those who have the “proper” enhancements and those who do not. For simplicity’s sake, this analysis leaves aside questions related to genetic enhancement and considers only changes made for therapeutic purposes. Regardless, most of the censure of He Jiankui focuses on the results of human modification and often overlooks the prior question of how gene editing research itself conducted. Germline gene editing in humans is not safe or morally licit under current practices and technology, because of its reliance on technologies such as IVF, the danger to and destruction of the embryos used, and the unknown consequences of changing the germline.
APA, Harvard, Vancouver, ISO, and other styles
3

Cartier-Lacave, Nathalie, Robin Ali, Seppo Ylä-Herttuala, Kazuto Kato, Bernard Baetschi, Robin Lovell-Badge, Luigi Naldini, and Adrian Thrasher. "Debate on Germline Gene Editing." Human Gene Therapy Methods 27, no. 4 (August 2016): 135–42. http://dx.doi.org/10.1089/hgtb.2016.28999.deb.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Nalley, Catlin. "Germline Gene Editing for Deafness." Hearing Journal 73, no. 2 (February 2020): 28. http://dx.doi.org/10.1097/01.hj.0000654908.10936.e1.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Cwik, Bryan. "Intergenerational monitoring in clinical trials of germline gene editing." Journal of Medical Ethics 46, no. 3 (August 31, 2019): 183–87. http://dx.doi.org/10.1136/medethics-2019-105620.

Full text
Abstract:
Design of clinical trials for germline gene editing stretches current accepted standards for human subjects research. Among the challenges involved is a set of issues concerning intergenerational monitoring—long-term follow-up study of subjects and their descendants. Because changes made at the germline would be heritable, germline gene editing could have adverse effects on individuals’ health that can be passed on to future generations. Determining whether germline gene editing is safe and effective for clinical use thus may require intergenerational monitoring. The aim of this paper is to identify and argue for the significance of a set of ethical issues raised by intergenerational monitoring in future clinical trials of germline gene editing. Though long-term, multigenerational follow-up study of this kind is not without precedent, intergenerational monitoring in this context raises unique ethical challenges, challenges that go beyond existing protocols and standards for human subjects research. These challenges will need to be addressed if clinical trials of germline gene editing are ever pursued.
APA, Harvard, Vancouver, ISO, and other styles
6

Gyngell, Christopher, Thomas Douglas, and Julian Savulescu. "The Ethics of Germline Gene Editing." Journal of Applied Philosophy 34, no. 4 (November 9, 2016): 498–513. http://dx.doi.org/10.1111/japp.12249.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Ranisch, Robert, Tina Rudolph, Hans-Joachim Cremer, and Nikolaus Knoepffler. "Ordo-Responsibility for Germline Gene Editing." CRISPR Journal 3, no. 1 (February 1, 2020): 37–43. http://dx.doi.org/10.1089/crispr.2019.0040.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Cwik, Bryan. "Designing Ethical Trials of Germline Gene Editing." New England Journal of Medicine 377, no. 20 (November 16, 2017): 1911–13. http://dx.doi.org/10.1056/nejmp1711000.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Sharma, Akshay, Nickhill Bhakta, and Liza-Marie Johnson. "Germline Gene Editing for Sickle Cell Disease." American Journal of Bioethics 20, no. 8 (August 2, 2020): 46–49. http://dx.doi.org/10.1080/15265161.2020.1781970.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Koplin, Julian J., Christopher Gyngell, and Julian Savulescu. "Germline gene editing and the precautionary principle." Bioethics 34, no. 1 (June 27, 2019): 49–59. http://dx.doi.org/10.1111/bioe.12609.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Savvina, Olga V. "Genetic Modification of Human Embryos: Limits." Ethical Thought 22, no. 1 (2022): 124–34. http://dx.doi.org/10.21146/2074-4870-2022-22-1-124-134.

Full text
Abstract:
The article analyses the moral justification of human germline editing and the tendency to its legalization. The study is based on documents of international organizations, such as the World Health Organization (WHO), national bioethics committees and others that regulate the usage of technologies for human germline editing or issue related recommendations. The paper an­alyzes the impact of the introduction of new technologies on human germline editing recom­mendations. It is concluded that that the development of biotechnologies contributes to lib­eral attitude towards human germline editing, slowly canceling the technologies’ usage ban firstly for therapeutic purposes, and then for the human enhancement purposes. The article suggests that the development of biotechnologies makes it difficult to apply the old bioethics principles; and exacerbates the discussion about the boundaries of the new biotechnologies’ application. Despite the shock and condemnation of the first experiments that violate ban (as in the cases with CRISPR/Cas9 in 2015 and 2018 in China), the scientific community, international organizations and governments return to the issue concerning gene editing technologies limitation. The inability to be guided by the old bioethics principles forces to look for new ethical grounds for gene editing. Now old principles and values are applied with utilitarian approach in ethics, that cancel ban and raises the issue of human germline editing limitation. The article also describes the limits of permissible interventions in the is­sue of human germline editing at the end of 2021.
APA, Harvard, Vancouver, ISO, and other styles
12

Tan, Zi Ying, Taosheng Huang, and Joanne Ngeow. "65 YEARS OF THE DOUBLE HELIX: The advancements of gene editing and potential application to hereditary cancer." Endocrine-Related Cancer 25, no. 8 (August 2018): T141—T158. http://dx.doi.org/10.1530/erc-18-0039.

Full text
Abstract:
Hereditary cancer predisposition syndromes are associated with germline mutations that lead to increased vulnerability for an individual to develop cancers. Such germline mutations in tumour suppressor genes, oncogenes and genes encoding for proteins essential in DNA repair pathways and cell cycle control can cause overall chromosomal instability in the genome and increase risk in developing cancers. Gene correction of these germline mutations to restore normal protein functions is anticipated as a new therapeutic option. This can be achieved through disruption of gain-of-function pathogenic mutation, restoration of loss-of-function mutation, addition of a transgene essential for cell function and single nucleotide changes. Genome editing tools are applicable to precise gene correction. Development of genome editing tools comes in two waves. The first wave focuses on improving targeting specificity and editing efficiency of nucleases, and the second wave of gene editing draws on innovative engineering of fusion proteins combining deactivated nucleases and other enzymes that are able to create limitless functional molecular tools. This gene editing advancement is going to impact medicine, particularly in hereditary cancers. In this review, we discuss the application of gene editing as an early intervention and possible treatment for hereditary cancers, by highlighting a selection of highly penetrant cancer syndromes as examples of how this may be achieved in clinical practice.
APA, Harvard, Vancouver, ISO, and other styles
13

Monckton, Darren G. "Manage risk of accidental gene editing of germline." Nature 568, no. 7753 (April 2019): 458. http://dx.doi.org/10.1038/d41586-019-01284-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
14

Ssebunnya, Gerald Michael. "Towards an appropriate African framework for public engagement with human genome editing: a call to synergistic action." Wellcome Open Research 7 (December 12, 2022): 302. http://dx.doi.org/10.12688/wellcomeopenres.18579.1.

Full text
Abstract:
The CRISPR-Cas9 system has revolutionised the biotechnology of human genome editing. Human germline gene editing promises exponential benefits to many in Africa and elsewhere, especially those affected by the highly prevalent monogenic disorders - for which, thanks to CRISPR, a relatively safe heritable radical therapy is now possible. Africa evidently presents a unique opportunity for empirical research in human germline gene editing because of its high prevalence of monogenic disorders. Critically, however, germline gene editing has raised serious ethical concerns especially because of the significant risks of inadvertent and intentional misuse of its transgenerational heritability. Calls for due prudence have become even more pronounced in the wake of the 2018 case of He Jiankui’s ‘CRISPR’d babies’. Meanwhile, Africa is seriously lagging in articulating its position on human genome editing. Conspicuously, there has been little to no attempt at comprehensively engaging the African public in discussions on the promises and concerns about human genome editing. Thus, the echoing key question remains as to how Africa should prudently embrace and govern this revolutionary biotechnology. In this article, therefore, I lay the groundwork for the possible development of an appropriate African framework for public engagement with human genome editing and call upon all stakeholders to urgent synergistic action. I particularly highlight the World Health Organisation’s possible leadership role in promptly establishing the requisite expert working group for this urgent need.
APA, Harvard, Vancouver, ISO, and other styles
15

Charo, R. Alta. "Germline Engineering and Human Rights." AJIL Unbound 112 (2018): 344–49. http://dx.doi.org/10.1017/aju.2018.88.

Full text
Abstract:
With the ever-increasing range of medical technologies at our disposal to mediate the processes of life, from conception to death, comes an ever-increasing number of decision points about human control of fate. And as we debate altering our fate—whether dictated by a deity or by chance—the discussion frequently devolves into a question of whether we may alter not only our own fate, but also that of our children. The advent of genome editing, whether by older methods or the newer, often more easily used methods employing CRISPR, has only made debating the controversial possibility of heritable “germline” editing more urgent. The advent of genome editing, whether by older methods or the newer, often more easily used methods employing CRISPR, has only made debating the controversial possibility of heritable “germline” editing more urgent. On the eve of the Second International Summit on Human Genome Editing, held at the end of November 2018 in Hong Kong, a startling and disturbing story began circulating - a Chinese researcher announced the first births of children whose genomes had been edited at the embryonic stage. The work (assuming the claim can be verified) suffered from myriad problems, beginning with the lack of a compelling medical need, and including inadequate preclinical research, lack of peer review, flawed subject recruitment and consent procedures, and an apparent disregard for both formal and informal rules governing genetic manipulation of embryos. The summit's organizing committee issued a statement, distinguishing this experiment from what would be a responsible translational pathway forward. But not surprisingly, others around the world immediately called for a global, enforceable prohibition on such genetic engineering. On the occasion of the Universal Declaration on Human Rights (UDHR)’s seventieth anniversary, this essay argues that the current human rights law on germline editing misunderstands both the mechanisms of genetics and the moral basis for human rights, suggesting a more nuanced approach as we move forward and keep pace with new gene-editing technologies.
APA, Harvard, Vancouver, ISO, and other styles
16

Malmqvist, Erik. "Clinical trials of germline gene editing: The exploitation problem." Bioethics 35, no. 7 (June 15, 2021): 688–95. http://dx.doi.org/10.1111/bioe.12903.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

de Melo-Martin, Inmaculada. "Germline Gene Editing: Minding the Past and the Future." American Journal of Bioethics 20, no. 8 (August 2, 2020): 36–38. http://dx.doi.org/10.1080/15265161.2020.1782521.

Full text
APA, Harvard, Vancouver, ISO, and other styles
18

de Wert, Guido, Guido Pennings, Angus Clarke, Ursula Eichenlaub-Ritter, Carla G. van El, Francesca Forzano, Mariëtte Goddijn, et al. "Human germline gene editing: Recommendations of ESHG and ESHRE." European Journal of Human Genetics 26, no. 4 (January 12, 2018): 445–49. http://dx.doi.org/10.1038/s41431-017-0076-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
19

Lyon, Jeff. "Bioethics Panels Open Door Slightly to Germline Gene Editing." JAMA 318, no. 17 (November 7, 2017): 1639. http://dx.doi.org/10.1001/jama.2017.13962.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Lantos, John D. "Hopes, Fears, and Deja Vu Regarding Germline Gene Editing." JAMA Pediatrics 173, no. 5 (May 1, 2019): 411. http://dx.doi.org/10.1001/jamapediatrics.2019.0098.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Kühn, Ralf. "Genome engineering in rodents – status quo and perspectives." Laboratory Animals 56, no. 1 (October 21, 2021): 83–87. http://dx.doi.org/10.1177/00236772211051842.

Full text
Abstract:
The introduction of the CRISPR-Cas9 system in 2013 has revolutionized experimental genetics in mice and rats. This commentary gives an overview on the use of CRISPR either for gene editing in the germline or for editing and beyond editing in somatic cells. Future perspectives are opened by emerging CRISPR technologies that could enable genome engineering at larger scale.
APA, Harvard, Vancouver, ISO, and other styles
22

Schwartz, Matthew L., M. Wayne Davis, Matthew S. Rich, and Erik M. Jorgensen. "High-efficiency CRISPR gene editing in C. elegans using Cas9 integrated into the genome." PLOS Genetics 17, no. 11 (November 8, 2021): e1009755. http://dx.doi.org/10.1371/journal.pgen.1009755.

Full text
Abstract:
Gene editing in C. elegans using plasmid-based CRISPR reagents requires microinjection of many animals to produce a single edit. Germline silencing of plasmid-borne Cas9 is a major cause of inefficient editing. Here, we present a set of C. elegans strains that constitutively express Cas9 in the germline from an integrated transgene. These strains markedly improve the success rate for plasmid-based CRISPR edits. For simple, short homology arm GFP insertions, 50–100% of injected animals typically produce edited progeny, depending on the target locus. Template-guided editing from an extrachromosomal array is maintained over multiple generations. We have built strains with the Cas9 transgene on multiple chromosomes. Additionally, each Cas9 locus also contains a heatshock-driven Cre recombinase for selectable marker removal and a bright fluorescence marker for easy outcrossing. These integrated Cas9 strains greatly reduce the workload for producing individual genome edits.
APA, Harvard, Vancouver, ISO, and other styles
23

Nordgren, Anders. "Designing Preclinical Studies in Germline Gene Editing: Scientific and Ethical Aspects." Journal of Bioethical Inquiry 16, no. 4 (November 21, 2019): 559–70. http://dx.doi.org/10.1007/s11673-019-09947-9.

Full text
Abstract:
AbstractHuman germline gene editing is often debated in hypothetical terms: if it were safe and efficient, on what further conditions would it then be ethically acceptable? This paper takes another course. The key question is: how can scientists reduce uncertainty about safety and efficiency to a level that may justify initiation of first-time clinical trials? The only way to proceed is by well-designed preclinical studies. However, what kinds of investigation should preclinical studies include and what specific conditions should they satisfy in order to be considered well-designed? It is argued that multispecies and multigenerational animal studies are needed as well as human embryo editing without implantation. In order to be possible to translate to first-time clinical trials, animal studies need to satisfy strict conditions of validity. Moreover, embryo studies intended for translation to first-time clinical trials need to correspond to the animal studies in experimental design (with exception of implantation). Only in this way can uncertainty about risk for harm (safety) and prospect of benefit (efficiency) in first-time clinical trials be reduced to a modest level. If uncertainty is not reduced to such a level, first-time clinical trials in germline gene editing should not be initiated.
APA, Harvard, Vancouver, ISO, and other styles
24

Beriain, Iñigo de Miguel, Emilio Armaza Armaza, and Aliuska Duardo Sánchez. "Human germline editing is not prohibited by the Oviedo Convention: An argument." Medical Law International 19, no. 2-3 (June 2019): 226–32. http://dx.doi.org/10.1177/0968533219862590.

Full text
Abstract:
Human germline gene editing has ignited wide-ranging debates on the ethical and legal issues involved. The text of the Oviedo Convention is particularly relevant here, as it remains the only international legally binding instrument on the protection of human rights in the biomedical field which considers human genome modification. However, it is often misinterpreted. Indeed, most of the academic literature assumes that Article 13 forbids germline gene editing. This article seeks to demonstrate that this belief is mistaken. To this purpose, it develops a general analysis of the Convention, its Explanatory Report, and its historical background. As a result, it argues that the Convention does not veto genetic editing for basic research purposes, but only its clinical application on human embryos to be transferred into a womb. Nevertheless, it recommends a revision of the clause according to the original intention of the Convention.
APA, Harvard, Vancouver, ISO, and other styles
25

Johnston, Josephine. "Budgets versus Bans: How U.S. Law Restricts Germline Gene Editing." Hastings Center Report 50, no. 2 (March 2020): 4–5. http://dx.doi.org/10.1002/hast.1094.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

CWIK, BRYAN. "Moving Beyond ‘Therapy’ and ‘Enhancement’ in the Ethics of Gene Editing." Cambridge Quarterly of Healthcare Ethics 28, no. 04 (September 17, 2019): 695–707. http://dx.doi.org/10.1017/s0963180119000641.

Full text
Abstract:
Abstract:Since the advent of recombinant DNA technology, expectations (and trepidations) about the potential for altering genes and controlling our biology at the fundamental level have been sky high. These expectations have gone largely unfulfilled. But though the dream (or nightmare) of being able to control our biology is still far off, gene editing research has made enormous strides toward potential clinical use. This paper argues that when it comes to determining permissible uses of gene editing in one important medical context—germline intervention in reproductive medicine—issues about enhancement and eugenics are, for the foreseeable future, a red herring. Current translational goals for gene editing research involve a different kind of editing than would be required to achieve manipulation of complex traits such as intelligence, and there are more pressing (and unresolved) questions that need attention if clinical use of gene editing in reproductive medicine ever becomes a possibility.
APA, Harvard, Vancouver, ISO, and other styles
27

Annas, George J. "Genome Editing 2020: Ethics and Human Rights in Germline Editing in Humans and Gene Drives in Mosquitoes." American Journal of Law & Medicine 46, no. 2-3 (May 2020): 143–65. http://dx.doi.org/10.1177/0098858820933492.

Full text
Abstract:
The moon landing, now more than a half century in the past, has turned out to be the culmination of human space travel, rather than its beginning. Genetic engineering, especially applications of CRISPR, now presents the most publicly-discussed engineering challenges—and not just technical, but ethical as well. In this article, I will use the two most controversial genomic engineering applications to help identify the ethics and human rights implications of these research projects. Each of these techniques directly modifies the mechanisms of evolution, threatens to alter our views of ourselves as humans and our planet as our home, and presents novel informed consent and dual use challenges: human genome editing and gene drives in insects.I begin with a discussion of so far disastrously unsuccessful attempts to regulate germline editing in humans, including a summary of the first application of germline genome editing in humans and its aftermath. I then turn to a discussion of setting ethical standards for a genomic technology that has not yet been deployed in nature—gene drives. Finally, I end by suggesting that human rights can and should be directly applicable to defining the ethics of genomic research.
APA, Harvard, Vancouver, ISO, and other styles
28

Zhang, Di, and Reidar K. Lie. "Ethical issues in human germline gene editing: a perspective from China." Monash Bioethics Review 36, no. 1-4 (December 2018): 23–35. http://dx.doi.org/10.1007/s40592-018-0091-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

Coller, Barry S. "Ethics of Human Genome Editing." Annual Review of Medicine 70, no. 1 (January 27, 2019): 289–305. http://dx.doi.org/10.1146/annurev-med-112717-094629.

Full text
Abstract:
Advances in human genome editing, in particular the development of the clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 method, have led to increasing concerns about the ethics of editing the human genome. In response, the US National Academy of Sciences and the National Academy of Medicine constituted a multidisciplinary, international committee to review the current status and make recommendations. I was a member of that committee, and the core of this review reflects the committee's conclusions. The committee's report, issued in February 2017, recommends the application of current ethical and regulatory standards for gene therapy to somatic (nonheritable) human genome editing. It also recommends allowing experimental germline genome editing to proceed if ( a) it is restricted to preventing transmission of a serious disease or condition, ( b) the edit is a modification to a common DNA sequence known not to be associated with disease, and ( c) the research is conducted under a stringent set of ethical and regulatory requirements. Crossing the so-called red line of germline genome editing raises important bioethical issues, most importantly, serious concern about the potential negative impact on individuals with disabilities. This review highlights some of the major ethical considerations in human genome editing in light of the report's recommendations.
APA, Harvard, Vancouver, ISO, and other styles
30

Hurlbut, J. Benjamin. "The Demands of CRISPR’s World." Ethics & Medics 41, no. 4 (2016): 1–2. http://dx.doi.org/10.5840/em20164146.

Full text
Abstract:
A growing chorus of voices is declaring that CRISPR will revolutionize the ability to control life, including human life. As genetically altering future generations becomes technically realistic, it raises the prospect of genetic enhancement and the specter of eugenics. Prominent scientists are calling for international guidelines to govern human applications of gene-editing technology. They argue that the technical possibility of human germline gene editing makes ethical deliberation urgent. Now that the technology is upon us, the time has come to ask whether we want it. Human germline genetic engineering has long been marked as a morally significant boundary, and in numerous countries it is explicitly prohibited by law. The Oviedo Convention, a legally binding treaty among twenty-nine European countries, prohibits it as a violation of human rights and dignity. Nevertheless, numerous commentators argue that prohibitions made before it was technically possible meant little, and past proscriptions must now be revised.
APA, Harvard, Vancouver, ISO, and other styles
31

Hall, Jeanatan. "The Ethics of Human Tripronuclear Zygotes as Germline Editing Subjects." National Catholic Bioethics Quarterly 19, no. 3 (2019): 429–42. http://dx.doi.org/10.5840/ncbq201919332.

Full text
Abstract:
Despite great interest in the field of gene editing, sparked by the advent of CRISPR/Cas9-mediated applications, the personhood of tripronuclear zygotes has not been addressed appropriately. 3PN zygotes are discarded as medical waste, and their use as models for human genome editing is becoming increasing common. 3PN zygotes possess an extra set of chromosomes, which often leads to severe genetic abnormalities; they are dismissed as “nonviable embryos” and treated as an ethically acceptable alternative to human embryonic research. However, given the development cycle of 3PN zygotes and the qualifications for human personhood assessed, there is compelling evidence that 3PN zygotes are indeed human persons. Although genetically disadvantaged, they deserve the same respect as do genetically normal human zygotes.
APA, Harvard, Vancouver, ISO, and other styles
32

Evans, John H. "Setting ethical limits on human gene editing after the fall of the somatic/germline barrier." Proceedings of the National Academy of Sciences 118, no. 22 (April 30, 2021): e2004837117. http://dx.doi.org/10.1073/pnas.2004837117.

Full text
Abstract:
The ethical debate about what is now called human gene editing (HGE) has gone on for more than 50 y. For nearly that entire time, there has been consensus that a moral divide exists between somatic and germline HGE. Conceptualizing this divide as a barrier on a slippery slope, in this paper, I first describe the slope, what makes it slippery, and describe strong barriers that arrest the slippage down to the dystopian bottom of pervasive eugenic enhancement. I then show how the somatic/germline barrier in the debate has been weakened to the level of ineffectiveness, with no replacement below. I examine a number of possible barriers on the slope below the somatic/germline barrier, most of which lack sufficient strength. With the exception of the minority of people in the HGE debate who see the eugenic society as utopia, the majority will need a barrier on the slope to stop the slide to dystopia.
APA, Harvard, Vancouver, ISO, and other styles
33

Cussins, Jessica, and Leah Lowthorp. "Germline Modification and Policymaking: The Relationship between Mitochondrial Replacement and Gene Editing." New Bioethics 24, no. 1 (January 2, 2018): 74–94. http://dx.doi.org/10.1080/20502877.2018.1443409.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Johnston, Josephine. "Shaping the CRISPR Gene-Editing Debate: Questions About Enhancement and Germline Modification." Perspectives in Biology and Medicine 63, no. 1 (2020): 141–54. http://dx.doi.org/10.1353/pbm.2020.0011.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

Wolinetz, Carrie D., and Francis S. Collins. "NIH supports call for moratorium on clinical uses of germline gene editing." Nature 567, no. 7747 (March 2019): 175. http://dx.doi.org/10.1038/d41586-019-00814-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

Ranisch, Robert. "Germline Gene Editing and Genetic Enhancement: The Value of(Non-)Positional Goods." American Journal of Bioethics 19, no. 7 (June 25, 2019): 45–47. http://dx.doi.org/10.1080/15265161.2019.1618956.

Full text
APA, Harvard, Vancouver, ISO, and other styles
37

Ranisch, Robert. "‘Eugenics is Back’? Historic References in Current Discussions of Germline Gene Editing." NanoEthics 13, no. 3 (November 28, 2019): 209–22. http://dx.doi.org/10.1007/s11569-019-00351-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
38

Lala, Ismail. "Germ-Inating Solutions or Gene-Rating Problems: An Islamic Perspective on Human Germline Gene Editing." Journal of Religion and Health 59, no. 4 (February 18, 2019): 1855–69. http://dx.doi.org/10.1007/s10943-019-00770-5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
39

Пестрикова, Анастасия, and Елена Холопова. "FORMATION OF PRINCIPLES OF LEGAL REGULATION OF HUMAN GERMLINE GENOME EDITING." Rule-of-law state: theory and practice 16, no. 4-2 (April 1, 2020): 144–56. http://dx.doi.org/10.33184/pravgos-2020.4.31.

Full text
Abstract:
The article substantiates the need to establish legal principles for the use of gene modifications in human embryos and reproductive cells; highlights the problems of genome editing that require understanding by legal science, including at the international and national levels, and do not have a clear solution at the present stage. Purpose: based on the analysis of the legislation of foreign countries, the need for accessibility and openness to the international community of information on the safety of using technologies for gene editing of human germline, the identification of new forms of scientific activity focused on working with public opinion on the use of genetic technologies is proved. Methods: methods of comparative legal analysis, formal logic, description, statistical methods, and the method of legal norms interpretation are used. Results: at the present stage, the main task of the legal community is to develop legal principles for regulating public relations in the field of genetic engineering and biomedical technologies. At the same time, the most important component is the formation of public participation and active involvement of public institutions in the formation of the legal environment for creating a system of legal norms that meet the realities of scientific progress and protect the rights and interests of each person and humanity as a whole.
APA, Harvard, Vancouver, ISO, and other styles
40

Bi, Honglun, Xia Xu, Xiaowei Li, Yong Zhang, Yongping Huang, Kai Li, and Jun Xu. "CRISPR Disruption of BmOvo Resulted in the Failure of Emergence and Affected the Wing and Gonad Development in the Silkworm Bombyx mori." Insects 10, no. 8 (August 19, 2019): 254. http://dx.doi.org/10.3390/insects10080254.

Full text
Abstract:
The domesticated silkworm is an economically important insect that is widely used as a lepidopteran insect model. Although somatic sex determination in the silkworm is well characterized, germline sex determination is not. Here, we used the transgenic-based CRISPR/Cas9 genome editing system to study the function of the Ovo gene in Bombyx mori. BmOvo is the homolog of a factor important in germline sex determination in Drosophila melanogaster. BmOvo mutants had abnormally shaped eggs that were disordered in the ovarioles, and gonad development was abnormal. Interestingly, wing discs and wings did not develop properly, and most of the mutants failed to eclose. Gene expression analyses by qRT-PCR showed that BmOvo gene was highly expressed in the wing disc and epidermis. Genes involved in the WNT signaling pathway and wing development genes BmWCP10 and BmE74 were downregulated in the BmOvo mutants when compared with wild-type animals. These results demonstrate that the BmOvo gene product plays an important role in wing metamorphosis. Thus, this study provides new insights into the multiple functions of BmOvo beyond germline sex determination.
APA, Harvard, Vancouver, ISO, and other styles
41

Sutton, Agneta. "Editing della linea germinale: quali sono i rischi sociali e morali? / Germ-line gene editing: What are the social and moral risks?" Medicina e Morale 65, no. 2 (September 21, 2016): 123–30. http://dx.doi.org/10.4081/mem.2016.430.

Full text
Abstract:
Dovremmo accogliere tutti i possibili sviluppi dell’editing genetico? L’editing genetico delle cellule somatiche potrebbe essere considerato alla pari delle terapie convenzionali volte a trattare particolari patologie o ad alleviarne i sintomi. Tale intervento interesserebbe esclusivamente il singolo paziente trattato. Esso potrebbe quindi essere ben accolto come un nuovo tipo di trattamento per i tumori e le malattie del sangue, come ad esempio la beta-talassemia. Diversamente, l’editing della linea germinale avrebbe effetti ereditari. Ciò solleva preoccupazioni particolari riguardo al rischio medico. I rischi medici non sono, tuttavia, gli unici tipi di rischi che possono derivare dalla modificazione genetica della linea germinale. Nel contributo non vengono discussi i rischi medici, ma quelli sociali e morali correlati alla manipolazione genetica della linea-germinale. ---------- Should we welcome all developments in gene editing? Somatic cell gene editing would be on a par with conventional therapies aimed at treating particular conditions or alleviating symptoms. It would solely affect the individual patient treated. It could thus serve as a welcome new kind of treatment for cancers and blood diseases such as ß-thalassaemia. Germ-line gene editing, on the other hand, would have hereditary effects. This raises special concerns about medical mishaps. Medical risks are, however, not the only kinds of risks in the case of germline gene editing. Discussed here are not the medical risks, but the social and moral risks of germ-line-gene editing.
APA, Harvard, Vancouver, ISO, and other styles
42

Isa, Noor Munirah, Nurul Atiqah Zulkifli, and Saadan Man. "Islamic Perspectives on CRISPR/Cas9-Mediated Human Germline Gene Editing: A Preliminary Discussion." Science and Engineering Ethics 26, no. 1 (March 4, 2019): 309–23. http://dx.doi.org/10.1007/s11948-019-00098-z.

Full text
APA, Harvard, Vancouver, ISO, and other styles
43

Isa, Noor Munirah. "Human Germline Gene Editing from Maslahah Perspective: The Case of the World’s First Gene Edited Babies." Journal of Bioethical Inquiry 18, no. 2 (March 24, 2021): 349–55. http://dx.doi.org/10.1007/s11673-021-10101-7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
44

Xue, Yang, and Lijun Shang. "Governance of Heritable Human Gene Editing World-Wide and Beyond." International Journal of Environmental Research and Public Health 19, no. 11 (May 31, 2022): 6739. http://dx.doi.org/10.3390/ijerph19116739.

Full text
Abstract:
To date, the controversy surrounding the unknown risks and consequences of heritable genome editing has grown, with such work raising biosafety and ethical concerns for future generations. However, the current guideline of global governance is limited. In the context of the new framework for the governance of human genome editing developed by the World Health Organization (WHO) committee, this paper presents further analysis by highlighting predicaments of governance on germline engineering that merit the most attention: (1) building a scientific culture informed by a broader set of values and considerations in the internal scientific community at large, such as codes of ethics, and education, in addition to awareness-raising measures; and (2) reflecting on and institutionalizing policies in grassroots practice according to local conditions in external governance, such as the experimentalist governance, which is a multi-layered model of governance that establishes an open-ended framework from the top and offers stakeholders the freedom of discussion. The key to achieving these goals is more democratic deliberation between the public and the inclusive engagement of the global scientific community, which has been extensively used in the Biological and Toxin Weapons Convention (BTWC). On a global scale, we believe that practicing heritable human genome editing in accordance with the WHO and BTWC appears to be a good choice.
APA, Harvard, Vancouver, ISO, and other styles
45

Allyse, Megan, Yvonne Bombard, Rosario Isasi, Marsha Michie, Kiran Musunuru, and Kelly E. Ormond. "What do we do now?: Responding to claims of germline gene editing in humans." Genetics in Medicine 21, no. 10 (March 27, 2019): 2181–83. http://dx.doi.org/10.1038/s41436-019-0492-3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
46

Cadera, Emily J., Fengyi Wan, Rupesh H. Amin, Hector Nolla, Michael J. Lenardo, and Mark S. Schlissel. "NF-κB activity marks cells engaged in receptor editing." Journal of Experimental Medicine 206, no. 8 (July 6, 2009): 1803–16. http://dx.doi.org/10.1084/jem.20082815.

Full text
Abstract:
Because of the extreme diversity in immunoglobulin genes, tolerance mechanisms are necessary to ensure that B cells do not respond to self-antigens. One such tolerance mechanism is called receptor editing. If the B cell receptor (BCR) on an immature B cell recognizes self-antigen, it is down-regulated from the cell surface, and light chain gene rearrangement continues in an attempt to edit the autoreactive specificity. Analysis of a heterozygous mutant mouse in which the NF-κB–dependent IκBα gene was replaced with a lacZ (β-gal) reporter complementary DNA (cDNA; IκBα+/lacZ) suggests a potential role for NF-κB in receptor editing. Sorted β-gal+ pre–B cells showed increased levels of various markers of receptor editing. In IκBα+/lacZ reporter mice expressing either innocuous or self-specific knocked in BCRs, β-gal was preferentially expressed in pre–B cells from the mice with self-specific BCRs. Retroviral-mediated expression of a cDNA encoding an IκBα superrepressor in primary bone marrow cultures resulted in diminished germline κ and rearranged λ transcripts but similar levels of RAG expression as compared with controls. We found that IRF4 transcripts were up-regulated in β-gal+ pre–B cells. Because IRF4 is a target of NF-κB and is required for receptor editing, we suggest that NF-κB could be acting through IRF4 to regulate receptor editing.
APA, Harvard, Vancouver, ISO, and other styles
47

Carrington, Blake, Rachel N. Weinstein, and Raman Sood. "BE4max and AncBE4max Are Efficient in Germline Conversion of C:G to T:A Base Pairs in Zebrafish." Cells 9, no. 7 (July 14, 2020): 1690. http://dx.doi.org/10.3390/cells9071690.

Full text
Abstract:
The ease of use and robustness of genome editing by CRISPR/Cas9 has led to successful use of gene knockout zebrafish for disease modeling. However, it still remains a challenge to precisely edit the zebrafish genome to create single-nucleotide substitutions, which account for ~60% of human disease-causing mutations. Recently developed base editing nucleases provide an excellent alternate to CRISPR/Cas9-mediated homology dependent repair for generation of zebrafish with point mutations. A new set of cytosine base editors, termed BE4max and AncBE4max, demonstrated improved base editing efficiency in mammalian cells but have not been evaluated in zebrafish. Therefore, we undertook this study to evaluate their efficiency in converting C:G to T:A base pairs in zebrafish by somatic and germline analysis using highly active sgRNAs to twist and ntl genes. Our data demonstrated that these improved BE4max set of plasmids provide desired base substitutions at similar efficiency and without any indels compared to the previously reported BE3 and Target-AID plasmids in zebrafish. Our data also showed that AncBE4max produces fewer incorrect and bystander edits, suggesting that it can be further improved by codon optimization of its components for use in zebrafish.
APA, Harvard, Vancouver, ISO, and other styles
48

Ciccarelli, Michela, Mariana I. Giassetti, Deqiang Miao, Melissa J. Oatley, Colton Robbins, Blanca Lopez-Biladeau, Muhammad Salman Waqas, et al. "Donor-derived spermatogenesis following stem cell transplantation in sterile NANOS2 knockout males." Proceedings of the National Academy of Sciences 117, no. 39 (September 14, 2020): 24195–204. http://dx.doi.org/10.1073/pnas.2010102117.

Full text
Abstract:
Spermatogonial stem cell transplantation (SSCT) is an experimental technique for transfer of germline between donor and recipient males that could be used as a tool for biomedical research, preservation of endangered species, and dissemination of desirable genetics in food animal populations. To fully realize these potentials, recipient males must be devoid of endogenous germline but possess normal testicular architecture and somatic cell function capable of supporting allogeneic donor stem cell engraftment and regeneration of spermatogenesis. Here we show that male mice, pigs, goats, and cattle harboring knockout alleles of the NANOS2 gene generated by CRISPR-Cas9 editing have testes that are germline ablated but otherwise structurally normal. In adult pigs and goats, SSCT with allogeneic donor stem cells led to sustained donor-derived spermatogenesis. With prepubertal mice, allogeneic SSCT resulted in attainment of natural fertility. Collectively, these advancements represent a major step toward realizing the enormous potential of surrogate sires as a tool for dissemination and regeneration of germplasm in all mammalian species.
APA, Harvard, Vancouver, ISO, and other styles
49

Oatley, Jon M. "Recent advances for spermatogonial stem cell transplantation in livestock." Reproduction, Fertility and Development 30, no. 1 (2018): 44. http://dx.doi.org/10.1071/rd17418.

Full text
Abstract:
At the foundation of spermatogenesis are the actions of spermatogonial stem cells (SSCs), and a remarkable feature of these cells is the capacity to regenerate spermatogenesis following transplantation into testes of a recipient male that lacks endogenous germline. This ability could be exploited in livestock production as a breeding tool to enhance genetic gain. A key element to success is derivation of culture conditions that support proliferation of SSCs to provide sufficient numbers of cells for transfer into multiple recipient males. Using methodology devised for rodent cells as a foundation, advances in culturing cattle SSCs have occurred over the past few years and efforts are underway to extend this capability to pig cells. Another critical component to SSC transplantation is generation of males with germline ablation but intact somatic support cell function that can serve as surrogate sires for donor-derived spermatogenesis in a natural mating scheme. Recent advances in pigs using gene editing technologies have demonstrated that knockout of a key male germ cell-specific gene, namely NANOS2, leads to male-specific germline ablation but otherwise normal physiology, including intact seminiferous tubules. Together with recent advances in culturing spermatogonia of higher-order mammals, the now efficient means of producing germline-ablated recipient males have brought the application of SSC transplantation in livestock as a production tool closer to reality than ever before.
APA, Harvard, Vancouver, ISO, and other styles
50

Vats, Sanskriti, Surbhi Kumawat, Virender Kumar, Gunvant B. Patil, Trupti Joshi, Humira Sonah, Tilak Raj Sharma, and Rupesh Deshmukh. "Genome Editing in Plants: Exploration of Technological Advancements and Challenges." Cells 8, no. 11 (November 4, 2019): 1386. http://dx.doi.org/10.3390/cells8111386.

Full text
Abstract:
Genome-editing, a recent technological advancement in the field of life sciences, is one of the great examples of techniques used to explore the understanding of the biological phenomenon. Besides having different site-directed nucleases for genome editing over a decade ago, the CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein) based genome editing approach has become a choice of technique due to its simplicity, ease of access, cost, and flexibility. In the present review, several CRISPR/Cas based approaches have been discussed, considering recent advances and challenges to implicate those in the crop improvement programs. Successful examples where CRISPR/Cas approach has been used to improve the biotic and abiotic stress tolerance, and traits related to yield and plant architecture have been discussed. The review highlights the challenges to implement the genome editing in polyploid crop plants like wheat, canola, and sugarcane. Challenges for plants difficult to transform and germline-specific gene expression have been discussed. We have also discussed the notable progress with multi-target editing approaches based on polycistronic tRNA processing, Csy4 endoribonuclease, intron processing, and Drosha ribonuclease. Potential to edit multiple targets simultaneously makes it possible to take up more challenging tasks required to engineer desired crop plants. Similarly, advances like precision gene editing, promoter bashing, and methylome-editing will also be discussed. The present review also provides a catalog of available computational tools and servers facilitating designing of guide-RNA targets, construct designs, and data analysis. The information provided here will be useful for the efficient exploration of technological advances in genome editing field for the crop improvement programs.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography