1
|
Islam ST, Cheheltani S, Cheng C, Fowler VM. Disease-related non-muscle myosin IIA D1424N rod domain mutation, but not R702C motor domain mutation, disrupts mouse ocular lens fiber cell alignment and hexagonal packing. Cytoskeleton (Hoboken) 2024; 81:789-805. [PMID: 38516850 PMCID: PMC11416570 DOI: 10.1002/cm.21853] [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: 01/01/2024] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/23/2024]
Abstract
The mouse ocular lens is an excellent vertebrate model system for studying hexagonal cell packing and shape changes during tissue morphogenesis and differentiation. The lens is composed of two types of cells, epithelial and fiber cells. During the initiation of fiber cell differentiation, lens epithelial cells transform from randomly packed cells to hexagonally shaped and packed cells to form meridional row cells. The meridional row cells further differentiate and elongate into newly formed fiber cells that maintain hexagonal cell shape and ordered packing. In other tissues, actomyosin contractility regulates cell hexagonal packing geometry during epithelial tissue morphogenesis. Here, we use the mouse lens as a model to study the effect of two human disease-related non-muscle myosin IIA (NMIIA) mutations on lens cellular organization during fiber cell morphogenesis and differentiation. We studied genetic knock-in heterozygous mice with NMIIA-R702C motor domain or NMIIA-D1424N rod domain mutations. We observed that while one allele of NMIIA-R702C has no impact on lens meridional row epithelial cell shape and packing, one allele of the NMIIA-D1424N mutation can cause localized defects in cell hexagonal packing. Similarly, one allele of NMIIA-R702C motor domain mutation does not affect lens fiber cell organization while the NMIIA-D1424N mutant proteins disrupt fiber cell organization and packing. Our work demonstrates that disease-related NMIIA rod domain mutations (D1424N or E1841K) disrupt mouse lens fiber cell morphogenesis and differentiation.
Collapse
Affiliation(s)
- Sadia T. Islam
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Sepideh Cheheltani
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Catherine Cheng
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Velia M. Fowler
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
2
|
Greiling TM, Clark JM, Clark JI. The significance of growth shells in development of symmetry, transparency, and refraction of the human lens. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1434327. [PMID: 39100140 PMCID: PMC11294239 DOI: 10.3389/fopht.2024.1434327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/27/2024] [Indexed: 08/06/2024]
Abstract
Human visual function depends on the biological lens, a biconvex optical element formed by coordinated, synchronous generation of growth shells produced from ordered cells at the lens equator, the distal edge of the epithelium. Growth shells are comprised of straight (St) and S-shaped (SSh) lens fibers organized in highly symmetric, sinusoidal pattern which optimizes both the refractile, transparent structure and the unique microcirculation that regulates hydration and nutrition over the lifetime of an individual. The fiber cells are characterized by diversity in composition and age. All fiber cells remain interconnected in their growth shells throughout the life of the adult lens. As an optical element, cellular differentiation is constrained by the physical properties of light and its special development accounts for its characteristic symmetry, gradient of refractive index (GRIN), short range transparent order (SRO), and functional longevity. The complex sinusoidal structure is the basis for the lens microcirculation required for the establishment and maintenance of image formation.
Collapse
Affiliation(s)
- Teri M. Greiling
- Department of Dermatology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Judy M. Clark
- Department of Biological Structure, University of Washington, Seattle, WA, United States
| | - John I. Clark
- Department of Biological Structure, University of Washington, Seattle, WA, United States
- Department of Biological Structure & Ophthalmology, School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
3
|
Islam ST, Cheng C, Parreno J, Fowler VM. Nonmuscle Myosin IIA Regulates the Precise Alignment of Hexagonal Eye Lens Epithelial Cells During Fiber Cell Formation and Differentiation. Invest Ophthalmol Vis Sci 2023; 64:20. [PMID: 37070941 PMCID: PMC10123325 DOI: 10.1167/iovs.64.4.20] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/27/2023] [Indexed: 04/19/2023] Open
Abstract
Purpose Epithelial cells in the equatorial region of the ocular lens undergo a remarkable transition from randomly packed cells into precisely aligned and hexagon-shaped cells organized into meridional rows. We investigated the function of nonmuscle myosin IIA (encoded by Myh9) in regulating equatorial epithelial cell alignment to form meridional rows during secondary fiber cell morphogenesis. Methods We used genetic knock-in mice to study a common human Myh9 mutation, E1841K, in the rod domain. The E1841K mutation disrupts bipolar filament assembly. Lens shape, clarity, and stiffness were evaluated, and Western blots were used to determine the level of normal and mutant myosins. Cryosections and lens whole mounts were stained and imaged by confocal microscopy to investigate cell shape and organization. Results We observed no obvious changes in lens size, shape, and biomechanical properties (stiffness and resilience) between the control and nonmuscle myosin IIA-E1841K mutant mice at 2 months of age. Surprisingly, we found misalignment and disorder of fiber cells in heterozygous and homozygous mutant lenses. Further analysis revealed misshapen equatorial epithelial cells that cause disorientation of the meridional rows before fiber cell differentiation in homozygous mutant lenses. Conclusions Our data indicate that nonmuscle myosin IIA bipolar filament assembly is required for the precise alignment of the meridional rows at the lens equator and that the organization of lens fiber cells depends on the proper patterning of meridional row epithelial cells. These data also suggest that lens fiber cell organization and a hexagonal shape are not required for normal lens size, shape transparency, or biomechanical properties.
Collapse
Affiliation(s)
- Sadia T. Islam
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Catherine Cheng
- School of Optometry and Vision Science Program, Indiana University, Bloomington, Indiana, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Justin Parreno
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Velia M. Fowler
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| |
Collapse
|
4
|
Parreno J, Emin G, Vu MP, Clark JT, Aryal S, Patel SD, Cheng C. Methodologies to unlock the molecular expression and cellular structure of ocular lens epithelial cells. Front Cell Dev Biol 2022; 10:983178. [PMID: 36176273 PMCID: PMC9514789 DOI: 10.3389/fcell.2022.983178] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/16/2022] [Indexed: 01/25/2023] Open
Abstract
The transparent ocular lens in the anterior chamber of the eye is responsible for fine focusing of light onto the retina. The lens is entirely cellular with bulk of the tissue composed of fiber cells, and the anterior hemisphere of the lens is covered by a monolayer of epithelial cells. Lens epithelial cells are important for maintaining fiber cell homeostasis and for continual growth of the lens tissue throughout life. Cataracts, defined as any opacity in the lens, remain the leading cause of blindness in the world. Following cataract surgery, lens epithelial cells can undergo a process of epithelial-to-mesenchymal transition (EMT), leading to secondary cataracts due to posterior capsular opacification (PCO). Since the epithelial cells make up only a small fraction of the lens, specialized techniques are required to study lens epithelial cell biology and pathology. Studies using native lens epithelial cells often require pooling of samples to obtain enough cells to make sufficient samples for traditional molecular biology techniques. Here, we provide detailed protocols that enable the study of native mouse lens epithelial cells, including immunostaining of the native lens epithelium in flat mounts, extraction of RNA and proteins from pairs of lens epithelial monolayers, and isolation of lens epithelial cells for primary culture. These protocols will enable researchers to gain better insight on representative molecular expression and cellular structure of lens epithelial cells. We also provide comparative data between native, primary culture, and immortalized lens epithelial cells and discuss the advantages and disadvantages of each technique presented.
Collapse
Affiliation(s)
- Justin Parreno
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
- *Correspondence: Justin Parreno, ; Catherine Cheng,
| | - Grace Emin
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Michael P. Vu
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN, United States
| | - Jackson T. Clark
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN, United States
| | - Sandeep Aryal
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shaili D. Patel
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Catherine Cheng
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN, United States
- *Correspondence: Justin Parreno, ; Catherine Cheng,
| |
Collapse
|
5
|
Cheng C, Gao J, Sun X, Mathias RT. Eph-ephrin Signaling Affects Eye Lens Fiber Cell Intracellular Voltage and Membrane Conductance. Front Physiol 2021; 12:772276. [PMID: 34899394 PMCID: PMC8656704 DOI: 10.3389/fphys.2021.772276] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/27/2021] [Indexed: 01/07/2023] Open
Abstract
The avascular eye lens generates its own microcirculation that is required for maintaining lifelong lens transparency. The microcirculation relies on sodium ion flux, an extensive network of gap junction (GJ) plaques between lens fiber cells and transmembrane water channels. Disruption of connexin proteins, the building blocks of GJs, or aquaporins, which make up water and adhesion channels, lead to lens opacification or cataracts. Recent studies have revealed that disruption of Eph-ephrin signaling, in particular the receptor EphA2 and the ligand ephrin-A5, in humans and mice lead to congenital and age-related cataracts. We investigated whether changes in lens transparency in EphA2 or ephrin-A5 knockout (–/–) mice is related to changes in GJ coupling and lens fluid and ion homeostasis. Immunostaining revealed changes in connexin 50 (Cx50) subcellular localization in EphA2–/– peripheral lens fibers and alteration in aquaporin 0 (Aqp0) staining patterns in ephrin-A5–/– and EphA2–/– inner mature fiber cells. Surprisingly, there was no obvious change in GJ coupling in knockout lenses. However, there were changes in fiber cell membrane conductance and intracellular voltage in knockout lenses from 3-month-old mice. These knockout lenses displayed decreased conductance of mature fiber membranes and were hyperpolarized compared to control lenses. This is the first demonstration that the membrane conductance of lens fibers can be regulated. Together these data suggest that EphA2 may be needed for normal Cx50 localization to the cell membrane and that conductance of lens fiber cells requires normal Eph-ephrin signaling and water channel localization.
Collapse
Affiliation(s)
- Catherine Cheng
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN, United States
| | - Junyuan Gao
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, NY, United States
| | - Xiurong Sun
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, NY, United States
| | - Richard T Mathias
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, NY, United States
| |
Collapse
|
6
|
Karnam S, Maddala R, Stiber JA, Rao PV. Drebrin, an actin-binding protein, is required for lens morphogenesis and growth. Dev Dyn 2021; 250:1600-1617. [PMID: 33896079 PMCID: PMC8542647 DOI: 10.1002/dvdy.353] [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: 01/06/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Lens morphogenesis, architecture, and clarity are known to be critically dependent on actin cytoskeleton organization and cell adhesive interactions. There is limited knowledge, however regarding the identity and role of key proteins regulating actin cytoskeletal organization in the lens. This study investigated the role of drebrin, a developmentally regulated actin-binding protein, in mouse lens development by generating and characterizing a conditional knockout (cKO) mouse model using the Cre-LoxP recombination approach. RESULTS Drebrin E, a splice variant of DBN1 is a predominant isoform expressed in the mouse lens and exhibits a maturation-dependent downregulation. Drebrin co-distributes with actin in both epithelium and fibers. Conditional deficiency (both haploinsufficiency and complete absence) of drebrin results in disrupted lens morphogenesis leading to cataract and microphthalmia. The drebrin cKO lens reveals a dramatic decrease in epithelial height and width, E-cadherin, and proliferation, and increased apoptotic cell death and expression of α-smooth muscle actin, together with severely impaired fiber cell organization, polarity, and cell-cell adhesion. CONCLUSIONS This study demonstrates the requirement of drebrin in lens development and growth, with drebrin deficiency leading to impaired lens morphogenesis and microphthalmia.
Collapse
Affiliation(s)
- Shruthi Karnam
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
| | - Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
| | - Jonathan A Stiber
- Department of Medicine, Duke University School of Medicine, Durham, NC. USA
| | - Ponugoti V Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC. USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC. USA
| |
Collapse
|
7
|
Stopka W, Libby T, Lin S, Wang E, Xia CH, Gong X. Age-related changes of lens stiffness in wild-type and Cx46 knockout mice. Exp Eye Res 2021; 212:108777. [PMID: 34597677 PMCID: PMC10054276 DOI: 10.1016/j.exer.2021.108777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
We have investigated how connexin 46 (Cx46) regulates lens stiffness by studying different Cx46 knockout (Cx46KO) mice. A modified muscle lever system was used to determine the lens stiffness of wild-type (WT) and Cx46KO mice at the C57BL/6J (B6) and the 129SvJae (129) strain backgrounds according to total lens displacement at the point of maximum force when fresh lenses were compressed with a maximum of 2 mN of force. In comparison to B6-WT controls, young and old B6-Cx46KO lenses showed 23% and 28% reductions in lens displacement, respectively. Comparing to 129-WT controls, old 129-Cx46KO lenses showed 50% reduction in the lens displacement while young 129-Cx46KO lenses displayed similar displacement. Old B6-Cx46KO and old 129-Cx46KO lenses showed almost identical lens displacement, 128 μm versus 127 μm. Morphological data revealed unique changes of peripheral fiber cell shapes in young B6-WT lenses but not in young B6-Cx46KO, 129-WT and 129-Cx46KO lenses. This work reveals Cx46 deletion increases the lens stiffness in both young and old mice at B6 strain background but only in old mice at 129 strain background which contains intermediate filament CP49 gene deletion. Cx46 impairment increases old mouse lens stiffness and may contribute to the development of presbyopia.
Collapse
Affiliation(s)
- Wiktor Stopka
- UC Berkeley - UCSF Graduate Program in Bioengineering, University of California, Berkeley, CA, USA
| | - Tom Libby
- Center for Biological Inspiration in Education and Research (CiBER), University of California, Berkeley, CA, USA
| | - Stephanie Lin
- School of Optometry and Vision Science Program, University of California, Berkeley, CA, USA
| | - Eddie Wang
- School of Optometry and Vision Science Program, University of California, Berkeley, CA, USA
| | - Chun-Hong Xia
- School of Optometry and Vision Science Program, University of California, Berkeley, CA, USA
| | - Xiaohua Gong
- UC Berkeley - UCSF Graduate Program in Bioengineering, University of California, Berkeley, CA, USA; School of Optometry and Vision Science Program, University of California, Berkeley, CA, USA.
| |
Collapse
|
8
|
Martis RM, Li B, Donaldson PJ, Lim JCH. Early Onset of Age-Related Cataracts in Cystine/Glutamate Antiporter Knockout Mice. Invest Ophthalmol Vis Sci 2021; 62:23. [PMID: 34156426 PMCID: PMC8237109 DOI: 10.1167/iovs.62.7.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Purpose The purpose of this study was to determine the importance of the xCT is a subunit. The cystine/glutamate antiporter is actually system xc-xCT subunit of the cystine/glutamate antiporter in maintaining redox balance by investigating the effects of the loss of xCT on lens transparency and cystine/cysteine balance in the aqueous humour. Methods C57Bl/6 wild-type and xCT knockout mice at five age groups (6 weeks to 12 months) were used. Lens transparency was examined using a slit-lamp and morphological changes visualized by immunolabelling and confocal microscopy. Quantification of glutathione in lenses and cysteine and cystine levels in the aqueous was conducted by liquid chromatography tandem mass spectrometry (LC-MS/MS). Results Slit-lamp examinations revealed that 3-month-old wild-type mice and xCT knockout mice lenses exhibited an anterior localized cataract. The frequency of this cataract significantly increased in the knockout mice compared to the wild-type mice. Morphological studies revealed a localized swelling of the lens fiber cells at the anterior pole. Glutathione levels in whole lenses were similar between wild-type and knockout mice. However, glutathione levels were significantly decreased at 3 months in the knockout mice in the lens epithelium compared to the wild-type mice. Aqueous cysteine levels remained similar between wild-type and knockout mice at all age groups, whereas cystine levels were significantly increased in 3-, 9-, and 12-month-old knockout mice compared to wild-type mice. Conclusions Loss of xCT resulted in the depletion of glutathione in the epithelium and an oxidative shift in the cysteine/cystine ratio of the aqueous. Together, these oxidative changes may contribute to the accelerated development of an anterior cataract in knockout mice, which appears to be a normal feature of aging in wild-type mice.
Collapse
Affiliation(s)
- Renita Maria Martis
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Bo Li
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Paul James Donaldson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Julie Ching-Hsia Lim
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|
9
|
Cvekl A, Eliscovich C. Crystallin gene expression: Insights from studies of transcriptional bursting. Exp Eye Res 2021; 207:108564. [PMID: 33894228 DOI: 10.1016/j.exer.2021.108564] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/05/2021] [Accepted: 03/22/2021] [Indexed: 01/26/2023]
Abstract
Cellular differentiation is marked by temporally and spatially regulated gene expression. The ocular lens is one of the most powerful mammalian model system since it is composed from only two cell subtypes, called lens epithelial and fiber cells. Lens epithelial cells differentiate into fiber cells through a series of spatially and temporally orchestrated processes, including massive production of crystallins, cellular elongation and the coordinated degradation of nuclei and other organelles. Studies of transcriptional and posttranscriptional gene regulatory mechanisms in lens provide a wide range of opportunities to understand global molecular mechanisms of gene expression as steady-state levels of crystallin mRNAs reach very high levels comparable to globin genes in erythrocytes. Importantly, dysregulation of crystallin gene expression results in lens structural abnormalities and cataracts. The mRNA life cycle is comprised of multiple stages, including transcription, splicing, nuclear export into cytoplasm, stabilization, localization, translation and ultimate decay. In recent years, development of modern mRNA detection methods with single molecule and single cell resolution enabled transformative studies to visualize the mRNA life cycle to generate novel insights into the sequential regulatory mechanisms of gene expression during embryogenesis. This review is focused on recent major advancements in studies of transcriptional bursting in differentiating lens fiber cells, analysis of nascent mRNA expression from bi-directional promoters, transient nuclear accumulation of specific mRNAs, condensation of chromatin prior lens fiber cell denucleation, and outlines future studies to probe the interactions of individual mRNAs with specific RNA-binding proteins (RBPs) in the cytoplasm and regulation of translation and mRNA decay.
Collapse
Affiliation(s)
- Ales Cvekl
- Department of Ophthalmology and VIsual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Carolina Eliscovich
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| |
Collapse
|
10
|
Gupta A, Ruminski D, Jimenez Villar A, Duarte Toledo R, Manzanera S, Panezai S, Mompean J, Artal P, Grulkowski I. In vivo SS-OCT imaging of crystalline lens sutures. BIOMEDICAL OPTICS EXPRESS 2020; 11:5388-5400. [PMID: 33149958 PMCID: PMC7587285 DOI: 10.1364/boe.401254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 05/07/2023]
Abstract
We demonstrate in vivo three-dimensional (3-D) visualization of crystalline lens sutures in healthy eyes using swept source optical coherence tomography (SS-OCT). Volumetric data sets of the crystalline lenses were acquired and processed to obtain enhanced contrast projection images and to extract suture patterns in both anterior and posterior lens. The results presented different types of the sutures including Y-sutures, simple and complex star sutures. Age-related changes in suture arrangement were characterized quantitatively. Crystalline lens suture imaging with SS-OCT might be a useful tool in fundamental studies on development and ageing of human lens.
Collapse
Affiliation(s)
- Ashish Gupta
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, ul. Grudziądzka 5, 87-100 Toruń, Poland
| | - Daniel Ruminski
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, ul. Grudziądzka 5, 87-100 Toruń, Poland
| | - Alfonso Jimenez Villar
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, ul. Grudziądzka 5, 87-100 Toruń, Poland
| | - Raúl Duarte Toledo
- Laboratorio de Óptica, Instituto Universitario de Investigación en Óptica y Nanofísica, Universidad de Murcia, Campus de Espinardo, E-30100, Murcia, Spain
| | - Silvestre Manzanera
- Laboratorio de Óptica, Instituto Universitario de Investigación en Óptica y Nanofísica, Universidad de Murcia, Campus de Espinardo, E-30100, Murcia, Spain
| | - Spozmai Panezai
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, ul. Grudziądzka 5, 87-100 Toruń, Poland
| | - Juan Mompean
- Laboratorio de Óptica, Instituto Universitario de Investigación en Óptica y Nanofísica, Universidad de Murcia, Campus de Espinardo, E-30100, Murcia, Spain
| | - Pablo Artal
- Laboratorio de Óptica, Instituto Universitario de Investigación en Óptica y Nanofísica, Universidad de Murcia, Campus de Espinardo, E-30100, Murcia, Spain
| | - Ireneusz Grulkowski
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, ul. Grudziądzka 5, 87-100 Toruń, Poland
| |
Collapse
|
11
|
Barnum CE, Al Saai S, Patel SD, Cheng C, Anand D, Xu X, Dash S, Siddam AD, Glazewski L, Paglione E, Polson SW, Chuma S, Mason RW, Wei S, Batish M, Fowler VM, Lachke SA. The Tudor-domain protein TDRD7, mutated in congenital cataract, controls the heat shock protein HSPB1 (HSP27) and lens fiber cell morphology. Hum Mol Genet 2020; 29:2076-2097. [PMID: 32420594 PMCID: PMC7390939 DOI: 10.1093/hmg/ddaa096] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/10/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Mutations of the RNA granule component TDRD7 (OMIM: 611258) cause pediatric cataract. We applied an integrated approach to uncover the molecular pathology of cataract in Tdrd7-/- mice. Early postnatal Tdrd7-/- animals precipitously develop cataract suggesting a global-level breakdown/misregulation of key cellular processes. High-throughput RNA sequencing integrated with iSyTE-bioinformatics analysis identified the molecular chaperone and cytoskeletal modulator, HSPB1, among high-priority downregulated candidates in Tdrd7-/- lens. A protein fluorescence two-dimensional difference in-gel electrophoresis (2D-DIGE)-coupled mass spectrometry screen also identified HSPB1 downregulation, offering independent support for its importance to Tdrd7-/- cataractogenesis. Lens fiber cells normally undergo nuclear degradation for transparency, posing a challenge: how is their cell morphology, also critical for transparency, controlled post-nuclear degradation? HSPB1 functions in cytoskeletal maintenance, and its reduction in Tdrd7-/- lens precedes cataract, suggesting cytoskeletal defects may contribute to Tdrd7-/- cataract. In agreement, scanning electron microscopy (SEM) revealed abnormal fiber cell morphology in Tdrd7-/- lenses. Further, abnormal phalloidin and wheat germ agglutinin (WGA) staining of Tdrd7-/- fiber cells, particularly those exhibiting nuclear degradation, reveals distinct regulatory mechanisms control F-actin cytoskeletal and/or membrane maintenance in post-organelle degradation maturation stage fiber cells. Indeed, RNA immunoprecipitation identified Hspb1 mRNA in wild-type lens lysate TDRD7-pulldowns, and single-molecule RNA imaging showed co-localization of TDRD7 protein with cytoplasmic Hspb1 mRNA in differentiating fiber cells, suggesting that TDRD7-ribonucleoprotein complexes may be involved in optimal buildup of key factors. Finally, Hspb1 knockdown in Xenopus causes eye/lens defects. Together, these data uncover TDRD7's novel upstream role in elevation of stress-responsive chaperones for cytoskeletal maintenance in post-nuclear degradation lens fiber cells, perturbation of which causes early-onset cataracts.
Collapse
Affiliation(s)
- Carrie E Barnum
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Salma Al Saai
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Shaili D Patel
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Catherine Cheng
- School of Optometry, Indiana University, Bloomington, IN 47405, USA
| | - Deepti Anand
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Xiaolu Xu
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Soma Dash
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Archana D Siddam
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Lisa Glazewski
- Nemours Biomedical Research Department, Alfred I duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Emily Paglione
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Shawn W Polson
- Center for Bioinformatics & Computational Biology, University of Delaware, Newark, DE 19716, USA
| | - Shinichiro Chuma
- Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Robert W Mason
- Nemours Biomedical Research Department, Alfred I duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Mona Batish
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE 19716, USA
| | - Velia M Fowler
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Salil A Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Center for Bioinformatics & Computational Biology, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
12
|
Lu F, Cui D, Mu B, Zhao L, Mu P. Downregulation of TMOD1 promotes cell motility and cell proliferation in cervical cancer cells. Oncol Lett 2020; 19:3339-3348. [PMID: 32218869 DOI: 10.3892/ol.2020.11410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 01/23/2020] [Indexed: 12/30/2022] Open
Abstract
Tropomodulin-1 (TMOD1) is a key regulator of actin dynamics, which caps the pointed end of actin filaments. TMOD1 has been reported to be involved in several cellular processes, including neurite outgrowth, spine formation and cell migration. Increasing evidence demonstrates that TMOD1 is implicated in several aspects of cancer development. The present study aimed to investigate the role of TMOD1 in cervical cancer. HeLa and CaSki cell lines, derived from human cervical cancer, were used to evaluate the function of TMOD1. Cell motility was measured via a wound-healing assay, with the TMOD1 short hairpin (sh)RNAs transfected cells. Subsequently, cell proliferation was assessed using low serum cell culture condition, while cell cycle distribution was analyzed via flow cytometry. The results demonstrated that downregulated TMOD1 promoted cell motility and proliferation, which is attributed to promotion of G1/S phase transition in HeLa and CaSki cells. Furthermore, it was indicated that co-expression of shRNA resistant TMOD1 rescued these phenomena. The clinical data demonstrated that high TMOD1 expression is associated with good pathological status in patients with cervical cancer. Overall, the results of the present study indicated that TMOD1 may act as a tumor suppressor in cervical cancer, whereby its downregulated expression was demonstrated to have direct effects on cell motility and cell proliferation. These results provide new evidence for the prognostic prediction of cervical cancer, which may serve as a promising therapeutic strategy for patients with cervical cancer.
Collapse
Affiliation(s)
- Fangjin Lu
- Department of Pharmacology, Shenyang Medical College, Shenyang, Liaoning 110034, P.R. China
| | - Dandan Cui
- Department of Maternity, Shenyang Women and Children's Health Hospital, Shenyang, Liaoning 110014, P.R. China
| | - Bin Mu
- Shanghai Zhaohui Pharmaceutical Co., Ltd., Shanghai 201900, P.R. China
| | - Lu Zhao
- Department of Biochemistry and Molecular Biology, Basic Medical School, Shenyang Medical College, Shenyang, Liaoning 110034, P.R. China
| | - Ping Mu
- Department of Biochemistry and Molecular Biology, Basic Medical School, Shenyang Medical College, Shenyang, Liaoning 110034, P.R. China.,Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi 4660065, Japan
| |
Collapse
|
13
|
Kumari R, Jiu Y, Carman PJ, Tojkander S, Kogan K, Varjosalo M, Gunning PW, Dominguez R, Lappalainen P. Tropomodulins Control the Balance between Protrusive and Contractile Structures by Stabilizing Actin-Tropomyosin Filaments. Curr Biol 2020; 30:767-778.e5. [PMID: 32037094 DOI: 10.1016/j.cub.2019.12.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/06/2019] [Accepted: 12/16/2019] [Indexed: 02/08/2023]
Abstract
Eukaryotic cells have diverse protrusive and contractile actin filament structures, which compete with one another for a limited pool of actin monomers. Numerous actin-binding proteins regulate the dynamics of actin structures, including tropomodulins (Tmods), which cap the pointed end of actin filaments. In striated muscles, Tmods prevent actin filaments from overgrowing, whereas in non-muscle cells, their function has remained elusive. Here, we identify two Tmod isoforms, Tmod1 and Tmod3, as key components of contractile stress fibers in non-muscle cells. Individually, Tmod1 and Tmod3 can compensate for one another, but their simultaneous depletion results in disassembly of actin-tropomyosin filaments, loss of force-generating stress fibers, and severe defects in cell morphology. Knockout-rescue experiments reveal that Tmod's interaction with tropomyosin is essential for its role in the stabilization of actin-tropomyosin filaments in cells. Thus, in contrast to their role in muscle myofibrils, in non-muscle cells, Tmods bind actin-tropomyosin filaments to protect them from depolymerizing, not elongating. Furthermore, loss of Tmods shifts the balance from linear actin-tropomyosin filaments to Arp2/3 complex-nucleated branched networks, and this phenotype can be partially rescued by inhibiting the Arp2/3 complex. Collectively, the data reveal that Tmods are essential for the maintenance of contractile actomyosin bundles and that Tmod-dependent capping of actin-tropomyosin filaments is critical for the regulation of actin homeostasis in non-muscle cells.
Collapse
Affiliation(s)
- Reena Kumari
- HiLIFE Institute of Biotechnology, University of Helsinki, PO Box 56, 00014 Helsinki, Finland
| | - Yaming Jiu
- HiLIFE Institute of Biotechnology, University of Helsinki, PO Box 56, 00014 Helsinki, Finland; CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Life Science Research Building 320, Yueyang Road, Xuhui District, 200031 Shanghai, China; University of Chinese Academy of Sciences, Yuquan Road No.19(A), Shijingshan District, 100049 Beijing, China
| | - Peter J Carman
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 728 Clinical Research Bldg, 415 Curie Boulevard, Philadelphia, PA 19104, USA; Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sari Tojkander
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Agnes Sjöberginkatu 2, 00014 Helsinki, Finland
| | - Konstantin Kogan
- HiLIFE Institute of Biotechnology, University of Helsinki, PO Box 56, 00014 Helsinki, Finland
| | - Markku Varjosalo
- HiLIFE Institute of Biotechnology, University of Helsinki, PO Box 56, 00014 Helsinki, Finland
| | - Peter W Gunning
- School of Medical Sciences, UNSW, Sydney, Wallace Wurth Building, Sydney, NSW 2052, Australia
| | - Roberto Dominguez
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 728 Clinical Research Bldg, 415 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, PO Box 56, 00014 Helsinki, Finland.
| |
Collapse
|
14
|
Zhao Y, Wilmarth PA, Cheng C, Limi S, Fowler VM, Zheng D, David LL, Cvekl A. Proteome-transcriptome analysis and proteome remodeling in mouse lens epithelium and fibers. Exp Eye Res 2019; 179:32-46. [PMID: 30359574 PMCID: PMC6360118 DOI: 10.1016/j.exer.2018.10.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 08/31/2018] [Accepted: 10/20/2018] [Indexed: 12/21/2022]
Abstract
Epithelial cells and differentiated fiber cells represent distinct compartments in the ocular lens. While previous studies have revealed proteins that are preferentially expressed in epithelial vs. fiber cells, a comprehensive proteomics library comparing the molecular compositions of epithelial vs. fiber cells is essential for understanding lens formation, function, disease and regenerative potential, and for efficient differentiation of pluripotent stem cells for modeling of lens development and pathology in vitro. To compare protein compositions between the lens epithelium and fibers, we employed tandem mass spectrometry (2D-LC/MS) analysis of microdissected mouse P0.5 lenses. Functional classifications of the top 525 identified proteins into gene ontology categories by molecular processes and subcellular localizations, were adapted for the lens. Expression levels of both epithelial and fiber proteomes were compared with whole lens proteome and mRNA levels using E14.5, E16.5, E18.5, and P0.5 RNA-Seq data sets. During this developmental time window, multiple complex biosynthetic and catabolic processes generate the molecular and structural foundation for lens transparency. As expected, crystallins showed a high correlation between their mRNA and protein levels. Comprehensive data analysis confirmed and/or predicted roles for transcription factors (TFs), RNA-binding proteins (e.g. Carhsp1), translational apparatus including ribosomal heterogeneity and initiation factors, microtubules, cytoskeletal [e.g. non-muscle myosin IIA heavy chain (Myh9) and βB2-spectrin (Sptbn2)] and membrane proteins in lens formation and maturation. Our data highlighted many proteins with unknown functions in the lens that were preferentially enriched in epithelium or fibers, setting the stage for future studies to further dissect the roles of these proteins in fiber cell differentiation vs. epithelial cell maintenance. In conclusion, the present proteomic datasets represent the first mouse lens epithelium and fiber cell proteomes, establish comparative analyses of protein and RNA-Seq data, and characterize the major proteome remodeling required to form the mature lens fiber cells.
Collapse
Affiliation(s)
- Yilin Zhao
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Phillip A Wilmarth
- Department of Biochemistry & Molecular Biology, Oregon Health Sciences University, 3181 Southwest Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Catherine Cheng
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Saima Limi
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Velia M Fowler
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Deyou Zheng
- Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Neurology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Larry L David
- Department of Biochemistry & Molecular Biology, Oregon Health Sciences University, 3181 Southwest Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Ales Cvekl
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
15
|
Cheng C, Nowak RB, Amadeo MB, Biswas SK, Lo WK, Fowler VM. Tropomyosin 3.5 protects the F-actin networks required for tissue biomechanical properties. J Cell Sci 2018; 131:jcs222042. [PMID: 30333143 PMCID: PMC6288072 DOI: 10.1242/jcs.222042] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022] Open
Abstract
Tropomyosins (Tpms) stabilize F-actin and regulate interactions with other actin-binding proteins. The eye lens changes shape in order to focus light to transmit a clear image, and thus lens organ function is tied to its biomechanical properties, presenting an opportunity to study Tpm functions in tissue mechanics. Mouse lenses contain Tpm3.5 (also known as TM5NM5), a previously unstudied isoform encoded by Tpm3, which is associated with F-actin on lens fiber cell membranes. Decreased levels of Tpm3.5 lead to softer and less mechanically resilient lenses that are unable to resume their original shape after compression. While cell organization and morphology appear unaffected, Tmod1 dissociates from the membrane in Tpm3.5-deficient lens fiber cells resulting in reorganization of the spectrin-F-actin and α-actinin-F-actin networks at the membrane. These rearranged F-actin networks appear to be less able to support mechanical load and resilience, leading to an overall change in tissue mechanical properties. This is the first in vivo evidence that a Tpm protein is essential for cell biomechanical stability in a load-bearing non-muscle tissue, and indicates that Tpm3.5 protects mechanically stable, load-bearing F-actin in vivoThis article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Catherine Cheng
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Roberta B Nowak
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael B Amadeo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sondip K Biswas
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30314, USA
| | - Woo-Kuen Lo
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30314, USA
| | - Velia M Fowler
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
16
|
Tropomodulin Isoform-Specific Regulation of Dendrite Development and Synapse Formation. J Neurosci 2018; 38:10271-10285. [PMID: 30301754 DOI: 10.1523/jneurosci.3325-17.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 11/21/2022] Open
Abstract
Neurons of the CNS elaborate highly branched dendritic arbors that host numerous dendritic spines, which serve as the postsynaptic platform for most excitatory synapses. The actin cytoskeleton plays an important role in dendrite development and spine formation, but the underlying mechanisms remain incompletely understood. Tropomodulins (Tmods) are a family of actin-binding proteins that cap the slow-growing (pointed) end of actin filaments, thereby regulating the stability, length, and architecture of complex actin networks in diverse cell types. Three members of the Tmod family, Tmod1, Tmod2, and Tmod3 are expressed in the vertebrate CNS, but their function in neuronal development is largely unknown. In this study, we present evidence that Tmod1 and Tmod2 exhibit distinct roles in regulating spine development and dendritic arborization, respectively. Using rat hippocampal tissues from both sexes, we find that Tmod1 and Tmod2 are expressed with distinct developmental profiles: Tmod2 is expressed early during hippocampal development, whereas Tmod1 expression coincides with synaptogenesis. We then show that knockdown of Tmod2, but not Tmod1, severely impairs dendritic branching. Both Tmod1 and Tmod2 are localized to a distinct subspine region where they regulate local F-actin stability. However, the knockdown of Tmod1, but not Tmod2, disrupts spine morphogenesis and impairs synapse formation. Collectively, these findings demonstrate that regulation of the actin cytoskeleton by different members of the Tmod family plays an important role in distinct aspects of dendrite and spine development.SIGNIFICANCE STATEMENT The Tropomodulin family of molecules is best known for controlling the length and stability of actin myofilaments in skeletal muscles. While several Tropomodulin members are expressed in the brain, fundamental knowledge about their role in neuronal function is limited. In this study, we show the unique expression profile and subcellular distribution of Tmod1 and Tmod2 in hippocampal neurons. While both Tmod1 and Tmod2 regulate F-actin stability, we find that they exhibit isoform-specific roles in dendrite development and synapse formation: Tmod2 regulates dendritic arborization, whereas Tmod1 is required for spine development and synapse formation. These findings provide novel insight into the actin regulatory mechanisms underlying neuronal development, thereby shedding light on potential pathways disrupted in a number of neurological disorders.
Collapse
|
17
|
A zebrafish model of foxe3 deficiency demonstrates lens and eye defects with dysregulation of key genes involved in cataract formation in humans. Hum Genet 2018; 137:315-328. [PMID: 29713869 DOI: 10.1007/s00439-018-1884-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/16/2018] [Indexed: 12/14/2022]
Abstract
The Forkhead box E3 (FOXE3) gene encodes a transcription factor with a forkhead/winged helix domain that is critical for development of the lens and anterior segment of the eye. Monoallelic and biallelic deleterious sequence variants in FOXE3 cause aphakia, cataracts, sclerocornea and microphthalmia in humans. We used clustered regularly interspaced short palindromic repeats/Cas9 injections to target the foxe3 transcript in zebrafish in order to create an experimental model of loss of function for this gene. Larvae that were homozygous for an indel variant, c.296_300delTGCAG, predicting p.(Val99Alafs*2), demonstrated severe eye defects, including small or absent lenses and microphthalmia. The lenses of the homozygous foxe3 indel mutants showed more intense staining with zl-1 antibody compared to control lenses, consistent with increased lens fiber cell differentiation. Whole genome transcriptome analysis (RNA-Seq) on RNA isolated from wildtype larvae and larvae with eye defects that were putative homozygotes for the foxe3 indel variant found significant dysregulation of genes expressed in the lens and eye whose orthologues are associated with cataracts in human patients, including cryba2a, cryba1l1, mipa and hsf4. Comparative analysis of this RNA-seq data with iSyTE data identified several lens-enriched genes to be down-regulated in foxe3 indel mutants. We also noted upregulation of lgsn and crygmxl2 and downregulation of fmodb and cx43.4, genes that are expressed in the zebrafish lens, but that are not yet associated with an eye phenotype in humans. These findings demonstrate that this new zebrafish foxe3 mutant model is highly relevant to the study of the gene regulatory networks conserved in vertebrate lens and eye development.
Collapse
|
18
|
Cheng C, Nowak RB, Biswas SK, Lo WK, FitzGerald PG, Fowler VM. Tropomodulin 1 Regulation of Actin Is Required for the Formation of Large Paddle Protrusions Between Mature Lens Fiber Cells. Invest Ophthalmol Vis Sci 2017; 57:4084-99. [PMID: 27537257 PMCID: PMC4986768 DOI: 10.1167/iovs.16-19949] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose To elucidate the proteins required for specialized small interlocking protrusions and large paddle domains at lens fiber cell tricellular junctions (vertices), we developed a novel method to immunostain single lens fibers and studied changes in cell morphology due to loss of tropomodulin 1 (Tmod1), an F-actin pointed end–capping protein. Methods We investigated F-actin and F-actin–binding protein localization in interdigitations of Tmod1+/+ and Tmod1−/− single mature lens fibers. Results F-actin–rich small protrusions and large paddles were present along cell vertices of Tmod1+/+ mature fibers. In contrast, Tmod1−/− mature fiber cells lack normal paddle domains, while small protrusions were unaffected. In Tmod1+/+ mature fibers, Tmod1, β2-spectrin, and α-actinin are localized in large puncta in valleys between paddles; but in Tmod1−/− mature fibers, β2-spectrin was dispersed while α-actinin was redistributed at the base of small protrusions and rudimentary paddles. Fimbrin and Arp3 (actin-related protein 3) were located in puncta at the base of small protrusions, while N-cadherin and ezrin outlined the cell membrane in both Tmod1+/+ and Tmod1−/− mature fibers. Conclusions These results suggest that distinct F-actin organizations are present in small protrusions versus large paddles. Formation and/or maintenance of large paddle domains depends on a β2-spectrin–actin network stabilized by Tmod1. α-Actinin–crosslinked F-actin bundles are enhanced in absence of Tmod1, indicating altered cytoskeleton organization. Formation of small protrusions is likely facilitated by Arp3-branched and fimbrin-bundled F-actin networks, which do not depend on Tmod1. This is the first work to reveal the F-actin–associated proteins required for the formation of paddles between lens fibers.
Collapse
Affiliation(s)
- Catherine Cheng
- Department of Cell and Molecular Biology The Scripps Research Institute, La Jolla, California, United States
| | - Roberta B Nowak
- Department of Cell and Molecular Biology The Scripps Research Institute, La Jolla, California, United States
| | - Sondip K Biswas
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia, United States
| | - Woo-Kuen Lo
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia, United States
| | - Paul G FitzGerald
- Department of Cell Biology and Human Anatomy, University of California, Davis, California, United States
| | - Velia M Fowler
- Department of Cell and Molecular Biology The Scripps Research Institute, La Jolla, California, United States
| |
Collapse
|
19
|
Donaldson PJ, Grey AC, Maceo Heilman B, Lim JC, Vaghefi E. The physiological optics of the lens. Prog Retin Eye Res 2017; 56:e1-e24. [DOI: 10.1016/j.preteyeres.2016.09.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/09/2016] [Accepted: 09/13/2016] [Indexed: 11/17/2022]
|
20
|
Abstract
PURPOSE OF REVIEW This article discusses recent advances and unsolved questions in our understanding of actin filament organization and dynamics in the red blood cell (RBC) membrane skeleton, a two-dimensional quasi-hexagonal network consisting of (α1β1)2-spectrin tetramers interconnecting short actin filament-based junctional complexes. RECENT FINDINGS In contrast to the long-held view that RBC actin filaments are static structures that do not exchange subunits with the cytosol, RBC actin filaments are dynamic structures that undergo subunit exchange and turnover, as evidenced by monomer incorporation experiments with rhodamine-actin and filament disruption experiments with actin-targeting drugs. The malaria-causing parasite, Plasmodium falciparum, co-opts RBC actin dynamics to construct aberrantly branched actin filament networks. Even though RBC actin filaments are dynamic, RBC actin filament lengths are highly uniform (∼37 nm). RBC actin filament lengths are thought to be stabilized by the capping proteins, tropomodulin-1 and αβ-adducin, as well as the side-binding protein tropomyosin, present in an equimolar combination of two isoforms, TM5b (Tpm1.9) and TM5NM1 (Tpm3.1). SUMMARY New evidence indicates that RBC actin filaments are not simply passive cytolinkers, but rather dynamic structures whose assembly and disassembly play important roles in RBC membrane function.
Collapse
|
21
|
Mujoo K, Hunt CR, Pandita TK. Nuclear functions of β2-Spectrin in genomic stability. Aging (Albany NY) 2016; 8:3151-3152. [PMID: 27978509 PMCID: PMC5270656 DOI: 10.18632/aging.101147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Indexed: 11/25/2022]
Affiliation(s)
- Kalpana Mujoo
- Department of Radiation Oncology, Weill Cornell Medical College, The Methodist Hospital Research Institute, Houston, TX 77030, . USA
| | - Clayton R Hunt
- Department of Radiation Oncology, Weill Cornell Medical College, The Methodist Hospital Research Institute, Houston, TX 77030, . USA
| | - Tej K Pandita
- Department of Radiation Oncology, Weill Cornell Medical College, The Methodist Hospital Research Institute, Houston, TX 77030, . USA
| |
Collapse
|
22
|
Vasilevska J, De Souza GA, Stensland M, Skrastina D, Zhulenvovs D, Paplausks R, Kurena B, Kozlovska T, Zajakina A. Comparative protein profiling of B16 mouse melanoma cells susceptible and non-susceptible to alphavirus infection: Effect of the tumor microenvironment. Cancer Biol Ther 2016; 17:1035-1050. [PMID: 27636533 DOI: 10.1080/15384047.2016.1219813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Alphavirus vectors are promising tools for cancer treatment. However, relevant entry mechanisms and interactions with host cells are still not clearly understood. The first step toward a more effective therapy is the identification of novel intracellular alterations that could be associated with cancer aggressiveness and could affect the therapeutic potential of these vectors. In this study, we observed that alphaviruses efficiently infected B16 mouse melanoma tumors/tumor cells in vivo, whereas their transduction efficiency in B16 cells under in vitro conditions was blocked. Therefore, we further aimed to understand the mechanisms pertaining to the differential transduction efficacy of alphaviruses in B16 tumor cells under varying growth conditions. We hypothesized that the tumor microenvironment might alter gene expression in B16 cells, leading to an up-regulation of the expression of virus-binding receptors or factors associated with virus entry and replication. To test our hypothesis, we performed a proteomics analysis of B16 cells cultured in vitro and of B16 cells isolated from tumors, and we identified 277 differentially regulated proteins. A further in-depth analysis to identify the biological and molecular functions of the detected proteins revealed a set of candidate genes that could affect virus infectivity. Importantly, we observed a decrease in the expression of interferon α (IFN-α) in tumor-isolated cells that resulted in the suppression of several IFN-regulated genes, thereby abrogating host cell antiviral defense. Additionally, differences in the expression of genes that regulate cytoskeletal organization caused significant alterations in cell membrane elasticity. Taken together, our findings demonstrated favorable intracellular conditions for alphavirus transduction/replication that occurred during tumor transformation. These results pave the way for optimizing the development of strategies for the application of alphaviral vectors as a potent cancer therapy.
Collapse
Affiliation(s)
- Jelena Vasilevska
- a Department of Protein Engineering , Biomedical Research and Study Center , Riga , Latvia
| | | | - Maria Stensland
- b Department of Immunology , Oslo University Hospital , Oslo , Norway
| | - Dace Skrastina
- a Department of Protein Engineering , Biomedical Research and Study Center , Riga , Latvia
| | - Dmitry Zhulenvovs
- a Department of Protein Engineering , Biomedical Research and Study Center , Riga , Latvia
| | | | - Baiba Kurena
- a Department of Protein Engineering , Biomedical Research and Study Center , Riga , Latvia
| | - Tatjana Kozlovska
- a Department of Protein Engineering , Biomedical Research and Study Center , Riga , Latvia
| | - Anna Zajakina
- a Department of Protein Engineering , Biomedical Research and Study Center , Riga , Latvia
| |
Collapse
|
23
|
Rao PV, Maddala R. Ankyrin-B in lens architecture and biomechanics: Just not tethering but more. BIOARCHITECTURE 2016; 6:39-45. [PMID: 27044909 DOI: 10.1080/19490992.2016.1156284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The ankyrins are a family of well-characterized metazoan adaptor proteins that play a key role in linking various membrane-spanning proteins to the underlying spectrin-actin cytoskeleton; a mechanistic understanding of their role in tissue architecture and mechanics, however, remains elusive. Here we comment on a recent study demonstrating a key role for ankyrin-B in maintaining the hexagonal shape and radial alignment of ocular lens fiber cells by regulating the membrane organization of periaxin, dystrophins/dystroglycan, NrCAM and spectrin-actin network of proteins, and revealing that ankyrin-B deficiency impairs fiber cell shape and mechanical properties of the ocular lens. These observations indicate that ankyrin-B plays an important role in maintaining tissue cytoarchitecture, cell shape and biomechanical properties via engaging in key protein: protein interactions required for membrane anchoring and organization of the spectrin-actin skeleton, scaffolding proteins and cell adhesive proteins.
Collapse
Affiliation(s)
- Ponugoti Vasantha Rao
- a Department of Ophthalmology , Duke University School of Medicine , Durham , NC , USA.,b Department of Pharmacology & Cancer Biology , Duke University School of Medicine , Durham , NC , USA
| | - Rupalatha Maddala
- a Department of Ophthalmology , Duke University School of Medicine , Durham , NC , USA
| |
Collapse
|
24
|
Audette DS, Scheiblin DA, Duncan MK. The molecular mechanisms underlying lens fiber elongation. Exp Eye Res 2016; 156:41-49. [PMID: 27015931 DOI: 10.1016/j.exer.2016.03.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 03/14/2016] [Accepted: 03/16/2016] [Indexed: 12/28/2022]
Abstract
Lens fiber cells are highly elongated cells with complex membrane morphologies that are critical for the transparency of the ocular lens. Investigations into the molecular mechanisms underlying lens fiber cell elongation were first reported in the 1960s, however, our understanding of the process is still poor nearly 50 years later. This review summarizes what is currently hypothesized about the regulation of lens fiber cell elongation along with the available experimental evidence, and how this information relates to what is known about the regulation of cell shape/elongation in other cell types, particularly neurons.
Collapse
Affiliation(s)
- Dylan S Audette
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - David A Scheiblin
- 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
|
25
|
Cheng C, Nowak RB, Fowler VM. The lens actin filament cytoskeleton: Diverse structures for complex functions. Exp Eye Res 2016; 156:58-71. [PMID: 26971460 DOI: 10.1016/j.exer.2016.03.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 03/01/2016] [Accepted: 03/07/2016] [Indexed: 01/05/2023]
Abstract
The eye lens is a transparent and avascular organ in the front of the eye that is responsible for focusing light onto the retina in order to transmit a clear image. A monolayer of epithelial cells covers the anterior hemisphere of the lens, and the bulk of the lens is made up of elongated and differentiated fiber cells. Lens fiber cells are very long and thin cells that are supported by sophisticated cytoskeletal networks, including actin filaments at cell junctions and the spectrin-actin network of the membrane skeleton. In this review, we highlight the proteins that regulate diverse actin filament networks in the lens and discuss how these actin cytoskeletal structures assemble and function in epithelial and fiber cells. We then discuss methods that have been used to study actin in the lens and unanswered questions that can be addressed with novel techniques.
Collapse
Affiliation(s)
- Catherine Cheng
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Roberta B Nowak
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Velia M Fowler
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
26
|
Maddala R, Walters M, Brophy PJ, Bennett V, Rao PV. Ankyrin-B directs membrane tethering of periaxin and is required for maintenance of lens fiber cell hexagonal shape and mechanics. Am J Physiol Cell Physiol 2015; 310:C115-26. [PMID: 26538089 DOI: 10.1152/ajpcell.00111.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/31/2015] [Indexed: 12/22/2022]
Abstract
Periaxin (Prx), a PDZ domain protein expressed preferentially in myelinating Schwann cells and lens fibers, plays a key role in membrane scaffolding and cytoarchitecture. Little is known, however, about how Prx is anchored to the plasma membrane. Here we report that ankyrin-B (AnkB), a well-characterized adaptor protein involved in linking the spectrin-actin cytoskeleton to integral membrane proteins, is required for membrane association of Prx in lens fibers and colocalizes with Prx in hexagonal fiber cells. Under AnkB haploinsufficiency, Prx accumulates in the soluble fraction with a concomitant loss from the membrane-enriched fraction of mouse lenses. Moreover, AnkB haploinsufficiency induced age-dependent disruptions in fiber cell hexagonal geometry and radial alignment and decreased compressive stiffness in mouse lenses parallel to the changes observed in Prx null mouse lens. Both AnkB- and Prx-deficient mice exhibit disruptions in membrane organization of the spectrin-actin network and the dystrophin-glycoprotein complex in lens fiber cells. Taken together, these observations reveal that AnkB is required for Prx membrane anchoring and for maintenance of lens fiber cell hexagonal geometry, membrane skeleton organization, and biomechanics.
Collapse
Affiliation(s)
- Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina
| | - Mark Walters
- Department of Mechanical Engineering and Materials Science, Pratt School of Engineering, Duke University, Durham, North Carolina
| | - Peter J Brophy
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
| | - Vann Bennett
- Howard Hughes Medical Institute, Chevy Chase, Maryland; Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina; and
| | - Ponugoti V Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
27
|
Cvekl A, McGreal R, Liu W. Lens Development and Crystallin Gene Expression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 134:129-67. [PMID: 26310154 DOI: 10.1016/bs.pmbts.2015.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The eye and lens represent excellent models to understand embryonic development at cellular and molecular levels. Initial 3D formation of the eye depends on a reciprocal invagination of the lens placode/optic vesicle to form the eye primordium, i.e., the optic cup partially surrounding the lens vesicle. Subsequently, the anterior part of the lens vesicle gives rise to the lens epithelium, while the posterior cells of the lens vesicle differentiate into highly elongated lens fibers. Lens fiber differentiation involves cytoskeletal rearrangements, cellular elongation, accumulation of crystallin proteins, production of extracellular matrix for the lens capsule, and degradation of organelles. This chapter summarizes recent advances in lens development and provides insights into the regulatory mechanisms and differentiation at the level of chromatin structure and dynamics, the emerging field of noncoding RNAs, and novel strategies to fill the gaps in our understanding of lens development.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA.
| | - Rebecca McGreal
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Wei Liu
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
28
|
Hejtmancik JF, Riazuddin SA, McGreal R, Liu W, Cvekl A, Shiels A. Lens Biology and Biochemistry. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 134:169-201. [PMID: 26310155 DOI: 10.1016/bs.pmbts.2015.04.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The primary function of the lens resides in its transparency and ability to focus light on the retina. These require both that the lens cells contain high concentrations of densely packed lens crystallins to maintain a refractive index constant over distances approximating the wavelength of the light to be transmitted, and a specific arrangement of anterior epithelial cells and arcuate fiber cells lacking organelles in the nucleus to avoid blocking transmission of light. Because cells in the lens nucleus have shed their organelles, lens crystallins have to last for the lifetime of the organism, and are specifically adapted to this function. The lens crystallins comprise two major families: the βγ-crystallins are among the most stable proteins known and the α-crystallins, which have a chaperone-like function. Other proteins and metabolic activities of the lens are primarily organized to protect the crystallins from damage over time and to maintain homeostasis of the lens cells. Membrane protein channels maintain osmotic and ionic balance across the lens, while the lens cytoskeleton provides for the specific shape of the lens cells, especially the fiber cells of the nucleus. Perhaps most importantly, a large part of the metabolic activity in the lens is directed toward maintaining a reduced state, which shelters the lens crystallins and other cellular components from damage from UV light and oxidative stress. Finally, the energy requirements of the lens are met largely by glycolysis and the pentose phosphate pathway, perhaps in response to the avascular nature of the lens. Together, all these systems cooperate to maintain lens transparency over time.
Collapse
Affiliation(s)
- J Fielding Hejtmancik
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - S Amer Riazuddin
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebecca McGreal
- Department of Genetics and Ophthalmology, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Wei Liu
- Department of Genetics and Ophthalmology, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ales Cvekl
- Department of Genetics and Ophthalmology, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Alan Shiels
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
29
|
Cheng C, Nowak RB, Gao J, Sun X, Biswas SK, Lo WK, Mathias RT, Fowler VM. Lens ion homeostasis relies on the assembly and/or stability of large connexin 46 gap junction plaques on the broad sides of differentiating fiber cells. Am J Physiol Cell Physiol 2015; 308:C835-47. [PMID: 25740157 PMCID: PMC4436989 DOI: 10.1152/ajpcell.00372.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/03/2015] [Indexed: 12/31/2022]
Abstract
The eye lens consists of layers of tightly packed fiber cells, forming a transparent and avascular organ that is important for focusing light onto the retina. A microcirculation system, facilitated by a network of gap junction channels composed of connexins 46 and 50 (Cx46 and Cx50), is hypothesized to maintain and nourish lens fiber cells. We measured lens impedance in mice lacking tropomodulin 1 (Tmod1, an actin pointed-end capping protein), CP49 (a lens-specific intermediate filament protein), or both Tmod1 and CP49. We were surprised to find that simultaneous loss of Tmod1 and CP49, which disrupts cytoskeletal networks in lens fiber cells, results in increased gap junction coupling resistance, hydrostatic pressure, and sodium concentration. Protein levels of Cx46 and Cx50 in Tmod1(-/-);CP49(-/-) double-knockout (DKO) lenses were unchanged, and electron microscopy revealed normal gap junctions. However, immunostaining and quantitative analysis of three-dimensional confocal images showed that Cx46 gap junction plaques are smaller and more dispersed in DKO differentiating fiber cells. The localization and sizes of Cx50 gap junction plaques in DKO fibers were unaffected, suggesting that Cx46 and Cx50 form homomeric channels. We also demonstrate that gap junction plaques rest in lacunae of the membrane-associated actin-spectrin network, suggesting that disruption of the actin-spectrin network in DKO fibers may interfere with gap junction plaque accretion into micrometer-sized domains or alter the stability of large plaques. This is the first work to reveal that normal gap junction plaque localization and size are associated with normal lens coupling conductance.
Collapse
Affiliation(s)
- Catherine Cheng
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California
| | - Roberta B Nowak
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California
| | - Junyuan Gao
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, New York; and
| | - Xiurong Sun
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, New York; and
| | - Sondip K Biswas
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Woo-Kuen Lo
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Richard T Mathias
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, New York; and
| | - Velia M Fowler
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California;
| |
Collapse
|
30
|
Regulation of actin polymerization by tropomodulin-3 controls megakaryocyte actin organization and platelet biogenesis. Blood 2015; 126:520-30. [PMID: 25964668 DOI: 10.1182/blood-2014-09-601484] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 05/04/2015] [Indexed: 01/02/2023] Open
Abstract
The actin cytoskeleton is important for platelet biogenesis. Tropomodulin-3 (Tmod3), the only Tmod isoform detected in platelets and megakaryocytes (MKs), caps actin filament (F-actin) pointed ends and binds tropomyosins (TMs), regulating actin polymerization and stability. To determine the function of Tmod3 in platelet biogenesis, we studied Tmod3(-/-) embryos, which are embryonic lethal by E18.5. Tmod3(-/-) embryos often show hemorrhaging at E14.5 with fewer and larger platelets, indicating impaired platelet biogenesis. MK numbers are moderately increased in Tmod3(-/-) fetal livers, with only a slight increase in the 8N population, suggesting that MK differentiation is not significantly affected. However, Tmod3(-/-) MKs fail to develop a normal demarcation membrane system (DMS), and cytoplasmic organelle distribution is abnormal. Moreover, cultured Tmod3(-/-) MKs exhibit impaired proplatelet formation with a wide range of proplatelet bud sizes, including abnormally large proplatelet buds containing incorrect numbers of von Willebrand factor-positive granules. Tmod3(-/-) MKs exhibit F-actin disturbances, and Tmod3(-/-) MKs spreading on collagen fail to polymerize F-actin into actomyosin contractile bundles. Tmod3 associates with TM4 and the F-actin cytoskeleton in wild-type MKs, and confocal microscopy reveals that Tmod3, TM4, and F-actin partially colocalize near the membrane of proplatelet buds. In contrast, the abnormally large proplatelets from Tmod3(-/-) MKs show increased F-actin and redistribution of F-actin and TM4 from the cortex to the cytoplasm, but normal microtubule coil organization. We conclude that F-actin capping by Tmod3 regulates F-actin organization in mouse fetal liver-derived MKs, thereby controlling MK cytoplasmic morphogenesis, including DMS formation and organelle distribution, as well as proplatelet formation and sizing.
Collapse
|
31
|
Altered ubiquitin causes perturbed calcium homeostasis, hyperactivation of calpain, dysregulated differentiation, and cataract. Proc Natl Acad Sci U S A 2015; 112:1071-6. [PMID: 25583491 DOI: 10.1073/pnas.1404059112] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Although the ocular lens shares many features with other tissues, it is unique in that it retains its cells throughout life, making it ideal for studies of differentiation/development. Precipitation of proteins results in lens opacification, or cataract, the major blinding disease. Lysines on ubiquitin (Ub) determine fates of Ub-protein substrates. Information regarding ubiquitin proteasome systems (UPSs), specifically of K6 in ubiquitin, is undeveloped. We expressed in the lens a mutant Ub containing a K6W substitution (K6W-Ub). Protein profiles of lenses that express wild-type ubiquitin (WT-Ub) or K6W-Ub differ by only ∼2%. Despite these quantitatively minor differences, in K6W-Ub lenses and multiple model systems we observed a fourfold Ca(2+) elevation and hyperactivation of calpain in the core of the lens, as well as calpain-associated fragmentation of critical lens proteins including Filensin, Fodrin, Vimentin, β-Crystallin, Caprin family member 2, and tudor domain containing 7. Truncations can be cataractogenic. Additionally, we observed accumulation of gap junction Connexin43, and diminished Connexin46 levels in vivo and in vitro. These findings suggest that mutation of Ub K6 alters UPS function, perturbs gap junction function, resulting in Ca(2+) elevation, hyperactivation of calpain, and associated cleavage of substrates, culminating in developmental defects and a cataractous lens. The data show previously unidentified connections between UPS and calpain-based degradative systems and advance our understanding of roles for Ub K6 in eye development. They also inform about new approaches to delay cataract and other protein precipitation diseases.
Collapse
|
32
|
Rumyantseva YV, Ryabchikova EI, Fursova AZ, Kolosova NG. Ameliorative effects of SkQ1 eye drops on cataractogenesis in senescence-accelerated OXYS rats. Graefes Arch Clin Exp Ophthalmol 2014; 253:237-48. [PMID: 25267419 DOI: 10.1007/s00417-014-2806-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/06/2014] [Accepted: 09/15/2014] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Antioxidant supplements have been suggested as a strategy to decrease the risk of age-related cataract, but there is no evidence that antioxidants can reduce the signs of the disease. Recently, we showed that the mitochondrial antioxidant SkQ1 can partially reverse cataract signs in senescence-accelerated OXYS rats. The aim of the present study was the histomorphological examination of the influence of SkQ1 eye drops on the cataract development in OXYS rats. METHODS OXYS rats received SkQ1 eye drops (250 nM) from 9 to 12 months of age. Ophthalmoscopic examination was carried out before and after treatment. Light and electron microscopy were used for histomorphological examination. Expression of the Cryaa and Cryab genes was determined using real-time PCR. αB-crystallin expression was detected using Western blotting. RESULTS SkQ1 completely prevented the cataract development in OXYS rats, and in some of the animals diminished the signs of the disease. Light and electron microscopy showed that SkQ1 attenuated the (typical for cataract) alterations in the lens capsule and epithelial cells, ameliorated disturbances of the hexagonal packing geometry of lens fibers, and improved ultrastructure of the epithelial cells. The levels of mRNA of α-crystallins genes which encode small heat shock proteins αA- and αB-crystallin that play a central role in maintaining lens transparency were significantly lower in the OXYS rats' lenses than in Wistar rats (control). SkQ1 normalized the level of mRNA of Cryaa, and significantly increased the level of Cryab mRNA as well as αB-crystallin protein in the lens of OXYS rats to the level of the control Wistar rats. CONCLUSION SkQ1 eye drops hold promise as a treatment of cataract.
Collapse
Affiliation(s)
- Yuliya V Rumyantseva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090, Acad. Lavrentjev 10, Novosibirsk, Russia,
| | | | | | | |
Collapse
|
33
|
Yamashiro S, Gokhin DS, Sui Z, Bergeron SE, Rubenstein PA, Fowler VM. Differential actin-regulatory activities of Tropomodulin1 and Tropomodulin3 with diverse tropomyosin and actin isoforms. J Biol Chem 2014; 289:11616-11629. [PMID: 24644292 DOI: 10.1074/jbc.m114.555128] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Tropomodulins (Tmods) are F-actin pointed end capping proteins that interact with tropomyosins (TMs) and cap TM-coated filaments with higher affinity than TM-free filaments. Here, we tested whether differences in recognition of TM or actin isoforms by Tmod1 and Tmod3 contribute to the distinct cellular functions of these Tmods. We found that Tmod3 bound ~5-fold more weakly than Tmod1 to α/βTM, TM5b, and TM5NM1. However, surprisingly, Tmod3 was as effective as Tmod1 at capping pointed ends of skeletal muscle α-actin (αsk-actin) filaments coated with α/βTM, TM5b, or TM5NM1. Tmod3 only capped TM-coated αsk-actin filaments more weakly than Tmod1 in the presence of recombinant αTM2, which is unacetylated at its NH2 terminus, binds F-actin weakly, and has a disabled Tmod-binding site. Moreover, both Tmod1 and Tmod3 were similarly effective at capping pointed ends of platelet β/cytoplasmic γ (γcyto)-actin filaments coated with TM5NM1. In the absence of TMs, both Tmod1 and Tmod3 had similarly weak abilities to nucleate β/γcyto-actin filament assembly, but only Tmod3 could sequester cytoplasmic β- and γcyto-actin (but not αsk-actin) monomers and prevent polymerization under physiological conditions. Thus, differences in TM binding by Tmod1 and Tmod3 do not appear to regulate the abilities of these Tmods to cap TM-αsk-actin or TM-β/γcyto-actin pointed ends and, thus, are unlikely to determine selective co-assembly of Tmod, TM, and actin isoforms in different cell types and cytoskeletal structures. The ability of Tmod3 to sequester β- and γcyto-actin (but not αsk-actin) monomers in the absence of TMs suggests a novel function for Tmod3 in regulating actin remodeling or turnover in cells.
Collapse
Affiliation(s)
- Sawako Yamashiro
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; Laboratory of Single-Molecule Cell Biology, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - David S Gokhin
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037
| | - Zhenhua Sui
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037
| | - Sarah E Bergeron
- Department of Biochemistry, University of Iowa, Iowa City, Iowa 52242
| | | | - Velia M Fowler
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037.
| |
Collapse
|
34
|
Scheiblin DA, Gao J, Caplan JL, Simirskii VN, Czymmek KJ, Mathias RT, Duncan MK. Beta-1 integrin is important for the structural maintenance and homeostasis of differentiating fiber cells. Int J Biochem Cell Biol 2014; 50:132-45. [PMID: 24607497 DOI: 10.1016/j.biocel.2014.02.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 02/04/2014] [Accepted: 02/21/2014] [Indexed: 11/19/2022]
Abstract
β1-Integrin is a heterodimeric transmembrane protein that has roles in both cell-extra-cellular matrix and cell-cell interactions. Conditional deletion of β1-integrin from all lens cells during embryonic development results in profound lens defects, however, it is less clear whether this reflects functions in the lens epithelium alone or whether this protein plays a role in lens fibers. Thus, a conditional approach was used to delete β1-integrin solely from the lens fiber cells. This deletion resulted in two distinct phenotypes with some lenses exhibiting cataracts while others were clear, albeit with refractive defects. Analysis of "clear" conditional knockout lenses revealed that they had profound defects in fiber cell morphology associated with the loss of the F-actin network. Physiological measurements found that the lens fiber cells had a twofold increase in gap junctional coupling, perhaps due to differential localization of connexins 46 and 50, as well as increased water permeability. This would presumably facilitate transport of ions and nutrients through the lens, and may partially explain how lenses with profound structural abnormalities can maintain transparency. In summary, β1-integrin plays a role in maintaining the cellular morphology and homeostasis of the lens fiber cells.
Collapse
Affiliation(s)
- David A Scheiblin
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Junyuan Gao
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, New York, NY 11794-8661, United States
| | - Jeffrey L Caplan
- Delaware Biotechnology Institute, University of Delaware, Newark, DE 19716, United States
| | - Vladimir N Simirskii
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Kirk J Czymmek
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Richard T Mathias
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, New York, NY 11794-8661, United States
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
35
|
McKeown CR, Nowak RB, Gokhin DS, Fowler VM. Tropomyosin is required for cardiac morphogenesis, myofibril assembly, and formation of adherens junctions in the developing mouse embryo. Dev Dyn 2014; 243:800-17. [PMID: 24500875 DOI: 10.1002/dvdy.24115] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 01/31/2014] [Accepted: 02/03/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND We explored a function for tropomyosin (TM) in mammalian myofibril assembly and cardiac development by analyzing a deletion in the mouse TPM1 gene targeting αTM1, the major striated muscle TM isoform. RESULTS Mice lacking αTM1 are embryonic lethal at E9.5 with enlarged, misshapen, and non-beating hearts characterized by an abnormally thin myocardium and reduced trabeculae. αTM1-deficient cardiomyocytes do not assemble striated myofibrils, instead displaying aberrant non-striated F-actin fibrils with α-actinin puncta dispersed irregularly along their lengths. αTM1's binding partner, tropomodulin1 (Tmod1), is also disorganized, and both myomesin-containing thick filaments as well as titin Z1Z2 fail to assemble in a striated pattern. Adherens junctions are reduced in size in αTM1-deficient cardiomyocytes, α-actinin/F-actin adherens belts fail to assemble at apical cell-cell contacts, and cell contours are highly irregular, resulting in abnormal cell shapes and a highly folded cardiac surface. In addition, Tmod1-deficient cardiomyocytes exhibit failure of α-actinin/F-actin adherens belt assembly. CONCLUSIONS Absence of αTM1 resulting in unstable F-actin may preclude sarcomere formation and/or lead to degeneration of partially assembled sarcomeres due to unregulated actomyosin interactions. Our data also identify a novel αTM1/Tmod1-based pathway stabilizing F-actin at cell-cell junctions, which may be required for maintenance of cell shapes during embryonic cardiac morphogenesis.
Collapse
Affiliation(s)
- Caroline R McKeown
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California
| | | | | | | |
Collapse
|
36
|
Matsuyama M, Tanaka H, Inoko A, Goto H, Yonemura S, Kobori K, Hayashi Y, Kondo E, Itohara S, Izawa I, Inagaki M. Defect of mitotic vimentin phosphorylation causes microophthalmia and cataract via aneuploidy and senescence in lens epithelial cells. J Biol Chem 2013; 288:35626-35. [PMID: 24142690 PMCID: PMC3861614 DOI: 10.1074/jbc.m113.514737] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vimentin, a type III intermediate filament (IF) protein, is phosphorylated predominantly in mitosis. The expression of a phosphorylation-compromised vimentin mutant in T24 cultured cells leads to cytokinetic failure, resulting in binucleation (multinucleation). The physiological significance of intermediate filament phosphorylation during mitosis for organogenesis and tissue homeostasis was uncertain. Here, we generated knock-in mice expressing vimentin that have had the serine sites phosphorylated during mitosis substituted by alanine residues. Homozygotic mice (VIM(SA/SA)) presented with microophthalmia and cataracts in the lens, whereas heterozygotic mice (VIM(WT/SA)) were indistinguishable from WT (VIM(WT/WT)) mice. In VIM(SA/SA) mice, lens epithelial cell number was not only reduced but the cells also exhibited chromosomal instability, including binucleation and aneuploidy. Electron microscopy revealed fiber membranes that were disorganized in the lenses of VIM(SA/SA), reminiscent of similar characteristic changes seen in age-related cataracts. Because the mRNA level of the senescence (aging)-related gene was significantly elevated in samples from VIM(SA/SA), the lens phenotype suggests a possible causal relationship between chromosomal instability and premature aging.
Collapse
|
37
|
Cheng C, Ansari MM, Cooper JA, Gong X. EphA2 and Src regulate equatorial cell morphogenesis during lens development. Development 2013; 140:4237-45. [PMID: 24026120 DOI: 10.1242/dev.100727] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
High refractive index and transparency of the eye lens require uniformly shaped and precisely aligned lens fiber cells. During lens development, equatorial epithelial cells undergo cell-to-cell alignment to form meridional rows of hexagonal cells. The mechanism that controls this morphogenesis from randomly packed cuboidal epithelial cells to highly organized hexagonal fiber cells remains unknown. In Epha2(-/-) mouse lenses, equatorial epithelial cells fail to form precisely aligned meridional rows; moreover, the lens fulcrum, where the apical tips of elongating epithelial cells constrict to form an anchor point before fiber cell differentiation and elongation at the equator, is disrupted. Phosphorylated Src-Y424 and cortactin-Y466, actin and EphA2 cluster at the vertices of wild-type hexagonal epithelial cells in organized meridional rows. However, phosphorylated Src and phosphorylated cortactin are not detected in disorganized Epha2(-/-) cells with altered F-actin distribution. E-cadherin junctions, which are normally located at the basal-lateral ends of equatorial epithelial cells and are diminished in newly differentiating fiber cells, become widely distributed in the apical, lateral and basal sides of epithelial cells and persist in differentiating fiber cells in Epha2(-/-) lenses. Src(-/-) equatorial epithelial cells also fail to form precisely aligned meridional rows and lens fulcrum. These results indicate that EphA2/Src signaling is essential for the formation of the lens fulcrum. EphA2 also regulates Src/cortactin/F-actin complexes at the vertices of hexagonal equatorial cells for cell-to-cell alignment. This mechanistic information explains how EphA2 mutations lead to disorganized lens cells that subsequently contribute to altered refractive index and cataracts in humans and mice.
Collapse
Affiliation(s)
- Catherine Cheng
- School of Optometry and Vision Science Program, University of California Berkeley, Berkeley, CA 94720, USA
| | | | | | | |
Collapse
|
38
|
Bardet PL, Guirao B, Paoletti C, Serman F, Léopold V, Bosveld F, Goya Y, Mirouse V, Graner F, Bellaïche Y. PTEN controls junction lengthening and stability during cell rearrangement in epithelial tissue. Dev Cell 2013; 25:534-46. [PMID: 23707736 DOI: 10.1016/j.devcel.2013.04.020] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 03/12/2013] [Accepted: 04/22/2013] [Indexed: 12/18/2022]
Abstract
Planar cell rearrangements control epithelial tissue morphogenesis and cellular pattern formation. They lead to the formation of new junctions whose length and stability determine the cellular pattern of tissues. Here, we show that during Drosophila wing development the loss of the tumor suppressor PTEN disrupts cell rearrangements by preventing the lengthening of newly formed junctions that become unstable and keep on rearranging. We demonstrate that the failure to lengthen and to stabilize is caused by the lack of a decrease of Myosin II and Rho-kinase concentration at the newly formed junctions. This defect results in a heterogeneous cortical contractility at cell junctions that disrupts regular hexagonal pattern formation. By identifying PTEN as a specific regulator of junction lengthening and stability, our results uncover how a homogenous distribution of cortical contractility along the cell cortex is restored during cell rearrangement to control the formation of epithelial cellular pattern.
Collapse
Affiliation(s)
- Pierre-Luc Bardet
- Polarity Division and Morphogenesis Team, Institut Curie, CNRS UMR 3215, INSERM U934, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Marcinkevicius E, Zallen JA. Regulation of cytoskeletal organization and junctional remodeling by the atypical cadherin Fat. Development 2013; 140:433-43. [PMID: 23250217 DOI: 10.1242/dev.083949] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The atypical cadherin Fat is a conserved regulator of planar cell polarity, but the mechanisms by which Fat controls cell shape and tissue structure are not well understood. Here, we show that Fat is required for the planar polarized organization of actin denticle precursors, adherens junction proteins and microtubules in the epidermis of the late Drosophila embryo. In wild-type embryos, spatially regulated cell-shape changes and rearrangements organize cells into highly aligned columns. Junctional remodeling is suppressed at dorsal and ventral cell boundaries, where adherens junction proteins accumulate. By contrast, adherens junction proteins fail to accumulate to the wild-type extent and all cell boundaries are equally engaged in junctional remodeling in fat mutants. The effects of loss of Fat on cell shape and junctional localization, but not its role in denticle organization, are recapitulated by mutations in Expanded, an upstream regulator of the conserved Hippo pathway, and mutations in Hippo and Warts, two kinases in the Hippo kinase cascade. However, the cell shape and planar polarity defects in fat mutants are not suppressed by removing the transcriptional co-activator Yorkie, suggesting that these roles of Fat are independent of Yorkie-mediated transcription. The effects of Fat on cell shape, junctional remodeling and microtubule localization are recapitulated by expression of activated Notch. These results demonstrate that cell shape, junctional localization and cytoskeletal planar polarity in the Drosophila embryo are regulated by a common signal provided by the atypical cadherin Fat and suggest that Fat influences tissue organization through its role in polarized junctional remodeling.
Collapse
Affiliation(s)
- Emily Marcinkevicius
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | | |
Collapse
|
40
|
Kubo E, Hasanova N, Fatma N, Sasaki H, Singh DP. Elevated tropomyosin expression is associated with epithelial-mesenchymal transition of lens epithelial cells. J Cell Mol Med 2012. [PMID: 23205574 PMCID: PMC3560320 DOI: 10.1111/j.1582-4934.2012.01654.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Injury to lens epithelial cells (LECs) leads to epithelial–mesenchymal transition (EMT) with resultant fibrosis. The tropomyosin (Tpm) family of cytoskeleton proteins is involved in regulating and stabilizing actin microfilaments. Aberrant expression of Tpms leads to abnormal morphological changes with disintegration of epithelial integrity. The EMT of LECs has been proposed as a major cause of posterior capsule opacification (PCO) after cataract surgery. Using in vivo rodent PCO and human cataractous LECs, we demonstrated that the aberrant expression of rat Tpm and human Tpm1α/2β suggested their association in remodelling of the actin cytoskeleton during EMT of LECs. Expression analysis from abnormally growing LECs after lens extraction revealed elevated expression of α-smooth muscle actin (α-SMA), a marker for EMT. Importantly, these cells displayed increased expression of Tpm1α/2β following EMT/PCO formation. Expression of Tpm1α/2β was up-regulated in LECs isolated from cataractous lenses of Shumiya Cataract Rats (SCRs), compared with non-cataractous lenses. Also, LECs from human patients with nuclear cataract and anterior subcapsular fibrosis (ASF) displayed significantly increased expression of Tpm2β mRNA, suggesting that similar signalling invokes the expression of these molecules in LECs of cataractous SCR and human lenses. EMT was observed in LECs overexpressed with Tpm1α/2β, as evidenced by increased expression of α-SMA. These conditions were correlated with remodelling of actin filaments, possibly leading to EMT/PCO and ASF. The present findings may help clarify the condition of the actin cytoskeleton during morphogenetic EMT, and may contribute to development of Tpm-based inhibitors for postponing PCO and cataractogenesis.
Collapse
Affiliation(s)
- Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kahoku, Ishikawa, Japan.
| | | | | | | | | |
Collapse
|
41
|
Gokhin DS, Nowak RB, Kim NE, Arnett EE, Chen AC, Sah RL, Clark JI, Fowler VM. Tmod1 and CP49 synergize to control the fiber cell geometry, transparency, and mechanical stiffness of the mouse lens. PLoS One 2012; 7:e48734. [PMID: 23144950 PMCID: PMC3492431 DOI: 10.1371/journal.pone.0048734] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 09/28/2012] [Indexed: 11/25/2022] Open
Abstract
The basis for mammalian lens fiber cell organization, transparency, and biomechanical properties has contributions from two specialized cytoskeletal systems: the spectrin-actin membrane skeleton and beaded filament cytoskeleton. The spectrin-actin membrane skeleton predominantly consists of α2β2-spectrin strands interconnecting short, tropomyosin-coated actin filaments, which are stabilized by pointed-end capping by tropomodulin 1 (Tmod1) and structurally disrupted in the absence of Tmod1. The beaded filament cytoskeleton consists of the intermediate filament proteins CP49 and filensin, which require CP49 for assembly and contribute to lens transparency and biomechanics. To assess the simultaneous physiological contributions of these cytoskeletal networks and uncover potential functional synergy between them, we subjected lenses from mice lacking Tmod1, CP49, or both to a battery of structural and physiological assays to analyze fiber cell disorder, light scattering, and compressive biomechanical properties. Findings show that deletion of Tmod1 and/or CP49 increases lens fiber cell disorder and light scattering while impairing compressive load-bearing, with the double mutant exhibiting a distinct phenotype compared to either single mutant. Moreover, Tmod1 is in a protein complex with CP49 and filensin, indicating that the spectrin-actin network and beaded filament cytoskeleton are biochemically linked. These experiments reveal that the spectrin-actin membrane skeleton and beaded filament cytoskeleton establish a novel functional synergy critical for regulating lens fiber cell geometry, transparency, and mechanical stiffness.
Collapse
Affiliation(s)
- David S. Gokhin
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Roberta B. Nowak
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Nancy E. Kim
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Ernest E. Arnett
- Department of Biological Structure, University of Washington, Seattle, Washington, United States of America
| | - Albert C. Chen
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Robert L. Sah
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - John I. Clark
- Department of Biological Structure, University of Washington, Seattle, Washington, United States of America
| | - Velia M. Fowler
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
42
|
Yamashiro S, Gokhin DS, Kimura S, Nowak RB, Fowler VM. Tropomodulins: pointed-end capping proteins that regulate actin filament architecture in diverse cell types. Cytoskeleton (Hoboken) 2012; 69:337-70. [PMID: 22488942 DOI: 10.1002/cm.21031] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 03/23/2012] [Accepted: 03/26/2012] [Indexed: 01/31/2023]
Abstract
Tropomodulins are a family of four proteins (Tmods 1-4) that cap the pointed ends of actin filaments in actin cytoskeletal structures in a developmentally regulated and tissue-specific manner. Unique among capping proteins, Tmods also bind tropomyosins (TMs), which greatly enhance the actin filament pointed-end capping activity of Tmods. Tmods are defined by a TM-regulated/Pointed-End Actin Capping (TM-Cap) domain in their unstructured N-terminal portion, followed by a compact, folded Leucine-Rich Repeat/Pointed-End Actin Capping (LRR-Cap) domain. By inhibiting actin monomer association and dissociation from pointed ends, Tmods regulate actin dynamics and turnover, stabilizing actin filament lengths and cytoskeletal architecture. In this review, we summarize the genes, structural features, molecular and biochemical properties, actin regulatory mechanisms, expression patterns, and cell and tissue functions of Tmods. By understanding Tmods' functions in the context of their molecular structure, actin regulation, binding partners, and related variants (leiomodins 1-3), we can draw broad conclusions that can explain the diverse morphological and functional phenotypes that arise from Tmod perturbation experiments in vitro and in vivo. Tmod-based stabilization and organization of intracellular actin filament networks provide key insights into how the emergent properties of the actin cytoskeleton drive tissue morphogenesis and physiology.
Collapse
Affiliation(s)
- Sawako Yamashiro
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
43
|
Ruiz-Llorente S, Carrillo Santa de Pau E, Sastre-Perona A, Montero-Conde C, Gómez-López G, Fagin JA, Valencia A, Pisano DG, Santisteban P. Genome-wide analysis of Pax8 binding provides new insights into thyroid functions. BMC Genomics 2012; 13:147. [PMID: 22531031 PMCID: PMC3403905 DOI: 10.1186/1471-2164-13-147] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 04/24/2012] [Indexed: 11/25/2022] Open
Abstract
Background The transcription factor Pax8 is essential for the differentiation of thyroid cells. However, there are few data on genes transcriptionally regulated by Pax8 other than thyroid-related genes. To better understand the role of Pax8 in the biology of thyroid cells, we obtained transcriptional profiles of Pax8-silenced PCCl3 thyroid cells using whole genome expression arrays and integrated these signals with global cis-regulatory sequencing studies performed by ChIP-Seq analysis Results Exhaustive analysis of Pax8 immunoprecipitated peaks demonstrated preferential binding to intragenic regions and CpG-enriched islands, which suggests a role of Pax8 in transcriptional regulation of orphan CpG regions. In addition, ChIP-Seq allowed us to identify Pax8 partners, including proteins involved in tertiary DNA structure (CTCF) and chromatin remodeling (Sp1), and these direct transcriptional interactions were confirmed in vivo. Moreover, both factors modulate Pax8-dependent transcriptional activation of the sodium iodide symporter (Nis) gene promoter. We ultimately combined putative and novel Pax8 binding sites with actual target gene expression regulation to define Pax8-dependent genes. Functional classification suggests that Pax8-regulated genes may be directly involved in important processes of thyroid cell function such as cell proliferation and differentiation, apoptosis, cell polarity, motion and adhesion, and a plethora of DNA/protein-related processes. Conclusion Our study provides novel insights into the role of Pax8 in thyroid biology, exerted through transcriptional regulation of important genes involved in critical thyrocyte processes. In addition, we found new transcriptional partners of Pax8, which functionally cooperate with Pax8 in the regulation of thyroid gene transcription. Besides, our data demonstrate preferential location of Pax8 in non-promoter CpG regions. These data point to an orphan CpG island-mediated mechanism that represents a novel role of Pax8 in the transcriptional output of the thyrocyte.
Collapse
Affiliation(s)
- Sergio Ruiz-Llorente
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-CSIC y Universidad Autónoma de Madrid-UAM, C/Arturo Duperier 4, Madrid 28029, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Nowak RB, Fowler VM. Tropomodulin 1 constrains fiber cell geometry during elongation and maturation in the lens cortex. J Histochem Cytochem 2012; 60:414-27. [PMID: 22473940 DOI: 10.1369/0022155412440881] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lens fiber cells exhibit a high degree of hexagonal packing geometry, determined partly by tropomodulin 1 (Tmod1), which stabilizes the spectrin-actin network on lens fiber cell membranes. To ascertain whether Tmod1 is required during epithelial cell differentiation to fiber cells or during fiber cell elongation and maturation, the authors quantified the extent of fiber cell disorder in the Tmod1-null lens and determined locations of disorder by confocal microscopy and computational image analysis. First, nearest neighbor analysis of fiber cell geometry in Tmod1-null lenses showed that disorder is confined to focal patches. Second, differentiating epithelial cells at the equator aligned into ordered meridional rows in Tmod1-null lenses, with disordered patches first observed in elongating fiber cells. Third, as fiber cells were displaced inward in Tmod1-null lenses, total disordered area increased due to increased sizes (but not numbers) of individual disordered patches. The authors conclude that Tmod1 is required first to coordinate fiber cell shapes and interactions during tip migration and elongation and second to stabilize ordered fiber cell geometry during maturation in the lens cortex. An unstable spectrin-actin network without Tmod1 may result in imbalanced forces along membranes, leading to fiber cell rearrangements during elongation, followed by propagation of disorder as fiber cells mature.
Collapse
|
45
|
Tropomodulin capping of actin filaments in striated muscle development and physiology. J Biomed Biotechnol 2011; 2011:103069. [PMID: 22013379 PMCID: PMC3196151 DOI: 10.1155/2011/103069] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/18/2011] [Indexed: 11/17/2022] Open
Abstract
Efficient striated muscle contraction requires precise assembly and regulation of diverse actin filament systems, most notably the sarcomeric thin filaments of the contractile apparatus. By capping the pointed ends of actin filaments, tropomodulins (Tmods) regulate actin filament assembly, lengths, and stability. Here, we explore the current understanding of the expression patterns, localizations, and functions of Tmods in both cardiac and skeletal muscle. We first describe the mechanisms by which Tmods regulate myofibril assembly and thin filament lengths, as well as the roles of closely related Tmod family variants, the leiomodins (Lmods), in these processes. We also discuss emerging functions for Tmods in the sarcoplasmic reticulum. This paper provides abundant evidence that Tmods are key structural regulators of striated muscle cytoarchitecture and physiology.
Collapse
|
46
|
Maddala R, Skiba NP, Lalane R, Sherman DL, Brophy PJ, Rao PV. Periaxin is required for hexagonal geometry and membrane organization of mature lens fibers. Dev Biol 2011; 357:179-90. [PMID: 21745462 PMCID: PMC3164832 DOI: 10.1016/j.ydbio.2011.06.036] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/14/2011] [Accepted: 06/14/2011] [Indexed: 01/06/2023]
Abstract
Transparency of the ocular lens depends on symmetric packing and membrane organization of highly elongated hexagonal fiber cells. These cells possess an extensive, well-ordered cortical cytoskeleton to maintain cell shape and to anchor membrane components. Periaxin (Prx), a PDZ domain protein involved in myelin sheath stabilization, is also a component of adhaerens plaques in lens fiber cells. Here we show that Prx is expressed in lens fibers and exhibits maturation dependent redistribution, clustering discretely at the tricellular junctions in mature fiber cells. Prx exists in a macromolecular complex with proteins involved in membrane organization including ankyrin-B, spectrin, NrCAM, filensin, ezrin and desmoyokin. Importantly, Prx knockout mouse lenses were found to be softer and more easily deformed than normal lenses, revealing disruptions in fiber cell hexagonal packing, membrane skeleton and membrane stability. These observations suggest a key role for Prx in maturation, packing, and membrane organization of lens fiber cells. Hence, there may be functional parallels between the roles of Prx in membrane stabilization of the myelin sheath and the lens fiber cell.
Collapse
Affiliation(s)
- Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, NC. USA
| | - Nikolai P. Skiba
- Department of Ophthalmology, Duke University School of Medicine, NC. USA
| | - Robert Lalane
- Department of Ophthalmology, Duke University School of Medicine, NC. USA
| | - Diane L. Sherman
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Peter J. Brophy
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Ponugoti V. Rao
- Department of Ophthalmology, Duke University School of Medicine, NC. USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, NC. USA
| |
Collapse
|
47
|
Grieve AG, Daniels RD, Sanchez-Heras E, Hayes MJ, Moss SE, Matter K, Lowe M, Levine TP. Lowe Syndrome protein OCRL1 supports maturation of polarized epithelial cells. PLoS One 2011; 6:e24044. [PMID: 21901156 PMCID: PMC3162020 DOI: 10.1371/journal.pone.0024044] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Accepted: 08/04/2011] [Indexed: 12/24/2022] Open
Abstract
Mutations in the inositol polyphosphate 5-phosphatase OCRL1 cause Lowe Syndrome, leading to cataracts, mental retardation and renal failure. We noted that cell types affected in Lowe Syndrome are highly polarized, and therefore we studied OCRL1 in epithelial cells as they mature from isolated individual cells into polarized sheets and cysts with extensive communication between neighbouring cells. We show that a proportion of OCRL1 targets intercellular junctions at the early stages of their formation, co-localizing both with adherens junctional components and with tight junctional components. Correlating with this distribution, OCRL1 forms complexes with junctional components α-catenin and zonula occludens (ZO)-1/2/3. Depletion of OCRL1 in epithelial cells growing as a sheet inhibits maturation; cells remain flat, fail to polarize apical markers and also show reduced proliferation. The effect on shape is reverted by re-expressed OCRL1 and requires the 5'-phosphatase domain, indicating that down-regulation of 5-phosphorylated inositides is necessary for epithelial development. The effect of OCRL1 in epithelial maturation is seen more strongly in 3-dimensional cultures, where epithelial cells lacking OCRL1 not only fail to form a central lumen, but also do not have the correct intracellular distribution of ZO-1, suggesting that OCRL1 functions early in the maturation of intercellular junctions when cells grow as cysts. A role of OCRL1 in junctions of polarized cells may explain the pattern of organs affected in Lowe Syndrome.
Collapse
Affiliation(s)
- Adam G. Grieve
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Rachel D. Daniels
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Elena Sanchez-Heras
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Matthew J. Hayes
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Stephen E. Moss
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Karl Matter
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Martin Lowe
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Timothy P. Levine
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
Wang Z, Schey KL. Aquaporin-0 interacts with the FERM domain of ezrin/radixin/moesin proteins in the ocular lens. Invest Ophthalmol Vis Sci 2011; 52:5079-87. [PMID: 21642618 DOI: 10.1167/iovs.10-6998] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
PURPOSE Aquaporin 0 (AQP0) is the major intrinsic protein in the lens and is essential for establishing proper fiber cell structure and organization. Cytoskeletal proteins that directly interact with the C terminus of AQP0 are identified herein. METHODS The water-insoluble fraction of lens fiber cells was chemically cross-linked, and cross-linked peptides with the C terminus of AQP0 were identified by mass spectrometry. Coimmunoprecipitation and AQP0 C-terminal peptide pulldown experiments were used to confirm the protein-protein interaction. RESULTS Unexpectedly, AQP0 was found to directly associate with ezrin/radixin/moesin (ERM) family members, proteins that are involved in linkage of actin filaments to the plasma membrane. Cross-linked peptides were detected between AQP0 and degenerate sequences of ezrin and radixin; however, AQP0 interaction with ezrin is believed to play a more significant function in the lens because of its higher level of expression and observed ezrin-specific cross-linking. The interaction was found to occur between the C terminus of AQP0 and subdomains F1 and F3 of ERM proteins. The interaction between AQP0 and ezrin was confirmed by coimmunoprecipitation and AQP0 C-terminal peptide pulldown experiments. CONCLUSIONS Considering the important known functions of the cellular actin cytoskeleton in fiber cell differentiation, the interaction of AQP0 and ERM proteins may play an important role in fiber cell morphology, elongation, and organization.
Collapse
Affiliation(s)
- Zhen Wang
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8575, USA
| | | |
Collapse
|
49
|
Gokhin DS, Fowler VM. Cytoplasmic gamma-actin and tropomodulin isoforms link to the sarcoplasmic reticulum in skeletal muscle fibers. ACTA ACUST UNITED AC 2011; 194:105-20. [PMID: 21727195 PMCID: PMC3135406 DOI: 10.1083/jcb.201011128] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Tropomodulins, cytoplasmic γ-actin, and small ankyrin 1.5 mechanically stabilize the sarcoplasmic reticulum and maintain myofibril alignment in skeletal muscle fibers. The sarcoplasmic reticulum (SR) serves as the Ca2+ reservoir for muscle contraction. Tropomodulins (Tmods) cap filamentous actin (F-actin) pointed ends, bind tropomyosins (Tms), and regulate F-actin organization. In this paper, we use a genetic targeting approach to examine the effect of Tmod1 deletion on the organization of cytoplasmic γ-actin (γcyto-actin) in the SR of skeletal muscle. In wild-type muscle fibers, γcyto-actin and Tmod3 defined an SR microdomain that was distinct from another Z line–flanking SR microdomain containing Tmod1 and Tmod4. The γcyto-actin/Tmod3 microdomain contained an M line complex composed of small ankyrin 1.5 (sAnk1.5), γcyto-actin, Tmod3, Tm4, and Tm5NM1. Tmod1 deletion caused Tmod3 to leave its SR compartment, leading to mislocalization and destabilization of the Tmod3–γcyto-actin–sAnk1.5 complex. This was accompanied by SR morphological defects, impaired Ca2+ release, and an age-dependent increase in sarcomere misalignment. Thus, Tmod3 regulates SR-associated γcyto-actin architecture, mechanically stabilizes the SR via a novel cytoskeletal linkage to sAnk1.5, and maintains the alignment of adjacent myofibrils.
Collapse
Affiliation(s)
- David S Gokhin
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
50
|
Gokhin DS, Fowler VM. The sarcoplasmic reticulum: Actin and tropomodulin hit the links. BIOARCHITECTURE 2011; 1:175-179. [PMID: 22069510 DOI: 10.4161/bioa.1.4.17533] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 07/22/2011] [Accepted: 07/26/2011] [Indexed: 12/29/2022]
Abstract
Skeletal muscle exhibits strikingly regular intracellular sorting of actin and tropomodulin (Tmod) isoforms, which are essential for efficient muscle contraction. A recent study from our laboratory demonstrates that the skeletal muscle sarcoplasmic reticulum (SR) is associated with cytoplasmic γ-actin (γ(cyto)-actin) filaments, which are predominantly capped by Tmod3. When Tmod3 is experimentally induced to vacate its SR compartment, the cytoskeletal organization of SR-associated γ(cyto)-actin is perturbed, leading to SR swelling, depressed SR Ca(2+) release and myofibril misalignment. Based on these findings, Tmod3-capped γ(cyto)-actin filaments mechanically stabilize SR structure and regulate SR function via a novel lateral linkage. Furthermore, by placing these findings in the context of studies in nonmuscle cells, we conclude that Tmodcapped actin filaments are emerging as critical regulators of membrane stability and physiology in a broad assortment of cell types.
Collapse
Affiliation(s)
- David S Gokhin
- Department of Cell Biology; The Scripps Research Institute; La Jolla, CA USA
| | | |
Collapse
|