1
|
Coomson SY, Lachke SA. Bringing signaling complexity into focus. eLife 2025; 14:e106519. [PMID: 40167030 PMCID: PMC11961117 DOI: 10.7554/elife.106519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
A study in mice reveals key interactions between proteins involved in fibroblast growth factor signaling and how they contribute to distinct stages of eye lens development.
Collapse
Affiliation(s)
- Sarah Y Coomson
- Department of Biological Sciences, University of DelawareNewarkUnited States
| | - Salil A Lachke
- Department of Biological Sciences and Center for Bioinformatics and Computational Biology, University of DelawareNewarkUnited States
| |
Collapse
|
2
|
Le T, Htun S, Pandey MK, Sun Y, Magnusen AF, Ullah E, Lauzon J, Beres S, Lee C, Guan B, Hufnagel RB, Brooks BP, Baranzini SE, Slavotinek A. A zebrafish model of crim1 loss of function has small and misshapen lenses with dysregulated clic4 and fgf1b expression. Front Cell Dev Biol 2025; 13:1522094. [PMID: 40114969 PMCID: PMC11922885 DOI: 10.3389/fcell.2025.1522094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/22/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Heterozygous deletions predicting haploinsufficiency for the Cysteine Rich Motor Neuron 1 (CRIM1) gene have been identified in two families with macrophthalmia, colobomatous, with microcornea (MACOM), an autosomal dominant trait. Crim1 encodes a type I transmembrane protein that is expressed at the cell membrane of lens epithelial and fiber cells at the stage of lens pit formation. Decreased Crim1 expression in the mouse reduced the number of lens epithelial cells and caused defective adhesion between lens epithelial cells and between the epithelial and fiber cells. Methods We present three patients with heterozygous deletions and truncating variants predicted to result in haploinsufficiency for CRIM1 as further evidence for the role of this gene in eye defects, including retinal coloboma, optic pallor, and glaucoma. We used Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 to make a stable Danio rerio model of crim1 deficiency, generating zebrafish that were homozygous for a 2 basepair deletion, c.339_340delCT p.Leu112Leufs*, in crim1. Results Homozygous, crim1 -/- larvae demonstrated smaller eyes and small and misshapen lenses compared to controls, but we did not observe colobomas. Bulk RNA-Seq using dissected eyes from crim1 -/- larvae and controls at 72 h post fertilization showed significant downregulation of crim1 and chloride intracellular channel 4 (clic4) and upregulation of fibroblast growth factor 1b (fgf1b) and complement component 1, q subcomponent (c1q), amongst other dysregulated genes. Discussion Our work strengthens the association between haploinsufficiency for CRIM1 and eye defects and characterizes a stable model of crim1 loss of function for future research.
Collapse
Affiliation(s)
- Tien Le
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Stephanie Htun
- Division of Medical Genetics, Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States
| | - Manoj Kumar Pandey
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Yihui Sun
- Department of Neurology, Weill Institute for Neurosciences., University of California San Francisco, San Francisco, CA, United States
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA, United States
| | - Albert Frank Magnusen
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Ehsan Ullah
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Julie Lauzon
- Department of Medical Genetics and Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| | - Shannon Beres
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, United States
- Department of Neurology and Neurosciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Chung Lee
- Stanford University Pediatrics/Medical Genetics, Stanford University, Stanford, CA, United States
| | - Bin Guan
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sergio E Baranzini
- Department of Neurology, Weill Institute for Neurosciences., University of California San Francisco, San Francisco, CA, United States
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA, United States
| | - Anne Slavotinek
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Division of Medical Genetics, Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
3
|
Wang Q, Li H, Mao Y, Garg A, Park ES, Wu Y, Chow A, Peregrin J, Zhang X. Shc1 cooperates with Frs2 and Shp2 to recruit Grb2 in FGF-induced lens development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.20.619055. [PMID: 39484547 PMCID: PMC11527007 DOI: 10.1101/2024.10.20.619055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Fibroblast growth factor (FGF) signaling elicits multiple downstream pathways, most notably the Ras/MAPK cascade facilitated by the adaptor protein Grb2. However, the mechanism by which Grb2 is recruited to the FGF signaling complex remains unresolved. Here we showed that genetic ablation of FGF signaling prevented lens induction by disrupting transcriptional regulation and actin cytoskeletal arrangements, which could be reproduced by deleting the juxtamembrane region of the FGF receptor and rescued by Kras activation. Conversely, mutations affecting the Frs2-binding site on the FGF receptor or the deletion of Frs2 and Shp2 primarily impact later stages of lens vesicle development involving lens fiber cell differentiation. Our study further revealed that the loss of Grb2 abolished MAPK signaling, resulting in a profound arrest of lens development. However, removing Grb2's putative Shp2 dephosphorylation site (Y209) neither produced a detectable phenotype nor impaired MAPK signaling during lens development. Furthermore, the catalytically inactive Shp2 mutation (C459S) only modestly impaired FGF signaling, whereas replacing Shp2's C-terminal phosphorylation sites (Y542/Y580) previously implicated in Grb2 binding only caused placental defects, perinatal lethality, and reduced lacrimal gland branching without impacting lens development, suggesting that Shp2 only partially mediates Grb2 recruitment. In contrast, we observed that FGF signaling is required for the phosphorylation of the Grb2-binding sites on Shc1 and the deletion of Shc1 exacerbates the lens vesicle defect caused by Frs2 and Shp2 deletion. These findings establish Shc1 as a critical collaborator with Frs2 and Shp2 in targeting Grb2 during FGF signaling.
Collapse
Affiliation(s)
- Qian Wang
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Hongge Li
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Yingyu Mao
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Ankur Garg
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Eun Sil Park
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Yihua Wu
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Alyssa Chow
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - John Peregrin
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Xin Zhang
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
4
|
Yin H, Staples SCR, Pickering JG. The fundamentals of fibroblast growth factor 9. Differentiation 2024; 139:100731. [PMID: 37783652 DOI: 10.1016/j.diff.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/07/2023] [Accepted: 09/17/2023] [Indexed: 10/04/2023]
Abstract
Fibroblast growth factor 9 (FGF9) was first identified during a screen for factors acting on cells of the central nervous system (CNS). Research over the subsequent two decades has revealed this protein to be a critically important and elegantly regulated growth factor. A hallmark control feature is reciprocal compartmentalization, particularly during development, with epithelium as a dominant source and mesenchyme a prime target. This mesenchyme selectivity is accomplished by the high affinity of FGF9 to the IIIc isoforms of FGFR1, 2, and 3. FGF9 is expressed widely in the embryo, including the developing heart and lungs, and more selectively in the adult, including the CNS and kidneys. Global Fgf9-null mice die shortly after birth due to respiratory failure from hypoplastic lungs. As well, their hearts are dilated and poorly vascularized, the skeleton is small, the intestine is shortened, and male-to-female sex reversal can be found. Conditional Fgf9-null mice have revealed CNS phenotypes, including ataxia and epilepsy. In humans, FGF9 variants have been found to underlie multiple synostoses syndrome 3, a syndrome characterized by multiple joint fusions. Aberrant FGF9 signaling has also been implicated in differences of sex development and cancer, whereas vascular stabilizing effects of FGF9 could benefit chronic diseases. This primer reviews the attributes of this vital growth factor.
Collapse
Affiliation(s)
- Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Sabrina C R Staples
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada; Department of Medical Biophysics, Western University, London, Canada
| | - J Geoffrey Pickering
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada; Department of Medical Biophysics, Western University, London, Canada; Department of Biochemistry, Western University, London, Canada; Department of Medicine, Western University, London, Canada; London Health Sciences Centre, London, Canada.
| |
Collapse
|
5
|
Upreti A, Padula SL, Weaver JM, Wagner BD, Kneller AM, Petulla AL, Lachke SA, Robinson ML. A Transcriptomics Analysis of the Regulation of Lens Fiber Cell Differentiation in the Absence of FGFRs and PTEN. Cells 2024; 13:1222. [PMID: 39056803 PMCID: PMC11274593 DOI: 10.3390/cells13141222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/28/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Adding 50% vitreous humor to the media surrounding lens explants induces fiber cell differentiation and a significant immune/inflammatory response. While Fgfr loss blocks differentiation in lens epithelial explants, this blockage is partially reversed by deleting Pten. To investigate the functions of the Fgfrs and Pten during lens fiber cell differentiation, we utilized a lens epithelial explant system and conducted RNA sequencing on vitreous humor-exposed explants lacking Fgfrs, or Pten or both Fgfrs and Pten. We found that Fgfr loss impairs both vitreous-induced differentiation and inflammation while the additional loss of Pten restores these responses. Furthermore, transcriptomic analysis suggested that PDGFR-signaling in FGFR-deficient explants is required to mediate the rescue of vitreous-induced fiber differentiation in explants lacking both Fgfrs and Pten. The blockage of β-crystallin induction in explants lacking both Fgfrs and Pten in the presence of a PDGFR inhibitor supports this hypothesis. Our findings demonstrate that a wide array of genes associated with fiber cell differentiation are downstream of FGFR-signaling and that the vitreous-induced immune responses also depend on FGFR-signaling. Our data also demonstrate that many of the vitreous-induced gene-expression changes in Fgfr-deficient explants are rescued in explants lacking both Fgfrs and Pten.
Collapse
Affiliation(s)
- Anil Upreti
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA; (A.U.); (S.L.P.); (J.M.W.)
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Stephanie L. Padula
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA; (A.U.); (S.L.P.); (J.M.W.)
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Jacob M. Weaver
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA; (A.U.); (S.L.P.); (J.M.W.)
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Brad D. Wagner
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Allison M. Kneller
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Anthony L. Petulla
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Salil A. Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA
| | - Michael L. Robinson
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA; (A.U.); (S.L.P.); (J.M.W.)
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| |
Collapse
|
6
|
VanSlyke JK, Boswell BA, Musil LS. TGFβ overcomes FGF-induced transinhibition of EGFR in lens cells to enable fibrotic secondary cataract. Mol Biol Cell 2024; 35:ar75. [PMID: 38598298 PMCID: PMC11238076 DOI: 10.1091/mbc.e24-01-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/29/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
To cause vision-disrupting fibrotic secondary cataract (PCO), lens epithelial cells that survive cataract surgery must migrate to the posterior of the lens capsule and differentiate into myofibroblasts. During this process, the cells become exposed to the FGF that diffuses out of the vitreous body. In normal development, such relatively high levels of FGF induce lens epithelial cells to differentiate into lens fiber cells. It has been a mystery as to how lens cells could instead undergo a mutually exclusive cell fate, namely epithelial to myofibroblast transition, in the FGF-rich environment of the posterior capsule. We and others have reported that the ability of TGFβ to induce lens cell fibrosis requires the activity of endogenous ErbBs. We show here that lens fiber-promoting levels of FGF induce desensitization of ErbB1 (EGFR) that involves its phosphorylation on threonine 669 mediated by both ERK and p38 activity. Transinhibition of ErbB1 by FGF is overcome by a time-dependent increase in ErbB1 levels induced by TGFβ, the activation of which is increased after cataract surgery. Our studies provide a rationale for why TGFβ upregulates ErbB1 in lens cells and further support the receptor as a therapeutic target for PCO.
Collapse
Affiliation(s)
- Judy K. VanSlyke
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239
| | - Bruce A. Boswell
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239
| | - Linda S. Musil
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
7
|
Cvekl A, Camerino MJ. Generation of Lens Progenitor Cells and Lentoid Bodies from Pluripotent Stem Cells: Novel Tools for Human Lens Development and Ocular Disease Etiology. Cells 2022; 11:3516. [PMID: 36359912 PMCID: PMC9658148 DOI: 10.3390/cells11213516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
In vitro differentiation of human pluripotent stem cells (hPSCs) into specialized tissues and organs represents a powerful approach to gain insight into those cellular and molecular mechanisms regulating human development. Although normal embryonic eye development is a complex process, generation of ocular organoids and specific ocular tissues from pluripotent stem cells has provided invaluable insights into the formation of lineage-committed progenitor cell populations, signal transduction pathways, and self-organization principles. This review provides a comprehensive summary of recent advances in generation of adenohypophyseal, olfactory, and lens placodes, lens progenitor cells and three-dimensional (3D) primitive lenses, "lentoid bodies", and "micro-lenses". These cells are produced alone or "community-grown" with other ocular tissues. Lentoid bodies/micro-lenses generated from human patients carrying mutations in crystallin genes demonstrate proof-of-principle that these cells are suitable for mechanistic studies of cataractogenesis. Taken together, current and emerging advanced in vitro differentiation methods pave the road to understand molecular mechanisms of cataract formation caused by the entire spectrum of mutations in DNA-binding regulatory genes, such as PAX6, SOX2, FOXE3, MAF, PITX3, and HSF4, individual crystallins, and other genes such as BFSP1, BFSP2, EPHA2, GJA3, GJA8, LIM2, MIP, and TDRD7 represented in human cataract patients.
Collapse
Affiliation(s)
- Aleš Cvekl
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michael John Camerino
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
8
|
He J, Xie P, Ouyang J. Circ_0122396 Protects Human Lens Epithelial Cells from Hydrogen Peroxide-induced Injury by Binding to miR-15a-5p to Stimulate FGF1 Expression. Curr Eye Res 2021; 47:246-255. [PMID: 34486899 DOI: 10.1080/02713683.2021.1978100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Circular_0122396 (circ_0122396) has been reported to be downregulated in age-related cataract (ARC); however, the underlying mechanism remains unknown. The study aimed to reveal the role of circ_0122396 in ARC progression and underneath mechanism. METHODS Hydrogen peroxide (H2O2) was employed to induce lens epithelial cells (SRA01/04) injury. The RNA expression of circ_0122396, microRNA-15a-5p (miR-15a-5p) and fibroblast growth factor 1 (FGF1) was detected by quantitative real-time polymerase chain reaction. Protein expression was checked by western blot. Cell viability, proliferation and apoptosis were investigated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, 5-Ethynyl-29-deoxyuridine and flow cytometry analysis, respectively. Oxidative stress was evaluated by superoxide dismutase and catalase activity assay kits and lipid peroxidation malondialdehyde assay kit. Online databases and mechanism assays were used to predict and identify the relationship between miR-15a-5p and circ_0122396 or FGF1. RESULTS Circ_0122396 and FGF1 expression were significantly downregulated, but miR-15a-5p expression was upregulated in ARC tissues or/and H2O2-treated SRA01/04 cells in comparison with control groups. H2O2 treatment repressed cell proliferation and induced cell apoptosis and oxidative stress, which was attenuated after circ_0122396 overexpression. MiR-15a-5p, a target mRNA of circ_0122396, was found to participate in H2O2-triggered cell damage by interacting with circ_0122396. Additionally, FGF1 silencing attenuated miR-15a-5p inhibitors-mediated action. Importantly, circ_0122396 regulated FGF1 expression by interaction with miR-15a-5p in H2O2-treated SRA01/04 cells. CONCLUSION Circ_0122396 ameliorated H2O2-triggered cell injury by inducing FGF1 through sponging miR-15a-5p, providing a potential target for ARC therapy.
Collapse
Affiliation(s)
- Jing He
- Department of Ophthalmology, Jiujiang No.1 People's Hospital, Jiujiang City, Jiangxi Provincial, China
| | - Ping Xie
- Department of Ophthalmology, Jiujiang No.1 People's Hospital, Jiujiang City, Jiangxi Provincial, China
| | - Jun Ouyang
- Department of Ophthalmology, Jiujiang No.1 People's Hospital, Jiujiang City, Jiangxi Provincial, China
| |
Collapse
|
9
|
Lens fiber cell differentiation occurs independently of fibroblast growth factor receptor signaling in the absence of Pten. Dev Biol 2020; 467:1-13. [PMID: 32858001 DOI: 10.1016/j.ydbio.2020.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 11/21/2022]
Abstract
Fibroblast growth factor receptor (FGFR) signaling patterns multiple tissues in both vertebrates and invertebrates, largely through the activation of intracellular kinases. Recent studies have demonstrated that the phosphatase, PTEN negatively regulates FGFR signaling, such that the loss of PTEN can compensate for reduced FGFR signaling to rescue aspects of normal development. In the developing mouse lens, FGFR signaling promotes cell survival and fiber cell differentiation, and the loss of Pten largely compensates for the loss of Fgfr2 during lens development. To explore this regulatory relationship further, we focused on the phenotypic consequences of Pten loss on lens development and fiber cell differentiation in the absence of all FGFR signaling, both in vivo and in lens epithelial explants. Pten deletion partially rescues primary fiber cell elongation and γ-crystallin accumulation in FGFR-deficient lenses in vivo but fails to rescue cell survival or proliferation. However, in lens epithelial explants, where cells survive without FGFR signaling, Pten deletion rescues vitreous humor-induced lens fiber cell differentiation in the combined absence of Fgfr1, Fgfr2 and Fgfr3. This represents the first evidence that vitreous-initiated signaling cascades, independent of FGFR signaling, can drive mammalian lens fiber cell differentiation, when freed from repression by PTEN.
Collapse
|
10
|
Wang J, Zhang J, Xiong Y, Li J, Li X, Zhao J, Zhu G, He H, Mayinuer Y, Wan X. TGF-β regulation of microRNA miR-497-5p and ocular lens epithelial cell mesenchymal transition. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1928-1937. [PMID: 32399769 DOI: 10.1007/s11427-019-1603-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/05/2019] [Indexed: 12/15/2022]
Abstract
The purpose of this study was to investigate the role of a human lens microRNA (miR-497-5p) in regulating epithelialmesenchymal transition (EMT) under the control of transforming growth factor beta (TGF-β). A microRNA array was used to evaluate the microRNA profiles of untreated and TGF-β-treated human lens epithelial cells in culture. This showed that TGF-β treatment led to the upregulation of 96 microRNAs and downregulation of 39 microRNAs. Thirteen microRNAs were predicted to be involved in the pathogenesis of posterior capsule opacification (PCO). Meanwhile, overexpression of miR-497-5p suppressed cell proliferation and EMT 48 h post-transfection, and inhibition of miR-497-5p accelerated cell proliferation and EMT. Treatment with TGF-β inhibited the expression of miR-497-5p, but not cell proliferation. miR-497-5p was also found to regulate the level of CCNE1 and FGF7, which are reported to be actively involved in EMT. CCNE1 and FGF7 were bona fide targets of miR-497-5p. The results suggest that miR-497-5p participates in the direct regulation of lens epithelial cell EMT and is regulated by TGF-β. miR-497-5p may be a novel target for PCO therapy.
Collapse
Affiliation(s)
- Jinda Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing Institute of Ophthalmology, Beijing, 100005, China
| | - Jingshang Zhang
- Beijing Tongren Eye Center, Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing Institute of Ophthalmology, Beijing, 100005, China
| | - Ying Xiong
- Beijing Tongren Hospital of Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Eye Center, Beijing, 100005, China
| | - Jing Li
- Beijing Tongren Hospital of Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Eye Center, Beijing, 100005, China
| | - Xiaoxia Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing Institute of Ophthalmology, Beijing, 100005, China
| | - Jing Zhao
- Beijing Tongren Eye Center, Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing Institute of Ophthalmology, Beijing, 100005, China
| | - Guyu Zhu
- Beijing Tongren Eye Center, Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing Institute of Ophthalmology, Beijing, 100005, China
| | - Hailong He
- Beijing Tongren Eye Center, Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing Institute of Ophthalmology, Beijing, 100005, China
| | - Yusufu Mayinuer
- Beijing Tongren Eye Center, Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing Institute of Ophthalmology, Beijing, 100005, China
| | - Xiuhua Wan
- Beijing Tongren Eye Center, Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing Institute of Ophthalmology, Beijing, 100005, China.
| |
Collapse
|
11
|
Lens differentiation is controlled by the balance between PDGF and FGF signaling. PLoS Biol 2019; 17:e3000133. [PMID: 30716082 PMCID: PMC6375662 DOI: 10.1371/journal.pbio.3000133] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 02/14/2019] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
How multiple receptor tyrosine kinases coordinate cell fate determination is yet to be elucidated. We show here that the receptor for platelet-derived growth factor (PDGF) signaling recruits the p85 subunit of Phosphoinositide 3-kinase (PI3K) to regulate mammalian lens development. Activation of PI3K signaling not only prevents B-cell lymphoma 2 (BCL2)-Associated X (Bax)- and BCL2 Antagonist/Killer (Bak)-mediated apoptosis but also promotes Notch signaling to prevent premature cell differentiation. Reducing PI3K activity destabilizes the Notch intracellular domain, while the constitutive activation of Notch reverses the PI3K deficiency phenotype. In contrast, fibroblast growth factor receptors (FGFRs) recruit Fibroblast Growth Factor Receptor Substrate 2 (Frs2) and Rous sarcoma oncogene (Src) Homology Phosphatase 2 (Shp2) to activate Mitogen-Activated Protein Kinase (MAPK) signaling, which induces the Notch ligand Jagged 1 (Jag1) and promotes cell differentiation. Inactivation of Shp2 restored the proper timing of differentiation in the p85 mutant lens, demonstrating the antagonistic interaction between FGF-induced MAPK and PDGF-induced PI3K signaling. By selective activation of PI3K and MAPK, PDGF and FGF cooperate with and oppose each other to balance progenitor cell maintenance and differentiation. A central aim in understanding cell signaling is to decode the cellular logic that underlies the functional specificity of growth factors. Although these factors are known to activate a common set of intracellular pathways, they nevertheless play specific roles in development and physiology. Using lens development in mice as a model, we show that fibroblast growth factor (FGF) and platelet-derived growth factor (PDGF) antagonize each other through their intrinsic biases toward distinct downstream targets. While FGF primarily induces the Ras–Mitogen-Activated Protein Kinase (MAPK) axis to promote lens cell differentiation, PDGF preferentially stimulates Phosphoinositide 3-kinase (PI3K) to enhance Notch signaling, which is necessary for maintaining the lens progenitor cell pool. By revealing the intricate interactions between PDGF, FGF, and Notch, we present a paradigm for how signaling crosstalk enables balanced growth and differentiation in multicellular organisms.
Collapse
|
12
|
Roles of TGF β and FGF Signals in the Lens: Tropomyosin Regulation for Posterior Capsule Opacity. Int J Mol Sci 2018; 19:ijms19103093. [PMID: 30304871 PMCID: PMC6212802 DOI: 10.3390/ijms19103093] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 01/16/2023] Open
Abstract
Transforming growth factor (TGF) β and fibroblast growth factor (FGF) 2 are related to the development of posterior capsule opacification (PCO) after lens extraction surgery and other processes of epithelial–mesenchymal transition (EMT). Oxidative stress seems to activate TGF β1 largely through reactive oxygen species (ROS) production, which in turn alters the transcription of several survival genes, including lens epithelium-cell derived growth factor (LEDGF). Higher ROS levels attenuate LEDGF function, leading to down-regulation of peroxiredoxin 6 (Prdx6). TGF β is regulated by ROS in Prdx6 knock-out lens epithelial cells (LECs) and induces the up-regulation of tropomyosins (Tpms) 1/2, and EMT of LECs. Mouse and rat PCO are accompanied by elevated expression of Tpm2. Further, the expression of Tpm1/2 is induced by TGF β2 in LECs. Importantly, we previously showed that TGF β2 and FGF2 play regulatory roles in LECs in a contrasting manner. An injury-induced EMT of a mouse lens as a PCO model was attenuated in the absence of Tpm2. In this review, we present findings regarding the roles of TGF β and FGF2 in the differential regulation of EMT in the lens. Tpms may be associated with TGF β2- and FGF2-related EMT and PCO development.
Collapse
|
13
|
Spred negatively regulates lens growth by modulating epithelial cell proliferation and fiber differentiation. Exp Eye Res 2018; 178:160-175. [PMID: 30290165 DOI: 10.1016/j.exer.2018.09.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/28/2018] [Accepted: 09/28/2018] [Indexed: 12/27/2022]
Abstract
Spred, like Sprouty (Spry) and also Sef proteins, have been identified as important regulators of receptor tyrosine kinase (RTK)-mediated MAPK/ERK-signaling in various developmental systems, controlling cellular processes such as proliferation, migration and differentiation. Spreds are widely expressed during early embryogenesis, and in the eye lens, become more localised in the lens epithelium with later development, overlapping with other antagonists including Spry. Given the synexpression of Spreds and Spry in lens, in order to gain a better understanding of their specific roles in regulating growth factor mediated-signaling and cell behavior, we established and characterised lines of transgenic mice overexpressing Spred1 or Spred2, specifically in the lens. This overexpression of Spreds resulted in a small lens phenotype during ocular morphogenesis, retarding its growth by compromising epithelial cell proliferation and fiber differentiation. These in situ findings were shown to be dependent on the ability of Spreds to suppress MAPK-signaling, in particular FGF-induced ERK1/2-signaling in lens cells. This was validated in vitro using lens epithelial explants, that highlighted the overlapping role of Spreds with Spry2, but not Spry1. This study provides insights into the putative function of Spreds and Spry in situ, some overlapping and some distinct, and their importance in regulating lens cell proliferation and fiber differentiation contributing to lens and eye growth.
Collapse
|
14
|
Gao X, Yao X, Yang H, Deng K, Guo Y, Zhang T, Zhang G, Wang F. Role of FGF9 in sheep testis steroidogenesis during sexual maturation. Anim Reprod Sci 2018; 197:177-184. [DOI: 10.1016/j.anireprosci.2018.08.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/28/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022]
|
15
|
Molecular regulation of ocular gland development. Semin Cell Dev Biol 2018; 91:66-74. [PMID: 30266427 DOI: 10.1016/j.semcdb.2018.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 06/01/2018] [Accepted: 07/24/2018] [Indexed: 01/19/2023]
Abstract
The tear film is produced by two ocular glands, the lacrimal glands, which produce the aqueous component of this film, and the meibomian glands, which secrete the lipidic component that is key to reduce evaporation of the watery film at the surface of the eye. Embryonic development of these exocrine glands has been mostly studied in mice, which also develop Harderian glands, a third type of ocular gland whose role is still not well understood. This review provides an update on the signalling pathways, transcription factors andextracellular matrix components that have been shown to play a role in ocular gland development.
Collapse
|
16
|
Tam OH, Pennisi D, Wilkinson L, Little MH, Wazin F, Wan VL, Lovicu FJ. Crim1 is required for maintenance of the ocular lens epithelium. Exp Eye Res 2018; 170:58-66. [PMID: 29458060 DOI: 10.1016/j.exer.2018.02.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 12/28/2022]
Abstract
The development and growth of the vertebrate ocular lens is dependent on the regulated proliferation of an anterior monolayer of epithelial cells, and their subsequent differentiation into elongate fiber cells. The growth factor rich ocular media that bathes the lens mediates these cellular processes, and their respective intracellular signaling pathways are in turn regulated to ensure that the proper lens architecture is maintained. Recent studies have proposed that Cysteine Rich Motor Neuron 1 (Crim1), a transmembrane protein involved in organogenesis of many tissues, might influence cell adhesion, polarity and proliferation in the lens by regulating integrin-signaling. Here, we characterise the lens and eyes of the Crim1KST264 mutant mice, and show that the loss of Crim1 function in the ocular tissues results in inappropriate differentiation of the lens epithelium into fiber cells. Furthermore, restoration of Crim1 levels in just the lens tissue of Crim1KST264 mice is sufficient to ameliorate most of the dysgenesis observed in the mutant animals. Based on our findings, we propose that tight regulation of Crim1 activity is required for maintenance of the lens epithelium, and its depletion leads to ectopic differentiation into fiber cells, dramatically altering lens structure and ultimately leading to microphthalmia and aphakia.
Collapse
Affiliation(s)
- Oliver H Tam
- Save Sight Institute and Anatomy & Histology, Bosch Institute, The University of Sydney, NSW 2006, Australia
| | - David Pennisi
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | - Lorine Wilkinson
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | - Melissa H Little
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | - Fatima Wazin
- Save Sight Institute and Anatomy & Histology, Bosch Institute, The University of Sydney, NSW 2006, Australia
| | - Victor L Wan
- Save Sight Institute and Anatomy & Histology, Bosch Institute, The University of Sydney, NSW 2006, Australia
| | - Frank J Lovicu
- Save Sight Institute and Anatomy & Histology, Bosch Institute, The University of Sydney, NSW 2006, Australia.
| |
Collapse
|
17
|
Abstract
The eye lens grows by systematic proliferation of its epithelial cells and their differentiation into fibre cells. The anterior aqueous humour regulates lens epithelial cell proliferation whereas posteriorly, the vitreous stimulates lens fibre differentiation. Vitreous-derived members of the fibroblast growth factor (FGF) family induce fibre differentiation, with added support for FGFs as putative regulators of aqueous-induced lens cell proliferation. To further characterize this, given FGFs' known affinity for proteoglycans, we compared the effect of proteoglycan sulphation in growth factor- and aqueous-induced lens cell proliferation. Disruption of proteoglycan sulphation in lens cells specifically impacted on aqueous- and FGF-induced MAPK/ERK1/2-signalling, but not on that induced by other mitogens such as PDGF; however, cell proliferation was reduced in all treatment groups, regardless of the mitogen. Overall, by disrupting proteoglycan activity, we further highlight the significant role of FGFs in aqueous-induced ERK1/2 phosphorylation leading to lens cell proliferation.
Collapse
Affiliation(s)
- Laxmi Iyengar
- a Save Sight Institute and Discipline of Anatomy and Histology, Bosch Institute, University of Sydney , Sydney , Australia
| | - Frank J Lovicu
- a Save Sight Institute and Discipline of Anatomy and Histology, Bosch Institute, University of Sydney , Sydney , Australia
| |
Collapse
|
18
|
Cvekl A, Zhang X. Signaling and Gene Regulatory Networks in Mammalian Lens Development. Trends Genet 2017; 33:677-702. [PMID: 28867048 DOI: 10.1016/j.tig.2017.08.001] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 11/16/2022]
Abstract
Ocular lens development represents an advantageous system in which to study regulatory mechanisms governing cell fate decisions, extracellular signaling, cell and tissue organization, and the underlying gene regulatory networks. Spatiotemporally regulated domains of BMP, FGF, and other signaling molecules in late gastrula-early neurula stage embryos generate the border region between the neural plate and non-neural ectoderm from which multiple cell types, including lens progenitor cells, emerge and undergo initial tissue formation. Extracellular signaling and DNA-binding transcription factors govern lens and optic cup morphogenesis. Pax6, c-Maf, Hsf4, Prox1, Sox1, and a few additional factors regulate the expression of the lens structural proteins, the crystallins. Extensive crosstalk between a diverse array of signaling pathways controls the complexity and order of lens morphogenetic processes and lens transparency.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
19
|
Abstract
Specialized areas in the vertebrate retina are critical for high-acuity vision, yet the molecular mechanisms driving the development of high-acuity areas (HAAs) remain largely unknown. In Developmental Cell, da Silva and Cepko (2017) show that restricted degradation of retinoic acid and elevated FGF8 signaling give rise to the chick HAA.
Collapse
Affiliation(s)
- Adele Tufford
- Cellular Neurobiology Research Unit, Montreal Clinical Research Institute (IRCM), Montreal, QC H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B4, Canada
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Montreal Clinical Research Institute (IRCM), Montreal, QC H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B4, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
20
|
Lai MS, Wang CY, Yang SH, Wu CC, Sun HS, Tsai SJ, Chuang JI, Chen YC, Huang BM. The expression profiles of fibroblast growth factor 9 and its receptors in developing mice testes. Organogenesis 2016; 12:61-77. [PMID: 27078042 DOI: 10.1080/15476278.2016.1171448] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
An expressional lack of fibroblast growth factor 9 (FGF9) would cause male-to-female sex reversal in the mouse, implying the essential role of FGF9 in testicular organogenesis and maturation. However, the temporal expression of FGF9 and its receptors during testicular development remains elusive. In this study, immunohistochemistry was used to identify the localization of FGF9 and its receptors at different embryonic and postnatal stages in mice testes. Results showed that FGF9 continuously expressed in the testis during development. FGF9 had highest expression in the interstitial region at 17-18 d post coitum (dpc) and in the spermatocytes, spermatids and Leydig cell on postnatal days (pnd) 35-65. Regarding receptor expression, FGFR1 and FGFR4 were evenly expressed in the whole testis during the embryonic and postnatal stages. However, FGFR2 and FGFR3 were widely expressed during the embryonic testis development with higher FGFR2 expression in seminiferous tubules at 16-18 dpc and higher FGFR3 expression in interstitial region at 17-18 dpc. In postnatal stage, FGFR2 extensively expressed with higher expression at spermatids and Leydig cells on 35-65 pnd and FGFR3 widely expressed in the whole testis. Taken together, these results strongly suggest that FGF9 is correlated with the temporal expression profiles of FGFR2 and FGFR3 and possibly associated with testis development.
Collapse
Affiliation(s)
- Meng-Shao Lai
- a Institute of Basic Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China
| | - Chia-Yih Wang
- a Institute of Basic Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China.,b Department of Cell Biology and Anatomy , College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China
| | - Shang-Hsun Yang
- a Institute of Basic Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China.,c Department of Physiology , College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China
| | - Chia-Ching Wu
- a Institute of Basic Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China.,b Department of Cell Biology and Anatomy , College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China
| | - H Sunny Sun
- a Institute of Basic Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China.,d Institute of Molecular Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China
| | - Shaw-Jenq Tsai
- a Institute of Basic Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China.,c Department of Physiology , College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China
| | - Jih-Ing Chuang
- a Institute of Basic Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China.,c Department of Physiology , College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China
| | - Yung-Chia Chen
- e Department of Anatomy , School of Medicine, Kaohsiung Medical University , Kaohsiung , Taiwan , Republic of China
| | - Bu-Miin Huang
- a Institute of Basic Medicine, College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China.,b Department of Cell Biology and Anatomy , College of Medicine, National Cheng Kung University , Tainan , Taiwan , Republic of China
| |
Collapse
|
21
|
Chaffee BR, Hoang TV, Leonard MR, Bruney DG, Wagner BD, Dowd JR, Leone G, Ostrowski MC, Robinson ML. FGFR and PTEN signaling interact during lens development to regulate cell survival. Dev Biol 2016; 410:150-163. [PMID: 26764128 DOI: 10.1016/j.ydbio.2015.12.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 12/31/2015] [Accepted: 12/31/2015] [Indexed: 12/13/2022]
Abstract
Lens epithelial cells express many receptor tyrosine kinases (RTKs) that stimulate PI3K-AKT and RAS-RAF-MEK-ERK intracellular signaling pathways. These pathways ultimately activate the phosphorylation of key cellular transcription factors and other proteins that control proliferation, survival, metabolism, and differentiation in virtually all cells. Among RTKs in the lens, only stimulation of fibroblast growth factor receptors (FGFRs) elicits a lens epithelial cell to fiber cell differentiation response in mammals. Moreover, although the lens expresses three different Fgfr genes, the isolated removal of Fgfr2 at the lens placode stage inhibits both lens cell survival and fiber cell differentiation. Phosphatase and tensin homolog (PTEN), commonly known as a tumor suppressor, inhibits ERK and AKT activation and initiates both apoptotic pathways, and cell cycle arrest. Here, we show that the combined deletion of Fgfr2 and Pten rescues the cell death phenotype associated with Fgfr2 loss alone. Additionally, Pten removal increased AKT and ERK activation, above the levels of controls, in the presence or absence of Fgfr2. However, isolated deletion of Pten failed to stimulate ectopic fiber cell differentiation, and the combined deletion of Pten and Fgfr2 failed to restore differentiation-specific Aquaporin0 and DnaseIIβ expression in the lens fiber cells.
Collapse
Affiliation(s)
- Blake R Chaffee
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Thanh V Hoang
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Melissa R Leonard
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Devin G Bruney
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Brad D Wagner
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Joseph Richard Dowd
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Gustavo Leone
- Department of Molecular Virology, Immunology and Medical Genetics, Department of Molecular Genetics, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Michael C Ostrowski
- Department of Molecular Virology, Immunology and Medical Genetics, Department of Molecular Genetics, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Michael L Robinson
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA.
| |
Collapse
|
22
|
Lovicu FJ, Shin EH, McAvoy JW. Fibrosis in the lens. Sprouty regulation of TGFβ-signaling prevents lens EMT leading to cataract. Exp Eye Res 2015; 142:92-101. [PMID: 26003864 DOI: 10.1016/j.exer.2015.02.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/22/2015] [Accepted: 02/03/2015] [Indexed: 12/22/2022]
Abstract
Cataract is a common age-related condition that is caused by progressive clouding of the normally clear lens. Cataract can be effectively treated by surgery; however, like any surgery, there can be complications and the development of a secondary cataract, known as posterior capsule opacification (PCO), is the most common. PCO is caused by aberrant growth of lens epithelial cells that are left behind in the capsular bag after surgical removal of the fiber mass. An epithelial-to-mesenchymal transition (EMT) is central to fibrotic PCO and forms of fibrotic cataract, including anterior/posterior polar cataracts. Transforming growth factor β (TGFβ) has been shown to induce lens EMT and consequently research has focused on identifying ways of blocking its action. Intriguingly, recent studies in animal models have shown that EMT and cataract developed when a class of negative-feedback regulators, Sprouty (Spry)1 and Spry2, were conditionally deleted from the lens. Members of the Spry family act as general antagonists of the receptor tyrosine kinase (RTK)-mediated MAPK signaling pathway that is involved in many physiological and developmental processes. As the ERK/MAPK signaling pathway is a well established target of Spry proteins, and overexpression of Spry can block aberrant TGFβ-Smad signaling responsible for EMT and anterior subcapsular cataract, this indicates a role for the ERK/MAPK pathway in TGFβ-induced EMT. Given this and other supporting evidence, a case is made for focusing on RTK antagonists, such as Spry, for cataract prevention. In addition, and looking to the future, this review also looks at possibilities for supplanting EMT with normal fiber differentiation and thereby promoting lens regenerative processes after cataract surgery. Whilst it is now known that the epithelial to fiber differentiation process is driven by FGF, little is known about factors that coordinate the precise assembly of fibers into a functional lens. However, recent research provides key insights into an FGF-activated mechanism intrinsic to the lens that involves interactions between the Wnt-Frizzled and Jagged/Notch signaling pathways. This reciprocal epithelial-fiber cell interaction appears to be critical for the assembly and maintenance of the highly ordered three-dimensional architecture that is central to lens function. This information is fundamental to defining the specific conditions and stimuli needed to recapitulate developmental programs and promote regeneration of lens structure and function after cataract surgery.
Collapse
Affiliation(s)
- F J Lovicu
- Discipline of Anatomy and Histology, Bosch Institute, School of Medical Sciences, University of Sydney, 2006, NSW, Australia; Save Sight Institute, University of Sydney, Sydney 2001, NSW, Australia.
| | - E H Shin
- Discipline of Anatomy and Histology, Bosch Institute, School of Medical Sciences, University of Sydney, 2006, NSW, Australia
| | - J W McAvoy
- Save Sight Institute, University of Sydney, Sydney 2001, NSW, Australia
| |
Collapse
|
23
|
Boswell BA, Musil LS. Synergistic interaction between the fibroblast growth factor and bone morphogenetic protein signaling pathways in lens cells. Mol Biol Cell 2015; 26:2561-72. [PMID: 25947138 PMCID: PMC4571308 DOI: 10.1091/mbc.e15-02-0117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/01/2015] [Indexed: 12/12/2022] Open
Abstract
Relatively little is known about how receptor tyrosine kinase ligands can positively cooperate with BMP signaling. Primary cultures of lens cells were used to reveal an unprecedented type of cross-talk between the canonical FGF and BMP signaling pathways that regulates lens cell differentiation and intercellular coupling. Fibroblast growth factors (FGFs) play a central role in two processes essential for lens transparency—fiber cell differentiation and gap junction–mediated intercellular communication (GJIC). Using serum-free primary cultures of chick lens epithelial cells (DCDMLs), we investigated how the FGF and bone morphogenetic protein (BMP) signaling pathways positively cooperate to regulate lens development and function. We found that culturing DCDMLs for 6 d with the BMP blocker noggin inhibits the canonical FGF-to-ERK pathway upstream of FRS2 activation and also prevents FGF from stimulating FRS2- and ERK-independent gene expression, indicating that BMP signaling is required at the level of FGF receptors. Other experiments revealed a second type of BMP/FGF interaction by which FGF promotes expression of BMP target genes as well as of BMP4. Together these studies reveal a novel mode of cooperation between the FGF and BMP pathways in which BMP keeps lens cells in an optimally FGF-responsive state and, reciprocally, FGF enhances BMP-mediated gene expression. This interaction provides a mechanistic explanation for why disruption of either FGF or BMP signaling in the lens leads to defects in lens development and function.
Collapse
Affiliation(s)
- Bruce A Boswell
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239
| | - Linda S Musil
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239 )
| |
Collapse
|
24
|
Lai MS, Cheng YS, Chen PR, Tsai SJ, Huang BM. Fibroblast growth factor 9 activates akt and MAPK pathways to stimulate steroidogenesis in mouse leydig cells. PLoS One 2014; 9:e90243. [PMID: 24603862 PMCID: PMC3946167 DOI: 10.1371/journal.pone.0090243] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 01/27/2014] [Indexed: 12/22/2022] Open
Abstract
Fibroblast growth factor 9 (FGF9) is a multifunctional polypeptide belonging to the FGF family and has functions related to bone formation, lens-fiber differentiation, nerve development, gap-junction formation and sex determination. In a previous study, we demonstrated that FGF9 stimulates the production of testosterone in mouse Leydig cells. In the present study, we used both primary mouse Leydig cells and MA-10 mouse Leydig tumor cells to further investigate the molecular mechanism of FGF9-stimulated steroidogenesis. Results showed that FGF9 significantly activated steroidogenesis in both mouse primary and tumor Leydig cells (p<0.05). Furthermore, FGF9 significantly induced the expression of phospho-Akt at 0.5 and 24 hr, phospho-JNK at 0.25, 0.5, and 24 hr, phospho-p38 at 0.5 hr, and phospho-ERK1/2 from 0.25 to 24 hr in primary Leydig cells (p<0.05). Also, FGF9 significantly up-regulated the expression of phospho-Akt at 3 hr, phospho-JNK at 0.25 hr, and phospho-ERK1/2 at 1 and 3 hr in MA-10 cells (p<0.05). Using specific inhibitors of Akt, JNK, p38, and ERK1/2, we further demonstrated that the inhibitors of Akt and ERK1/2 significantly suppressed the stimulatory effect of FGF9 on steroidogenesis in mouse Leydig cells. In conclusion, FGF9 specifically activated the Akt and ERK1/2 in normal mouse Leydig cells and the Akt, JNK and ERK1/2 in MA-10 mouse Leydig tumor cells to stimulate steroidogenesis.
Collapse
Affiliation(s)
- Meng-Shao Lai
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Yu-Sheng Cheng
- Department of Urology, National Cheng Kung University Hospital Douliou Branch, Yunlin, Taiwan, Republic of China
| | - Pei-Rong Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Shaw-Jenq Tsai
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Bu-Miin Huang
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
25
|
Song X, Tanaka H, Ohta K. Multiple roles of Equarin during lens development. Dev Growth Differ 2014; 56:199-205. [DOI: 10.1111/dgd.12121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 01/07/2014] [Accepted: 01/07/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Xiaohong Song
- Division of Developmental Neurobiology Faculty of Life Sciences Kumamoto University 1‐1‐1 Honjo Chuo‐ku Kumamoto 860‐8556 Japan
| | - Hideaki Tanaka
- Division of Developmental Neurobiology Faculty of Life Sciences Kumamoto University 1‐1‐1 Honjo Chuo‐ku Kumamoto 860‐8556 Japan
| | - Kunimasa Ohta
- Division of Developmental Neurobiology Faculty of Life Sciences Kumamoto University 1‐1‐1 Honjo Chuo‐ku Kumamoto 860‐8556 Japan
| |
Collapse
|
26
|
Stem Cells and the Ocular Lens: Implications for Cataract Research and Therapy. STEM CELL BIOLOGY AND REGENERATIVE MEDICINE 2014. [DOI: 10.1007/978-1-4939-0787-8_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Li H, Tao C, Cai Z, Hertzler-Schaefer K, Collins TN, Wang F, Feng GS, Gotoh N, Zhang X. Frs2α and Shp2 signal independently of Gab to mediate FGF signaling in lens development. J Cell Sci 2013; 127:571-82. [PMID: 24284065 DOI: 10.1242/jcs.134478] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Fibroblast growth factor (FGF) signaling requires a plethora of adaptor proteins to elicit downstream responses, but the functional significances of these docking proteins remain controversial. In this study, we used lens development as a model to investigate Frs2α and its structurally related scaffolding proteins, Gab1 and Gab2, in FGF signaling. We show that genetic ablation of Frs2α alone has a modest effect, but additional deletion of tyrosine phosphatase Shp2 causes a complete arrest of lens vesicle development. Biochemical evidence suggests that this Frs2α-Shp2 synergy reflects their epistatic relationship in the FGF signaling cascade, as opposed to compensatory or parallel functions of these two proteins. Genetic interaction experiments further demonstrate that direct binding of Shp2 to Frs2α is necessary for activation of ERK signaling, whereas constitutive activation of either Shp2 or Kras signaling can compensate for the absence of Frs2α in lens development. By contrast, knockout of Gab1 and Gab2 failed to disrupt FGF signaling in vitro and lens development in vivo. These results establish the Frs2α-Shp2 complex as the key mediator of FGF signaling in lens development.
Collapse
Affiliation(s)
- Hongge Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Antosova B, Smolikova J, Borkovcova R, Strnad H, Lachova J, Machon O, Kozmik Z. Ectopic activation of Wnt/β-catenin signaling in lens fiber cells results in cataract formation and aberrant fiber cell differentiation. PLoS One 2013; 8:e78279. [PMID: 24205179 PMCID: PMC3813504 DOI: 10.1371/journal.pone.0078279] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 09/09/2013] [Indexed: 12/04/2022] Open
Abstract
The Wnt/β-catenin signaling pathway controls many processes during development, including cell proliferation, cell differentiation and tissue homeostasis, and its aberrant regulation has been linked to various pathologies. In this study we investigated the effect of ectopic activation of Wnt/β-catenin signaling during lens fiber cell differentiation. To activate Wnt/β-catenin signaling in lens fiber cells, the transgenic mouse referred to as αA-CLEF was generated, in which the transactivation domain of β-catenin was fused to the DNA-binding protein LEF1, and expression of the transgene was controlled by αA-crystallin promoter. Constitutive activation of Wnt/β-catenin signaling in lens fiber cells of αA-CLEF mice resulted in abnormal and delayed fiber cell differentiation. Moreover, adult αA-CLEF mice developed cataract, microphthalmia and manifested downregulated levels of γ-crystallins in lenses. We provide evidence of aberrant expression of cell cycle regulators in embryonic lenses of αA-CLEF transgenic mice resulting in the delay in cell cycle exit and in the shift of fiber cell differentiation to the central fiber cell compartment. Our results indicate that precise regulation of the Wnt/β-catenin signaling activity during later stages of lens development is essential for proper lens fiber cell differentiation and lens transparency.
Collapse
Affiliation(s)
- Barbora Antosova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- Department of Genetics and Microbiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Jana Smolikova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Romana Borkovcova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Hynek Strnad
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jitka Lachova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Ondrej Machon
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Zbynek Kozmik
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
29
|
Wolf L, Harrison W, Huang J, Xie Q, Xiao N, Sun J, Kong L, Lachke SA, Kuracha MR, Govindarajan V, Brindle PK, Ashery-Padan R, Beebe DC, Overbeek PA, Cvekl A. Histone posttranslational modifications and cell fate determination: lens induction requires the lysine acetyltransferases CBP and p300. Nucleic Acids Res 2013; 41:10199-214. [PMID: 24038357 PMCID: PMC3905850 DOI: 10.1093/nar/gkt824] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
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.
Collapse
Affiliation(s)
- Louise Wolf
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children's Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Henry JJ, Thomas AG, Hamilton PW, Moore L, Perry KJ. Cell signaling pathways in vertebrate lens regeneration. Curr Top Microbiol Immunol 2013; 367:75-98. [PMID: 23224710 DOI: 10.1007/82_2012_289] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Certain vertebrates are capable of regenerating parts of the eye, including the lens. Depending on the species, two principal forms of in vivo lens regeneration have been described wherein the new lens arises from either the pigmented epithelium of the dorsal iris or the cornea epithelium. These forms of lens regeneration are triggered by retinal factors present in the eye. Studies have begun to illuminate the nature of the signals that support lens regeneration. This review describes evidence for the involvement of specific signaling pathways in lens regeneration, including the FGF, retinoic acid, TGF-beta, Wnt, and Hedgehog pathways.
Collapse
Affiliation(s)
- Jonathan J Henry
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61801, USA.
| | | | | | | | | |
Collapse
|
31
|
Madakashira BP, Kobrinski DA, Hancher AD, Arneman EC, Wagner BD, Wang F, Shin H, Lovicu FJ, Reneker LW, Robinson ML. Frs2α enhances fibroblast growth factor-mediated survival and differentiation in lens development. Development 2012; 139:4601-12. [PMID: 23136392 DOI: 10.1242/dev.081737] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Most growth factor receptor tyrosine kinases (RTKs) signal through similar intracellular pathways, but they often have divergent biological effects. Therefore, elucidating the mechanism of channeling the intracellular effect of RTK stimulation to facilitate specific biological responses represents a fundamental biological challenge. Lens epithelial cells express numerous RTKs with the ability to initiate the phosphorylation (activation) of Erk1/2 and PI3-K/Akt signaling. However, only Fgfr stimulation leads to lens fiber cell differentiation in the developing mammalian embryo. Additionally, within the lens, only Fgfrs activate the signal transduction molecule Frs2α. Loss of Frs2α in the lens significantly increases apoptosis and decreases phosphorylation of both Erk1/2 and Akt. Also, Frs2α deficiency decreases the expression of several proteins characteristic of lens fiber cell differentiation, including Prox1, p57(KIP2), aquaporin 0 and β-crystallins. Although not normally expressed in the lens, the RTK TrkC phosphorylates Frs2α in response to binding the ligand NT3. Transgenic lens epithelial cells expressing both TrkC and NT3 exhibit several features characteristic of lens fiber cells. These include elongation, increased Erk1/2 and Akt phosphorylation, and the expression of β-crystallins. All these characteristics of NT3-TrkC transgenic lens epithelial cells depend on Frs2α. Therefore, tyrosine phosphorylation of Frs2α mediates Fgfr-dependent lens cell survival and provides a mechanistic basis for the unique fiber-differentiating capacity of Fgfs on mammalian lens epithelial cells.
Collapse
|
32
|
New insights into the mechanism of lens development using zebra fish. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 296:1-61. [PMID: 22559937 DOI: 10.1016/b978-0-12-394307-1.00001-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
On the basis of recent advances in molecular biology, genetics, and live-embryo imaging, direct comparisons between zebra fish and human lens development are being made. The zebra fish has numerous experimental advantages for investigation of fundamental biomedical problems that are often best studied in the lens. The physical characteristics of visible light can account for the highly coordinated cell differentiation during formation of a beautifully transparent, refractile, symmetric optical element, the biological lens. The accessibility of the zebra fish lens for direct investigation during rapid development will result in new knowledge about basic functional mechanisms of epithelia-mesenchymal transitions, cell fate, cell-matrix interactions, cytoskeletal interactions, cytoplasmic crowding, membrane transport, cell adhesion, cell signaling, and metabolic specialization. The lens is well known as a model for characterization of cell and molecular aging. We review the recent advances in understanding vertebrate lens development conducted with zebra fish.
Collapse
|
33
|
Sousounis K, Tsonis PA. Patterns of gene expression in microarrays and expressed sequence tags from normal and cataractous lenses. Hum Genomics 2012; 6:14. [PMID: 23244575 PMCID: PMC3563465 DOI: 10.1186/1479-7364-6-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/14/2012] [Indexed: 11/30/2022] Open
Abstract
In this contribution, we have examined the patterns of gene expression in normal and cataractous lenses as presented in five different papers using microarrays and expressed sequence tags. The purpose was to evaluate unique and common patterns of gene expression during development, aging and cataracts.
Collapse
Affiliation(s)
- Konstantinos Sousounis
- Department of Biology and Center for Tissue Regeneration and Engineering, University of Dayton, Dayton, OH 45469-2320, USA
| | | |
Collapse
|
34
|
Equarin is involved as an FGF signaling modulator in chick lens differentiation. Dev Biol 2012; 368:109-17. [DOI: 10.1016/j.ydbio.2012.05.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/28/2012] [Accepted: 05/23/2012] [Indexed: 11/20/2022]
|
35
|
Primary cultures of embryonic chick lens cells as a model system to study lens gap junctions and fiber cell differentiation. J Membr Biol 2012; 245:357-68. [PMID: 22797938 DOI: 10.1007/s00232-012-9458-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/20/2012] [Indexed: 01/31/2023]
Abstract
A major limitation in lens gap junction research has been the lack of experimentally tractable ex vivo systems to study the formation and regulation of fiber-type gap junctions. Although immortalized lens-derived cell lines are amenable to both gene transfection and siRNA-mediated knockdown, to our knowledge none are capable of undergoing appreciable epithelial-to-fiber differentiation. Lens central epithelial explants have the converse limitation. A key advance in the field was the development of a primary embryonic chick lens cell culture system by Drs. Sue Menko and Ross Johnson. Unlike central epithelial explants, these cultures also include cells from the peripheral (preequatorial and equatorial) epithelium, which is the most physiologically relevant population for the study of fiber-type gap junction formation. We have modified the Menko/Johnson system and refer to our cultures as dissociated cell-derived monolayer cultures (DCDMLs). We culture DCDMLs without serum to mimic the avascular lens environment and on laminin, the major matrix component of the lens capsule. Here, I review the features of the DCDML system and how we have used it to study lens gap junctions and fiber cell differentiation. Our results demonstrate the power of DCDMLs to generate new findings germane to the mammalian lens and how these cultures can be exploited to conduct experiments that would be impossible, prohibitively expensive and/or difficult to interpret using transgenic animals in vivo.
Collapse
|
36
|
Jarrin M, Pandit T, Gunhaga L. A balance of FGF and BMP signals regulates cell cycle exit and Equarin expression in lens cells. Mol Biol Cell 2012; 23:3266-74. [PMID: 22718906 PMCID: PMC3418319 DOI: 10.1091/mbc.e12-01-0075] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The roles of BMP and FGF during the transition of proliferating lens epithelial cells to differentiated primary lens fiber cells are examined. The results show that proliferation, cell cycle exit, and early differentiation of primary lens fiber cells are regulated by counterbalancing BMP and FGF signals. In embryonic and adult lenses, a balance of cell proliferation, cell cycle exit, and differentiation is necessary to maintain physical function. The molecular mechanisms regulating the transition of proliferating lens epithelial cells to differentiated primary lens fiber cells are poorly characterized. To investigate this question, we used gain- and loss-of-function analyses to modulate fibroblast growth factor (FGF) and/or bone morphogenetic protein (BMP) signals in chick lens/retina explants. Here we show that FGF activity plays a key role for proliferation independent of BMP signals. Moreover, a balance of FGF and BMP signals regulates cell cycle exit and the expression of Ccdc80 (also called Equarin), which is expressed at sites where differentiation of lens fiber cells occurs. BMP activity promotes cell cycle exit and induces Equarin expression in an FGF-dependent manner. In contrast, FGF activity is required but not sufficient to induce cell cycle exit or Equarin expression. Furthermore, our results show that in the absence of BMP activity, lens cells have increased cell cycle length or are arrested in the cell cycle, which leads to decreased cell cycle exit. Taken together, these findings suggest that proliferation, cell cycle exit, and early differentiation of primary lens fiber cells are regulated by counterbalancing BMP and FGF signals.
Collapse
Affiliation(s)
- Miguel Jarrin
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
37
|
The lens in focus: a comparison of lens development in Drosophila and vertebrates. Mol Genet Genomics 2011; 286:189-213. [PMID: 21877135 DOI: 10.1007/s00438-011-0643-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/04/2011] [Indexed: 12/24/2022]
Abstract
The evolution of the eye has been a major subject of study dating back centuries. The advent of molecular genetics offered the surprising finding that morphologically distinct eyes rely on conserved regulatory gene networks for their formation. While many of these advances often stemmed from studies of the compound eye of the fruit fly, Drosophila melanogaster, and later translated to discoveries in vertebrate systems, studies on vertebrate lens development far outnumber those in Drosophila. This may be largely historical, since Spemann and Mangold's paradigm of tissue induction was discovered in the amphibian lens. Recent studies on lens development in Drosophila have begun to define molecular commonalities with the vertebrate lens. Here, we provide an overview of Drosophila lens development, discussing intrinsic and extrinsic factors controlling lens cell specification and differentiation. We then summarize key morphological and molecular events in vertebrate lens development, emphasizing regulatory factors and networks strongly associated with both systems. Finally, we provide a comparative analysis that highlights areas of research that would help further clarify the degree of conservation between the formation of dioptric systems in invertebrates and vertebrates.
Collapse
|
38
|
Puk O, Möller G, Geerlof A, Krowiorz K, Ahmad N, Wagner S, Adamski J, de Angelis MH, Graw J. The pathologic effect of a novel neomorphic Fgf9(Y162C) allele is restricted to decreased vision and retarded lens growth. PLoS One 2011; 6:e23678. [PMID: 21858205 PMCID: PMC3157460 DOI: 10.1371/journal.pone.0023678] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 07/25/2011] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factor (Fgf) signalling plays a crucial role in many developmental processes. Among the Fgf pathway ligands, Fgf9 (UniProt: P54130) has been demonstrated to participate in maturation of various organs and tissues including skeleton, testes, lung, heart, and eye. Here we establish a novel Fgf9 allele, discovered in a dominant N-ethyl-N-nitrosourea (ENU) screen for eye-size abnormalities using the optical low coherence interferometry technique. The underlying mouse mutant line Aca12 was originally identified because of its significantly reduced lens thickness. Linkage studies located Aca12 to chromosome 14 within a 3.6 Mb spanning interval containing the positional candidate genes Fgf9 (MGI: 104723), Gja3 (MGI: 95714), and Ift88 (MGI: 98715). While no sequence differences were found in Gja3 and Ift88, we identified an A→G missense mutation at cDNA position 770 of the Fgf9 gene leading to an Y162C amino acid exchange. In contrast to previously described Fgf9 mutants, Fgf9Y162C carriers were fully viable and did not reveal reduced body-size, male-to-female sexual reversal or skeletal malformations. The histological analysis of the retina as well as its basic functional characterization by electroretinography (ERG) did not show any abnormality. However, the analysis of head-tracking response of the Fgf9Y162C mutants in a virtual drum indicated a gene-dosage dependent vision loss of almost 50%. The smaller lenses in Fgf9Y162C suggested a role of Fgf9 during lens development. Histological investigations showed that lens growth retardation starts during embryogenesis and continues after birth. Young Fgf9Y162C lenses remained transparent but developed age-related cataracts. Taken together, Fgf9Y162C is a novel neomorphic allele that initiates microphakia and reduced vision without effects on organs and tissues outside the eye. Our data point to a role of Fgf9 signalling in primary and secondary lens fiber cell growth. The results underline the importance of allelic series to fully understand multiple functions of a gene.
Collapse
MESH Headings
- Alleles
- Amino Acid Sequence
- Animals
- Base Sequence
- Binding, Competitive
- Cataract/genetics
- Female
- Fibroblast Growth Factor 9/chemistry
- Fibroblast Growth Factor 9/genetics
- Fibroblast Growth Factor 9/metabolism
- Genotype
- Haplotypes
- Heparin/metabolism
- Lens, Crystalline/embryology
- Lens, Crystalline/growth & development
- Lens, Crystalline/metabolism
- Male
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Models, Molecular
- Molecular Sequence Data
- Mutation, Missense
- Protein Binding
- Protein Structure, Tertiary
- Sequence Homology, Amino Acid
- Vision, Ocular/genetics
- Visual Acuity/genetics
Collapse
Affiliation(s)
- Oliver Puk
- German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sugiyama Y, Lovicu FJ, McAvoy JW. Planar cell polarity in the mammalian eye lens. Organogenesis 2011; 7:191-201. [PMID: 22027540 DOI: 10.4161/org.7.3.18421] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The major role of the eye lens is to transmit and focus images onto the retina. For this function, the lens needs to develop and maintain the correct shape, notably, the precise curvature and high-level order and organization of its elements. The lens is mainly comprised of highly elongated fiber cells with hexagonal cross-sectional profiles that facilitate regular packing. Collectively, they form concentrically arranged layers around the anterior-posterior polar axis, and their convex curvature contributes to the spheroidal shape of the lens. Although the lens has been a popular system for developmental studies, little is known about the mechanism(s) that underlies the development of its exquisite three-dimensional cellular architecture. In this review, we will describe our recent work, which shows how planar cell polarity (PCP) operates in lens and contributes to its morphogenesis. We believe that the lens will be a useful model system to study PCP in general and gain insights into mechanisms that generate high-level cellular order during development.
Collapse
|
40
|
Lovicu FJ, McAvoy JW, de Iongh RU. Understanding the role of growth factors in embryonic development: insights from the lens. Philos Trans R Soc Lond B Biol Sci 2011; 366:1204-18. [PMID: 21402581 PMCID: PMC3061110 DOI: 10.1098/rstb.2010.0339] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Growth factors play key roles in influencing cell fate and behaviour during development. The epithelial cells and fibre cells that arise from the lens vesicle during lens morphogenesis are bathed by aqueous and vitreous, respectively. Vitreous has been shown to generate a high level of fibroblast growth factor (FGF) signalling that is required for secondary lens fibre differentiation. However, studies also show that FGF signalling is not sufficient and roles have been identified for transforming growth factor-β and Wnt/Frizzled families in regulating aspects of fibre differentiation. In the case of the epithelium, key roles for Wnt/β-catenin and Notch signalling have been demonstrated in embryonic development, but it is not known if other factors are required for its formation and maintenance. This review provides an overview of current knowledge about growth factor regulation of differentiation and maintenance of lens cells. It also highlights areas that warrant future study.
Collapse
Affiliation(s)
- F. J. Lovicu
- Discipline of Anatomy and Histology, Bosch Institute, University of Sydney, NSW 2006, Australia
- Save Sight Institute, University of Sydney, Sydney, NSW 2001, Australia
- Vision Cooperative Research Centre, Sydney, Australia
| | - J. W. McAvoy
- Save Sight Institute, University of Sydney, Sydney, NSW 2001, Australia
- Vision Cooperative Research Centre, Sydney, Australia
| | - R. U. de Iongh
- Anatomy and Cell Biology, University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
41
|
Qu X, Hertzler K, Pan Y, Grobe K, Robinson ML, Zhang X. Genetic epistasis between heparan sulfate and FGF-Ras signaling controls lens development. Dev Biol 2011; 355:12-20. [PMID: 21536023 DOI: 10.1016/j.ydbio.2011.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/14/2011] [Accepted: 04/01/2011] [Indexed: 02/06/2023]
Abstract
Vertebrate lens development depends on a complex network of signaling molecules to coordinate cell proliferation, migration and differentiation. In this study, we have investigated the role of heparan sulfate in lens specific signaling by generating a conditional ablation of heparan sulfate modification genes, Ndst1 and Ndst2. In this mutant, N-sulfation of heparan sulfate was disrupted after the lens induction stage, resulting in reduced lens cell proliferation, increased cell death and defective lens fiber differentiation in later lens development. The loss of Ndst function also prevented the assembly of Fgf/Fgfr complexes on the lens cell surface and disrupted ERK signaling within the lens. We further demonstrated that Ndst mutation completely inhibited the FGF1 and Fgf3 overexpression phenotypes, but Kras reactivation was sufficient to reverse the Ndst deficient lens differentiation defect. The epistatic relationship between Ndst and FGF-Ras signaling demonstrates that FGF signaling is the predominant signaling pathway controlled by Ndst in lens development.
Collapse
Affiliation(s)
- Xiuxia Qu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | |
Collapse
|
42
|
Weigelt C, Haas R, Kobbe G. Pharmacokinetic evaluation of palifermin for mucosal protection from chemotherapy and radiation. Expert Opin Drug Metab Toxicol 2011; 7:505-15. [PMID: 21417820 DOI: 10.1517/17425255.2011.566556] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Oral mucositis, one of the major side effects of chemotherapy and irradiation, is still a burden of modern oncology. The keratinocyte growth factor (KGF) palifermin has been approved as a new, targeted therapy for the prevention of severe oral mucositis. AREAS COVERED The authors review the literature on pharmacokintetics and clinical use of palifermin in patients with hematological malignancies and solid tumors for the prevention of chemo- and radiation-induced mucositis by using the PubMed database and additional literature where applicable. The article includes in vitro data, clinical trials as well as case reports regarding dosage, administration schedule, efficacy and adverse events. EXPERT OPINION There is sufficient data for a beneficial effect of palifermin prophylaxis for patients with hematological cancers receiving high-dose chemotherapy and total body irradiation as well as patients with head and neck cancer receiving combined irradiation and chemotherapy. In less mucotoxic regimens, dose and schedule of palifermin to achieve protection from mucositis are less well defined. The balance of benefit and unwanted effects has to be evaluated and weighed for individual chemotherapy regimens and patient groups. Further research on the prevention of mucositis should aim to determine the patient's individual risk to develop severe mucositis.
Collapse
Affiliation(s)
- Christian Weigelt
- Department of Hematology, Oncology and Clinical Immunology of the University Hospital of Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | | | | |
Collapse
|
43
|
Martinez G, de Iongh R. The lens epithelium in ocular health and disease. Int J Biochem Cell Biol 2010; 42:1945-63. [PMID: 20883819 DOI: 10.1016/j.biocel.2010.09.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 09/19/2010] [Accepted: 09/20/2010] [Indexed: 01/11/2023]
|
44
|
Mailankot M, Howell S, Nagaraj RH. Kynurenine inhibits fibroblast growth factor 2-mediated expression of crystallins and MIP26 in lens epithelial cells. Biochim Biophys Acta Mol Basis Dis 2010; 1802:609-20. [PMID: 20478381 DOI: 10.1016/j.bbadis.2010.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 04/16/2010] [Accepted: 05/10/2010] [Indexed: 10/19/2022]
Abstract
Fibroblast growth factor-2 (FGF2)-mediated signaling plays an important role in fiber cell differentiation in eye lens. We had previously shown that kynurenine (KYN) produced from the overexpression of indoleamine 2,3-dioxygenase (IDO) causes defects in the differentiation of fiber cells, induces fiber cell apoptosis and cataract formation in the mouse lens, and leads to cell cycle arrest in cultured mouse lens epithelial cells (mLEC). In this study, we demonstrate that exogenous KYN reduces FGF2-mediated expression of alpha-, beta-, and gamma-crystallin and MIP26 in mLEC. We show that endogenously produced KYN in mLEC of IDO transgenic animals causes similar defects in FGF2-induced protein expression and that a competitive inhibitor of IDO prevents such defects. Our data also show that KYN inhibits FGF2-induced Akt and ERK1/2 phosphorylation in mLEC, which are required for crystallin and MIP26 expression in the lens. KYN does not inhibit FGF2 binding to cells but inhibit phosphorylation of FGFR1in mLEC. Together our data suggest that KYN might inhibit FGF2-mediated fiber cell differentiation by preventing expression of crystallins and MIP26. Our studies provide a novel mechanism by which KYN can exert deleterious effects in cells.
Collapse
Affiliation(s)
- Maneesh Mailankot
- Department of Ophthalmology & Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
45
|
Pan Y, Carbe C, Powers A, Feng GS, Zhang X. Sprouty2-modulated Kras signaling rescues Shp2 deficiency during lens and lacrimal gland development. Development 2010; 137:1085-93. [PMID: 20215346 DOI: 10.1242/dev.042820] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Shp2/Ptpn11 tyrosine phosphatase is a general regulator of the RTK pathways. By genetic ablation, we demonstrate that Shp2 is required for lacrimal gland budding, lens cell proliferation, survival and differentiation. Shp2 deletion disrupted ERK signaling and cell cycle regulation, which could be partially compensated by activated Kras signaling, confirming that Ras signaling was the main downstream target of Shp2 in lens and lacrimal gland development. We also showed that Sprouty2, a general suppressor of Ras signaling, was regulated by Shp2 positively at the transcriptional level and negatively at the post-translational level. Only in the absence of Sprouty2 could activated Kras signaling robustly rescue the lens proliferation and lacrimal-gland-budding defects in the Shp2 mutants. We propose that the dynamic regulation of Sprouty by Shp2 might be important not only for modulating Ras signaling in lens and lacrimal gland development, but also for RTK signaling in general.
Collapse
Affiliation(s)
- Yi Pan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
46
|
West-Mays JA, Pino G, Lovicu FJ. Development and use of the lens epithelial explant system to study lens differentiation and cataractogenesis. Prog Retin Eye Res 2010; 29:135-43. [PMID: 20006728 PMCID: PMC2964862 DOI: 10.1016/j.preteyeres.2009.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Over the last two decades much progress has been made in identifying and characterizing many of the molecules involved in understanding normal lens biology and its pathology. Much of this has been made possible through the establishment and use of the lens epithelial explant system. This simplistic tissue culture model, comprised of a sheet of lens epithelium on its native substratum, has been used effectively to study many cellular processes, including lens epithelial cell proliferation, fiber cell differentiation, cell apoptosis as well as epithelial-to-mesenchymal transformation of cells. In doing so, a number of key growth factors and cytokines, including members of the FGF, Wnt and TGFbeta family have been shown to play essential roles in many of these cellular events. This has led to further studies exploring the signaling pathways downstream of these molecules in the lens, paving the way for the development of a number of in situ models (primarily transgenic mouse lines) to further explore in more detail the nature of these molecular and cellular interactions. To reciprocate, the lens epithelial explant system is increasingly being used to further characterize the nature of many complex phenotypes and pathologies observed in these in situ models, allowing us to selectively isolate and examine the direct impact of an individual molecule on a specific cellular response in lens cells. There is no question that the lens epithelial explant system has served as a powerful tool to further our understanding of lens biology and pathology, and there is no doubt that it will continue to serve in such a capacity, as new developments are realized and putative treatments for aberrant lens cell behavior are to be trialed.
Collapse
Affiliation(s)
- Judith A West-Mays
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| | | | | |
Collapse
|
47
|
Burgess D, Zhang Y, Siefker E, Vaca R, Kuracha MR, Reneker L, Overbeek PA, Govindarajan V. Activated Ras alters lens and corneal development through induction of distinct downstream targets. BMC DEVELOPMENTAL BIOLOGY 2010; 10:13. [PMID: 20105280 PMCID: PMC2828409 DOI: 10.1186/1471-213x-10-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 01/27/2010] [Indexed: 01/04/2023]
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.
Collapse
Affiliation(s)
- Daniel Burgess
- Department of Surgery, 2500 California Plaza, Creighton University, Omaha, NE 68178, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Rivera C, Yamben IF, Shatadal S, Waldof M, Robinson ML, Griep AE. Cell-autonomous requirements for Dlg-1 for lens epithelial cell structure and fiber cell morphogenesis. Dev Dyn 2009; 238:2292-308. [PMID: 19623611 DOI: 10.1002/dvdy.22036] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Cell polarity and adhesion are thought to be key determinants in organismal development. In Drosophila, discs large (dlg) has emerged as an important regulator of epithelial cell proliferation, adhesion, and polarity. Herein, we investigated the role of the mouse homolog of dlg (Dlg-1) in the development of the mouse ocular lens. Tissue-specific ablation of Dlg-1 throughout the lens early in lens development led to an expansion and disorganization of the epithelium that correlated with changes in the distribution of adhesion and polarity factors. In the fiber cells, differentiation defects were observed. These included alterations in cell structure and the disposition of cell adhesion/cytoskeletal factors, delay in denucleation, and reduced levels of alpha-catenin, pERK1/2, and MIP26. These fiber cell defects were recapitulated when Dlg-1 was disrupted only in fiber cells. These results suggest that Dlg-1 acts in a cell autonomous manner to regulate epithelial cell structure and fiber cell differentiation.
Collapse
Affiliation(s)
- Charlene Rivera
- Department of Anatomy, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706, USA
| | | | | | | | | | | |
Collapse
|
49
|
Lin YM, Tsai CC, Chung CL, Chen PR, Sunny Sun H, Tsai SJ, Huang BM. Fibroblast growth factor 9 stimulates steroidogenesis in postnatal Leydig cells. ACTA ACUST UNITED AC 2009; 33:545-53. [DOI: 10.1111/j.1365-2605.2009.00966.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
50
|
Saravanamuthu SS, Gao CY, Zelenka PS. Notch signaling is required for lateral induction of Jagged1 during FGF-induced lens fiber differentiation. Dev Biol 2009; 332:166-76. [PMID: 19481073 DOI: 10.1016/j.ydbio.2009.05.566] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/19/2009] [Accepted: 05/20/2009] [Indexed: 01/08/2023]
Abstract
Previous studies of the developing lens have shown that Notch signaling regulates differentiation of lens fiber cells by maintaining a proliferating precursor pool in the anterior epithelium. However, whether Notch signaling is further required after the onset of fiber cell differentiation is not clear. This work investigates the role of Notch2 and Jagged1 (Jag1) in secondary fiber cell differentiation using rat lens epithelial explants undergoing FGF-2 dependent differentiation in vitro. FGF induced Jag1 expression and Notch2 signaling (as judged by the appearance of activated Notch2 Intracellular Domain (N2ICD)) within 12-24 h. These changes were correlated with induction of the Notch effector, Hes5, upregulation of N-cadherin (N-cad), and downregulation of E-cadherin (E-cad), a cadherin switch characteristic of fiber cell differentiation. Induction of Jag1 was efficiently blocked by U0126, a specific inhibitor of MAPK/ERK signaling, indicating a requirement for signaling through this pathway downstream of the FGF receptor. Other growth factors that activate MAPK/ERK signaling (EGF, PDGF, IGF) did not induce Jag1. Inhibition of Notch signaling using gamma secretase inhibitors DAPT and L-685,458 or anti-Jag1 antibody markedly decreased FGF-dependent expression of Jag1 demonstrating Notch-dependent lateral induction. In addition, inhibition of Notch signaling reduced expression of N-cad, and the cyclin dependent kinase inhibitor, p57Kip2, indicating a direct role for Notch signaling in secondary fiber cell differentiation. These results demonstrate that Notch-mediated lateral induction of Jag1 is an essential component of FGF-dependent lens fiber cell differentiation.
Collapse
Affiliation(s)
- Senthil S Saravanamuthu
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|