Human birth defects can be caused by disruption of molecular pathways that regulate cell shape and movements. Cell shape changes and movements are responsible for helping to establish and separate the three primary germ layers at gastrulation, for reification of these germ layers into different tissue types, and for combinin tissues into organs with unique internal architecture and anatomical relationships. Multiple signaling pathways contribute to cell shape and movement, including the Wnt Planar Cell Polarity (PCP) pathway upstream of convergent-extension (CE) movements in vertebrates. In the prior funded period we used an engineered knock-out at the mouse Dapper/Frodo (Dact1) locus to explore how its loss causes a spectrum of developmental defects in mice resembling a major spectrum of birth defects. Through this project we discovered that Dact1 regulates Van Gogh-like (Vangl) transmembrane proteins central to PCP during Epithelial-Mesenchymal Transition (EMT) at the Primitive Streak (PS) during gastrulation. We have now engineered novel knock-out mouse lines for the two remaining Dact family members in mammals (Dact2 and Dact3) as well as for Sestd1, which encodes a Dact-interacting protein identified in my lab. These animals very closely phenocopy Dact1 mutants, strongly supporting the hypothesis that major developmental functions of Sestd1 are allied closely with Dact1. Intriguingly, Sestd1 has previously been shown to help regulate calcium flux through TRPC channel proteins; moreover its closest molecular relative has been implicated in transmembrane protein trafficking. This agrees with evidence suggesting that the molecular pathway emerging from work in my lab acts via localization and/or levels of a subset of transmembrane proteins at the intersection of several important signaling cascades. In the renewed funding period I propose to address these Specific Aims: (1) Do Sestd1 & Dact1 form a genetic pathway with Scribble, Celsr1 & Vangl2 during EMT at the PS; with Dvl2 during CE; and with TRPC4/5 during calcium signaling? (2) Are both the PCP & Calcium pathways biochemically disrupted in posterior embryonic tissues of these mutants? (3) Are membrane localization & levels of pathway components linked to PCP & Calcium signaling in affected embryonic tissues? As in the prior funded period, the research strategy draws on deep mouse genetic, embryonic, cell biologic and biochemical tools custom-designed and assembled in my laboratory expressly to explore this pathway.

Public Health Relevance

Understanding the causes of birth defects is a high priority for public health that is particularly relevant to the mission of the National Institute of Child Healt and Human Development. The research funded by this grant employs genetically-engineered mouse and cell lines to investigate molecular and embryonic origins of complex birth defects in humans. This project will help uncover biological causes of developmental defects leading to many miscarriages, still-births, infant deaths and neonatal surgeries. It will thereby point the wa toward new and improved prenatal diagnostic and therapeutic interventions to minimize miscarriages and birth defects in the United States. Simultaneously, the molecular insights gained through this research may be relevant to some very significant diseases of adults, including in the cardiac and nervous systems.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
5R01HD055300-10
Application #
9265875
Study Section
Genetics of Health and Disease Study Section (GHD)
Program Officer
Toyama, Reiko
Project Start
2007-05-01
Project End
2018-04-30
Budget Start
2017-05-01
Budget End
2018-04-30
Support Year
10
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of California San Francisco
Department
Psychiatry
Type
Schools of Medicine
DUNS #
094878337
City
San Francisco
State
CA
Country
United States
Zip Code
94118
Yang, Xiao Yong; Stanley, Robert E; Ross, Adam P et al. (2018) Sestd1 Encodes a Developmentally Dynamic Synapse Protein That Complexes With BCR Rac1-GAP to Regulate Forebrain Dendrite, Spine and Synapse Formation. Cereb Cortex :
Martin, P-M; Stanley, R E; Ross, A P et al. (2018) DIXDC1 contributes to psychiatric susceptibility by regulating dendritic spine and glutamatergic synapse density via GSK3 and Wnt/?-catenin signaling. Mol Psychiatry 23:467-475
Okerlund, Nathan D; Stanley, Robert E; Cheyette, Benjamin N R (2016) The Planar Cell Polarity Transmembrane Protein Vangl2 Promotes Dendrite, Spine and Glutamatergic Synapse Formation in the Mammalian Forebrain. Mol Neuropsychiatry 2:107-14
Yang, Xiaoyong; Fisher, Daniel A; Cheyette, Benjamin Nr (2013) SEC14 and Spectrin Domains 1 (Sestd1), Dishevelled 2 (Dvl2) and Dapper Antagonist of Catenin-1 (Dact1) co-regulate the Wnt/Planar Cell Polarity (PCP) pathway during mammalian development. Commun Integr Biol 6:e26834
Yang, XiaoYong; Cheyette, Benjamin N R (2013) SEC14 and spectrin domains 1 (Sestd1) and Dapper antagonist of catenin 1 (Dact1) scaffold proteins cooperatively regulate the Van Gogh-like 2 (Vangl2) four-pass transmembrane protein and planar cell polarity (PCP) pathway during embryonic development in m J Biol Chem 288:20111-20
Kivimäe, Saul; Yang, Xiao Yong; Cheyette, Benjamin N R (2011) All Dact (Dapper/Frodo) scaffold proteins dimerize and exhibit conserved interactions with Vangl, Dvl, and serine/threonine kinases. BMC Biochem 12:33
Kettunen, Paivi; Kivimae, Saul; Keshari, Pankaj et al. (2010) Dact1-3 mRNAs exhibit distinct expression domains during tooth development. Gene Expr Patterns 10:140-3
Suriben, Rowena; Kivimäe, Saul; Fisher, Daniel A C et al. (2009) Posterior malformations in Dact1 mutant mice arise through misregulated Vangl2 at the primitive streak. Nat Genet 41:977-85
Louie, Sarah H; Yang, Xiao Yong; Conrad, William H et al. (2009) Modulation of the beta-catenin signaling pathway by the dishevelled-associated protein Hipk1. PLoS One 4:e4310
Jiang, Xia; Tan, Jing; Li, Jingsong et al. (2008) DACT3 is an epigenetic regulator of Wnt/beta-catenin signaling in colorectal cancer and is a therapeutic target of histone modifications. Cancer Cell 13:529-41