Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 721: 150025, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38768546

ABSTRACT

The causes of Alzheimer's disease (AD) are poorly understood, although many genes are known to be involved in this pathology. To gain insights into the underlying molecular mechanisms, it is essential to identify the relationships between individual AD genes. Previous work has shown that the splice variant E of KLC1 (KLC1_vE) promotes AD, and that the CELF1 gene, which encodes an RNA-binding protein involved in splicing regulation, is at a risk locus for AD. Here, we identified a functional link between CELF1 and KLC1 in AD pathogenesis. Transcriptomic data from human samples from different ethnic groups revealed that CELF1 mRNA levels are low in AD brains, and the splicing pattern of KLC1 is strongly correlated with CELF1 expression levels. Specifically, KLC1_vE is negatively correlated with CELF1. Depletion and overexpression experiments in cultured cells demonstrated that the CELF1 protein down-regulates KLC1_vE. In a cross-linking and immunoprecipitation sequencing (CLIP-seq) database, CELF1 directly binds to KLC1 RNA, following which it likely modulates terminal exon usage, hence KLC1_vE formation. These findings reveal a new pathogenic pathway where a risk allele of CELF1 is associated with reduced CELF1 expression, which up-regulates KLC1_vE to promote AD.


Subject(s)
Alternative Splicing , Alzheimer Disease , CELF1 Protein , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Brain/metabolism , CELF1 Protein/metabolism , CELF1 Protein/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
2.
Cells ; 12(20)2023 10 18.
Article in English | MEDLINE | ID: mdl-37887322

ABSTRACT

Cataract, the opacification of the lens, is the leading cause of blindness worldwide. Although effective, cataract surgery is costly and can lead to complications. Toward identifying alternate treatments, it is imperative to develop organoid models relevant for lens studies and drug screening. Here, we demonstrate that by culturing mouse lens epithelial cells under defined three-dimensional (3D) culture conditions, it is possible to generate organoids that display optical properties and recapitulate many aspects of lens organization and biology. These organoids can be rapidly produced in large amounts. High-throughput RNA sequencing (RNA-seq) on specific organoid regions isolated via laser capture microdissection (LCM) and immunofluorescence assays demonstrate that these lens organoids display a spatiotemporal expression of key lens genes, e.g., Jag1, Pax6, Prox1, Hsf4 and Cryab. Further, these lens organoids are amenable to the induction of opacities. Finally, the knockdown of a cataract-linked RNA-binding protein encoding gene, Celf1, induces opacities in these organoids, indicating their use in rapidly screening for genes that are functionally relevant to lens biology and cataract. In sum, this lens organoid model represents a compelling new tool to advance the understanding of lens biology and pathology and can find future use in the rapid screening of compounds aimed at preventing and/or treating cataracts.


Subject(s)
Cataract , Lens, Crystalline , Animals , Mice , Lens, Crystalline/metabolism , Cataract/metabolism , Transcription Factors/metabolism , RNA-Binding Proteins/metabolism , Organoids/metabolism
3.
bioRxiv ; 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37503005

ABSTRACT

The ocular lens, along with the cornea, focuses light on the retina to generate sharp images. Opacification of the lens, or cataract, is the leading cause of blindness worldwide. Presently, the best approach for cataract treatment is to surgically remove the diseased lens and replace it with an artificial implant. Although effective, this is costly and can have post-surgical complications. Toward identifying alternate treatments, it is imperative to develop organoid models relevant for lens studies and anti-cataract drug screening. Here, we demonstrate that by culturing mouse lens epithelial cells under defined 3-dimensional (3D) culture conditions, it is possible to generate organoids that display optical properties and recapitulate many aspects of lens organization at the tissue, cellular and transcriptomic levels. These 3D cultured lens organoids can be rapidly produced in large amounts. High-throughput RNA-sequencing (RNA-seq) on specific organoid regions isolated by laser capture microdissection (LCM) and immunofluorescence assays demonstrate that these lens organoids display spatiotemporal expression of key lens genes, e.g. , Jag1 , Pax6 , Prox1 , Hsf4 and Cryab . Further, these lens organoids are amenable to induction of opacities. Finally, knockdown of a cataract-linked RNA-binding protein encoding gene, Celf1 , induces opacities in these organoids, indicating their use in rapidly screening for genes functionally relevant to lens biology and cataract. In sum, this lens organoid model represents a compelling new tool to advance the understanding of lens biology and pathology, and can find future use in the rapid screening of compounds aimed at preventing and/or treating cataract.

4.
Hum Genet ; 139(12): 1541-1554, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32594240

ABSTRACT

The homeodomain transcription factors (TFs) Pax6 (OMIM: 607108) and Prox1 (OMIM: 601546) critically regulate gene expression in lens development. While PAX6 mutations in humans can cause cataract, aniridia, microphthalmia, and anophthalmia, among other defects, Prox1 deletion in mice causes severe lens abnormalities, in addition to other organ defects. Furthermore, the optimal dosage/spatiotemporal expression of these key TFs is essential for development. In lens development, Pax6 expression is elevated in cells of the anterior epithelium compared to fiber cells, while Prox1 exhibits the opposite pattern. Whether post-transcriptional regulatory mechanisms control these precise TF expression patterns is unknown. Here, we report the unprecedented finding that the cataract-linked RNA-binding protein (RBP), Celf1 (OMIM: 601074), post-transcriptionally regulates Pax6 and Prox1 protein expression in lens development. Immunostaining shows that Celf1 lens-specific conditional knockout (Celf1cKO) mice exhibit abnormal elevation of Pax6 protein in fiber cells and abnormal Prox1 protein levels in epithelial cells-directly opposite to their normal expression patterns in development. Furthermore, RT-qPCR shows no change in Pax6 and Prox1 transcript levels in Celf1cKO lenses, suggesting that Celf1 regulates these TFs on the translational level. Indeed, RNA-immunoprecipitation assays using Celf1 antibody indicate that Celf1 protein binds to Pax6 and Prox1 transcripts. Furthermore, reporter assays in Celf1 knockdown and Celf1-overexpression cells demonstrate that Celf1 negatively controls Pax6 and Prox1 translation via their 3' UTRs. These data define a new mechanism of RBP-based post-transcriptional regulation that enables precise control over spatiotemporal expression of Pax6 and Prox1 in lens development, thereby uncovering a new etiological mechanism for Celf1 deficiency-based cataract.


Subject(s)
CELF1 Protein/genetics , Cataract/genetics , Homeodomain Proteins/genetics , Lens, Crystalline/metabolism , PAX6 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Animals , CELF1 Protein/antagonists & inhibitors , CELF1 Protein/deficiency , Cataract/pathology , Cell Differentiation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Eye Proteins/antagonists & inhibitors , Eye Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Lens, Crystalline/growth & development , Mice , Mice, Knockout , RNA-Binding Proteins/genetics
5.
Dev Dyn ; 249(5): 610-621, 2020 05.
Article in English | MEDLINE | ID: mdl-31872467

ABSTRACT

BACKGROUND: Ocular lens clouding is termed as cataract, which depending on the onset, is classified as congenital or age-related. Developing new cataract treatments requires new models. Thus far, Xenopus embryos have not been evaluated as a system for studying cataract. RESULTS: We characterized the developmental process of lens formation in Xenopus laevis tailbuds and tadpoles, and we disrupted the orthologues of three mammalian cataract-linked genes in F0 by CRISPR/Cas9. We assessed the consequences of gene inactivation by combining external examination with histochemical analyses and functional vision assays. Inactivating the key metazoan eye development transcription factor gene pax6 produces a strong eye phenotype including an absence of eye tissue. Inactivating the genes for gap-junction protein and a nuclease, gja8 and dnase2b, produces lens defects that share several features of human cataracts, including impaired vision acuity, nuclei retention in lens fiber cells, and actin fibers disorganization. We tested the potential improvement of the visual acuity of gja8 crispant tadpoles upon treatment with the molecular chaperone 4-phenylbutyrate. CONCLUSION: Xenopus is a valuable model organism to understand the molecular pathology of congenital eye defects, including cataracts, and to screen molecules with a potential to prevent or reverse cataracts.


Subject(s)
Xenopus laevis/physiology , Animals , Cataract/physiopathology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Humans , Lens, Crystalline/physiology
6.
PLoS Genet ; 14(3): e1007278, 2018 03.
Article in English | MEDLINE | ID: mdl-29565969

ABSTRACT

Opacification of the ocular lens, termed cataract, is a common cause of blindness. To become transparent, lens fiber cells undergo degradation of their organelles, including their nuclei, presenting a fundamental question: does signaling/transcription sufficiently explain differentiation of cells progressing toward compromised transcriptional potential? We report that a conserved RNA-binding protein Celf1 post-transcriptionally controls key genes to regulate lens fiber cell differentiation. Celf1-targeted knockout mice and celf1-knockdown zebrafish and Xenopus morphants have severe eye defects/cataract. Celf1 spatiotemporally down-regulates the cyclin-dependent kinase (Cdk) inhibitor p27Kip1 by interacting with its 5' UTR and mediating translation inhibition. Celf1 deficiency causes ectopic up-regulation of p21Cip1. Further, Celf1 directly binds to the mRNA of the nuclease Dnase2b to maintain its high levels. Together these events are necessary for Cdk1-mediated lamin A/C phosphorylation to initiate nuclear envelope breakdown and DNA degradation in fiber cells. Moreover, Celf1 controls alternative splicing of the membrane-organization factor beta-spectrin and regulates F-actin-crosslinking factor Actn2 mRNA levels, thereby controlling fiber cell morphology. Thus, we illustrate new Celf1-regulated molecular mechanisms in lens development, suggesting that post-transcriptional regulatory RNA-binding proteins have evolved conserved functions to control vertebrate oculogenesis.


Subject(s)
CELF1 Protein/physiology , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Endodeoxyribonucleases/genetics , Lens, Crystalline/growth & development , RNA-Binding Proteins/physiology , Xenopus Proteins/physiology , Zebrafish Proteins/physiology , Animals , Cell Line , Gene Expression Regulation , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Mice , Xenopus laevis , Zebrafish
7.
Dev Biol ; 426(2): 449-459, 2017 06 15.
Article in English | MEDLINE | ID: mdl-27546377

ABSTRACT

Regulation of alternative splicing is an important process for cell differentiation and development. Down-regulation of Ptbp1, a regulatory RNA-binding protein, leads to developmental skin defects in Xenopus laevis. To identify Ptbp1-dependent splicing events potentially related to the phenotype, we conducted RNAseq experiments following Ptbp1 depletion. We systematically compared exon-centric and junction-centric approaches to detect differential splicing events. We showed that the junction-centric approach performs far better than the exon-centric approach in Xenopus laevis. We carried out the same comparisons using simulated data in human, which led us to propose that the better performances of the junction-centric approach in Xenopus laevis essentially relies on an incomplete exonic annotation associated with a correct transcription unit annotation. We assessed the capacity of the exon-centric and junction-centric approaches to retrieve known and to discover new Ptbp1-dependent splicing events. Notably, the junction-centric approach identified Ptbp1-controlled exons in agfg1, itga6, actn4, and tpm4 mRNAs, which were independently confirmed. We conclude that the junction-centric approach allows for a more complete and informative description of splicing events, and we propose that this finding might hold true for other species with incomplete annotations.


Subject(s)
Alternative Splicing , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Polypyrimidine Tract-Binding Protein/physiology , Xenopus Proteins/physiology , Xenopus laevis/genetics , Animals , Computer Simulation , Embryo, Nonmammalian/metabolism , Exons/genetics , Gene Library , Models, Genetic , Molecular Sequence Annotation , Morpholinos/pharmacology , RNA, Messenger/genetics , Sequence Alignment , Sequence Analysis, RNA , Xenopus laevis/embryology
8.
Dev Biol ; 409(2): 489-501, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26546114

ABSTRACT

In humans, genetic diseases affecting skin integrity (genodermatoses) are generally caused by mutations in a small number of genes that encode structural components of the dermal-epidermal junctions. In this article, we first show that inactivation of both exosc9, which encodes a component of the RNA exosome, and ptbp1, which encodes an RNA-binding protein abundant in Xenopus embryonic skin, impairs embryonic Xenopus skin development, with the appearance of dorsal blisters along the anterior part of the fin. However, histological and electron microscopy analyses revealed that the two phenotypes are distinct. Exosc9 morphants are characterized by an increase in the apical surface of the goblet cells, loss of adhesion between the sensorial and peridermal layers, and a decrease in the number of ciliated cells within the blisters. Ptbp1 morphants are characterized by an altered goblet cell morphology. Gene expression profiling by deep RNA sequencing showed that the expression of epidermal and genodermatosis-related genes is also differentially affected in the two morphants, indicating that alterations in post-transcriptional regulations can lead to skin developmental defects through different routes. Therefore, the developing larval epidermis of Xenopus will prove to be a useful model for dissecting the post-transcriptional regulatory network involved in skin development and stability with significant implications for human diseases.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/genetics , Gene Knockdown Techniques , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Polypyrimidine Tract-Binding Protein/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Skin/embryology , Skin/pathology , Xenopus Proteins/genetics , Xenopus laevis/embryology , Animal Fins/embryology , Animals , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/pathology , Embryo, Nonmammalian/ultrastructure , Epidermis/drug effects , Epidermis/pathology , Epidermis/ultrastructure , Exosome Multienzyme Ribonuclease Complex/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Gene Regulatory Networks/drug effects , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , In Situ Hybridization , Morpholinos/pharmacology , Polypyrimidine Tract-Binding Protein/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Xenopus Proteins/metabolism
9.
Mol Cell Biol ; 35(18): 3244-53, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26169831

ABSTRACT

CELF1 is a multifunctional RNA-binding protein that controls several aspects of RNA fate. The targeted disruption of the Celf1 gene in mice causes male infertility due to impaired spermiogenesis, the postmeiotic differentiation of male gametes. Here, we investigated the molecular reasons that underlie this testicular phenotype. By measuring sex hormone levels, we detected low concentrations of testosterone in Celf1-null mice. We investigated the effect of Celf1 disruption on the expression levels of steroidogenic enzyme genes, and we observed that Cyp19a1 was upregulated. Cyp19a1 encodes aromatase, which transforms testosterone into estradiol. Administration of testosterone or the aromatase inhibitor letrozole partly rescued the spermiogenesis defects, indicating that a lack of testosterone associated with excessive aromatase contributes to the testicular phenotype. In vivo and in vitro interaction assays demonstrated that CELF1 binds to Cyp19a1 mRNA, and reporter assays supported the conclusion that CELF1 directly represses Cyp19a1 translation. We conclude that CELF1 downregulates Cyp19a1 (Aromatase) posttranscriptionally to achieve high concentrations of testosterone compatible with spermiogenesis completion. We discuss the implications of these findings with respect to reproductive defects in men, including patients suffering from isolated hypogonadotropic hypogonadism and myotonic dystrophy type I.


Subject(s)
Aromatase/genetics , CELF1 Protein/genetics , Cytochrome P-450 CYP1A1/metabolism , Hypogonadism/genetics , Testosterone/metabolism , Animals , Aromatase Inhibitors/pharmacology , CELF1 Protein/metabolism , Cytochrome P-450 CYP1A1/biosynthesis , Down-Regulation , Estradiol/biosynthesis , Hypogonadism/etiology , Hypogonadism/pathology , Letrozole , Mice , Mice, Knockout , Myotonic Dystrophy/etiology , Nitriles/pharmacology , Protein Binding , Protein Biosynthesis , Spermatogenesis/drug effects , Spermatogenesis/physiology , Testosterone/blood , Triazoles/pharmacology , Up-Regulation
10.
Mol Cell Biol ; 35(4): 758-68, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25512611

ABSTRACT

The output of alternative splicing depends on the cooperative or antagonistic activities of several RNA-binding proteins (RBPs), like Ptbp1 and Esrp1 in Xenopus. Fine-tuning of the RBP abundance is therefore of prime importance to achieve tissue- or cell-specific splicing patterns. Here, we addressed the mechanisms leading to the high expression of the ptbp1 gene, which encodes Ptbp1, in Xenopus epidermis. Two splice isoforms of ptbp1 mRNA differ by the presence of an alternative exon 11, and only the isoform including exon 11 can be translated to a full-length protein. In vivo minigene assays revealed that the nonproductive isoform was predominantly produced. Knockdown experiments demonstrated that Esrp1, which is specific to the epidermis, strongly stimulated the expression of ptbp1 by favoring the productive isoform. Consequently, knocking down esrp1 phenocopied ptbp1 inactivation. Conversely, Ptbp1 repressed the expression of its own gene by favoring the nonproductive isoform. Hence, a complex posttranscriptional mechanism controls Ptbp1 abundance in Xenopus epidermis: skipping of exon 11 is the default splicing pattern, but Esrp1 stimulates ptbp1 expression by favoring the inclusion of exon 11 up to a level that is limited by Ptbp1 itself. These results decipher a posttranscriptional mechanism that achieves various abundances of the ubiquitous RBP Ptbp1 in different tissues.


Subject(s)
Amphibian Proteins/genetics , Epidermis/metabolism , Polypyrimidine Tract-Binding Protein/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Xenopus laevis/genetics , Alternative Splicing , Amphibian Proteins/antagonists & inhibitors , Amphibian Proteins/metabolism , Animals , Embryo, Nonmammalian , Epidermis/growth & development , Exons , Genotype , Introns , Phenotype , Polypyrimidine Tract-Binding Protein/antagonists & inhibitors , Polypyrimidine Tract-Binding Protein/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Xenopus laevis/growth & development , Xenopus laevis/metabolism
11.
Exp Cell Res ; 317(13): 1922-34, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21570965

ABSTRACT

The nuclear zinc finger protein ZFPIP/Zfp462 is an important factor involved in cell division during the early embryonic development of vertebrates. In pluripotent P19 cells, ZFPIP/Zfp462 takes part in cell proliferation, likely via its role in maintaining chromatin structure. To further define the function of ZFPIP/Zfp462 in the mechanisms of pluripotency and cell differentiation, we constructed a stable P19 cell line in which ZFPIP/Zfp462 knockdown is inducible. We report that ZFPIP/Zfp462 was vital for mitosis and self-renewal in pluripotent P19 cells. Its depletion induced substantial decreases in the expression of the pluripotency genes Nanog, Oct4 and Sox2 and was associated with the transient expression of specific neuronal differentiation markers. We also demonstrated that ZFPIP/Zfp462 expression appears to be unnecessary after neuronal differentiation is induced in P19 cells. Taken together, our results strongly suggest that ZFPIP/Zfp462 is a key chromatin factor involved in maintaining P19 pluripotency and in the early mechanisms of neural differentiation but that it is dispensable in differentiated P19 cells.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins , Mice , Nerve Tissue Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...