1
|
Heffer A, Lee C, Mayernik JP, Holt JC, Kiernan AE. Notch1 is Required to Maintain Supporting Cell Identity and Vestibular Function during Maturation of the Mammalian Balance Organs. J Neurosci 2025; 45:e1365242024. [PMID: 39779370 PMCID: PMC11867012 DOI: 10.1523/jneurosci.1365-24.2024] [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: 07/15/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The inner ear houses both hearing and balance sensory modalities. The hearing and balance organs consist of similar cell types, including sensory hair cells and associated supporting cells. Previously we showed that Notch1 is required for maintaining supporting cell survival during cochlear maturation. To understand the role of Notch during vestibular maturation, we deleted Notch1 from the vestibular organs of both male and female mice at birth. Histological analyses showed a reduction of supporting cells accompanied by an increase in type II hair cells, indicating a conversion of supporting cells to hair cells. Analysis of mature sensory organs indicate the converted hair cells survive, despite a severe reduction of supporting cells. Vestibular sensory evoked potentials (VsEPs), thought to be generated within the striola regions of the maculae, were absent, indicating that NOTCH1 is critical for striolar function. Specialized type I hair cells in the striola failed to develop the complex calyces typical of these cells. Notch1 mutants did not exhibit vestibular behaviors such as circling and head shaking but showed difficulties with tests of balance and swimming. These results indicate that, unlike the cochlea, supporting cells in balance organs retain the plasticity to convert to hair cells which can survive into adulthood. Despite hair cell survival, vestibular function is compromised likely due to the loss of supporting cells and altered innervation.
Collapse
MESH Headings
- Animals
- Receptor, Notch1/genetics
- Receptor, Notch1/physiology
- Receptor, Notch1/deficiency
- Receptor, Notch1/metabolism
- Mice
- Female
- Male
- Postural Balance/physiology
- Vestibule, Labyrinth/growth & development
- Vestibule, Labyrinth/physiology
- Vestibule, Labyrinth/cytology
- Hair Cells, Auditory/physiology
- Hair Cells, Vestibular/physiology
- Mice, Knockout
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Alison Heffer
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York 14642
| | - Choongheon Lee
- Departments of Otolaryngology, University of Rochester, Rochester, New York 14642
- Mechanical Engineering, University of Rochester, Rochester, New York 14642
| | - Joseph P Mayernik
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York 14642
| | - Joseph C Holt
- Departments of Otolaryngology, University of Rochester, Rochester, New York 14642
- Neuroscience, University of Rochester, Rochester, New York 14642
| | - Amy E Kiernan
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York 14642
| |
Collapse
|
2
|
Liu Y, Wang Z, Ma T, Gao Y, Chen W, Ye Z, Li Z. Differentiation of mesenchymal stem cells towards lens epithelial stem cells based on three-dimensional bio-printed matrix. Front Cell Dev Biol 2025; 12:1526943. [PMID: 39834393 PMCID: PMC11743933 DOI: 10.3389/fcell.2024.1526943] [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: 11/12/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
The high risks of traumatic cataract treatments promoted the development of the concept of autologous lens regeneration. Biochemical cues can influence the cellular behavior of stem cells, and in this case, biophysical cues may be the important factors in producing rapid activation of cellular behavior. Here we bio-printed mesenchymal stem cells (MSCs) using a commonly used bioink sodium alginate-gelatin blends, and investigated the induction effect of MSC differentiation towards lens epithelial stem cells (LESCs) under a combination of biochemical cues and biophysical cues. It was found that biochemical cues in the porous three-dimensional (3D) matrix constructed using bioink sodium alginate-gelatin blends for bio-printing did not reduce the cell viability of loaded MSCs in the matrix by scanning electron microscope (SEM) observation and cell viability detection. Loaded MSCs in the matrix were consistently upregulated in the expression of proteins and genes involved in phenotypes and development signaling pathways of LESCs, as detected by polymerase chain reaction (PCR) with the support of biochemical cues. These results indicated that biophysical cues could rapidly activate the cellular behavior of MSCs differentiation, and biochemical cues could continuously induce MSCs differentiation towards LESCs.
Collapse
Affiliation(s)
| | | | | | | | | | - Zi Ye
- Senior Department of Ophthalmology, The Third Medical Center of PLA General Hospital, PLA General Hospital and PLA Medical College, Beijing, China
| | - Zhaohui Li
- Senior Department of Ophthalmology, The Third Medical Center of PLA General Hospital, PLA General Hospital and PLA Medical College, Beijing, China
| |
Collapse
|
3
|
Novo SG, Faranda AP, D'Antin JC, Wang Y, Shihan M, Barraquer RI, Michael R, Duncan MK. Human lens epithelial cells induce the inflammatory response when placed into the lens capsular bag model of posterior capsular opacification. Mol Vis 2024; 30:348-367. [PMID: 39959166 PMCID: PMC11829793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 10/05/2024] [Indexed: 02/18/2025] Open
Abstract
Purpose Cataracts are typically treated by phacoemulsification followed by intraocular lens implantation. Studies of mouse models of cataract surgery have revealed that lens epithelial cells rapidly remodel their transcriptome to express proinflammatory cytokines after lens fiber cell removal, but it is currently unknown whether this response is conserved in human lenses. This study seeks to fill this knowledge gap. Methods Human cadaver eyes from 70 to 89 year old individuals were prepared for the human capsular bag model of cataract surgery. The central epithelium was preserved in RNAlater during culture preparation, then the equatorial epithelium was either immediately preserved in RNAlater after the culture was created, or 24 h later. Gene expression profiles were generated by bulk sequencing of RNA isolated from these tissue samples. The transcriptomic response of human cadaver-derived lens epithelial cells to culture in this "capsular bag" model was characterized by bioinformatic analysis. The human response was directly compared to that of 24-month-old mouse lens epithelial cells subjected to fiber cell removal surgery. Results Human lens epithelial cells remodel approximately a third of their transcriptome by 24 h after surgery, and like mice, this response consists of induction of proinflammatory cytokine genes, upregulation of fibrotic markers and downregulation of genes controlling the lens epithelial phenotype. Conclusions These observations demonstrate that humans and mice have similar responses to cataract surgery and support the use of mice to study the response of lens epithelial cells to cataract surgery, suggesting that identified injury response mechanisms can be leveraged to elucidate new approaches to improve the outcomes of cataract surgery.
Collapse
Affiliation(s)
- Samuel G Novo
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Adam P Faranda
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Justin C D'Antin
- Centro de Oftalmología Barraquer, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Yan Wang
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Mahbubul Shihan
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Rafael I Barraquer
- Centro de Oftalmología Barraquer, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ralph Michael
- Centro de Oftalmología Barraquer, Barcelona, Spain
- Institute for Medical Informatics, Statistics and Epidemiology, University Leipzig, Leipzig, Germany
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE
| |
Collapse
|
4
|
DeCotiis-Mauro J, Han SM, Mello H, Goyeneche C, Marchesini-Tovar G, Jin L, Bellofatto V, Lukac DM. The cellular Notch1 protein promotes KSHV reactivation in an Rta-dependent manner. J Virol 2024; 98:e0078824. [PMID: 38975769 PMCID: PMC11334469 DOI: 10.1128/jvi.00788-24] [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: 05/06/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
The cellular Notch signal transduction pathway is intimately associated with infections by Kaposi's sarcoma-associated herpesvirus (KSHV) and other gamma-herpesviruses. RBP-Jk, the cellular DNA binding component of the canonical Notch pathway, is the key Notch downstream effector protein in virus-infected and uninfected animal cells. Reactivation of KSHV from latency requires the viral lytic switch protein, Rta, to form complexes with RBP-Jk on numerous sites within the viral DNA. Constitutive Notch activity is essential for KSHV pathophysiology in models of Kaposi's sarcoma (KS) and Primary Effusion Lymphoma (PEL), and we demonstrate that Notch1 is also constitutively active in infected Vero cells. Although the KSHV genome contains >100 RBP-Jk DNA motifs, we show that none of the four isoforms of activated Notch can productively reactivate the virus from latency in a highly quantitative trans-complementing reporter virus system. Nevertheless, Notch contributed positively to reactivation because broad inhibition of Notch1-4 with gamma-secretase inhibitor (GSI) or expression of dominant negative mastermind-like1 (dnMAML1) coactivators severely reduced production of infectious KSHV from Vero cells. Reduction of KSHV production is associated with gene-specific reduction of viral transcription in both Vero and PEL cells. Specific inhibition of Notch1 by siRNA partially reduces the production of infectious KSHV, and NICD1 forms promoter-specific complexes with viral DNA during reactivation. We conclude that constitutive Notch activity is required for the robust production of infectious KSHV, and our results implicate activated Notch1 as a pro-viral member of a MAML1/RBP-Jk/DNA complex during viral reactivation. IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) manipulates the host cell oncogenic Notch signaling pathway for viral reactivation from latency and cell pathogenesis. KSHV reactivation requires that the viral protein Rta functionally interacts with RBP-Jk, the DNA-binding component of the Notch pathway, and with promoter DNA to drive transcription of productive cycle genes. We show that the Notch pathway is constitutively active during KSHV reactivation and is essential for robust production of infectious virus progeny. Inhibiting Notch during reactivation reduces the expression of specific viral genes yet does not affect the growth of the host cells. Although Notch cannot reactivate KSHV alone, the requisite expression of Rta reveals a previously unappreciated role for Notch in reactivation. We propose that activated Notch cooperates with Rta in a promoter-specific manner that is partially programmed by Rta's ability to redistribute RBP-Jk DNA binding to the virus during reactivation.
Collapse
Affiliation(s)
- Jennifer DeCotiis-Mauro
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Health Science Campus at Newark, Rutgers University, Newark, New Jersey, USA
| | - Sun M. Han
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Health Science Campus at Newark, Rutgers University, Newark, New Jersey, USA
| | - Helena Mello
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Health Science Campus at Newark, Rutgers University, Newark, New Jersey, USA
| | - Corey Goyeneche
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Health Science Campus at Newark, Rutgers University, Newark, New Jersey, USA
| | - Giuseppina Marchesini-Tovar
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Health Science Campus at Newark, Rutgers University, Newark, New Jersey, USA
| | - Lianhua Jin
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Health Science Campus at Newark, Rutgers University, Newark, New Jersey, USA
| | - Vivian Bellofatto
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Health Science Campus at Newark, Rutgers University, Newark, New Jersey, USA
| | - David M. Lukac
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Health Science Campus at Newark, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
5
|
Heffer A, Lee C, Holt JC, Kiernan AE. Notch1 is required to maintain supporting cell identity and vestibular function during maturation of the mammalian balance organs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600098. [PMID: 38948821 PMCID: PMC11212955 DOI: 10.1101/2024.06.21.600098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The inner ear houses two sensory modalities: the hearing organ, located in the cochlea, and the balance organs, located throughout the vestibular regions of the ear. Both hearing and vestibular sensory regions are composed of similar cell types, including hair cells and associated supporting cells. Recently, we showed that Notch1 is required for maintaining supporting cell survival postnatally during cochlear maturation. However, it is not known whether Notch1 plays a similar role in the balance organs of the inner ear. To characterize the role of Notch during vestibular maturation, we conditionally deleted Notch1 from Sox2-expressing cells of the vestibular organs in the mouse at P0/P1. Histological analyses showed a dramatic loss of supporting cells accompanied by an increase in type II hair cells without cell death, indicating the supporting cells are converting to hair cells in the maturing vestibular regions. Analysis of 6-week old animals indicate that the converted hair cells survive, despite the reduction of supporting cells. Interestingly, measurements of vestibular sensory evoked potentials (VsEPs), known to be generated in the striolar regions of the vestibular afferents in the maculae, failed to show a response, indicating that NOTCH1 expression is critical for striolar function postnatally. Consistent with this, we find that the specialized type I hair cells in the striola fail to develop the complex calyces typical of these cells. These defects are likely due to the reduction in supporting cells, which have previously been shown to express factors critical for the striolar region. Similar to other mutants that lack proper striolar development, Notch1 mutants do not exhibit typical vestibular behaviors such as circling and head shaking, but do show difficulties in some vestibular tests, including the balance beam and forced swim test. These results indicate that, unlike the hearing organ in which the supporting cells undergo cell death, supporting cells in the balance regions retain the ability to convert to hair cells during maturation, which survive into adulthood despite the reduction in supporting cells.
Collapse
Affiliation(s)
- Alison Heffer
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, 14642, USA
| | - Choongheon Lee
- Department of Otolaryngology, University of Rochester, Rochester, NY, 14642, USA
| | - Joseph C. Holt
- Department of Otolaryngology, University of Rochester, Rochester, NY, 14642, USA
- Dept. of Neuroscience, University of Rochester, Rochester, New York 14642, USA
| | - Amy E. Kiernan
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, 14642, USA
| |
Collapse
|
6
|
Liu M, Li M, Ruan J, Jia J, Ge C, Cao W. Analysis of microRNA Expression Profiles in Broiler Muscle Tissues by Feeding Different Levels of Guanidinoacetic Acid. Curr Issues Mol Biol 2024; 46:3713-3728. [PMID: 38666961 PMCID: PMC11048799 DOI: 10.3390/cimb46040231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The aim of this study was to explore the molecular mechanisms through which different levels of GAA affect chicken muscle development by influencing miRNA expression, to lay a theoretical foundation for the identification of key functional small RNAs related to poultry muscle development, and to provide new insights into the regulatory mechanisms of GAA on muscle development and meat quality in broilers. It provides a new theoretical basis for using GAA as a feed additive to improve feed performance. Small RNA sequencing technology was utilized to obtain the expression profiles of miRNA in the broiler pectoral muscle fed with different levels of GAA (0 g/kg, 1.2 g/kg and 3.6 g/kg). An analysis of differentially expressed miRNAs revealed 90 such miRNAs in the three combination comparisons, with gga-miR-130b-5p exhibiting significant differences across all three combinations. Furthermore, three of the differentially expressed miRNAs were performed by RT-qPCR verification, yielding results consistent with those obtained from small RNA sequencing. Target gene prediction, as well as the GO and KEGG enrichment analysis of differentially expressed miRNAs, indicated their involvement in muscle cell differentiation and other processes, particularly those associated with the MAPK signaling pathway. This study has, thus, provided valuable insights and resources for the further exploration of the miRNA molecular mechanism underlying the influence of guanidine acetic acid on broiler muscle development. Combined with previous studies and small RNA sequencing, adding 1.2 g/kg GAA to the diet can better promote the muscle development of broilers.
Collapse
Affiliation(s)
- Mengqian Liu
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (M.L.); (M.L.); (J.R.)
| | - Mengyuan Li
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (M.L.); (M.L.); (J.R.)
| | - Jinrui Ruan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (M.L.); (M.L.); (J.R.)
| | - Junjing Jia
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (M.L.); (M.L.); (J.R.)
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming 650201, China
| | - Changrong Ge
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (M.L.); (M.L.); (J.R.)
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming 650201, China
| | - Weina Cao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (M.L.); (M.L.); (J.R.)
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
7
|
Duot M, Viel R, Viet J, Le Goff-Gaillard C, Paillard L, Lachke SA, Gautier-Courteille C, Reboutier D. Eye Lens Organoids Made Simple: Characterization of a New Three-Dimensional Organoid Model for Lens Development and Pathology. Cells 2023; 12:2478. [PMID: 37887322 PMCID: PMC10605248 DOI: 10.3390/cells12202478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Cataract, the opacification of the lens, is the leading cause of blindness worldwide. Although effective, cataract surgery is costly and can lead to complications. Toward identifying alternate treatments, it is imperative to develop organoid models relevant for lens studies and drug screening. Here, we demonstrate that by culturing mouse lens epithelial cells under defined three-dimensional (3D) culture conditions, it is possible to generate organoids that display optical properties and recapitulate many aspects of lens organization and biology. These organoids can be rapidly produced in large amounts. High-throughput RNA sequencing (RNA-seq) on specific organoid regions isolated via laser capture microdissection (LCM) and immunofluorescence assays demonstrate that these lens organoids display a spatiotemporal expression of key lens genes, e.g., Jag1, Pax6, Prox1, Hsf4 and Cryab. Further, these lens organoids are amenable to the induction of opacities. Finally, the knockdown of a cataract-linked RNA-binding protein encoding gene, Celf1, induces opacities in these organoids, indicating their use in rapidly screening for genes that are functionally relevant to lens biology and cataract. In sum, this lens organoid model represents a compelling new tool to advance the understanding of lens biology and pathology and can find future use in the rapid screening of compounds aimed at preventing and/or treating cataracts.
Collapse
Affiliation(s)
- Matthieu Duot
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Roselyne Viel
- CNRS, Inserm UMS Biosit, H2P2 Core Facility, Université de Rennes, 35000 Rennes, France
| | - Justine Viet
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| | - Catherine Le Goff-Gaillard
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| | - Luc Paillard
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| | - 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
| | - Carole Gautier-Courteille
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| | - David Reboutier
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, 35000 Rennes, France
| |
Collapse
|
8
|
Duot M, Viel R, Viet J, Le Goff-Gaillard C, Paillard L, Lachke SA, Gautier-Courteille C, Reboutier D. Eye lens organoids going simple: characterization of a new 3-dimensional organoid model for lens development and pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548679. [PMID: 37503005 PMCID: PMC10370037 DOI: 10.1101/2023.07.12.548679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The ocular lens, along with the cornea, focuses light on the retina to generate sharp images. Opacification of the lens, or cataract, is the leading cause of blindness worldwide. Presently, the best approach for cataract treatment is to surgically remove the diseased lens and replace it with an artificial implant. Although effective, this is costly and can have post-surgical complications. Toward identifying alternate treatments, it is imperative to develop organoid models relevant for lens studies and anti-cataract drug screening. Here, we demonstrate that by culturing mouse lens epithelial cells under defined 3-dimensional (3D) culture conditions, it is possible to generate organoids that display optical properties and recapitulate many aspects of lens organization at the tissue, cellular and transcriptomic levels. These 3D cultured lens organoids can be rapidly produced in large amounts. High-throughput RNA-sequencing (RNA-seq) on specific organoid regions isolated by laser capture microdissection (LCM) and immunofluorescence assays demonstrate that these lens organoids display spatiotemporal expression of key lens genes, e.g. , Jag1 , Pax6 , Prox1 , Hsf4 and Cryab . Further, these lens organoids are amenable to induction of opacities. Finally, knockdown of a cataract-linked RNA-binding protein encoding gene, Celf1 , induces opacities in these organoids, indicating their use in rapidly screening for genes functionally relevant to lens biology and cataract. In sum, this lens organoid model represents a compelling new tool to advance the understanding of lens biology and pathology, and can find future use in the rapid screening of compounds aimed at preventing and/or treating cataract.
Collapse
|
9
|
Disatham J, Brennan L, Cvekl A, Kantorow M. Multiomics Analysis Reveals Novel Genetic Determinants for Lens Differentiation, Structure, and Transparency. Biomolecules 2023; 13:693. [PMID: 37189439 PMCID: PMC10136076 DOI: 10.3390/biom13040693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/13/2023] [Accepted: 04/16/2023] [Indexed: 05/17/2023] Open
Abstract
Recent advances in next-generation sequencing and data analysis have provided new gateways for identification of novel genome-wide genetic determinants governing tissue development and disease. These advances have revolutionized our understanding of cellular differentiation, homeostasis, and specialized function in multiple tissues. Bioinformatic and functional analysis of these genetic determinants and the pathways they regulate have provided a novel basis for the design of functional experiments to answer a wide range of long-sought biological questions. A well-characterized model for the application of these emerging technologies is the development and differentiation of the ocular lens and how individual pathways regulate lens morphogenesis, gene expression, transparency, and refraction. Recent applications of next-generation sequencing analysis on well-characterized chicken and mouse lens differentiation models using a variety of omics techniques including RNA-seq, ATAC-seq, whole-genome bisulfite sequencing (WGBS), chip-seq, and CUT&RUN have revealed a wide range of essential biological pathways and chromatin features governing lens structure and function. Multiomics integration of these data has established new gene functions and cellular processes essential for lens formation, homeostasis, and transparency including the identification of novel transcription control pathways, autophagy remodeling pathways, and signal transduction pathways, among others. This review summarizes recent omics technologies applied to the lens, methods for integrating multiomics data, and how these recent technologies have advanced our understanding ocular biology and function. The approach and analysis are relevant to identifying the features and functional requirements of more complex tissues and disease states.
Collapse
Affiliation(s)
- Joshua Disatham
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA; (J.D.); (L.B.)
| | - Lisa Brennan
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA; (J.D.); (L.B.)
| | - Ales Cvekl
- Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Marc Kantorow
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA; (J.D.); (L.B.)
| |
Collapse
|
10
|
Upreti A, Padula SL, Tangeman JA, Wagner BD, O’Connell MJ, Jaquish TJ, Palko RK, Mantz CJ, Anand D, Lovicu FJ, Lachke SA, Robinson ML. Lens Epithelial Explants Treated with Vitreous Humor Undergo Alterations in Chromatin Landscape with Concurrent Activation of Genes Associated with Fiber Cell Differentiation and Innate Immune Response. Cells 2023; 12:501. [PMID: 36766843 PMCID: PMC9914805 DOI: 10.3390/cells12030501] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Lens epithelial explants are comprised of lens epithelial cells cultured in vitro on their native basement membrane, the lens capsule. Biologists have used lens epithelial explants to study many different cellular processes including lens fiber cell differentiation. In these studies, fiber differentiation is typically measured by cellular elongation and the expression of a few proteins characteristically expressed by lens fiber cells in situ. Chromatin and RNA was collected from lens epithelial explants cultured in either un-supplemented media or media containing 50% bovine vitreous humor for one or five days. Chromatin for ATAC-sequencing and RNA for RNA-sequencing was prepared from explants to assess regions of accessible chromatin and to quantitatively measure gene expression, respectively. Vitreous humor increased chromatin accessibility in promoter regions of genes associated with fiber differentiation and, surprisingly, an immune response, and this was associated with increased transcript levels for these genes. In contrast, vitreous had little effect on the accessibility of the genes highly expressed in the lens epithelium despite dramatic reductions in their mRNA transcripts. An unbiased analysis of differentially accessible regions revealed an enrichment of cis-regulatory motifs for RUNX, SOX and TEAD transcription factors that may drive differential gene expression in response to vitreous.
Collapse
Affiliation(s)
- Anil Upreti
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Stephanie L. Padula
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Jared A. Tangeman
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Brad D. Wagner
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | | | - Tycho J. Jaquish
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Raye K. Palko
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Courtney J. Mantz
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Deepti Anand
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Frank J. Lovicu
- Molecular and Cellular Biomedicine, School of Medical Sciences, and Save Sight Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - 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
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| |
Collapse
|
11
|
Loss of Hes1 in embryonic stem cells caused developmental disorders in retinal pigment epithelium morphogenesis and specification. Biochem Biophys Res Commun 2022; 632:76-84. [DOI: 10.1016/j.bbrc.2022.09.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 08/23/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022]
|
12
|
Disatham J, Brennan L, Jiao X, Ma Z, Hejtmancik JF, Kantorow M. Changes in DNA methylation hallmark alterations in chromatin accessibility and gene expression for eye lens differentiation. Epigenetics Chromatin 2022; 15:8. [PMID: 35246225 PMCID: PMC8897925 DOI: 10.1186/s13072-022-00440-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/16/2022] [Indexed: 12/13/2022] Open
Abstract
Background Methylation at cytosines (mCG) is a well-known regulator of gene expression, but its requirements for cellular differentiation have yet to be fully elucidated. A well-studied cellular differentiation model system is the eye lens, consisting of a single anterior layer of epithelial cells that migrate laterally and differentiate into a core of fiber cells. Here, we explore the genome-wide relationships between mCG methylation, chromatin accessibility and gene expression during differentiation of eye lens epithelial cells into fiber cells. Results Whole genome bisulfite sequencing identified 7621 genomic loci exhibiting significant differences in mCG levels between lens epithelial and fiber cells. Changes in mCG levels were inversely correlated with the differentiation state-specific expression of 1285 genes preferentially expressed in either lens fiber or lens epithelial cells (Pearson correlation r = − 0.37, p < 1 × 10–42). mCG levels were inversely correlated with chromatin accessibility determined by assay for transposase-accessible sequencing (ATAC-seq) (Pearson correlation r = − 0.86, p < 1 × 10–300). Many of the genes exhibiting altered regions of DNA methylation, chromatin accessibility and gene expression levels in fiber cells relative to epithelial cells are associated with lens fiber cell structure, homeostasis and transparency. These include lens crystallins (CRYBA4, CRYBB1, CRYGN, CRYBB2), lens beaded filament proteins (BFSP1, BFSP2), transcription factors (HSF4, SOX2, HIF1A), and Notch signaling pathway members (NOTCH1, NOTCH2, HEY1, HES5). Analysis of regions exhibiting cell-type specific alterations in DNA methylation revealed an overrepresentation of consensus sequences of multiple transcription factors known to play key roles in lens cell differentiation including HIF1A, SOX2, and the MAF family of transcription factors. Conclusions Collectively, these results link DNA methylation with control of chromatin accessibility and gene expression changes required for eye lens differentiation. The results also point to a role for DNA methylation in the regulation of transcription factors previously identified to be important for lens cell differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-022-00440-z.
Collapse
Affiliation(s)
- Joshua Disatham
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Lisa Brennan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Xiaodong Jiao
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhiwei Ma
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - J Fielding Hejtmancik
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marc Kantorow
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA.
| |
Collapse
|
13
|
Makrides N, Wang Q, Tao C, Schwartz S, Zhang X. Jack of all trades, master of each: the diversity of fibroblast growth factor signalling in eye development. Open Biol 2022; 12:210265. [PMID: 35016551 PMCID: PMC8753161 DOI: 10.1098/rsob.210265] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A central question in development biology is how a limited set of signalling pathways can instruct unlimited diversity of multicellular organisms. In this review, we use three ocular tissues as models of increasing complexity to present the astounding versatility of fibroblast growth factor (FGF) signalling. In the lacrimal gland, we highlight the specificity of FGF signalling in a one-dimensional model of budding morphogenesis. In the lens, we showcase the dynamics of FGF signalling in altering functional outcomes in a two-dimensional space. In the retina, we present the prolific utilization of FGF signalling from three-dimensional development to homeostasis. These examples not only shed light on the cellular basis for the perfection and complexity of ocular development, but also serve as paradigms for the diversity of FGF signalling.
Collapse
Affiliation(s)
- Neoklis Makrides
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Qian Wang
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Chenqi Tao
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Samuel Schwartz
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Xin Zhang
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
14
|
Disatham J, Brennan L, Chauss D, Kantorow J, Afzali B, Kantorow M. A functional map of genomic HIF1α-DNA complexes in the eye lens revealed through multiomics analysis. BMC Genomics 2021; 22:497. [PMID: 34215186 PMCID: PMC8254356 DOI: 10.1186/s12864-021-07795-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND During eye lens development the embryonic vasculature regresses leaving the lens without a direct oxygen source. Both embryonically and throughout adult life, the lens contains a decreasing oxygen gradient from the surface to the core that parallels the natural differentiation of immature surface epithelial cells into mature core transparent fiber cells. These properties of the lens suggest a potential role for hypoxia and the master regulator of the hypoxic response, hypoxia-inducible transcription factor 1 (HIF1), in the regulation of genes required for lens fiber cell differentiation, structure and transparency. Here, we employed a multiomics approach combining CUT&RUN, RNA-seq and ATACseq analysis to establish the genomic complement of lens HIF1α binding sites, genes activated or repressed by HIF1α and the chromatin states of HIF1α-regulated genes. RESULTS CUT&RUN analysis revealed 8375 HIF1α-DNA binding complexes in the chick lens genome. One thousand one hundred ninety HIF1α-DNA binding complexes were significantly clustered within chromatin accessible regions (χ2 test p < 1 × 10- 55) identified by ATACseq. Formation of the identified HIF1α-DNA complexes paralleled the activation or repression of 526 genes, 116 of which contained HIF1α binding sites within 10kB of the transcription start sites. Some of the identified HIF1α genes have previously established lens functions while others have novel functions never before examined in the lens. GO and pathway analysis of these genes implicate HIF1α in the control of a wide-variety of cellular pathways potentially critical for lens fiber cell formation, structure and function including glycolysis, cell cycle regulation, chromatin remodeling, Notch and Wnt signaling, differentiation, development, and transparency. CONCLUSIONS These data establish the first functional map of genomic HIF1α-DNA complexes in the eye lens. They identify HIF1α as an important regulator of a wide-variety of genes previously shown to be critical for lens formation and function and they reveal a requirement for HIF1α in the regulation of a wide-variety of genes not yet examined for lens function. They support a requirement for HIF1α in lens fiber cell formation, structure and function and they provide a basis for understanding the potential roles and requirements for HIF1α in the development, structure and function of more complex tissues.
Collapse
Affiliation(s)
- Joshua Disatham
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd., Boca Raton, FL 33431 USA
| | - Lisa Brennan
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd., Boca Raton, FL 33431 USA
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD 20892 USA
| | | | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD 20892 USA
| | - Marc Kantorow
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd., Boca Raton, FL 33431 USA
| |
Collapse
|
15
|
Gu Y, Yao K, Fu Q. Lens regeneration: scientific discoveries and clinical possibilities. Mol Biol Rep 2021; 48:4911-4923. [PMID: 34143397 DOI: 10.1007/s11033-021-06489-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022]
Abstract
In the process of exploring new methods for cataract treatment, lens regeneration is an ideal strategy for effectively restoring accommodative vision and avoiding postoperative complications and has great clinical potential. Lens regeneration, which is not a simple repetition of lens development, depends on the complex regulatory network comprising the FGF, BMP/TGF-β, Notch, and Wnt signaling pathways. Current research mainly focuses on in situ and in vitro lens regeneration. On the one hand, the possibility of the autologous stem cell in situ regeneration of functional lenses has been confirmed; on the other hand, both embryonic stem cells and induced pluripotent stem cells have been induced into lentoid bodies in vitro which are similar to the natural lens to a certain extent. This article will briefly summarize the regulatory mechanisms of lens development, describe the recent progress of lens regeneration, explore the key molecular signaling pathways, and, more importantly, discuss the prospects and challenges of their clinical applications to provide reference for clinical transformations.
Collapse
Affiliation(s)
- Yuzhou Gu
- Eye Center of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang Province, China.,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, 310009, Zhejiang Province, China
| | - Ke Yao
- Eye Center of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang Province, China. .,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, 310009, Zhejiang Province, China.
| | - Qiuli Fu
- Eye Center of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang Province, China. .,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, 310009, Zhejiang Province, China.
| |
Collapse
|
16
|
Huang D, Wang R. Exploring the mechanism of pancreatic cell fate decisions via cell-cell communication. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:2401-2424. [PMID: 33892552 DOI: 10.3934/mbe.2021122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The endocrine and exocrine cells in pancreas originate initially from a group of apparently identical endoderm cells in the early gut. The endocrine and exocrine tissues are composed of islet/acinar and duct cells respectively. To explore the mechanism of pancreas cell fate decisions, we first construct a minimal mathematical model related to pancreatic regulations. The regulatory mechanism of acinar-to-islet cell conversion is revealed by bifurcation analysis of the model. In addition, Notch signaling is critical in determining the fate of endocrine and exocrine in the developing pancreas and it is a typical mediator of lateral inhibition which instructs adjacent cells to make different fate decisions. Next, we construct a multicellular model of cell-cell communication mediated by Notch signaling with trans-activation and cis-inhibition. The roles of Notch signaling in regulating fate decisions of endocrine and exocrine cells during the differentiation of pancreatic cells are explored. The results indicate that high (or low) level of Notch signaling drive cells to select the fate of exocrine (or endocrine) progenitor cells. The networks and the models presented here might be good candidates for providing qualitative mechanisms of pancreatic cell fate decisions. These results can also provide some insight on choosing perturbation strategies for further experimental analysis.
Collapse
Affiliation(s)
- Dasong Huang
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| | - Ruiqi Wang
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| |
Collapse
|
17
|
Zhao Y, Liu S, Li X, Xu Z, Hao L, Cui Z, Bi K, Zhang Y, Liu Z. Cross-talk of Signaling Pathways in the Pathogenesis of Allergic Asthma and Cataract. Protein Pept Lett 2021; 27:810-822. [PMID: 32031062 DOI: 10.2174/0929866527666200207113439] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 12/13/2022]
Abstract
Allergic asthma is a chronic inflammatory disease, which involves many cellular and cellular components. Cataract is a condition that affects the transparency of the lens, which the opacity of the lens caused by any innate or acquired factor degrades its transparency or changes in color. Both of them belong to diseases induced by immune disorders or inflammation. We want to confirm the signaling pathways involved in the regulation of asthma and cataract simultaneously, and provide reference for the later related experiments. So we conducted a scoping review of many databases and searched for studies (Academic research published in Wiley, Springer and Bentham from 2000 to 2019) about the possible relationship between asthma and cataract. It was found that during the onset of asthma and cataract, Rho/Rock signaling pathway, Notch signaling pathway, Wnt/β-catenin signaling pathway, PI3K/AKT signaling pathway, JAK/STAT signaling pathway, MAPK signaling pathway, TGF-β1/Smad signaling pathway and NF-κB signaling pathway are all active, so they may have a certain correlation in pathogenesis. Asthma may be associated with cataract through the eight signaling pathways, causing inflammation or immune imbalance based on allergy that can lead to cataract. According to these studies, we speculated that the three most likely signaling pathways are PI3K/AKT, MAPK and NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yang Zhao
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Sumei Liu
- Department of Stomatology, No. 2 Hospital of Baoding, Baoding 071002, China
| | - Xiangsheng Li
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Zhenzhen Xu
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Lifang Hao
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Zhe Cui
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Kewei Bi
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Yanfen Zhang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China,Offices of Science and Technology, Hebei University, Baoding 071002, China
| | - Zhongcheng Liu
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| |
Collapse
|
18
|
González S, Halabi M, Ju D, Tsai M, Deng SX. Role of Jagged1-mediated Notch Signaling Activation in the Differentiation and Stratification of the Human Limbal Epithelium. Cells 2020; 9:cells9091945. [PMID: 32842657 PMCID: PMC7564045 DOI: 10.3390/cells9091945] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022] Open
Abstract
The Notch signaling pathway plays a key role in proliferation and differentiation. We investigated the effect of Jagged 1 (Jag1)-mediated Notch signaling activation in the human limbal stem/progenitor cell (LSC) population and the stratification of the limbal epithelium in vitro. After Notch signaling activation, there was a reduction in the amount of the stem/progenitor cell population, epithelial stratification, and expression of proliferation markers. There was also an increase of the corneal epithelial differentiation. In the presence of Jag1, asymmetric divisions were decreased, and the expression pattern of the polarity protein Par3, normally present at the apical-lateral membrane of basal cells, was dispersed in the cells. We propose a mechanism in which Notch activation by Jag1 decreases p63 expression at the basal layer, which in turn reduces stratification by decreasing the number of asymmetric divisions and increases differentiation.
Collapse
|
19
|
Garg A, Hannan A, Wang Q, Makrides N, Zhong J, Li H, Yoon S, Mao Y, Zhang X. Etv transcription factors functionally diverge from their upstream FGF signaling in lens development. eLife 2020; 9:e51915. [PMID: 32043969 PMCID: PMC7069720 DOI: 10.7554/elife.51915] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
The signal regulated transcription factors (SRTFs) control the ultimate transcriptional output of signaling pathways. Here, we examined a family of FGF-induced SRTFs - Etv1, Etv 4, and Etv 5 - in murine lens development. Contrary to FGF receptor mutants that displayed loss of ERK signaling and defective cell differentiation, Etv deficiency augmented ERK phosphorylation without disrupting the normal lens fiber gene expression. Instead, the transitional zone for lens differentiation was shifted anteriorly as a result of reduced Jag1-Notch signaling. We also showed that Etv proteins suppresses mTOR activity by promoting Tsc2 expression, which is necessary for the nuclei clearance in mature lens. These results revealed the functional divergence between Etv and FGF in lens development, demonstrating that these SRTFs can operate outside the confine of their upstream signaling.
Collapse
Affiliation(s)
- Ankur Garg
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Abdul Hannan
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Qian Wang
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Neoklis Makrides
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Jian Zhong
- Burke Neurological Institute and Feil Family Brain and Mind Research Institute, Weill Cornell MedicineWhite PlainsUnited States
| | - Hongge Li
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Sungtae Yoon
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Yingyu Mao
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Xin Zhang
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| |
Collapse
|
20
|
Notch Signalling: The Multitask Manager of Inner Ear Development and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:129-157. [DOI: 10.1007/978-3-030-34436-8_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
21
|
Vujovic F, Hunter N, Farahani RM. Notch pathway: a bistable inducer of biological noise? Cell Commun Signal 2019; 17:133. [PMID: 31640734 PMCID: PMC6805690 DOI: 10.1186/s12964-019-0453-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/04/2019] [Indexed: 12/11/2022] Open
Abstract
Notch signalling pathway is central to development of metazoans. The pathway codes a binary fate switch. Upon activation, downstream signals contribute to resolution of fate dichotomies such as proliferation/differentiation or sub-lineage differentiation outcome. There is, however, an interesting paradox in the Notch signalling pathway. Despite remarkable predictability of fate outcomes instructed by the Notch pathway, the associated transcriptome is versatile and plastic. This inconsistency suggests the presence of an interface that compiles input from the plastic transcriptome of the Notch pathway but communicates only a binary output in biological decisions. Herein, we address the interface that determines fate outcomes. We provide an alternative hypothesis for the Notch pathway as a biological master switch that operates by induction of genetic noise and bistability in order to facilitate resolution of dichotomous fate outcomes in development.
Collapse
Affiliation(s)
- Filip Vujovic
- IDR/Westmead Institute for Medical Research, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145 Australia
| | - Neil Hunter
- IDR/Westmead Institute for Medical Research, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145 Australia
| | - Ramin M. Farahani
- IDR/Westmead Institute for Medical Research, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145 Australia
| |
Collapse
|
22
|
Wopat S, Bagwell J, Sumigray KD, Dickson AL, Huitema LFA, Poss KD, Schulte-Merker S, Bagnat M. Spine Patterning Is Guided by Segmentation of the Notochord Sheath. Cell Rep 2019; 22:2026-2038. [PMID: 29466731 PMCID: PMC5860813 DOI: 10.1016/j.celrep.2018.01.084] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 01/10/2018] [Accepted: 01/26/2018] [Indexed: 01/05/2023] Open
Abstract
The spine is a segmented axial structure made of alternating vertebral bodies (centra) and intervertebral discs (IVDs) assembled around the notochord. Here, we show that, prior to centra formation, the outer epithelial cell layer of the zebrafish notochord, the sheath, segments into alternating domains corresponding to the prospective centra and IVD areas. This process occurs sequentially in an anteroposterior direction via the activation of Notch signaling in alternating segments of the sheath, which transition from cartilaginous to mineralizing domains. Subsequently, osteoblasts are recruited to the mineralized domains of the notochord sheath to form mature centra. Tissue-specific manipulation of Notch signaling in sheath cells produces notochord segmentation defects that are mirrored in the spine. Together, our findings demonstrate that notochord sheath segmentation provides a template for vertebral patterning in the zebrafish spine.
Collapse
Affiliation(s)
- Susan Wopat
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jennifer Bagwell
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kaelyn D Sumigray
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA
| | - Amy L Dickson
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Leonie F A Huitema
- Hubrecht Institute - KNAW & UMC Utrecht, 3584 CT, Utrecht, the Netherlands
| | - Kenneth D Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Stefan Schulte-Merker
- Hubrecht Institute - KNAW & UMC Utrecht, 3584 CT, Utrecht, the Netherlands; Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, 48149 Münster, Germany; CiM Cluster of Excellence (EXC1003-CiM), 48149 Münster, Germany
| | - Michel Bagnat
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
23
|
Abstract
Notch signaling regulates a multitude of cellular processes. During ocular lens development this pathway is required for lens progenitor growth, differentiation and maintenance of the transition zone. After ligand-receptor binding, the receptor proteins are processed, first by ADAM proteases, then by γ-secretase cleavage. This results in the release of a Notch intracellular domain (N-ICD), which is recruited into a nuclear transcription factor complex that activates Notch target genes. Previous in vitro studies showed that the Delta-like and Jagged ligand proteins can also be cleaved by the γ-secretase complex, but it remains unknown whether such processing occurs during in vivo vertebrate development. Here we show that mouse and human lens progenitor cells endogenously express multiple Jagged1 protein isoforms, including a Jagged1 intracellular domain. We also found that pharmacologic blockage of γ-secretase activity in vitro resulted in an accumulation of Jagged1 polypeptide intermediates. Finally, overexpression of an epitope-tagged Jagged1 intracellular domain displayed nuclear localization and induced the upregulation of endogenous JAG1 mRNA expression. These findings support the idea that along with its classical role as a Notch pathway ligand, Jagged1 is regulated post-translationally, to produce multiple active protein isoforms. Summary: The Notch pathway ligand protein Jagged1 undergoes multiple catalytic cleavages, regulated by Adam proteases and the gamma-secretase complex, during mammalian lens development, similar to Notch receptor proteins.
Collapse
Affiliation(s)
- Mina Azimi
- Department of Cell Biology & Human Anatomy, University of California, 1 Shields Avenue, Davis, CA 95616, USA
| | - Nadean L Brown
- Department of Cell Biology & Human Anatomy, University of California, 1 Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
24
|
Toward understanding cancer stem cell heterogeneity in the tumor microenvironment. Proc Natl Acad Sci U S A 2019; 116:148-157. [PMID: 30587589 PMCID: PMC6320545 DOI: 10.1073/pnas.1815345116] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) formation are two paramount processes driving tumor progression, therapy resistance, and cancer metastasis. Recent experiments show that cells with varying EMT and CSC phenotypes are spatially segregated in the primary tumor. The underlying mechanisms generating such spatiotemporal dynamics in the tumor microenvironment, however, remain largely unexplored. Here, we show through a mechanism-based dynamical model that the diffusion of EMT-inducing signals such as TGF-β, together with noncell autonomous control of EMT and CSC decision making via the Notch signaling pathway, can explain experimentally observed disparate localization of subsets of CSCs with varying EMT phenotypes in the tumor. Our simulations show that the more mesenchymal CSCs lie at the invasive edge, while the hybrid epithelial/mesenchymal (E/M) CSCs reside in the tumor interior. Further, motivated by the role of Notch-Jagged signaling in mediating EMT and stemness, we investigated the microenvironmental factors that promote Notch-Jagged signaling. We show that many inflammatory cytokines such as IL-6 that can promote Notch-Jagged signaling can (i) stabilize a hybrid E/M phenotype, (ii) increase the likelihood of spatial proximity of hybrid E/M cells, and (iii) expand the fraction of CSCs. To validate the predicted connection between Notch-Jagged signaling and stemness, we knocked down JAG1 in hybrid E/M SUM149 human breast cancer cells in vitro. JAG1 knockdown significantly restricted tumor organoid formation, confirming the key role that Notch-Jagged signaling can play in tumor progression. Together, our integrated computational-experimental framework reveals the underlying principles of spatiotemporal dynamics of EMT and CSCs.
Collapse
|
25
|
Jolly MK, Somarelli JA, Sheth M, Biddle A, Tripathi SC, Armstrong AJ, Hanash SM, Bapat SA, Rangarajan A, Levine H. Hybrid epithelial/mesenchymal phenotypes promote metastasis and therapy resistance across carcinomas. Pharmacol Ther 2018; 194:161-184. [PMID: 30268772 DOI: 10.1016/j.pharmthera.2018.09.007] [Citation(s) in RCA: 224] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer metastasis and therapy resistance are the major unsolved clinical challenges, and account for nearly all cancer-related deaths. Both metastasis and therapy resistance are fueled by epithelial plasticity, the reversible phenotypic transitions between epithelial and mesenchymal phenotypes, including epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET). EMT and MET have been largely considered as binary processes, where cells detach from the primary tumor as individual units with many, if not all, traits of a mesenchymal cell (EMT) and then convert back to being epithelial (MET). However, recent studies have demonstrated that cells can metastasize in ways alternative to traditional EMT paradigm; for example, they can detach as clusters, and/or occupy one or more stable hybrid epithelial/mesenchymal (E/M) phenotypes that can be the end point of a transition. Such hybrid E/M cells can integrate various epithelial and mesenchymal traits and markers, facilitating collective cell migration. Furthermore, these hybrid E/M cells may possess higher tumor-initiation and metastatic potential as compared to cells on either end of the EMT spectrum. Here, we review in silico, in vitro, in vivo and clinical evidence for the existence of one or more hybrid E/M phenotype(s) in multiple carcinomas, and discuss their implications in tumor-initiation, tumor relapse, therapy resistance, and metastasis. Together, these studies drive the emerging notion that cells in a hybrid E/M phenotype may occupy 'metastatic sweet spot' in multiple subtypes of carcinomas, and pathways linked to this (these) hybrid E/M state(s) may be relevant as prognostic biomarkers as well as a promising therapeutic targets.
Collapse
Affiliation(s)
- Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
| | - Jason A Somarelli
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Maya Sheth
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Adrian Biddle
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Satyendra C Tripathi
- Department of Clinical Cancer Prevention, UT MD Anderson Cancer Center, Houston, USA
| | - Andrew J Armstrong
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, UT MD Anderson Cancer Center, Houston, USA
| | - Sharmila A Bapat
- National Center for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Annapoorni Rangarajan
- Department of Molecular Reproduction, Development & Genetics, Indian Institute of Science, Bangalore, India
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
| |
Collapse
|
26
|
Ihermann-Hella A, Hirashima T, Kupari J, Kurtzeborn K, Li H, Kwon HN, Cebrian C, Soofi A, Dapkunas A, Miinalainen I, Dressler GR, Matsuda M, Kuure S. Dynamic MAPK/ERK Activity Sustains Nephron Progenitors through Niche Regulation and Primes Precursors for Differentiation. Stem Cell Reports 2018; 11:912-928. [PMID: 30220628 PMCID: PMC6178244 DOI: 10.1016/j.stemcr.2018.08.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 08/20/2018] [Accepted: 08/20/2018] [Indexed: 12/24/2022] Open
Abstract
The in vivo niche and basic cellular properties of nephron progenitors are poorly described. Here we studied the cellular organization and function of the MAPK/ERK pathway in nephron progenitors. Live-imaging of ERK activity by a Förster resonance energy transfer biosensor revealed a dynamic activation pattern in progenitors, whereas differentiating precursors exhibited sustained activity. Genetic experiments demonstrate that MAPK/ERK activity controls the thickness, coherence, and integrity of the nephron progenitor niche. Molecularly, MAPK/ERK activity regulates niche organization and communication with extracellular matrix through PAX2 and ITGA8, and is needed for CITED1 expression denoting undifferentiated status. MAPK/ERK activation in nephron precursors propels differentiation by priming cells for distal and proximal fates induced by the Wnt and Notch pathways. Thus, our results demonstrate a mechanism through which MAPK/ERK activity controls both progenitor maintenance and differentiation by regulating a distinct set of targets, which maintain the biomechanical milieu of tissue-residing progenitors and prime precursors for nephrogenesis.
Collapse
Affiliation(s)
| | - Tsuyoshi Hirashima
- Department of Pathology and Biology of Diseases, Graduate School of Medicine & Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Jussi Kupari
- HiLIFE and Medicum, University of Helsinki, Helsinki FIN-00014, Finland
| | | | - Hao Li
- HiLIFE and Medicum, University of Helsinki, Helsinki FIN-00014, Finland
| | - Hyuk Nam Kwon
- HiLIFE and Medicum, University of Helsinki, Helsinki FIN-00014, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Abdul Soofi
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arvydas Dapkunas
- Medicum and Meilahti Clinical Proteomics Core Facility/HiLIFE, University of Helsinki, Helsinki FIN-00014, Finland
| | - Ilkka Miinalainen
- Department of Pathology (Biocenter Oulu), University of Oulu, Oulu 90220, Finland
| | - Gregory R Dressler
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine & Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Satu Kuure
- HiLIFE and Medicum, University of Helsinki, Helsinki FIN-00014, Finland; GM-Unit, LAC/ HiLIFE, and Medicum, University of Helsinki, Helsinki FIN-00014, Finland.
| |
Collapse
|
27
|
Azimi M, Le TT, Brown NL. Presenilin gene function and Notch signaling feedback regulation in the developing mouse lens. Differentiation 2018; 102:40-52. [PMID: 30059908 DOI: 10.1016/j.diff.2018.07.003] [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: 05/04/2018] [Revised: 07/06/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022]
Abstract
Presenilins (Psen1 and Psen2 in mice) are polytopic transmembrane proteins that act in the γ-secretase complex to make intra-membrane cleavages of their substrates, including the well-studied Notch receptors. Such processing releases the Notch intracellular domain, allowing it to physically relocate from the cell membrane to the nucleus where it acts in a transcriptional activating complex to regulate downstream genes in the signal-receiving cell. Previous studies of Notch pathway mutants for Jagged1, Notch2, and Rbpj demonstrated that canonical signaling is a necessary component of normal mouse lens development. However, the central role of Psens within the γ-secretase complex has never been explored in any developing eye tissue or cell type. By directly comparing Psen single and double mutant phenotypes during mouse lens development, we found a stronger requirement for Psen1, although both genes are needed for progenitor cell growth and to prevent apoptosis. We also uncovered a novel genetic interaction between Psen1 and Jagged1. By quantifying protein and mRNA levels of key Notch pathway genes in Psen1/2 or Jagged1 mutant lenses, we identified multiple points in the overall signaling cascade where feedback regulation can occur. Our data are consistent with the loss of particular genes indirectly influencing the transcription level of another. However, we conclude that regulating Notch2 protein levels is particularly important during normal signaling, supporting the importance of post-translational regulatory mechanisms in this tissue.
Collapse
Affiliation(s)
- Mina Azimi
- Department of Cell Biology & Human Anatomy; University of California, Davis One Shields Avenue, Davis, CA 95616, USA
| | - Tien T Le
- Division of Developmental Biology, Cincinnati Childrens Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Nadean L Brown
- Department of Cell Biology & Human Anatomy; University of California, Davis One Shields Avenue, Davis, CA 95616, USA; Division of Developmental Biology, Cincinnati Childrens Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
28
|
Dawes LJ, Shelley EJ, McAvoy JW, Lovicu FJ. A role for Hippo/YAP-signaling in FGF-induced lens epithelial cell proliferation and fibre differentiation. Exp Eye Res 2018; 169:122-133. [PMID: 29355736 DOI: 10.1016/j.exer.2018.01.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/14/2018] [Accepted: 01/16/2018] [Indexed: 12/19/2022]
Abstract
Recent studies indicate an important role for the transcriptional co-activator Yes-associated protein (YAP), and its regulatory pathway Hippo, in controlling cell growth and fate during lens development; however, the exogenous factors that promote this pathway are yet to be identified. Given that fibroblast growth factor (FGF)-signaling is an established regulator of lens cell behavior, the current study investigates the relationship between this pathway and Hippo/YAP-signaling during lens cell proliferation and fibre differentiation. Rat lens epithelial explants were cultured with FGF2 to induce epithelial cell proliferation or fibre differentiation. Immunolabeling methods were used to detect the expression of Hippo-signaling components, Total and Phosphorylated YAP, as well as fibre cell markers, Prox-1 and β-crystallin. FGF-induced lens cell proliferation was associated with a strong nuclear localisation of Total-YAP and low-level immuno-staining for phosphorylated-YAP. FGF-induced lens fibre differentiation was associated with a significant increase in cytoplasmic phosphorylated YAP (inactive state) and enhanced expression of core Hippo-signaling components. Inhibition of YAP with Verteporfin suppressed FGF-induced lens cell proliferation and ablated cell elongation during lens fibre differentiation. Inhibition of either FGFR- or MEK/ERK-signaling suppressed FGF-promoted YAP nuclear translocation. Here we propose that FGF promotes Hippo/YAP-signaling during lens cell proliferation and differentiation, with FGF-induced nuclear-YAP expression playing an essential role in promoting the proliferation of lens epithelial cells. An FGF-induced switch from proliferation to differentiation, hence regulation of lens growth, may play a key role in mediating Hippo suppression of YAP transcriptional activity.
Collapse
Affiliation(s)
- L J Dawes
- Save Sight Institute, The University of Sydney, NSW, Australia.
| | - E J Shelley
- Save Sight Institute, The University of Sydney, NSW, Australia
| | - J W McAvoy
- Save Sight Institute, The University of Sydney, NSW, Australia
| | - F J Lovicu
- Save Sight Institute, The University of Sydney, NSW, Australia; Discipline of Anatomy and Histology, Bosch Institute, The University of Sydney, NSW, Australia
| |
Collapse
|
29
|
Luna-Escalante JC, Formosa-Jordan P, Ibañes M. Redundancy and cooperation in Notch intercellular signaling. Development 2018; 145:dev.154807. [PMID: 29242285 DOI: 10.1242/dev.154807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 11/27/2017] [Indexed: 12/25/2022]
Abstract
During metazoan development, Notch signaling drives spatially coordinated differentiation by establishing communication between adjacent cells. This occurs through either lateral inhibition, in which adjacent cells acquire distinct fates, or lateral induction, in which all cells become equivalent. Notch signaling is commonly activated by several distinct ligands, each of which drives signaling with a different efficiency upon binding to the Notch receptor of adjacent cells. Moreover, these ligands can also be distinctly regulated by Notch signaling. Under such complex circumstances, the overall spatial coordination becomes elusive. Here, we address this issue through both mathematical and computational analyses. Our results show that when two ligands have distinct efficiencies and compete for the same Notch receptor, they cooperate to drive new signaling states, thereby conferring additional robustness and evolvability to Notch signaling. Counterintuitively, whereas antagonistically regulated ligands cooperate to drive and enhance the response that is expected from the more efficient ligand, equivalently regulated ligands coordinate emergent spatial responses that are dependent on both ligands. Our study highlights the importance of ligand efficiency in multi-ligand scenarios, and can explain previously reported complex phenotypes.
Collapse
Affiliation(s)
- Juan C Luna-Escalante
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona 08028, Spain
| | - Pau Formosa-Jordan
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona 08028, Spain
| | - Marta Ibañes
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona 08028, Spain .,Universitat de Barcelona Institute of Complex Systems (UBICS), Universitat de Barcelona, Barcelona 08028, Spain
| |
Collapse
|
30
|
Prasasya RD, Mayo KE. Notch Signaling Regulates Differentiation and Steroidogenesis in Female Mouse Ovarian Granulosa Cells. Endocrinology 2018; 159:184-198. [PMID: 29126263 PMCID: PMC5761600 DOI: 10.1210/en.2017-00677] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/02/2017] [Indexed: 01/04/2023]
Abstract
The Notch pathway is a highly conserved juxtacrine signaling mechanism that is important for many cellular processes during development, including differentiation and proliferation. Although Notch is important during ovarian follicle formation and early development, its functions during the gonadotropin-dependent stages of follicle development are largely unexplored. We observed positive regulation of Notch activity and expression of Notch ligands and receptors following activation of the luteinizing hormone-receptor in prepubertal mouse ovary. JAG1, the most abundantly expressed Notch ligand in mouse ovary, revealed a striking shift in localization from oocytes to somatic cells following hormone stimulation. Using primary cultures of granulosa cells, we investigated the functions of Jag1 using small interfering RNA knockdown. The loss of JAG1 led to suppression of granulosa cell differentiation as marked by reduced expression of enzymes and factors involved in steroid biosynthesis, and in steroid secretion. Jag1 knockdown also resulted in enhanced cell proliferation. These phenotypes were replicated, although less robustly, following knockdown of the obligate canonical Notch transcription factor RBPJ. Intracellular signaling analysis revealed increased activation of the mitogenic phosphatidylinositol 3-kinase/protein kinase B and mitogen-activated protein kinase/extracellular signal-regulated kinase pathways following Notch knockdown, with a mitogen-activated protein kinase kinase inhibitor blocking the enhanced proliferation observed in Jag1 knockdown granulosa cells. Activation of YB-1, a known regulator of granulosa cell differentiation genes, was suppressed by Jag1 knockdown. Overall, this study reveals a role of Notch signaling in promoting the differentiation of preovulatory granulosa cells, adding to the diverse functions of Notch in the mammalian ovary.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Chorionic Gonadotropin/pharmacology
- Estradiol/metabolism
- Female
- Gene Expression Regulation, Developmental/drug effects
- Genes, Reporter/drug effects
- Gonadotropins, Equine/pharmacology
- Granulosa Cells/cytology
- Granulosa Cells/drug effects
- Granulosa Cells/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/antagonists & inhibitors
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism
- Jagged-1 Protein/antagonists & inhibitors
- Jagged-1 Protein/genetics
- Jagged-1 Protein/metabolism
- MAP Kinase Signaling System/drug effects
- Mice, Inbred Strains
- Mice, Transgenic
- Progesterone/metabolism
- RNA Interference
- Receptor, Notch2/agonists
- Receptor, Notch2/genetics
- Receptor, Notch2/metabolism
- Receptor, Notch3/agonists
- Receptor, Notch3/genetics
- Receptor, Notch3/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Rexxi D. Prasasya
- Department of Molecular Biosciences and Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| | - Kelly E. Mayo
- Department of Molecular Biosciences and Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
31
|
Fan F, Zhuang J, Zhou P, Liu X, Luo Y. MicroRNA-34a promotes mitochondrial dysfunction-induced apoptosis in human lens epithelial cells by targeting Notch2. Oncotarget 2017; 8:110209-110220. [PMID: 29299142 PMCID: PMC5746377 DOI: 10.18632/oncotarget.22597] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/18/2017] [Indexed: 02/06/2023] Open
Abstract
Purpose Human lens epithelial cell (HLEC) apoptosis is a common pathogenic mechanism in age-related cataracts (ARC). While the function of microRNAs (miRNAs) in the eye is beginning to be explored using miRNA expression array, the role of miR-34a in regulating HLEC apoptosis remains unknown and requires further investigation. Methods Quantitative reverse-transcript polymerase chain reaction (RT-PCR) was used to determine the expression level of miR-34a in cataractous and control samples. MiR-34a mimics and small interfering RNAs were transfected into SRA01/04. Cell apoptosis and oxidative stress were assessed by flow cytometry. The Dual-Luciferase Reporter Assay System was used to confirm whether miR-34a bound to the 3'-UTR of the target gene and blocked its activity. The potential roles of the identified target genes in apoptosis and mitochondria dysfunction were also evaluated. Results The expression of miR-34a increased in lens epithelial samples of ARC compared with the transparent group (cataract 2.41±0.81 vs. control 1.20±0.44, P=0.005). In cultured SRA01/04, miR-34a increased reactive oxygen species production and induced apoptosis (early apoptosis: 45.55%±5.96% vs. 15.85%±4.93%, P<0.01; late apoptosis: 6.10%±2.67% vs. 0.95%±0.42%, P<0.01). Overexpression of miR-34a promoted mitochondria-mediated apoptosis through activation of caspase-9, disruption of the mitochondrial membrane potential, blocking of mitochondrial energy metabolism and enhancement of cytochrome C release. Furthermore, Notch1 and Notch2 were confirmed as putative targets of miR-34a, but only Notch2 was verified as the effector that triggered mitochondria-mediated apoptosis. Conclusion MicroRNA-34a is increased in the cataractous lens and triggers mitochondria-mediated apoptosis and oxidative stress by suppressing Notch2.
Collapse
Affiliation(s)
- Fan Fan
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Myopia Key Laboratory of The Health Ministry and Visual Impairment and Reconstruction Key Laboratory of Shanghai, Shanghai, China
| | - Jianhui Zhuang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Zhou
- Parkway Health Hongqiao Medical Center, Shanghai, China
| | - Xin Liu
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Myopia Key Laboratory of The Health Ministry and Visual Impairment and Reconstruction Key Laboratory of Shanghai, Shanghai, China
| | - Yi Luo
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Myopia Key Laboratory of The Health Ministry and Visual Impairment and Reconstruction Key Laboratory of Shanghai, Shanghai, China
| |
Collapse
|
32
|
Boareto M, Jolly MK, Goldman A, Pietilä M, Mani SA, Sengupta S, Ben-Jacob E, Levine H, Onuchic JN. Notch-Jagged signalling can give rise to clusters of cells exhibiting a hybrid epithelial/mesenchymal phenotype. J R Soc Interface 2017; 13:rsif.2015.1106. [PMID: 27170649 PMCID: PMC4892257 DOI: 10.1098/rsif.2015.1106] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/15/2016] [Indexed: 01/14/2023] Open
Abstract
Metastasis can involve repeated cycles of epithelial-to-mesenchymal transition (EMT) and its reverse mesenchymal-to-epithelial transition. Cells can also undergo partial transitions to attain a hybrid epithelial/mesenchymal (E/M) phenotype that allows the migration of adhering cells to form a cluster of circulating tumour cells. These clusters can be apoptosis-resistant and possess an increased metastatic propensity as compared to the cells that undergo a complete EMT (mesenchymal cells). Hence, identifying the key players that can regulate the formation and maintenance of such clusters may inform anti-metastasis strategies. Here, we devise a mechanism-based theoretical model that links cell–cell communication via Notch-Delta-Jagged signalling with the regulation of EMT. We demonstrate that while both Notch-Delta and Notch-Jagged signalling can induce EMT in a population of cells, only Jagged-dominated Notch signalling, but not Delta-dominated signalling, can lead to the formation of clusters containing hybrid E/M cells. Our results offer possible mechanistic insights into the role of Jagged in tumour progression, and offer a framework to investigate the effects of other microenvironmental signals during metastasis.
Collapse
Affiliation(s)
- Marcelo Boareto
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005-1827, USA Institute of Physics, University of Sao Paulo, Sao Paulo 05508, Brazil
| | - Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005-1827, USA Department of Bioengineering, Rice University, Houston, TX 77005-1827, USA
| | - Aaron Goldman
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Mika Pietilä
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA Metastasis Research Center, MD Anderson Cancer Center, Houston, TX 77025, USA
| | - Shiladitya Sengupta
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Eshel Ben-Jacob
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005-1827, USA School of Physics and Astronomy and The Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005-1827, USA Department of Bioengineering, Rice University, Houston, TX 77005-1827, USA Department of Physics and Astronomy, Rice University, Houston, TX 77005-1827, USA Department of Biosciences, Rice University, Houston, TX 77005-1827, USA
| | - Jose' N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005-1827, USA Department of Chemistry, Rice University, Houston, TX 77005-1827, USA Department of Physics and Astronomy, Rice University, Houston, TX 77005-1827, USA Department of Biosciences, Rice University, Houston, TX 77005-1827, USA
| |
Collapse
|
33
|
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
|
34
|
Bassnett S, Šikić H. The lens growth process. Prog Retin Eye Res 2017; 60:181-200. [PMID: 28411123 DOI: 10.1016/j.preteyeres.2017.04.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 01/17/2023]
Abstract
The factors that regulate the size of organs to ensure that they fit within an organism are not well understood. A simple organ, the ocular lens serves as a useful model with which to tackle this problem. In many systems, considerable variance in the organ growth process is tolerable. This is almost certainly not the case in the lens, which in addition to fitting comfortably within the eyeball, must also be of the correct size and shape to focus light sharply onto the retina. Furthermore, the lens does not perform its optical function in isolation. Its growth, which continues throughout life, must therefore be coordinated with that of other tissues in the optical train. Here, we review the lens growth process in detail, from pioneering clinical investigations in the late nineteenth century to insights gleaned more recently in the course of cell and molecular studies. During embryonic development, the lens forms from an invagination of surface ectoderm. Consequently, the progenitor cell population is located at its surface and differentiated cells are confined to the interior. The interactions that regulate cell fate thus occur within the obligate ellipsoidal geometry of the lens. In this context, mathematical models are particularly appropriate tools with which to examine the growth process. In addition to identifying key growth determinants, such models constitute a framework for integrating cell biological and optical data, helping clarify the relationship between gene expression in the lens and image quality at the retinal plane.
Collapse
Affiliation(s)
- Steven Bassnett
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, USA.
| | - Hrvoje Šikić
- Department of Mathematics, Faculty of Science, University of Zagreb, Croatia
| |
Collapse
|
35
|
Fine-Tuning of the Kaposi's Sarcoma-Associated Herpesvirus Life Cycle in Neighboring Cells through the RTA-JAG1-Notch Pathway. PLoS Pathog 2016; 12:e1005900. [PMID: 27760204 PMCID: PMC5070770 DOI: 10.1371/journal.ppat.1005900] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/27/2016] [Indexed: 12/16/2022] Open
Abstract
Kaposi’s sarcoma (KS)-associated herpesvirus (KSHV) is an oncogenic pathogen that displays latent and lytic life cycles. In KS lesions, infiltrated immune cells, secreted viral and/or cellular cytokines, and hypoxia orchestrate a chronic pro-lytic microenvironment that can promote KSHV reactivation. However, only a small subset of viruses spontaneously undergoes lytic replication in this pro-lytic microenvironment while the majority remains in latency. Here, we show that the expression of the Notch ligand JAG1 is induced by KSHV-encoded replication and transcription activator (RTA) during reactivation. JAG1 up-regulation activates Notch signaling in neighboring cells and prevents viral lytic replication. The suppression of JAG1 and Notch1 with inhibitors or small interfering RNA promotes lytic replication in the presence of RTA induction or under conditions of hypoxia. The underlying mechanism involves the Notch downstream effector hairy and enhancer of split 1 (Hes1), which directly binds lytic gene promoters and attenuates viral lytic gene expression. RTA interacts with lymphoid enhancer-binding factor 1 (LEF1), disrupts LEF1/Groucho/TLE suppressive complexes and releases LEF1 to activate JAG1 expression. Taken together, our results suggest that cells with viral lytic replication can inhibit KSHV reactivation in neighboring cells through an RTA-JAG1-Notch pathway. These data provide insight into the mechanism by which the virus maintains the balance between lytic and latent infection in the pro-lytic tumor microenvironment. KSHV infected cells display significant heterogeneity in viral lytic replication within the universal pro-lytic inflammatory milieu, suggesting that the balance between latency and reactivation is carefully regulated. This fine-tuned regulatory mechanism is essential for KSHV to persist in the host and drive cells to malignancy. In the present study, we show that KSHV can usurp the Notch signaling pathway to inhibit the viral lytic life cycle in neighboring cells. Notch signaling in surrounding cells can be activated through an RTA-JAG1-Notch pathway initiated by cells in which KSHV is reactivated. Activated Notch inhibits KSHV reactivation through its downstream effector Hes1. These findings suggest that the ability of Notch to determine the fate of adjacent cells is hijacked by KSHV to maintain its life cycle, providing a mechanistic explanation for the phenomenon by which only a small fraction of viruses enters lytic replication in the common pro-lytic microenvironment.
Collapse
|
36
|
Pathania M, Wang Y, Simirskii VN, Duncan MK. β1-integrin controls cell fate specification in early lens development. Differentiation 2016; 92:133-147. [PMID: 27596755 PMCID: PMC5159248 DOI: 10.1016/j.diff.2016.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/05/2016] [Accepted: 08/09/2016] [Indexed: 02/03/2023]
Abstract
Integrins are heterodimeric cell surface molecules that mediate cell-extracellular matrix (ECM) adhesion, ECM assembly, and regulation of both ECM and growth factor induced signaling. However, the developmental context of these diverse functions is not clear. Loss of β1-integrin from the lens vesicle (mouse E10.5) results in abnormal exit of anterior lens epithelial cells (LECs) from the cell cycle and their aberrant elongation toward the presumptive cornea by E12.5. These cells lose expression of LEC markers and initiate expression of the Maf (also known as c-Maf) and Prox1 transcription factors as well as other lens fiber cell markers. β1-integrin null LECs also upregulate the ERK, AKT and Smad1/5/8 phosphorylation indicative of BMP and FGF signaling. By E14.5, β1-integrin null lenses have undergone a complete conversion of all lens epithelial cells into fiber cells. These data suggest that shortly after lens vesicle closure, β1-integrin blocks inappropriate differentiation of the lens epithelium into fibers, potentially by inhibiting BMP and/or FGF receptor activation. Thus, β1-integrin has an important role in fine-tuning the response of the early lens to the gradient of growth factors that regulate lens fiber cell differentiation.
Collapse
Affiliation(s)
- Mallika Pathania
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Yan Wang
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Vladimir N Simirskii
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
37
|
Intrinsic and extrinsic regulatory mechanisms are required to form and maintain a lens of the correct size and shape. Exp Eye Res 2016; 156:34-40. [PMID: 27109030 DOI: 10.1016/j.exer.2016.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 04/13/2016] [Accepted: 04/16/2016] [Indexed: 02/04/2023]
Abstract
Understanding how tissues and organs acquire and maintain an appropriate size and shape remains one of the most challenging areas in developmental biology. The eye lens represents an excellent system to provide insights into regulatory mechanisms because in addition to its relative simplicity in cellular composition (being made up of only two forms of cells, epithelial and fiber cells), these cells must become organized to generate the precise spheroidal arrangement that delivers normal lens function. Epithelial and fiber cells also represent spatially distinct proliferation and differentiation compartments, respectively, and an ongoing balance between these domains must be tightly regulated so that the lens achieves and maintains appropriate dimensions during growth and ageing. Recent research indicates that reciprocal inductive interactions mediated by Wnt-Frizzled and Notch-Jagged signaling pathways are important for maintaining and organizing these compartments. The Hippo-Yap pathway has also been implicated in maintaining the epithelial progenitor compartment and regulating growth processes. Thus, whilst some molecules and mechanisms have been identified, further work in this important area is needed to provide a clearer understanding of how lens size and shape is regulated.
Collapse
|
38
|
Murta D, Batista M, Silva E, Trindade A, Henrique D, Duarte A, Lopes-da-Costa L. Notch signaling in the epididymal epithelium regulates sperm motility and is transferred at a distance within epididymosomes. Andrology 2016; 4:314-27. [DOI: 10.1111/andr.12144] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 10/28/2015] [Accepted: 11/09/2015] [Indexed: 12/17/2022]
Affiliation(s)
- D. Murta
- Reproduction and Development; CIISA; Faculty of Veterinary Medicine; University of Lisbon; Lisbon Portugal
- CBIOS - Research Centre for Biosciences and Health Technologies; Faculty of Veterinary Medicine; Lusófona University of Humanities and Technologies; Lisboa Portugal
| | - M. Batista
- Reproduction and Development; CIISA; Faculty of Veterinary Medicine; University of Lisbon; Lisbon Portugal
| | - E. Silva
- Reproduction and Development; CIISA; Faculty of Veterinary Medicine; University of Lisbon; Lisbon Portugal
| | - A. Trindade
- Reproduction and Development; CIISA; Faculty of Veterinary Medicine; University of Lisbon; Lisbon Portugal
- Gulbenkian Institute of Science; Oeiras Portugal
| | - D. Henrique
- Institute of Molecular Medicine; Faculty of Medicine; University of Lisbon; Lisbon Portugal
| | - A. Duarte
- Reproduction and Development; CIISA; Faculty of Veterinary Medicine; University of Lisbon; Lisbon Portugal
- Gulbenkian Institute of Science; Oeiras Portugal
| | - L. Lopes-da-Costa
- Reproduction and Development; CIISA; Faculty of Veterinary Medicine; University of Lisbon; Lisbon Portugal
| |
Collapse
|
39
|
Li J, Zhao D, Guo C, Li J, Mi Y, Zhang C. Involvement of Notch signaling in early chick ovarian follicle development. Cell Biol Int 2015; 40:65-73. [DOI: 10.1002/cbin.10538] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/15/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Jun Li
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Dan Zhao
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Changquan Guo
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Jian Li
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Yuling Mi
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Caiqiao Zhang
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| |
Collapse
|
40
|
Sirakov M, Boussouar A, Kress E, Frau C, Lone IN, Nadjar J, Angelov D, Plateroti M. The thyroid hormone nuclear receptor TRα1 controls the Notch signaling pathway and cell fate in murine intestine. Development 2015; 142:2764-74. [PMID: 26286942 DOI: 10.1242/dev.121962] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Thyroid hormones control various aspects of gut development and homeostasis. The best-known example is in gastrointestinal tract remodeling during amphibian metamorphosis. It is well documented that these hormones act via the TR nuclear receptors, which are hormone-modulated transcription factors. Several studies have shown that thyroid hormones regulate the expression of several genes in the Notch signaling pathway, indicating a possible means by which they participate in the control of gut physiology. However, the mechanisms and biological significance of this control have remained unexplored. Using multiple in vivo and in vitro approaches, we show that thyroid hormones positively regulate Notch activity through the TRα1 receptor. From a molecular point of view, TRα1 indirectly controls Notch1, Dll1, Dll4 and Hes1 expression but acts as a direct transcriptional regulator of the Jag1 gene by binding to a responsive element in the Jag1 promoter. Our findings show that the TRα1 nuclear receptor plays a key role in intestinal crypt progenitor/stem cell biology by controlling the Notch pathway and hence the balance between cell proliferation and cell differentiation.
Collapse
Affiliation(s)
- Maria Sirakov
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Amina Boussouar
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Elsa Kress
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Carla Frau
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Imtiaz Nisar Lone
- Laboratoire de Biologie Moléculaire de la cellule, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon 69007, France
| | - Julien Nadjar
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Dimitar Angelov
- Laboratoire de Biologie Moléculaire de la cellule, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon 69007, France
| | - Michelina Plateroti
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| |
Collapse
|
41
|
Sampaziotis F, de Brito MC, Madrigal P, Bertero A, Saeb-Parsy K, Soares FAC, Schrumpf E, Melum E, Karlsen TH, Bradley JA, Gelson WTH, Davies S, Baker A, Kaser A, Alexander GJ, Hannan NR, Vallier L. Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation. Nat Biotechnol 2015; 33:845-852. [PMID: 26167629 PMCID: PMC4768345 DOI: 10.1038/nbt.3275] [Citation(s) in RCA: 291] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/05/2015] [Indexed: 12/20/2022]
Abstract
The study of biliary disease has been constrained by a lack of primary human cholangiocytes. Here we present an efficient, serum-free protocol for directed differentiation of human induced pluripotent stem cells into cholangiocyte-like cells (CLCs). CLCs show functional characteristics of cholangiocytes, including bile acids transfer, alkaline phosphatase activity, γ-glutamyl-transpeptidase activity and physiological responses to secretin, somatostatin and vascular endothelial growth factor. We use CLCs to model in vitro key features of Alagille syndrome, polycystic liver disease and cystic fibrosis (CF)-associated cholangiopathy. Furthermore, we use CLCs generated from healthy individuals and patients with polycystic liver disease to reproduce the effects of the drugs verapamil and octreotide, and we show that the experimental CF drug VX809 rescues the disease phenotype of CF cholangiopathy in vitro. Our differentiation protocol will facilitate the study of biological mechanisms controlling biliary development, as well as disease modeling and drug screening.
Collapse
Affiliation(s)
- Fotios Sampaziotis
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Miguel Cardoso de Brito
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Pedro Madrigal
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Alessandro Bertero
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Filipa A. C. Soares
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Elisabeth Schrumpf
- Norwegian PSC Research Center, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Melum
- Norwegian PSC Research Center, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tom H. Karlsen
- Norwegian PSC Research Center, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - J. Andrew Bradley
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - William TH Gelson
- Department of Hepatology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Susan Davies
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Alastair Baker
- Child Health Clinical Academic Grouping, King’s Health Partners, Denmark Hill Campus, London, United Kingdom
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Graeme J. Alexander
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas R.F. Hannan
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Ludovic Vallier
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
42
|
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
|
43
|
Su YX, Hou CC, Yang WX. Control of hair cell development by molecular pathways involving Atoh1, Hes1 and Hes5. Gene 2014; 558:6-24. [PMID: 25550047 DOI: 10.1016/j.gene.2014.12.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/23/2014] [Accepted: 12/25/2014] [Indexed: 01/14/2023]
Abstract
Atoh1, Hes1 and Hes5 are crucial for normal inner ear hair cell development. They regulate the expression of each other in a complex network, while they also interact with many other genes and pathways, such as Notch, FGF, SHH, WNT, BMP and RA. This paper summarized molecular pathways that involve Atoh1, Hes1, and Hes5. Some of the pathways and gene regulation mechanisms discussed here were studied in other tissues, yet they might inspire studies in inner ear hair cell development. Thereby, we presented a complex regulatory network involving these three genes, which might be crucial for proliferation and differentiation of inner ear hair cells.
Collapse
Affiliation(s)
- Yi-Xun Su
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cong-Cong Hou
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
44
|
Ware M, Hamdi-Rozé H, Dupé V. Notch signaling and proneural genes work together to control the neural building blocks for the initial scaffold in the hypothalamus. Front Neuroanat 2014; 8:140. [PMID: 25520625 PMCID: PMC4251447 DOI: 10.3389/fnana.2014.00140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/10/2014] [Indexed: 01/25/2023] Open
Abstract
The vertebrate embryonic prosencephalon gives rise to the hypothalamus, which plays essential roles in sensory information processing as well as control of physiological homeostasis and behavior. While patterning of the hypothalamus has received much attention, initial neurogenesis in the developing hypothalamus has mostly been neglected. The first differentiating progenitor cells of the hypothalamus will give rise to neurons that form the nucleus of the tract of the postoptic commissure (nTPOC) and the nucleus of the mammillotegmental tract (nMTT). The formation of these neuronal populations has to be highly controlled both spatially and temporally as these tracts will form part of the ventral longitudinal tract (VLT) and act as a scaffold for later, follower axons. This review will cumulate and summarize the existing data available describing initial neurogenesis in the vertebrate hypothalamus. It is well-known that the Notch signaling pathway through the inhibition of proneural genes is a key regulator of neurogenesis in the vertebrate central nervous system. It has only recently been proposed that loss of Notch signaling in the developing chick embryo causes an increase in the number of neurons in the hypothalamus, highlighting an early function of the Notch pathway during hypothalamus formation. Further analysis in the chick and mouse hypothalamus confirms the expression of Notch components and Ascl1 before the appearance of the first differentiated neurons. Many newly identified proneural target genes were also found to be expressed during neuronal differentiation in the hypothalamus. Given the critical role that hypothalamic neural circuitry plays in maintaining homeostasis, it is particularly important to establish the targets downstream of this Notch/proneural network.
Collapse
Affiliation(s)
- Michelle Ware
- Institut de Génétique et Développement de Rennes, Faculté de Médecine, CNRS UMR6290, Université de Rennes 1 Rennes, France
| | - Houda Hamdi-Rozé
- Institut de Génétique et Développement de Rennes, Faculté de Médecine, CNRS UMR6290, Université de Rennes 1 Rennes, France
| | - Valérie Dupé
- Institut de Génétique et Développement de Rennes, Faculté de Médecine, CNRS UMR6290, Université de Rennes 1 Rennes, France
| |
Collapse
|
45
|
Murta D, Batista M, Trindade A, Silva E, Henrique D, Duarte A, Lopes-da-Costa L. In vivo notch signaling blockade induces abnormal spermatogenesis in the mouse. PLoS One 2014; 9:e113365. [PMID: 25412258 PMCID: PMC4239051 DOI: 10.1371/journal.pone.0113365] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/22/2014] [Indexed: 12/19/2022] Open
Abstract
In a previous study we identified active Notch signaling in key cellular events occurring at adult spermatogenesis. In this study, we evaluated the function of Notch signaling in spermatogenesis through the effects of in vivo Notch blockade. Adult CD1 male mice were either submitted to a long term DAPT (?-secretase inhibitor) or vehicle treatment. Treatment duration was designed to attain one half the time (25 days) or the time (43 days) required to accomplish a complete cycle of spermatogenesis. Blockade of Notch signaling was depicted from decreased transcription of Notch effector genes. Notch signaling blockade disrupted the expression patterns of Notch components in the testis, induced male germ cell fate aberrations, and significantly increased germ cell apoptosis, mainly in the last stages of the spermatogenic cycle, and epididymis spermatozoa morphological defects. These effects were more pronounced following the 43 day than the 25 day DAPT treatment schedule. These results indicate a relevant regulatory role of Notch signaling in mammalian spermatogenesis.
Collapse
Affiliation(s)
- Daniel Murta
- Reproduction and Development, Interdisciplinary Centre of Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Marta Batista
- Reproduction and Development, Interdisciplinary Centre of Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Alexandre Trindade
- Reproduction and Development, Interdisciplinary Centre of Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Gulbenkian Institute of Science, Oeiras, Portugal
| | - Elisabete Silva
- Reproduction and Development, Interdisciplinary Centre of Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Domingos Henrique
- Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - António Duarte
- Reproduction and Development, Interdisciplinary Centre of Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Gulbenkian Institute of Science, Oeiras, Portugal
| | - Luís Lopes-da-Costa
- Reproduction and Development, Interdisciplinary Centre of Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- * E-mail:
| |
Collapse
|
46
|
Lee S, Griep AE. Loss of Dlg-1 in the mouse lens impairs fibroblast growth factor receptor signaling. PLoS One 2014; 9:e97470. [PMID: 24824078 PMCID: PMC4019587 DOI: 10.1371/journal.pone.0097470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/20/2014] [Indexed: 01/21/2023] Open
Abstract
Coordination of cell proliferation, differentiation and survival is essential for normal development and maintenance of tissues in the adult organism. Growth factor receptor tyrosine kinase signaling pathways and planar cell polarity pathways are two regulators of many developmental processes. We have previously shown through analysis of mice conditionally null in the lens for the planar cell polarity gene (PCP), Dlg-1, that Dlg-1 is required for fiber differentiation. Herein, we asked if Dlg-1 is a regulator of the Fibroblast growth factor receptor (Fgfr) signaling pathway, which is known to be required for fiber cell differentiation. Western blot analysis of whole fiber cell extracts from control and Dlg-1 deficient lenses showed that levels of the Fgfr signaling intermediates pErk, pAkt, and pFrs2α, the Fgfr target, Erm, and the fiber cell specific protein, Mip26, were reduced in the Dlg-1 deficient fiber cells. The levels of Fgfr2 were decreased in Dlg-1 deficient lenses compared to controls. Conversely, levels of Fgfr1 in Dlg-1 deficient lenses were increased compared to controls. The changes in Fgfr levels were found to be specifically in the triton insoluble, cytoskeletal associated fraction of Dlg-1 deficient lenses. Immunofluorescent staining of lenses from E13.5 embryos showed that expression levels of pErk were reduced in the transition zone, a region of the lens that exhibits PCP, in the Dlg-1 deficient lenses as compared to controls. In control lenses, immunofluorescent staining for Fgfr2 was observed in the epithelium, transition zone and fibers. By E13.5, the intensity of staining for Fgfr2 was reduced in these regions of the Dlg-1 deficient lenses. Thus, loss of Dlg-1 in the lens impairs Fgfr signaling and leads to altered levels of Fgfrs, suggesting that Dlg-1 is a modulator of Fgfr signaling pathway at the level of the receptors and that Dlg-1 regulates fiber cell differentiation through its role in PCP.
Collapse
Affiliation(s)
- SungKyoung Lee
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Anne E. Griep
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
47
|
Blockage of Notch signaling inhibits the migration and proliferation of retinal pigment epithelial cells. ScientificWorldJournal 2013; 2013:178708. [PMID: 24453806 PMCID: PMC3885266 DOI: 10.1155/2013/178708] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 11/12/2013] [Indexed: 12/03/2022] Open
Abstract
The Notch signaling is an evolutionarily conserved cell-cell communication pathway that plays critical roles in the proliferation, survival, apoptosis, and fate determination of mammalian cells. Retinal pigment epithelial (RPE) cells are responsible for supporting the function of the neural retina and maintaining vision. This study investigated the function of Notch signaling in RPE cells. We found that the members of the Notch signaling pathway components were differentially expressed in RPE cells. Furthermore, blockage of Notch signaling inhibited the migration and proliferation of RPE cells and reduced the expression levels of certain Notch signaling target genes, including HES1, MYC, HEY2, and SOX9. Our data reveal a critical role of Notch signaling in RPE cells, suggesting that targeting Notch signaling may provide a novel approach for the treatment of ophthalmic diseases related to RPE cells.
Collapse
|
48
|
Identification and characterization of FGF2-dependent mRNA: microRNA networks during lens fiber cell differentiation. G3-GENES GENOMES GENETICS 2013; 3:2239-55. [PMID: 24142921 PMCID: PMC3852386 DOI: 10.1534/g3.113.008698] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) and fibroblast growth factor (FGF) signaling regulate a wide range of cellular functions, including cell specification, proliferation, migration, differentiation, and survival. In lens, both these systems control lens fiber cell differentiation; however, a possible link between these processes remains to be examined. Herein, the functional requirement for miRNAs in differentiating lens fiber cells was demonstrated via conditional inactivation of Dicer1 in mouse (Mus musculus) lens. To dissect the miRNA-dependent pathways during lens differentiation, we used a rat (Rattus norvegicus) lens epithelial explant system, induced by FGF2 to differentiate, followed by mRNA and miRNA expression profiling. Transcriptome and miRNome analysis identified extensive FGF2-regulated cellular responses that were both independent and dependent on miRNAs. We identified 131 FGF2-regulated miRNAs. Seventy-six of these miRNAs had at least two in silico predicted and inversely regulated target mRNAs. Genes modulated by the greatest number of FGF-regulated miRNAs include DNA-binding transcription factors Nfib, Nfat5/OREBP, c-Maf, Ets1, and N-Myc. Activated FGF signaling influenced bone morphogenetic factor/transforming growth factor-β, Notch, and Wnt signaling cascades implicated earlier in lens differentiation. Specific miRNA:mRNA interaction networks were predicted for c-Maf, N-Myc, and Nfib (DNA-binding transcription factors); Cnot6, Cpsf6, Dicer1, and Tnrc6b (RNA to miRNA processing); and Ash1l, Med1/PBP, and Kdm5b/Jarid1b/Plu1 (chromatin remodeling). Three miRNAs, including miR-143, miR-155, and miR-301a, down-regulated expression of c-Maf in the 3′-UTR luciferase reporter assays. These present studies demonstrate for the first time global impact of activated FGF signaling in lens cell culture system and predicted novel gene regulatory networks connected by multiple miRNAs that regulate lens differentiation.
Collapse
|
49
|
Lassiter RNT, Stark MR, Zhao T, Zhou CJ. Signaling mechanisms controlling cranial placode neurogenesis and delamination. Dev Biol 2013; 389:39-49. [PMID: 24315854 DOI: 10.1016/j.ydbio.2013.11.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 11/22/2013] [Accepted: 11/23/2013] [Indexed: 01/17/2023]
Abstract
The neurogenic cranial placodes are a unique transient epithelial niche of neural progenitor cells that give rise to multiple derivatives of the peripheral nervous system, particularly, the sensory neurons. Placode neurogenesis occurs throughout an extended period of time with epithelial cells continually recruited as neural progenitor cells. Sensory neuron development in the trigeminal, epibranchial, otic, and olfactory placodes coincides with detachment of these neuroblasts from the encompassing epithelial sheet, leading to delamination and ingression into the mesenchyme where they continue to differentiate as neurons. Multiple signaling pathways are known to direct placodal development. This review defines the signaling pathways working at the finite spatiotemporal period when neuronal selection within the placodes occurs, and neuroblasts concomitantly delaminate from the epithelium. Examining neurogenesis and delamination after initial placodal patterning and specification has revealed a common trend throughout the neurogenic placodes, which suggests that both activated FGF and attenuated Notch signaling activities are required for neurogenesis and changes in epithelial cell adhesion leading to delamination. We also address the varying roles of other pathways such as the Wnt and BMP signaling families during sensory neurogenesis and neuroblast delamination in the differing placodes.
Collapse
Affiliation(s)
- Rhonda N T Lassiter
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children-Northern California, CA 95817, USA; Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Sacramento, CA 95817, USA.
| | - Michael R Stark
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Tianyu Zhao
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children-Northern California, CA 95817, USA; Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Chengji J Zhou
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children-Northern California, CA 95817, USA; Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Sacramento, CA 95817, USA; Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
50
|
Dawes LJ, Sugiyama Y, Lovicu FJ, Harris CG, Shelley EJ, McAvoy JW. Interactions between lens epithelial and fiber cells reveal an intrinsic self-assembly mechanism. Dev Biol 2013; 385:291-303. [PMID: 24211762 DOI: 10.1016/j.ydbio.2013.10.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 10/23/2013] [Accepted: 10/29/2013] [Indexed: 12/12/2022]
Abstract
How tissues and organs develop and maintain their characteristic three-dimensional cellular architecture is often a poorly understood part of their developmental program; yet, as is clearly the case for the eye lens, precise regulation of these features can be critical for function. During lens morphogenesis cells become organized into a polarized, spheroidal structure with a monolayer of epithelial cells overlying the apical tips of elongated fiber cells. Epithelial cells proliferate and progeny that shift below the lens equator differentiate into new fibers that are progressively added to the fiber mass. It is now known that FGF induces epithelial to fiber differentiation; however, it is not fully understood how these two forms of cells assemble into their characteristic polarized arrangement. Here we show that in FGF-treated epithelial explants, elongating fibers become polarized/oriented towards islands of epithelial cells and mimic their polarized arrangement in vivo. Epithelial explants secrete Wnt5 into the culture medium and we show that Wnt5 can promote directed behavior of lens cells. We also show that these explants replicate aspects of the Notch/Jagged signaling activity that has been shown to regulate proliferation of epithelial cells in vivo. Thus, our in vitro study identifies a novel mechanism, intrinsic to the two forms of lens cells, that facilitates self-assembly into the polarized arrangement characteristic of the lens in vivo. In this way the lens, with its relatively simple cellular composition, serves as a useful model to highlight the importance of such intrinsic self-assembly mechanisms in tissue developmental and regenerative processes.
Collapse
Affiliation(s)
- L J Dawes
- Save Sight Institute, University of Sydney, Australia
| | - Y Sugiyama
- Save Sight Institute, University of Sydney, Australia
| | - F J Lovicu
- Discipline of Anatomy and Histology, Bosch Institute, University of Sydney, Australia
| | - C G Harris
- Save Sight Institute, University of Sydney, Australia
| | - E J Shelley
- Save Sight Institute, University of Sydney, Australia
| | - J W McAvoy
- Save Sight Institute, University of Sydney, Australia.
| |
Collapse
|