Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
J Clin Invest ; 130(8): 4213-4217, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32369452

ABSTRACT

Molecular mechanisms governing the development of the mammalian cochlea, the hearing organ, remain largely unknown. Through genome sequencing in 3 subjects from 2 families with nonsyndromic cochlear aplasia, we identified homozygous 221-kb and 338-kb deletions in a noncoding region on chromosome 8 with an approximately 200-kb overlapping section. Genomic location of the overlapping deleted region started from approximately 350 kb downstream of GDF6, which codes for growth and differentiation factor 6. Otic lineage cells differentiated from induced pluripotent stem cells derived from an affected individual showed reduced expression of GDF6 compared with control cells. Knockout of Gdf6 in a mouse model resulted in cochlear aplasia, closely resembling the human phenotype. We conclude that GDF6 plays a necessary role in early cochlear development controlled by cis-regulatory elements located within an approximately 500-kb region of the genome in humans and that its disruption leads to deafness due to cochlear aplasia.


Subject(s)
Chromosomes, Human, Pair 8 , Cochlea , Cochlear Diseases , Growth Differentiation Factor 6 , Response Elements , Animals , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 8/metabolism , Cochlea/embryology , Cochlea/pathology , Cochlear Diseases/embryology , Cochlear Diseases/genetics , Cochlear Diseases/pathology , Female , Growth Differentiation Factor 6/biosynthesis , Growth Differentiation Factor 6/genetics , Humans , Male , Mice , Mice, Transgenic
2.
J Biol Chem ; 287(2): 1520-6, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22049084

ABSTRACT

Genome-wide association study (GWAS) has identified genetic variants in the promoter region of the high temperature requirement factor A1 (HTRA1) gene associated with age-related macular degeneration (AMD). As a secreted serine protease, HTRA1 has been reported to interact with members of the transforming growth factor-ß (TGF-ß) family and regulate their signaling pathways. Growth differentiation factor 6 (GDF6), a member of the TGF-ß family, is involved in ectoderm patterning and eye development. Mutations in GDF6 have been associated with abnormal eye development that may result in microphthalmia and anophthalmia. In this report, we identified a single nucleotide polymorphism (SNP) rs6982567 A/G near the GDF6 gene that is significantly associated with AMD (p value = 3.54 × 10(-8)). We demonstrated that the GDF6 AMD risk allele (rs6982567 A) is associated with decreased expression of the GDF6 and increased expression of HTRA1. Similarly, the HTRA1 AMD risk allele (rs10490924 T) is associated with decreased GDF6 and increased HTRA1 expression. We observed decreased vascular development in the retina and significant up-regulation of GDF6 gene in the RPE layer, retinal and brain tissues in HTRA1 knock-out (htra1(-/-)) mice as compared with the wild-type counterparts. Furthermore, we showed enhanced SMAD signaling in htra1(-/-) mice. Our data suggests a critical role of HTRA1 in the regulation of angiogenesis via TGF-ß signaling and identified GDF6 as a novel disease gene for AMD.


Subject(s)
Growth Differentiation Factor 6/biosynthesis , Macular Degeneration/metabolism , Neovascularization, Pathologic/metabolism , Polymorphism, Single Nucleotide , Serine Endopeptidases/biosynthesis , Aged , Alleles , Animals , Cohort Studies , Female , Gene Expression Regulation/genetics , Growth Differentiation Factor 6/genetics , High-Temperature Requirement A Serine Peptidase 1 , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Male , Mice , Mice, Knockout , Middle Aged , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Retina/metabolism , Retina/pathology , Risk Factors , Serine Endopeptidases/genetics , Signal Transduction/genetics , Smad Proteins/genetics , Smad Proteins/metabolism
3.
Growth Factors ; 29(4): 128-39, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21702718

ABSTRACT

Ectopic expression of recombinant human bone morphogenetic protein 2 (rhBMP2) induces osteogenesis, while ectopic expression of rhBMP12 and rhBMP13 induces the formation of tendon-like tissue. Despite their different in vivo activities, all three ligands bound to the type I bone morphogenic protein receptors (BMPRs), activin receptor-like kinase (ALK)-3 and ALK6, and to the type II BMPRs, activin receptor type-2A, activin receptor type-2B, and BMPR2, with similar affinities. Treatment of C3H10T1/2 cells with rhBMP2 activated SMAD signaling and induced expression of osteoblast markers including osteocalcin mRNA (Ocn). In contrast, treatment with rhBMP12 or rhBMP13 resulted in a dose-dependent induction of a tendon-specific gene (Thbs4) expression with no detectable activation of SMAD 1, 5, and 8. Differential regulation of Thbs4 and Ocn has potential utility as an in vitro biomarker for induction of tenogenic signaling. Such an assay also permits the ability to distinguish between the activities of different BMPs and may prove useful in studies on the molecular mechanisms of BMP tenogenic activity.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factor 6/metabolism , Growth Differentiation Factors/metabolism , Activin Receptors/metabolism , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Line , Growth Differentiation Factor 6/biosynthesis , Growth Differentiation Factor 6/pharmacology , Growth Differentiation Factors/pharmacology , Humans , Mice , Mice, Inbred C3H , Osteocalcin/biosynthesis , Osteocalcin/genetics , Osteogenesis/drug effects , Polymerase Chain Reaction , Protein Binding , RNA, Messenger/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Tendons/metabolism , Thrombospondins/biosynthesis
4.
Dev Biol ; 333(1): 37-47, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19545559

ABSTRACT

Dorsal-ventral patterning of the vertebrate retina is essential for accurate topographic mapping of retinal ganglion cell (RGC) axons to visual processing centers. Bone morphogenetic protein (Bmp) growth factors regulate dorsal retinal identity in vertebrate models, but the developmental timing of this signaling and the relative roles of individual Bmps remain unclear. In this study, we investigate the functions of two zebrafish Bmps, Gdf6a and Bmp4, during initiation of dorsal retinal identity, and subsequently during lens differentiation. Knockdown of zebrafish Gdf6a blocks initiation of retinal Smad phosphorylation and dorsal marker expression, while knockdown of Bmp4 produces no discernable retinal phenotype. These data, combined with analyses of embryos ectopically expressing Bmps, demonstrate that Gdf6a is necessary and sufficient for initiation of dorsal retinal identity. We note a profound expansion of ventral retinal identity in gdf6a morphants, demonstrating that dorsal BMP signaling antagonizes ventral marker expression. Finally, we demonstrate a role for Gdf6a in non-neural ocular tissues. Knockdown of Gdf6a leads to defects in lens-specific gene expression, and when combined with Bmp signaling inhibitors, disrupts lens fiber cell differentiation. Taken together, these data indicate that Gdf6a initiates dorsal retinal patterning independent of Bmp4, and regulates lens differentiation.


Subject(s)
Growth Differentiation Factor 6/physiology , Lens, Crystalline/embryology , Retina/embryology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Body Patterning/physiology , Bone Morphogenetic Protein 4/biosynthesis , Bone Morphogenetic Protein 4/genetics , Embryo, Nonmammalian/physiology , Gene Knockdown Techniques , Growth Differentiation Factor 6/biosynthesis , Growth Differentiation Factor 6/genetics , Lens, Crystalline/metabolism , Retina/metabolism , Signal Transduction/physiology , Smad Proteins/antagonists & inhibitors , Smad Proteins/physiology , T-Box Domain Proteins/genetics , Zebrafish , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...