Hostname: page-component-848d4c4894-4rdrl Total loading time: 0 Render date: 2024-07-07T18:50:35.010Z Has data issue: false hasContentIssue false

On recent advances in human engineering

Provocative trends in embryology, genetics, and regenerative medicine

Published online by Cambridge University Press:  22 December 2016

Roman Anton*
Affiliation:
Nürtingen-Geislingen University of Economics and Environment
*
Correspondence: Roman Anton, Nürtingen-Geislingen University of Economics and Environment, Neckarsteige 6-10, Nuertingen, Germany. Email: [email protected]
Get access

Abstract

Advances in embryology, genetics, and regenerative medicine regularly attract attention from scientists, scholars, journalists, and policymakers, yet implications of these advances may be broader than commonly supposed. Laboratories culturing human embryos, editing human genes, and creating human-animal chimeras have been working along lines that are now becoming intertwined. Embryogenic methods are weaving traditional in vivo and in vitro distinctions into a new “in vivitro” (in life in glass) fabric. These and other methods known to be in use or thought to be in development promise soon to bring society to startling choices and discomfiting predicaments, all in a global effort to supply reliably rejuvenating stem cells, to grow immunologically nonprovocative replacement organs, and to prevent, treat, cure, or even someday eradicate diseases having genetic or epigenetic mechanisms. With humanity’s human-engineering era now begun, procedural prohibitions, funding restrictions, institutional controls, and transparency rules are proving ineffective, and business incentives are migrating into the most basic life-sciences inquiries, wherein lie huge biomedical potentials and bioethical risks. Rights, health, and heritage are coming into play with bioethical presumptions and formal protections urgently needing reassessment.

Type
Articles
Copyright
© Association for Politics and the Life Sciences 2016 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Deglincerti, A., Croft, Gist F., Pietila, Lauren N., Zernicka-Goetz, Magdalena, Siggia, Eric D., and Brivanlou, Ali H., “Self-organization of the in vitro attached human embryo,” Nature , 2016, 533: 251254, doi:10.1038/nature17948.Google Scholar
Shahbazi, Marta A., Jedrusik, Agnieszka, Vuoristo, Sanna, Recher, Gaelle, Hupalowska, Anna, Bolton, Virginia, Fogarty, Norah M. E., Campbell, Alison, Devito, Liani G., Ilic, Dusko, Khalaf, Yakoub, Niakan, Kathy K., Fishel, Simon, and Zernicka-Goetz, Magdalena, “Self-organization of the human embryo in the absence of maternal tissues,” Nature Cell Biology , 2016, 18: 700708, doi:10.1038/ncb3347.Google Scholar
Bedzhov, I. and Zernicka-Goetz, M., “Self-organizing properties of mouse pluripotent cells initiate morphogenesis upon implantation,” Cell , 2014, 156: 10321044.Google Scholar
Anton, R., Kestler, H. A., and Kühl, M., “beta-Catenin signaling contributes to stemness and regulates early differentiation in murine embryonic stem cells,” Febs Letters , 2007, 581(27): 52475254, doi:10.1016/j.febslet.2007.10.012.Google Scholar
Deglincerti, A., Etoc, F., Ozair, M., and Brivanlou, A., “Self-organization of spatial patterning in human embryonic stem cells,” Current Topics in Developmental Biology , 2016, 116: 99113, doi:10.1016/bs.ctdb.2015.11.010.Google Scholar
Odling-Smee, L., Ledford, H., and Reardon, S., “Genome editing: 7 facts about a revolutionary technology,” Nature News , November 30, 2015, doi:10.1038/nature.2015.1886.Google Scholar
Mali, P., Yang, L., Esvelt, K. M., Aach, J., Guell, M., DiCarlo, J. E., Norville, J. E., and Church, G. M., “RNA-guided human genome engineering via Cas9,” Science , 2013, 339(6121): 823826, doi:10.1126/science.1232033.Google Scholar
Doudna, J. and Charpentier, E., “Genome editing. The new frontier of genome engineering with CRISPR-Cas9,” Science , 2014, 346(6213), doi:10.1126/science.1258096.Google Scholar
Takahashi, K. and Yamanaka, S., “Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors,” Cell , 2006, 126: 663676, doi:10.1016/j.cell.2006.07.024.Google Scholar
Xu, Yanwen, Zhang, Xiya, Ding, Chenhui, Huang, Rui, Zhang, Zhen, Lv, Jie, Xie, Xiaowei, Chen, Yuxi, Li, Yujing, Sun, Ying, Bai, Yaofu, Songyang, Zhou, Ma, Wenbin, Zhou, Canquan, and Huang, Junjiu, “CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes,” Protein and Cell , 2015, 6: 363, doi:10.1007/s13238-015-0153-5.Google Scholar
Hyun, I., Wilkerson, A., and Johnston, J., “Embryology policy: Revisit the 14-day rule,” Nature , 2016, 533: 169171, doi:10.1038/533169a.Google Scholar
Hammerman, M., “Classic and current opinion in embryonic organ transplantation,” Current Opinions in Organ Transplantation , 2014, 19: 133139.Google Scholar
Regalado, Antonio, “Human-animal chimeras are gestating on U.S. research farms: A radical new approach to generating human organs is to grow them inside pigs or sheep,” MIT Technology Review, January 6, 2016, https://www.technologyreview.com/s/545106/human-animal-chimeras-are-gestating-on-us-research-farms, accessed November 22, 2016.Google Scholar
Human Embyrology, “Module 8.1: Embryonic phase: The Carneigie stages,” http://www.embryology.ch/anglais/ iperiodembry/carnegie02.html, accessed November 22, 2016.Google Scholar
Glover, V., “The fetus may feel pain from 20 weeks,” Conscience , 2004–2005, 25(3): 3537.Google Scholar
“Abortion restrictions in states,” New York Times, June 17, 2013.Google Scholar
Raju, T., Mercer, B., Burchfield, D., and Joseph, G., “Periviable birth: Executive summary of a Joint Workshop by the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Society for Maternal-Fetal Medicine, American Academy of Pediatrics, and American College of Obstetricians and Gynecologists,” Journal of Perinatology , 2014, 34: 333342.Google Scholar
James, D., Noggle, S., Swigut, T., and Brivanlou, A., “Contribution of human embryonic stem cells to mouse blastocysts,” Developmental Biology , 2006, 295: 90102.Google Scholar
Lee, Y. L., Fong, S. W., Chen, A. C., Li, T., Yue, C., Lee, C. L., Ng, E. H., Yeung, W. S., and Lee, K. F., “Establishment of a novel human embryonic stem cell-derived trophoblastic spheroid implantation model,” Human Reproduction , 2015, 30(11): 26142626, doi:10.1093/humrep/dev223.Google Scholar
Savic, N. and Schwank, G., “Advances in therapeutic CRISPR/Cas9 genome editing,” Translational Research , 2016, 168: 1521, doi:10.1016/j.trsl.2015.09.008.Google Scholar
Spar, D., “The egg trade-making sense of the market for human oocytes,” New England Journal of Medicine , 2007, 356: 12891291.Google Scholar
Resnik, D. B., “Regulating the market for human eggs,” Bioethics , 2001, 15: 125, doi:10.1111/1467-8519.00209.Google Scholar
Isasi, R. M. and Knoppers, B. M., “Monetary payments for the procurement of oocytes for stem cell research: In search of ethical and political consistency,” Stem Cell Research , 2007, 1: 3744, doi:10.1016/j.scr2007.09.003.Google Scholar
Boeke, J Jef D., Church, George, Hessel, Andrew, Kelley, Nancy J., Arkin, Adam, Cai, Yizhi, Carlson, Rob, Chakravarti, Aravinda, Cornish, Virginia W., Holt, Liam, Isaacs, Farren J., Kuiken, Todd, Lajoie, Marc, Lessor, Tracy, Lunshof, Jeantine, Maurano, Matthew T., Mitchell, Leslie A., Rine, Jasper, Rosser, Susan, Sanjana, Neville E., Silver, Pamela A., Valle, David, Wang, Harris, Way, Jeffrey C., and Yang, Luhan, “The Genome Project — Write,” Science , 2016, 353: 126127, doi:10.1126/science.aaf6850.Google Scholar
Zetsche, Bernd, Gootenberg, Jonathan S., Abudayyeh, Omar O., Slaymaker, Ian M., Makarova, Kira S., Essletzbichler, Patrick, Volz, Sara E., Joung, Julia, van der Oost, John, Regev, Aviv, Koonin, Eugene V., and Zhang, Feng, “Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system,” Cell , 2015, 163(3): 759771, doi:10.1016/j.cell.2015.09.038.Google Scholar
Hilton, I. B., D’Ippolito, A. M., Vockley, C. M., Thakore, P. I., Crawford, G. E., Reddy, T. E., and Gersbach, C. A., “Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers,” Nature Biotechnology , 2015, 33(5): 510517, doi:10.1038/nbt.3199.Google Scholar
“Top 10 innovations 2015,” The Scientist, December 1, 2015, http://www.the-scientist.com/ ?articles.view/articleNo/44629/title/Top-10-Innovations-2015, accessed November 22, 2016.Google Scholar
Dayal, M. B., Gindoff, P. R., Sarhan, A. , Dubey, A., Peak, D., and Frankfurter, D., “Effects of triploidy after intracytoplasmic sperm injection on in vitro fertilization cycle outcome,” Fertility and Sterility , 2009, 91(1): 101105, Epub February 4, 2008, doi:10.1016/j.fertnstert.2007.11.033.Google Scholar
Kaminski, Rafal, Chen, Yilan, Fischer, Tracy, Tedaldi, Ellen, Napoli, Alessandro, Zhang, Yonggang, Karn, Jonathan, Hu, Wenhui, and Khalili, Kamel, “Elimination of HIV-1 genomes from human t-lymphoid cells by CRISPR/Cas9 gene editing,” Nature , 2016, 6: 22555, doi:10.1038/srep22555.Google Scholar
Lim, W. F., Inoue-Yokoo, T., Tan, K. S., Lai, M. I., and Sugiyama, D., “Hematopoietic cell differentiation from embryonic and induced pluripotent stem cells,” Stem Cell Research and Therapy , 2013, 4: 71, doi:10.1186/scrt222.CrossRefGoogle ScholarPubMed
Tam, P. P. and Rossant, J., “Mouse embryonic chimeras: Tools for studying mammalian development,” Development , 2003, 130(25): 61556163, doi:10.1242/dev.00893.Google Scholar
Kaiser, Jocelyn, “NIH moves to lift moratorium on animal-human chimera research,” Science , August 4, 2016, doi:10.1126/science.aag0732.Google Scholar
National Institutes of Health, “Guidelines on human stem cell research,” July 7, 2009, https://stemcells.nih.gov/ policy/2009-guidelines.htm, accessed November 22, 2016.Google Scholar
National Institutes of Health Office of Extramural Research, “NIH research involving introduction of human pluripotent cells into non-human vertebrate animal pre-gastrulation embryos,”Notice NOT-OD-15-158, September 23, 2015, https://grants.nih.gov/grants/guide/notice-files/NOT-OD-15-158.html, accessed November 22, 2016.Google Scholar
Hyoon, I., “Illusory fears must not stifle chimaera research,” Nature , 2016, 537: 281, doi:10.1038/537281a.Google Scholar