Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
J Biol Chem ; 295(47): 15840-15852, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32467232

ABSTRACT

Long noncoding RNAs (lncRNAs) have been shown to play key roles in a variety of biological activities of the cell. However, less is known about how lncRNAs respond to environmental cues and what transcriptional mechanisms regulate their expression. Studies from our laboratory have shown that the lncRNA Tug1 (taurine upregulated gene 1) is crucial for the progression of diabetic kidney disease, a major microvascular complication of diabetes. Using a combination of proximity labeling with the engineered soybean ascorbate peroxidase (APEX2), ChIP-qPCR, biotin-labeled oligonucleotide pulldown, and classical promoter luciferase assays in kidney podocytes, we extend our initial observations in the current study and now provide a detailed analysis on a how high-glucose milieu downregulates Tug1 expression in podocytes. Our results revealed an essential role for the transcription factor carbohydrate response element binding protein (ChREBP) in controlling Tug1 transcription in the podocytes in response to increased glucose levels. Along with ChREBP, other coregulators, including MAX dimerization protein (MLX), MAX dimerization protein 1 (MXD1), and histone deacetylase 1 (HDAC1), were enriched at the Tug1 promoter under high-glucose conditions. These observations provide the first characterization of the mouse Tug1 promoter's response to the high-glucose milieu. Our findings illustrate a molecular mechanism by which ChREBP can coordinate glucose homeostasis with the expression of the lncRNA Tug1 and further our understanding of dynamic transcriptional regulation of lncRNAs in a disease state.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Glucose/metabolism , Podocytes/metabolism , RNA, Long Noncoding/biosynthesis , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Glucose/genetics , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Mice , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
2.
Differentiation ; 112: 1-6, 2020.
Article in English | MEDLINE | ID: mdl-31830612

ABSTRACT

Defective anorectal and urogenital malformations are some of the most severe congenital anomalies encountered in children. Only a few molecular cues have been identified in early formation of the female urogenital system. Here we describe a novel long non-coding RNA molecule known as Leat1 (long non-coding RNA, EphrinB2 associated transcript 1). This lncRNA is syntenic with EfnB2 (which encodes EphrinB2) and expressed during embryonic development of the genital tubercle. While lncRNAs have varied functions, many are known to regulate their neighbouring genes. Eph/Ephrin bidirectional signaling molecules mediate many patterning pathways in early embryonic development, including cloacal septation and urethral development. Here we investigate the role of Leat1 and its possible regulation of EphrinB2 during development of the female reproductive tract. We show that a loss of Leat1 leads to reduced EfnB2 expression in the developing female genital tubercle, reduced anogenital distance and decreased fertility.


Subject(s)
Ephrin-B2/genetics , Organogenesis/genetics , RNA, Long Noncoding/genetics , Urogenital Abnormalities/genetics , Animals , Embryo, Mammalian , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Humans , Infertility, Female/genetics , Infertility, Female/pathology , Mice
3.
Hum Mol Genet ; 26(15): 2949-2960, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28486600

ABSTRACT

Ciliopathies form a group of inherited disorders sharing several clinical manifestations because of abnormal cilia formation or function, and few treatments have been successful against these disorders. Here, we report a mouse model with mutated Sclt1 gene, which encodes a centriole distal appendage protein important for ciliogenesis. Sodium channel and clathrin linker 1 (SCLT1) mutations were associated with the oral-facial-digital syndrome (OFD), an autosomal recessive ciliopathy. The Sclt1-/- mice exhibit typical ciliopathy phenotypes, including cystic kidney, cleft palate and polydactyly. Sclt1-loss decreases the number of cilia in kidney; increases proliferation and apoptosis of renal tubule epithelial cells; elevates protein kinase A, extracellular signal-regulated kinases, SMAD and signal transducer and activator of transcription 3 (STAT3) pathways; and enhances pro-inflammation and pro-fibrosis pathways with disease progression. Embryonic kidney cyst formation of Sclt1-/- mice was effectively reduced by an anti-STAT3 treatment using pyrimethamine. Overall, we reported a new mouse model for the OFD; and our data suggest that STAT3 inhibition may be a promising treatment for SCLT1-associated cystic kidney.


Subject(s)
STAT3 Transcription Factor/metabolism , Sodium Channels/metabolism , Animals , Cilia/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Cysts/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Kidney/metabolism , Kidney Diseases, Cystic/etiology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , MAP Kinase Signaling System , Mice , Mice, Transgenic , Models, Animal , Mutation , Phenotype , STAT3 Transcription Factor/genetics , Signal Transduction , Sodium Channels/genetics
4.
Kidney Int ; 92(5): 1282-1287, 2017 11.
Article in English | MEDLINE | ID: mdl-28754553

ABSTRACT

While increased mitochondrial reactive oxygen species have been commonly implicated in a variety of disease states, their in vivo role in the pathogenesis of diabetic nephropathy remains controversial. Using a two-photon imaging approach with a genetically encoded redox biosensor, we monitored mitochondrial redox state in the kidneys of experimental models of diabetes in real-time in vivo. Diabetic (db/db) mice that express a redox-sensitive Green Fluorescent Protein biosensor (roGFP) specifically in the mitochondrial matrix (db/dbmt-roGFP) were generated, allowing dynamic monitoring of redox changes in the kidneys. These db/dbmt-roGFP mice exhibited a marked increase in mitochondrial reactive oxygen species in the kidneys. Yeast NADH-dehydrogenase, a mammalian Complex I homolog, was ectopically expressed in cultured podocytes, and this forced expression in roGFP-expressing podocytes prevented high glucose-induced increases in mitochondrial reactive oxygen species. Thus, in vivo monitoring of mitochondrial roGFP in diabetic mice confirms increased production of mitochondrial reactive oxygen species in the kidneys.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/pathology , Kidney/pathology , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Animals , Biosensing Techniques , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Disease Models, Animal , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred Strains , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Mitochondria/metabolism , Mitochondria/ultrastructure , Oxidation-Reduction , Podocytes
5.
N Engl J Med ; 370(10): 943-949, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24597867

ABSTRACT

Premature ovarian failure is a major cause of female infertility. The genetic causes of this disorder remain unknown in most patients. Using whole-exome sequence analysis of a large consanguineous family with inherited premature ovarian failure, we identified a homozygous 1-bp deletion inducing a frameshift mutation in STAG3 on chromosome 7. STAG3 encodes a meiosis-specific subunit of the cohesin ring, which ensures correct sister chromatid cohesion. Female mice devoid of Stag3 are sterile, and their fetal oocytes are arrested at early prophase I, leading to oocyte depletion at 1 week of age.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Mutation , Nuclear Proteins/genetics , Primary Ovarian Insufficiency/genetics , Animals , Disease Models, Animal , Female , Humans , Infertility, Female/genetics , Mice , Pedigree , Cohesins
6.
PLoS Genet ; 10(1): e1004055, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24391519

ABSTRACT

The Notch signaling pathway is thought to regulate multiple stages of inner ear development. Mutations in the Notch signaling pathway cause disruptions in the number and arrangement of hair cells and supporting cells in sensory regions of the ear. In this study we identify an insertional mutation in the mouse Sfswap gene, a putative splicing factor, that results in mice with vestibular and cochlear defects that are consistent with disrupted Notch signaling. Homozygous Sfswap mutants display hyperactivity and circling behavior consistent with vestibular defects, and significantly impaired hearing. The cochlea of newborn Sfswap mutant mice shows a significant reduction in outer hair cells and supporting cells and ectopic inner hair cells. This phenotype most closely resembles that seen in hypomorphic alleles of the Notch ligand Jagged1 (Jag1). We show that Jag1; Sfswap compound mutants have inner ear defects that are more severe than expected from simple additive effects of the single mutants, indicating a genetic interaction between Sfswap and Jag1. In addition, expression of genes involved in Notch signaling in the inner ear are reduced in Sfswap mutants. There is increased interest in how splicing affects inner ear development and function. Our work is one of the first studies to suggest that a putative splicing factor has specific effects on Notch signaling pathway members and inner ear development.


Subject(s)
Alternative Splicing/genetics , Ear, Inner/growth & development , RNA-Binding Proteins/genetics , Receptors, Notch/genetics , Animals , Body Patterning/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cochlea/growth & development , Cochlea/pathology , Ear, Inner/metabolism , Ear, Inner/pathology , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation , RNA Splicing Factors , RNA-Binding Proteins/metabolism , Serrate-Jagged Proteins , Signal Transduction/genetics , Vestibule, Labyrinth/growth & development , Vestibule, Labyrinth/pathology
7.
Nucleic Acids Res ; 41(22): 10199-214, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24038357

ABSTRACT

Lens induction is a classical embryologic model to study cell fate determination. It has been proposed earlier that specific changes in core histone modifications accompany the process of cell fate specification and determination. The lysine acetyltransferases CBP and p300 function as principal enzymes that modify core histones to facilitate specific gene expression. Herein, we performed conditional inactivation of both CBP and p300 in the ectodermal cells that give rise to the lens placode. Inactivation of both CBP and p300 resulted in the dramatic discontinuation of all aspects of lens specification and organogenesis, resulting in aphakia. The CBP/p300(-/-) ectodermal cells are viable and not prone to apoptosis. These cells showed reduced expression of Six3 and Sox2, while expression of Pax6 was not upregulated, indicating discontinuation of lens induction. Consequently, expression of αB- and αA-crystallins was not initiated. Mutant ectoderm exhibited markedly reduced levels of histone H3 K18 and K27 acetylation, subtly increased H3 K27me3 and unaltered overall levels of H3 K9ac and H3 K4me3. Our data demonstrate that CBP and p300 are required to establish lens cell-type identity during lens induction, and suggest that posttranslational histone modifications are integral to normal cell fate determination in the mammalian lens.


Subject(s)
CREB-Binding Protein/physiology , E1A-Associated p300 Protein/physiology , Histones/metabolism , Lens, Crystalline/embryology , Acetylation , Animals , Apoptosis , CREB-Binding Protein/genetics , E1A-Associated p300 Protein/genetics , Embryonic Induction , Gene Expression , Lens, Crystalline/anatomy & histology , Lens, Crystalline/enzymology , Mice , Mutation , Protein Processing, Post-Translational , S Phase
8.
Development ; 138(20): 4487-97, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21937600

ABSTRACT

Correct development of the cerebellum requires coordinated sonic hedgehog (Shh) signaling from Purkinje to granule cells. How Shh expression is regulated in Purkinje cells is poorly understood. Using a novel tyrosinase minigene-tagged Sleeping Beauty transposon-mediated mutagenesis, which allows for coat color-based genotyping, we created mice in which the Ski/Sno family transcriptional co-repressor 2 (Skor2) gene is deleted. Loss of Skor2 leads to defective Purkinje cell development, a severe reduction of granule cell proliferation and a malformed cerebellum. Skor2 is specifically expressed in Purkinje cells in the brain, where it is required for proper expression of Shh. Skor2 overexpression suppresses BMP signaling in an HDAC-dependent manner and stimulates Shh promoter activity, suggesting that Skor2 represses BMP signaling to activate Shh expression. Our study identifies an essential function for Skor2 as a novel transcriptional regulator in Purkinje cells that acts upstream of Shh during cerebellum development.


Subject(s)
Cerebellum/growth & development , Cerebellum/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cerebellum/abnormalities , Gene Expression Regulation, Developmental , Genotype , Hair Color/genetics , Histone Deacetylases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Monophenol Monooxygenase/genetics , Mutagenesis, Insertional , Proto-Oncogene Proteins/deficiency , Purkinje Cells/cytology , Purkinje Cells/metabolism , Repressor Proteins/deficiency , Signal Transduction , Transforming Growth Factor beta/metabolism , Transposases/genetics
9.
Mamm Genome ; 25(3-4): 141-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24275887

ABSTRACT

Fertilization is the process that leads to the formation of a diploid zygote from two haploid gametes. This is achieved through a complex series of cell-to-cell interactions between a sperm and an egg. The final event of fertilization is the fusion of the gametes' membranes, which allows the delivery of the sperm genetic material into the egg cytoplasm. In vivo studies in the laboratory mouse have led to the discovery of membrane proteins that are essential for the fusion process in both the sperm and egg. Specifically, the sperm protein Izumo1 was shown to be necessary for normal fertility. Izumo1-deficient spermatozoa fail to fuse with the egg plasma membrane. Izumo1 is a member of the Immunoglobulin Superfamily of proteins, which are known to be involved in cell adhesion. Here, we describe BART97b, a new mouse line with a recessive mutation that displays a fertilization block associated with a failure of sperm fusion. BART97b mutants carry a deletion that inactivates Spaca6, a previously uncharacterized gene expressed in testis. Similar to Izumo1, Spaca6 encodes an immunoglobulin-like protein. We propose that the Spaca6 gene product may, together with Izumo1, mediate sperm fusion by binding an as yet unidentified egg membrane receptor.


Subject(s)
DNA Transposable Elements/genetics , Fertilization/genetics , Immunoglobulins/genetics , Membrane Proteins/genetics , Mice, Mutant Strains/genetics , Mice, Transgenic/genetics , Sperm-Ovum Interactions/genetics , Animals , Base Sequence , Chromosome Mapping , Female , Fertilization/physiology , Gene Deletion , Gene Silencing , Male , Mice , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Seminal Plasma Proteins/genetics , Sequence Analysis, DNA , Sperm-Ovum Interactions/physiology
10.
Res Sq ; 2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37461443

ABSTRACT

The novel long non-coding RNA (lncRNA) Leat1 is extraordinarily conserved in both its location (syntenic with EfnB2, an essential gene in anogenital patterning) and sequence. Here we show that Leat1 is upregulated following the testosterone surge from the developing testis and directly interacts with EfnB2, positively regulating its expression. Leat1 expression is suppressed by estrogen, which in turn suppresses the expression of EfnB2. Moreover, the loss of Leat1 leads to reduced EfnB2, resulting in a severe hypospadias phenotype. The human LEAT1 gene is also co-expressed with EFNB2 in the developing human penis suggesting a conserved function for this gene in urethral closure. Together our data identify Leat1 as a novel molecular regulator of urethral closure and implicate it as a target of endocrine disruption in the etiology of hypospadias.

11.
Cell Rep ; 36(6): 109510, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34380028

ABSTRACT

lncRNA taurine-upregulated gene 1 (Tug1) is a promising therapeutic target in the progression of diabetic nephropathy (DN), but the molecular basis of its protection remains poorly understood. Here, we generate a triple-mutant diabetic mouse model coupled with metabolomic profiling data to interrogate whether Tug1 interaction with peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α) is required for mitochondrial remodeling and progression of DN in vivo. We find that, compared with diabetic conditional deletion of Pgc1α in podocytes alone (db/db; Pgc1αPod-f/f), diabetic Pgc1α knockout combined with podocyte-specific Tug1 overexpression (db/db; TugPodTg; Pgc1αPod-f/f) reverses the protective phenotype of Tug1 overexpression, suggesting that PGC1α is required for the renoprotective effect of Tug1. Using unbiased metabolomic profiling, we find that altered urea cycle metabolites and mitochondrial arginase 2 play an important role in Tug1/PGC1α-induced mitochondrial remodeling. Our work identifies a functional role of the Tug1/PGC1α axis on mitochondrial metabolic homeostasis and urea cycle metabolites in experimental models of diabetes.


Subject(s)
Kidney/metabolism , Metabolome , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protective Agents/metabolism , RNA, Long Noncoding/metabolism , Urea/metabolism , Animals , Arginase/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Progression , Gene Deletion , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/deficiency , Podocytes/metabolism , RNA, Long Noncoding/genetics
12.
Dev Biol ; 333(1): 132-42, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19563798

ABSTRACT

Pax6 is a highly conserved transcription factor that controls the morphogenesis of various organs. Changes in Pax6 dosage have been shown to affect the formation of multiple tissues. PAX6 haploinsufficiency leads to aniridia, a pan-ocular disease primarily characterized by iris hypoplasia. Herein, we employ a modular system that includes null and overexpressed conditional alleles of Pax6. The use of the Tyrp2-Cre line, active in iris and ciliary body (CB) primordium, enabled us to investigate the effect of varying dosages of Pax6 on the development of these ocular sub-organs. Our findings show that a lack of Pax6 in these regions leads to dysgenesis of the iris and CB, while heterozygosity impedes growth of the iris and maturation of the iris sphincter. Overexpression of the canonical, but not the alternative splice variant of Pax6 results in severe structural aberrations of the CB and hyperplasia of the iris sphincter. A splice variant-specific rescue experiment revealed that both splice variants are able to correct iris hypoplasia, while only the canonical form rescues the sphincter. Overall, these findings demonstrate the dosage-sensitive roles of Pax6 in the formation of both the CB and the iris.


Subject(s)
Ciliary Body/embryology , Ciliary Body/growth & development , Eye Proteins/biosynthesis , Gene Dosage , Homeodomain Proteins/biosynthesis , Iris/embryology , Iris/growth & development , Paired Box Transcription Factors/biosynthesis , Repressor Proteins/biosynthesis , Alternative Splicing , Animals , Cell Differentiation , Ciliary Body/cytology , Ciliary Body/metabolism , Eye Proteins/genetics , Homeodomain Proteins/genetics , Iris/cytology , Iris/metabolism , Mice , Mice, Transgenic , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics
13.
BMC Dev Biol ; 10: 13, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20105280

ABSTRACT

BACKGROUND: Mammalian Ras genes regulate diverse cellular processes including proliferation and differentiation and are frequently mutated in human cancers. Tumor development in response to Ras activation varies between different tissues and the molecular basis for these variations are poorly understood. The murine lens and cornea have a common embryonic origin and arise from adjacent regions of the surface ectoderm. Activation of the fibroblast growth factor (FGF) signaling pathway induces the corneal epithelial cells to proliferate and the lens epithelial cells to exit the cell cycle. The molecular mechanisms that regulate the differential responses of these two related tissues have not been defined. We have generated transgenic mice that express a constitutively active version of human H-Ras in their lenses and corneas. RESULTS: Ras transgenic lenses and corneal epithelial cells showed increased proliferation with concomitant increases in cyclin D1 and D2 expression. This initial increase in proliferation is sustained in the cornea but not in the lens epithelial cells. Coincidentally, cdk inhibitors p27Kip1 and p57Kip2 were upregulated in the Ras transgenic lenses but not in the corneas. Phospho-Erk1 and Erk2 levels were elevated in the lens but not in the cornea and Spry 1 and Spry 2, negative regulators of Ras-Raf-Erk signaling, were upregulated more in the corneal than in the lens epithelial cells. Both lens and corneal differentiation programs were sensitive to Ras activation. Ras transgenic embryos showed a distinctive alteration in the architecture of the lens pit. Ras activation, though sufficient for upregulation of Prox1, a transcription factor critical for cell cycle exit and initiation of fiber differentiation, is not sufficient for induction of terminal fiber differentiation. Expression of Keratin 12, a marker of corneal epithelial differentiation, was reduced in the Ras transgenic corneas. CONCLUSIONS: Collectively, these results suggest that Ras activation a) induces distinct sets of downstream targets in the lens and cornea resulting in distinct cellular responses and b) is sufficient for initiation but not completion of lens fiber differentiation.


Subject(s)
Cornea/metabolism , Lens, Crystalline/metabolism , ras Proteins/metabolism , Animals , Cell Differentiation , Cell Proliferation , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mice, Transgenic
14.
Cleft Palate Craniofac J ; 47(6): 566-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20500056

ABSTRACT

Development of the secondary palate in mammals is a complex process that can be easily perturbed, leading to the common and distressing birth defect cleft palate. Animal models are particularly useful tools for dissecting underlying genetic components of cleft palate. We describe a new cleft palate model resulting from a transgene insertion mutation. Transgene insertional mutagenesis disrupts the genomic organization and expression of the Ap2ß1 gene located on chromosome 11. This gene encodes the ß2-adaptin subunit of the heterotetrameric adaptor protein 2 complex involved in clathrin-dependent endocytosis. Homozygous cleft palate mutant mice express no Ap2ß1 messenger RNA or ß2-adaptin protein and die during the perinatal period. Heterozygous mice are phenotypically normal despite expressing diminished ß2-adaptin messenger RNA and protein compared with wildtype. Remarkably, the paralogous ß1-adaptin subunit of the adaptor protein 1 complex partially substitutes for the missing ß2-adaptin in embryonic fibroblasts from homozygous mutant mice, resulting in assembly of reduced levels of an adaptor protein 2 complex bearing ß1-adaptin. This variant adaptor protein 2 complex is, therefore, apparently capable of maintaining viability of the homozygous mutant embryos until birth but insufficient to support palatogenesis. Nonsyndromic cleft palate in an animal model is associated with disruption of the Ap2ß1 gene.


Subject(s)
Cleft Palate/genetics , Transcription Factor AP-2/deficiency , Transcription Factor AP-2/genetics , Animals , Disease Models, Animal , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutagenesis, Insertional , Syndrome
15.
Dev Biol ; 324(2): 202-12, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18848538

ABSTRACT

It is widely accepted that vitreous humor-derived FGFs are required for the differentiation of anterior lens epithelial cells into crystallin-rich fibers. We show that BMP2, 4, and 7 can induce the expression of markers of fiber differentiation in primary lens cell cultures to an extent equivalent to FGF or medium conditioned by intact vitreous bodies (VBCM). Abolishing BMP2/4/7 signaling with noggin inhibited VBCM from upregulating fiber marker expression. Remarkably, noggin and anti-BMP antibodies also prevented purified FGF (but not unrelated stimuli) from upregulating the same fiber-specific proteins. This effect is attributable to inhibition of BMPs produced by the lens cells themselves. Although BMP signaling is required for FGF to enhance fiber differentiation, the converse is not true. Expression of noggin in the lenses of transgenic mice resulted in a postnatal block of epithelial-to-secondary fiber differentiation, with extension of the epithelial monolayer to the posterior pole of the organ. These results reveal the central importance of BMP in secondary fiber formation and show that although FGF may be necessary for this process, it is not sufficient. Differentiation of fiber cells, and thus proper vision, is dependent on cross-talk between the FGF and BMP signaling pathways.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Cell Differentiation , Fibroblast Growth Factors/metabolism , Lens, Crystalline/embryology , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins/genetics , Cells, Cultured , Chick Embryo , Culture Media, Conditioned , Epithelial Cells/metabolism , Eye Proteins/metabolism , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental , Intermediate Filament Proteins/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/drug effects , Mice , Mice, Transgenic , Vitreous Body/metabolism , delta-Crystallins/metabolism
16.
Dev Biol ; 323(1): 53-63, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18778700

ABSTRACT

The lens in the vertebrate eye has been shown to be critical for proper differentiation of the surrounding ocular tissues including the cornea, iris and ciliary body. In mice, previous investigators have assayed the consequences of molecular ablation of the lens. However, in these studies, lens ablation was initiated (and completed) after the cornea, retina, iris and ciliary body had initiated their differentiation programs thereby precluding analysis of the early role of the lens in fate determination of these tissues. In the present study, we have ablated the lens precursor cells of the surface ectoderm by generation of transgenic mice that express an attenuated version of diphtheria toxin (Tox176) linked to a modified Pax6 promoter that is active in the lens ectodermal precursors. In these mice, lens precursor cells fail to express Sox2, Prox1 and alphaA-crystallin and die before the formation of a lens placode. The Tox176 mice also showed profound alterations in the corneal differentiation program. The corneal epithelium displayed histological features of the skin, and expressed markers of skin differentiation such as Keratin 1 and 10 instead of Keratin 12, a marker of corneal epithelial differentiation. In the Tox176 mice, in the absence of the lens, extensive folding of the retina was seen. However, differentiation of the major cell types in the retina including the ganglion, amacrine, bipolar and horizontal cells was not affected. Unexpectedly, ectopic placement of the retinal pigmented epithelium was seen between the folds of the retina. Initial specification of the presumptive ciliary body and iris at the anterior margins of the retina was not altered in the Tox176 mice but their subsequent differentiation was blocked. Lacrimal and Harderian glands, which are derived from the Pax6-expressing surface ectodermal precursors, also failed to differentiate. These results suggest that, in mice, specification of the retina, ciliary body and iris occurs at the very outset of eye development and independent of the lens. In addition, our results also suggest that the lens cells of the surface ectoderm may be critical for the proper differentiation of the corneal epithelium.


Subject(s)
Aphakia/genetics , Gene Expression Regulation, Developmental , Genes, Dominant , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Animals , Crystallins/genetics , Diphtheria Toxin/genetics , Embryo, Mammalian , Endothelium, Corneal/abnormalities , Endothelium, Corneal/metabolism , Endothelium, Corneal/pathology , Immunohistochemistry , In Situ Hybridization , Lens, Crystalline/pathology , Lens, Crystalline/physiology , Mice , Mice, Transgenic , Transgenes
17.
Mol Vis ; 14: 602-14, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18385796

ABSTRACT

PURPOSE: The retinoblastoma (Rb) gene family member p130 binds preferentially to the E2F5 transcription factor and forms a repressive E2F5/p130 complex that inhibits cell cycle progression and tumor growth. It is unclear whether E2F5, either alone or in combination with p130, can interfere with the transcriptional activity of other E2F family members, such as E2F1 and E2F3a, in vivo. In this study, we used transgenic mice to test whether overexpression of E2F5 with/without p130 would be sufficient to inhibit E2F1 or E2F3a induced cell cycle reentry. METHODS: Transgenic mice were generated by microinjection of constructs containing different E2F cDNAs (E2F1, E2F3a, and E2F5) or the p130 cDNA linked to the mouse alphaA-crystallin promoter. The E2F5 single and E2F5/p130 double transgenic mice were cross-mated with E2F1 or E2F3a transgenic mice. The resulting double or triple transgenic mouse embryos were characterized by histology, in situ hybridization, immunohistochemistry, and BrdU incorporation assays. RESULTS: Overexpression of E2F5 alone was not sufficient to inhibit E2F1 or E2F3a induced cell cycle reentry in lens fiber cells. Transgenic mice coexpressing E2F5 and p130 in lens fiber cells did not show lens defects. However, coexpression of E2F5/p130 with E2F1 or E2F3a in lens fiber cells decreased the number of BrdU positive fiber cells induced by the E2F1 or E2F3a alone. Therefore, overexpression of E2F5/p130, but not E2F5 alone, can inhibit activator E2F-mediated cell proliferation in vivo, confirming that p130 plays a critical role in the repressive activity of E2F5/p130 complex. CONCLUSIONS: Overexpression of E2F5/p130 in post-mitotic lens fiber cells does not affect their normal differentiation program, but can inhibit inappropriate cell cycle reentry induced by the activator E2Fs. Since E2F5 alone cannot interfere with these E2F activities, we conclude that p130 is a key player in the inhibitory process.


Subject(s)
Cell Cycle/physiology , E2F5 Transcription Factor/metabolism , Lens, Crystalline/cytology , Retinoblastoma-Like Protein p130/metabolism , Animals , Cell Differentiation/physiology , E2F Transcription Factors/metabolism , E2F1 Transcription Factor/metabolism , E2F3 Transcription Factor/metabolism , Humans , Mice , Mice, Transgenic
18.
Mol Vis ; 13: 2289-300, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18199970

ABSTRACT

PURPOSE: The purpose of this study was to reassess the role of the lens as an "embryonic organizer" of ocular tissues. METHODS: We ablated the lens in mice by lens-specific expression of an attenuated version of diphtheria toxin A subunit(Tox176) driven by a modified crystallin promoter. Alterations in the differentiation programs of ocular tissues were examined by hematoxylin and eosin staining, in situ hybridization, and immunohistochemistry. RESULTS: Transgenic mice in the family OVE1757 exhibited severe microphakia. Apoptotic lens fibers were seen by embryonic day 15 (E15) and the lenses were completely ablated by post natal day 8. Multiple defects were seen in the anterior chamber. Corneal endothelial cells did not differentiate properly. The mesenchymal cells that would normally give rise to the endothelial layer were found to express N-cadherin, but they failed to form tight junctions and undergo a mesenchymal-to-epithelial transition. Although early specification of the presumptive ciliary body and iris was detected, subsequent differentiation of the iris was blocked. No dramatic changes were seen in the development of the retina. CONCLUSIONS: These results support the hypothesis that an intact lens is essential for proper differentiation of both the corneal endothelium and the iris and that the lens "organizes" the development of tissues in the anterior chamber.


Subject(s)
Anterior Chamber/abnormalities , Aphakia/congenital , Aphakia/complications , Lens, Crystalline/abnormalities , Animals , Animals, Newborn/abnormalities , Animals, Newborn/genetics , Cell Differentiation , Diphtheria Toxin , Endothelium, Corneal/abnormalities , Female , Fetal Organ Maturity/genetics , Iris/abnormalities , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Transgenic , Microphthalmos/etiology , Organogenesis/genetics , Pregnancy , Promoter Regions, Genetic , Retina/embryology
19.
Mol Vis ; 13: 397-407, 2007 Mar 26.
Article in English | MEDLINE | ID: mdl-17417601

ABSTRACT

PURPOSE: Insulin and insulin-like growth factors (IGFs) are putative regulators of cell proliferation and differentiation during lens development. Transgenic mice that overexpress IGF-1 in the lens have been previously described. To further understand the ocular functions of this growth factor family, the in vivo effects of insulin expression on lens development were investigated using transgenic mice. METHODS: Expression of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in mouse lens was examined by reverse-transcriptase-polymerase chain reaction (RT-PCR) and in situ hybridization. Transgenic mice that overexpress insulin in the lens were generated using two different promoters: a fiber-cell specific alphaA-crystallin (alphaA) promoter and a modified alphaA-promoter linked to the chicken delta1-crystallin enhancer (called the deltaenalphaA promoter). The deltaenalphaA promoter is active in both lens epithelial and fiber cells. The lens phenotypes were analyzed by histology and immunohistochemistry. Protein expression was examined by western blotting. RESULTS: Normal mouse lenses express both the insulin receptor (IR) and the IGF-1 receptor (IGF-1R), and their expression is highest at the lens periphery where the germinative and transitional zones are located. In transgenic mice, insulin expression in the lens induced cataract formation. The severity of the cataracts reflected the level of transgene expression, independent of the type of promoter used. In severely affected families, the spherical shape of the lens was altered and the lenses were smaller than normal. Histological analysis showed no evidence of premature differentiation of the anterior epithelial cells. In contrast to the IGF-1 mice, insulin transgenic mice exhibited an anterior shift in the location of the germinative and transitional zones, leading to a reduction of the lens epithelial compartment. Additional alterations included expansion of the lens transitional zone, variable nuclear positioning in the lens bow region, and inhibition of fiber cell denucleation and terminal differentiation. CONCLUSIONS: Elevated intraocular insulin does not enhance proliferation nor induce differentiation of mouse lens epithelial cells. Since an increase in IGF-1 causes a posterior shift of the lens geminative and transitional zones, while an increase in insulin causes an anterior shift of these zones, our results suggest that these two growth factors may work together to control the location of this structural domain during normal lens development. Our data also suggest that increased insulin-signaling activity in the lens can antagonize the endogenous signals that are responsible for fiber cell maturation and terminal differentiation.


Subject(s)
Insulin/metabolism , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Signal Transduction , Animals , Animals, Newborn , Cataract/etiology , Cataract/metabolism , Cataract/pathology , Cell Differentiation , Cell Proliferation , Cellular Senescence , Crystallins/metabolism , Embryo, Mammalian/metabolism , In Situ Hybridization , Insulin/genetics , Lens, Crystalline/pathology , Mice , Mice, Transgenic , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
20.
Biomed Opt Express ; 8(1): 407-419, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-28101427

ABSTRACT

Neural tube closure is a critical feature of central nervous system morphogenesis during embryonic development. Failure of this process leads to neural tube defects, one of the most common forms of human congenital defects. Although molecular and genetic studies in model organisms have provided insights into the genes and proteins that are required for normal neural tube development, complications associated with live imaging of neural tube closure in mammals limit efficient morphological analyses. Here, we report the use of optical coherence tomography (OCT) for dynamic imaging and quantitative assessment of cranial neural tube closure in live mouse embryos in culture. Through time-lapse imaging, we captured two neural tube closure mechanisms in different cranial regions, zipper-like closure of the hindbrain region and button-like closure of the midbrain region. We also used OCT imaging for phenotypic characterization of a neural tube defect in a mouse mutant. These results suggest that the described approach is a useful tool for live dynamic analysis of normal neural tube closure and neural tube defects in the mouse model.

SELECTION OF CITATIONS
SEARCH DETAIL