Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
Elife ; 92020 09 02.
Article En | MEDLINE | ID: mdl-32876567

Holoprosencephaly (HPE), a defect in midline patterning of the forebrain and midface, arises ~1 in 250 conceptions. It is associated with predisposing mutations in the Nodal and Hedgehog (HH) pathways, with penetrance and expressivity graded by genetic and environmental modifiers, via poorly understood mechanisms. CDON is a multifunctional co-receptor, including for the HH pathway. In mice, Cdon mutation synergizes with fetal alcohol exposure, producing HPE phenotypes closely resembling those seen in humans. We report here that, unexpectedly, Nodal signaling is a major point of synergistic interaction between Cdon mutation and fetal alcohol. Window-of-sensitivity, genetic, and in vitro findings are consistent with a model whereby brief exposure of Cdon mutant embryos to ethanol during gastrulation transiently and partially inhibits Nodal pathway activity, with consequent effects on midline patterning. These results illuminate mechanisms of gene-environment interaction in a multifactorial model of a common birth defect.


A common birth defect known as holoprosencephaly affects how the brain and face of a fetus develop in the womb. In many cases, the condition is so severe that the fetus dies before, or shortly after, birth. Mutations in certain genes that control how the fetus develops are associated with holoprosencephaly. For example, mutations in components of the Hedgehog and Nodal signaling pathways, which transmit information that help cells to become specialized, increase the risk that a fetus will develop holoprosencephaly. Environmental factors, such as exposure to alcohol in the womb, are also thought to contribute to this condition. A gene known as Cdon is a component of the Hedgehog signaling pathway. In 2012, a team of researchers reported that mice with a mutation in the Cdon gene exposed to alcohol in the womb develop symptoms similar to holoprosencephaly in humans. Here, Hong et al. ­ including some of the researchers involved in the previous work ­ set out to understand how Cdon and alcohol work together to cause holoprosencephaly in the mutant mice. First, the team exposed pregnant mice to alcohol at different times during gestation to find out when their young were sensitive to developing holoprosencephaly. This showed that the young mice were most sensitive in early pregnancy when the Nodal pathway was active in their growing bodies. Further experiments found that alcohol and mutations in Cdon change Nodal signaling in cells. Together, these findings demonstrate that exposure to alcohol in the womb works together with the mutant form of Cdon via the Nodal signaling pathway, rather than the Hedgehog pathway, to cause holoprosencephaly in mice. The causes of many common birth defects are complex and difficult to distinguish at the level of individual cases. The work of Hong et al. illuminates how multiple risk factors during pregnancy, which may not create any problems on their own, may work together to produce birth defects in the fetus. The findings also offer new ways to understand how exposure to alcohol in the womb affects the fetus. Ultimately, understanding how birth defects form could lead to new strategies to prevent them in the future.


Cell Adhesion Molecules , Ethanol/adverse effects , Holoprosencephaly , Mutation/genetics , Nodal Protein , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Female , Holoprosencephaly/chemically induced , Holoprosencephaly/genetics , Holoprosencephaly/pathology , Maternal Exposure , Mice , Nodal Protein/genetics , Nodal Protein/metabolism , Signal Transduction/drug effects
3.
Dev Cell ; 35(1): 36-48, 2015 Oct 12.
Article En | MEDLINE | ID: mdl-26439398

During forebrain development, LRP2 promotes morphogen signaling as an auxiliary SHH receptor. However, in the developing retina, LRP2 assumes the opposing function, mediating endocytic clearance of SHH and antagonizing morphogen action. LRP2-mediated clearance prevents spread of SHH activity from the central retina into the retinal margin to protect quiescent progenitor cells in this niche from mitogenic stimuli. Loss of LRP2 in mice increases the sensitivity of the retinal margin for SHH, causing expansion of the retinal progenitor cell pool and hyperproliferation of this tissue. Our findings document the ability of LRP2 to act, in a context-dependent manner, as activator or inhibitor of the SHH pathway. Our current findings uncovered LRP2 activity as the molecular mechanism imposing quiescence of the retinal margin in the mammalian eye and suggest SHH-induced proliferation of the retinal margin as cause of the large eye phenotype observed in mouse models and patients with LRP2 defects.


Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Hydrophthalmos/pathology , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Mitogens/pharmacology , Neurons/pathology , Retina/pathology , Animals , Cell Proliferation/drug effects , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Female , Humans , Hydrophthalmos/metabolism , Immunoenzyme Techniques , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Retina/drug effects , Retina/embryology , Retina/metabolism , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
4.
Dev Cell ; 22(2): 268-78, 2012 Feb 14.
Article En | MEDLINE | ID: mdl-22340494

Sonic hedgehog (SHH) is a regulator of forebrain development that acts through its receptor, patched 1. However, little is known about cellular mechanisms at neurulation, whereby SHH from the prechordal plate governs specification of the rostral diencephalon ventral midline (RDVM), a major forebrain organizer. We identified LRP2, a member of the LDL receptor gene family, as a component of the SHH signaling machinery in the RDVM. LRP2 acts as an apical SHH-binding protein that sequesters SHH in its target field and controls internalization and cellular trafficking of SHH/patched 1 complexes. Lack of LRP2 in mice and in cephalic explants results in failure to respond to SHH, despite functional expression of patched 1 and smoothened, whereas overexpression of LRP2 variants in cells increases SHH signaling capacity. Our data identify a critical role for LRP2 in SHH signaling and reveal the molecular mechanism underlying forebrain anomalies in mice and patients with Lrp2 defects.


Diencephalon/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Animals , Blotting, Western , Body Patterning , Bone Morphogenetic Protein 4/physiology , Diencephalon/embryology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Hedgehog Proteins/genetics , Immunoenzyme Techniques , Immunoprecipitation , In Situ Hybridization , Mice , Mice, Knockout , Patched Receptors , Patched-1 Receptor , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Smoothened Receptor
5.
Dev Dyn ; 240(6): 1567-77, 2011 Jun.
Article En | MEDLINE | ID: mdl-21455927

Low-density lipoprotein receptor-related protein 2 (LRP2) is a multifunctional cell surface receptor conserved from nematodes to humans. In mammals, it acts as regulator of sonic hedgehog and bone morphogenetic protein pathways in patterning of the embryonic forebrain and as a clearance receptor in the adult kidney. Little is known about activities of this LRP in other phyla. Here, we extend the functional elucidation of LRP2 to zebrafish as a model organism of receptor (dys)function. We demonstrate that expression of Lrp2 in embryonic and larval fish recapitulates the patterns seen in mammalian brain and kidney. Furthermore, we studied the consequence of receptor deficiencies in lrp2 and in lrp2b, a homologue unique to fish, using ENU mutagenesis or morpholino knockdown. While receptor-deficient zebrafish suffer from overt renal resorption deficiency, their brain development proceeds normally, suggesting evolutionary conservation of receptor functions in pronephric duct clearance but not in patterning of the teleost forebrain.


Kidney Tubules/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Metabolic Clearance Rate/genetics , Prosencephalon/embryology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Body Patterning/physiology , Gene Deletion , Gene Expression Regulation, Developmental , Kidney Tubules/embryology , Low Density Lipoprotein Receptor-Related Protein-2/chemistry , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Models, Biological , Phylogeny , Prosencephalon/metabolism , Protein Structure, Tertiary , Signal Transduction/genetics , Signal Transduction/physiology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/physiology
...