Understanding metabolites underlying eye development

2023-07-14
Aerobic glycolysis, the process by which cells transform glucose into lactate, is key for eye development in mammals, according to a new study. Aerobic glycolysis, the process by which cells transform glucose into lactate, is key for eye development in mammals, according to a new Northwestern Medicine study published in Nature Communications. While it has been well known that retinal cells use lactate during cell differentiation, the exact role that this process plays in early eye development was not previously understood. The findings further the field's understanding of the metabolic pathways underlying organ development, according to Guillermo Oliver, PhD, the Thomas D. Spies Professor of Lymphatic Metabolism, Director of the Feinberg Cardiovascular and Renal Research Institute Center for Vascular and Developmental Biology, and senior author of the study. "For a long time, my lab has been interested in developmental biology. In particular, to characterize the molecular and cellular steps regulating early eye morphogenesis," Oliver said. "For us, the question was: 'How do these remarkable and critical sensory organs we have in our face start to form?'" Nozomu Takata, PhD, a postdoctoral fellow in the Oliver lab and first author of the paper, initially approached this question by developing embryonic stem cell-derived eye organoids, which are organ-like tissues engineered in a petri dish. Intriguingly, he observed that early mouse eye progenitors display elevated glycolytic activity and production of lactate. After introducing a glycolysis inhibitor to the cultured organoids, normal optic vesicle development halted, according to the study, but adding back lactate allowed the organoids to resume normal eye morphogenesis, or development. Takata and his collaborators then compared those organoids to controls using genome-wide transcriptome and epigenetic analysis using RNA and ChIP sequencing. They found that inhibiting glycolysis and adding lactate to the organoids regulated the expression of certain critical and evolutionary conserved genes required for early eye development. To validate these findings, Takata deleted Glut1 and Ldha, genes known for regulating glucose transport and lactate production from developing retinas in mouse embryos. The deletion of these genes arrested normal glucose transport specifically in the eye-forming region, according to the study. "What we found was an ATP-independent role of the glycolytic pathway," Takata said. "Lactate, which is a metabolite known as a waste product before, is really doing something cool in eye morphogenesis. That really tells us that this metabolite is a key player in organ morphogenesis and in particular, eye morphogenesis. I see this discovery as having broader implications, as likely also being required in other organs and maybe in regeneration and disease as well." Following this discovery, Takata said he plans to continue to take advantage of traditional and emerging developmental biology's tools such as mouse genetics and stem cells-derived organoids to study the role of the glycolytic pathway and metabolism in the development of other organs. The findings could also be useful in better understanding the direct effect that metabolites could have in regulating gene expression during organ regeneration and tumor development, Oliver said. "Both regeneration and tumorigenesis involve developmental pathways that go awry in some occasions, or you need to reactivate," Oliver said. "For many developmental processes, you need very strict transcriptional regulation. A gene is on or off at certain times, and when that goes wrong, that could lead to developmental defects or promote tumorigenesis. Now that we know that there are specific metabolites responsible for normal or abnormal gene regulation, this can broaden our thinking on approaches to therapeutic treatments." Additional Feinberg faculty co-authors include Ali Shilatifard, PhD, the Robert Francis Furchgott Professor and chair of Biochemistry and Molecular Genetics and director of the Simpson Querrey Institute for Epigenetics, Alexander Misharin, MD, PhD, associate professor of Medicine in the Division of Pulmonary and Critical Care, Jason M. Miska, PhD, assistant professor of Neurological Surgery and Navdeep Chandel, PhD, the David W. Cugell, MD, Professor of Medicine in the Division of Pulmonary and Critical Care and of Biochemistry and Molecular Genetics. The study was supported by an Illumina Next Generation Sequencing award
更多内容,请访问原始网站
文中所述内容并不反映新药情报库及其所属公司任何意见及观点,如有版权侵扰或错误之处,请及时联系我们,我们会在24小时内配合处理。
适应症
靶点
立即开始免费试用!
智慧芽新药情报库是智慧芽专为生命科学人士构建的基于AI的创新药情报平台,助您全方位提升您的研发与决策效率。
立即开始数据试用!
智慧芽新药库数据也通过智慧芽数据服务平台,以API或者数据包形式对外开放,助您更加充分利用智慧芽新药情报信息。