1
|
Feng H, Yang Z, Bai X, Yang M, Fang Y, Zhang X, Guo Q, Ning H. Therapeutic potential of a dual mTORC1/2 inhibitor for the prevention of posterior capsule opacification: An in vitro study. Int J Mol Med 2018; 41:2099-2107. [PMID: 29344639 PMCID: PMC5810205 DOI: 10.3892/ijmm.2018.3398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 01/10/2018] [Indexed: 12/14/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) serves a central role in regulating cell growth and survival, and has been demonstrated to be involved in the pathological progression of posterior capsule opacification (PCO). In the present study, the potency of PP242, a novel dual inhibitor of mTOR complex 1/2 (mTORC1/2), in the suppression of the growth of human lens epithelial cells (HLECs) was investigated. Using a Cell Counting Kit‑8 and a wound healing assay, it was demonstrated that PP242 inhibited the proliferation and migration of HLECs. In addition, western blot analysis indicated that PP242 completely inhibited mTORC1 and mTORC2 downstream signaling activities, whereas rapamycin only partially inhibited mTORC1 activity within LECs. Furthermore, PP242 treatment led to an upregulation of the expression levels of p53 and B cell lymphoma‑2 (Bcl‑2)‑associated X and downregulation of Bcl‑2. In addition, flow cytometric analysis demonstrated that PP242 induced the cell cycle arrest at the G0/G1 phase, which may have caused apoptosis and induced autophagy within the LECs. The results of the present study suggested that administration of PP242 may potentially offer a novel therapeutic approach for the prevention of PCO.
Collapse
Affiliation(s)
- Hao Feng
- Department of Ophthalmology, The First Hospital of China Medical University
| | - Zhibo Yang
- Department of Ophthalmology, The Fourth People’s Hospital of Shenyang
| | - Xue Bai
- Department of Ophthalmology, The First Hospital of China Medical University
| | - Meirong Yang
- Department of Ophthalmology, The First Hospital of China Medical University
| | - Yuan Fang
- Department of Ophthalmology, The First Hospital of China Medical University
| | - Xiaonan Zhang
- Department of Ophthalmology, The First Hospital of China Medical University
| | - Qiqiang Guo
- Key Laboratory of Medical Cell Biology, College of Translational Medicine, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hong Ning
- Department of Ophthalmology, The First Hospital of China Medical University
| |
Collapse
|
2
|
Wolf L, Harrison W, Huang J, Xie Q, Xiao N, Sun J, Kong L, Lachke SA, Kuracha MR, Govindarajan V, Brindle PK, Ashery-Padan R, Beebe DC, Overbeek PA, Cvekl A. Histone posttranslational modifications and cell fate determination: lens induction requires the lysine acetyltransferases CBP and p300. Nucleic Acids Res 2013; 41:10199-214. [PMID: 24038357 PMCID: PMC3905850 DOI: 10.1093/nar/gkt824] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Lens induction is a classical embryologic model to study cell fate determination. It has been proposed earlier that specific changes in core histone modifications accompany the process of cell fate specification and determination. The lysine acetyltransferases CBP and p300 function as principal enzymes that modify core histones to facilitate specific gene expression. Herein, we performed conditional inactivation of both CBP and p300 in the ectodermal cells that give rise to the lens placode. Inactivation of both CBP and p300 resulted in the dramatic discontinuation of all aspects of lens specification and organogenesis, resulting in aphakia. The CBP/p300−/− ectodermal cells are viable and not prone to apoptosis. These cells showed reduced expression of Six3 and Sox2, while expression of Pax6 was not upregulated, indicating discontinuation of lens induction. Consequently, expression of αB- and αA-crystallins was not initiated. Mutant ectoderm exhibited markedly reduced levels of histone H3 K18 and K27 acetylation, subtly increased H3 K27me3 and unaltered overall levels of H3 K9ac and H3 K4me3. Our data demonstrate that CBP and p300 are required to establish lens cell-type identity during lens induction, and suggest that posttranslational histone modifications are integral to normal cell fate determination in the mammalian lens.
Collapse
Affiliation(s)
- Louise Wolf
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children's Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Wang WL, Li Q, Xu J, Cvekl A. Lens fiber cell differentiation and denucleation are disrupted through expression of the N-terminal nuclear receptor box of NCOA6 and result in p53-dependent and p53-independent apoptosis. Mol Biol Cell 2010; 21:2453-68. [PMID: 20484573 PMCID: PMC2903674 DOI: 10.1091/mbc.e09-12-1031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nuclear receptor coactivator 6 (NCOA6) is a multifunctional protein implicated in embryonic development, cell survival, and homeostasis. An 81-amino acid fragment, dnNCOA6, containing the N-terminal nuclear receptor box (LXXLL motif) of NCOA6, acts as a dominant-negative (dn) inhibitor of NCOA6. Here, we expressed dnNCOA6 in postmitotic transgenic mouse lens fiber cells. The transgenic lenses showed reduced growth; a wide spectrum of lens fiber cell differentiation defects, including reduced expression of gamma-crystallins; and cataract formation. Those lens fiber cells entered an alternate proapoptotic pathway, and the denucleation (karyolysis) process was stalled. Activation of caspase-3 at embryonic day (E)13.5 was followed by double-strand breaks (DSBs) formation monitored via a biomarker, gamma-H2AX. Intense terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) signals were found at E16.5. Thus, a window of approximately 72 h between these events suggested prolonged though incomplete apoptosis in the lens fiber cell compartment that preserved nuclei in its cells. Genetic experiments showed that the apoptotic-like processes in the transgenic lens were both p53-dependent and p53-independent. Lens-specific deletion of Ncoa6 also resulted in disrupted lens fiber cell differentiation. Our data demonstrate a cell-autonomous role of Ncoa6 in lens fiber cell differentiation and suggest novel insights into the process of lens fiber cell denucleation and apoptosis.
Collapse
Affiliation(s)
- Wei-Lin Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
4
|
Cvekl A, Wang WL. Retinoic acid signaling in mammalian eye development. Exp Eye Res 2009; 89:280-91. [PMID: 19427305 DOI: 10.1016/j.exer.2009.04.012] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 04/21/2009] [Accepted: 04/22/2009] [Indexed: 12/20/2022]
Abstract
Retinoic acid (RA) is a biologically active metabolite of vitamin A (retinol) that serves as a signaling molecule during a number of developmental and physiological processes. RA signaling plays multiple roles during embryonic eye development. RA signaling is initially required for reciprocal interactions between the optic vesicle and invaginating lens placode. RA signaling promotes normal development of the ventral retina and optic nerve through its activities in the neural crest cell-derived periocular mesenchyme. RA coordinates these processes by regulating biological activities of a family of non-steroid hormone receptors, RARalpha/beta/gamma, and RXRalpha/beta/gamma. These DNA-binding transcription factors recognize DNA as RAR/RXR heterodimers and recruit multiprotein transcriptional co-repressor complexes. RA-binding to RAR receptors induces a conformational change in the receptor, followed by the replacement of co-repressor with co-activator complexes. Inactivation of RARalpha/beta/gamma receptors in the periocular mesenchyme abrogates anterior eye segment formation. This review summarizes recent genetic studies of RA signaling and progress in understanding the molecular mechanism of transcriptional co-activators that function with RAR/RXR.
Collapse
Affiliation(s)
- Ales Cvekl
- The Department Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | |
Collapse
|
5
|
Cain S, Martinez G, Kokkinos MI, Turner K, Richardson RJ, Abud HE, Huelsken J, Robinson ML, de Iongh RU. Differential requirement for beta-catenin in epithelial and fiber cells during lens development. Dev Biol 2008; 321:420-33. [PMID: 18652817 DOI: 10.1016/j.ydbio.2008.07.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 06/27/2008] [Accepted: 07/01/2008] [Indexed: 11/18/2022]
Abstract
Recent studies implicate Wnt/beta-catenin signaling in lens differentiation (Stump, R. J., et al., 2003. A role for Wnt/beta-catenin signaling in lens epithelial differentiation. Dev Biol;259:48-61). Beta-catenin is a component of adherens junctions and functions as a transcriptional activator in canonical Wnt signaling. We investigated the effects of Cre/LoxP-mediated deletion of beta-catenin during lens development using two Cre lines that specifically deleted beta-catenin in whole lens or only in differentiated fibers, from E13.5. We found that beta-catenin was required in lens epithelium and during early fiber differentiation but appeared to be redundant in differentiated fiber cells. Complete loss of beta-catenin resulted in an abnormal and deficient epithelial layer with loss of E-cadherin and Pax6 expression as well as abnormal expression of c-Maf and p57(kip2) but not Prox1. There was also disrupted fiber cell differentiation, characterized by poor cell elongation, decreased beta-crystallin expression, epithelial cell cycle arrest at G(1)-S transition and premature cell cycle exit. Despite cell cycle arrest there was no induction of apoptosis. Mutant fiber cells displayed altered apical-basal polarity as evidenced by altered distribution of the tight junction protein, ZO1, disruption of apical actin filaments and abnormal deposition of extracellular matrix, resulting in a deficient lens capsule. Loss of beta-catenin also affected the formation of adhesion junctions as evidenced by dissociation of N-cadherin and F-actin localization in differentiating fiber cells. However, loss of beta-catenin from terminally differentiating fibers had no apparent effects on adhesion junctions between adjacent embryonic fibers. These data indicate that beta-catenin plays distinct functions during lens fiber differentiation and is involved in both Wnt signaling and adhesion-related mechanisms that regulate lens epithelium and early fiber differentiation.
Collapse
Affiliation(s)
- Sarah Cain
- Ocular Development Laboratory, Anatomy and Cell Biology Department, University of Melbourne, Victoria 3010, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Shui YB, Arbeit JM, Johnson RS, Beebe DC. HIF-1: an age-dependent regulator of lens cell proliferation. Invest Ophthalmol Vis Sci 2008; 49:4961-70. [PMID: 18586877 DOI: 10.1167/iovs.08-2118] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The lens grows throughout life, and lens size is a major risk factor for nuclear and cortical cataracts. A previous study showed that the hypoxic environment around the lens suppressed lens growth in older rats. The present study was conducted to investigate the mechanism responsible for the age-dependent decline in lens cell proliferation. METHODS Transgenic mice expressing Cre recombinase in the lens were bred to mice containing floxed Hif1a alleles. Transgenic mice expressing oxygen insensitive forms of HIF-1alpha in lens epithelial cells were exposed to room air or 60% oxygen. Proliferation was measured by BrdU labeling and cell death by using the TUNEL assay. Morphology was assessed in histologic sections. HIF-1alpha and p27(KIP1) levels were determined by Western blot. The expression of HIF-regulated genes was assessed on microarrays. RESULTS Lenses lacking Hif1a degenerated, precluding study in older animals. Breathing 60% oxygen reduced HIF-1alpha levels and HIF-1-regulated transcripts in lens epithelial cells from young and older lenses. Overexpression of oxygen-insensitive HIF-1alpha had no effect on lens size, but suppressed increased proliferation in response to oxygen. Systemic injection of the iron chelator, 1,10-phenanthroline prevented the degradation of HIF-1alpha and reduced oxygen-induced proliferation. Increasing oxygen decreased levels of p27(KIP1) in the epithelial cells of older mice, which was prevented by expressing oxygen-insensitive forms of HIF-1alpha. CONCLUSIONS HIF-1alpha is present and HIF-1 is transcriptionally active throughout life, but suppresses growth only in older lenses. Maintaining elevated levels of p27(KIP1) in older lenses requires HIF-1. p27(KIP1) and other growth regulators may selectively suppress the proliferation of older lens epithelial cells.
Collapse
Affiliation(s)
- Ying-Bo Shui
- Department of Ophthalmology and Visual Sciences, Washington University, St Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
7
|
Simirskii VN, Wang Y, Duncan MK. Conditional deletion of beta1-integrin from the developing lens leads to loss of the lens epithelial phenotype. Dev Biol 2007; 306:658-68. [PMID: 17493607 PMCID: PMC1950782 DOI: 10.1016/j.ydbio.2007.04.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Revised: 04/03/2007] [Accepted: 04/05/2007] [Indexed: 10/23/2022]
Abstract
Beta1-integrins are cell surface receptors that participate in sensing the cell's external environment. We used the Cre-lox system to delete beta1-integrin in all lens cells as the lens vesicle transitions into the lens. Adult mice lacking beta1-integrin in the lens are microphthalmic due to apoptosis of the lens epithelium and neonatal disintegration of the lens fibers. The first morphological alterations in beta1-integrin null lenses are seen at 16.5 dpc when the epithelium becomes disorganized and begins to upregulate the fiber cell markers beta- and gamma-crystallins, the transcription factors cMaf and Prox1 and downregulate Pax6 levels demonstrating that beta1-integrin is essential to maintain the lens epithelial phenotype. Furthermore, beta1-integrin null lens epithelial cells upregulate the expression of alpha-smooth muscle actin and nuclear Smad4 and downregulate Smad6 suggesting that beta1-integrin may brake TGFbeta family signaling leading to epithelial-mesenchymal transitions in the lens. In contrast, beta1-integrin null lens epithelial cells show increased E-cadherin immunoreactivity which supports the proposed role of beta1-integrins in mediating complete EMT in response to TGFbeta family members. Thus, beta1-integrin is required to maintain the lens epithelial phenotype and block inappropriate activation of some aspects of the lens fiber cell differentiation program.
Collapse
Affiliation(s)
| | | | - Melinda K. Duncan
- *To whom all the correspondence should be addressed: Melinda K. Duncan, Department of Biological Sciences, University of Delaware, Newark, DE 19716, Telephone: (302) 831-0533, Fax: (302) 831-2281, E-mail address:
| |
Collapse
|
8
|
Yang Y, Wolf LV, Cvekl A. Distinct embryonic expression and localization of CBP and p300 histone acetyltransferases at the mouse alphaA-crystallin locus in lens. J Mol Biol 2007; 369:917-26. [PMID: 17467007 PMCID: PMC2063435 DOI: 10.1016/j.jmb.2007.04.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 03/26/2007] [Accepted: 04/02/2007] [Indexed: 10/23/2022]
Abstract
Mouse alphaA-crystallin gene encodes the most abundant protein of the mammalian lens. Expression of alphaA-crystallin is regulated temporally and spatially during lens development with initial expression in the lens vesicle followed by strong upregulation in the differentiating primary lens fibers. Lens-specific expression of alphaA-crystallin is mediated by DNA-binding transcription factors Pax6, c-Maf and CREB bound to its promoter region. Its 5'-distal enhancer, DCR1, mediates regulation of alphaA-crystallin via FGF signaling, while its 3'-distal enhancer, DCR3, functions only in elongated primary lens fibers via other lens differentiation pathways. DCR1 and DCR3 establish outside borders of a lens-specific chromatin region marked by histone H3 K9 acetylation. Here, we identified CREB-binding protein (CBP) and p300 as major histone acetyltransferases (HATs) associated in vivo with the mouse alphaA-crystallin locus. Both HATs are expressed in embryonic lens. Expression of CBP in primary lens fiber cells coincides with alphaA-crystallin. In the chromatin of lens epithelial cells, chromatin immunoprecipitations (ChIPs) show that the alphaA-crystallin promoter is notably devoid of any significant presence of CBP and p300, though DCR1 and a few other regions show the presence of these HATs. In the chromatin obtained from newborn lens, CBP was localized specifically at the promoter region with about ten times higher abundance compared to the entire alphaA-crystallin locus. In contrast, p300 is distributed more evenly across the entire locus. Analysis of total histone H3 and H3 K9 acetylation revealed potential lower density of nucleosomes 2 kb upstream from the promoter region. Collectively, our data suggest that moderate level of alphaA-crystallin gene expression in lens epithelial cells does not require the presence of CBP and p300 in the promoter. However, the lens-specific chromatin domain contains both promoter localized CBP on the "background" of locus-spread presence of CBP and p300.
Collapse
Affiliation(s)
- Ying Yang
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA
| | | | | |
Collapse
|
9
|
Abstract
The ocular lens is a distinct system to study cell death for the following reasons. First, during animal development, the ocular lens is crafted into its unique shape. The crafting processes include cell proliferation, cell migration, and apoptosis. Moreover, the lens epithelial cells differentiate into lens fiber cells through a process, which utilizes the same regulators as those in apoptosis at multiple signaling steps. In addition, introduction of exogenous wild-type or mutant genes or knock-out of the endogenous genes leads to apoptosis of the lens epithelial cells followed by absence of the ocular lens or formation of abnormal lens. Finally, both in vitro and in vivo studies have shown that treatment of adult lens with stress factors induces apoptosis of lens epithelial cells, which is followed by cataractogenesis. The present review summarizes the current knowledge on apoptosis in the ocular lens with emphasis on its role in lens development and pathology.
Collapse
Affiliation(s)
- Qin Yan
- College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | | | | |
Collapse
|
10
|
Malecaze F, Decha A, Serre B, Penary M, Duboue M, Berg D, Levade T, Lubsen NH, Kremer EJ, Couderc B. Prevention of posterior capsule opacification by the induction of therapeutic apoptosis of residual lens cells. Gene Ther 2006; 13:440-8. [PMID: 16251995 DOI: 10.1038/sj.gt.3302667] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Posterior capsule opacification (PCO) is a common complication of cataract surgery. Using adenovirus(Ad)-mediated gene transfer, we overexpressed the proapoptotic molecules p53, procaspase 3, Bax, and TRAIL to induce therapeutic programmed cell death of residual lens cells to prevent PCO. Overexpressed TRAIL did not induce apoptosis in cultured rabbit lens cells or in human lens cells. Overexpressed p53 induced apoptosis of lens cells in vitro and ex vivo, but was unable to prevent PCO in vivo. Overexpressed procaspase 3 was associated with engagement of many components of the apoptotic pathway, including cleavage of intracellular caspase targets such as PARP and inter-nucleosome DNA fragmentation. Even when only slightly overexpressed, Bax caused apoptosis of transduced rabbit and human lens cells by engaging the mitochondrial pathway, including catalytic activation of the caspases. A single in vivo injection of Ad vectors expressing either Bax or procaspase 3 into the capsular bag at the end of phacoemulsification prevented PCO in rabbits. These experiments show that Ad-mediated Bax or procaspase 3 overexpression is capable of inducing therapeutic programmed cell death in vitro and in vivo in residual lens cells and preventing PCO in a rabbit model of PCO. Manipulation of proapoptotic molecule expression could be a novel gene therapy approach for prevention of PCO.
Collapse
Affiliation(s)
- F Malecaze
- INSERM U563, Department Ophtalmologie et Pathologie des épithéliums, Hôpital Purpan, Toulouse, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Couderc B, Penary M, Tohfe M, Pradines A, Casteignau A, Berg D, Favre G. Reversible inactivation of the transcriptional function of P53 protein by farnesylation. BMC Biotechnol 2006; 6:26. [PMID: 16732889 PMCID: PMC1481662 DOI: 10.1186/1472-6750-6-26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Accepted: 05/29/2006] [Indexed: 11/29/2022] Open
Abstract
Background The use of integrating viral vectors in Gene therapy clinical trials has pointed out the problem of the deleterous effect of the integration of the ectopic gene to the cellular genome and the safety of this strategy. We proposed here a way to induce the death of gene modified cells upon request by acting on a pro-apoptotic protein cellular localization and on the activation of its apoptotic function. Results We constructed an adenoviral vector coding a chimeric p53 protein by fusing p53 sequence with the 21 COOH term amino acids sequence of H-Ras. Indeed, the translation products of Ras genes are cytosolic proteins that become secondarily associated with membranes through a series of post-translational modifications initiated by a CAAX motif present at the C terminus of Ras proteins. The chimeric p53HRCaax protein was farnesylated efficiently in transduced human osteosarcoma p53-/- cell line. The farnesylated form of p53 resided mainly in the cytosol, where it is non-functional. Farnesyl transferase inhibitors (FTIs) specifically inhibited farnesyl isoprenoid lipid modification of proteins. Following treatment of the cells with an FTI, p53HRCaax underwent translocation into the nucleus where it retained transcription factor activity. Shifting p53 into the nucleus resulted in the induction of p21waf1/CIP1 and Bax transcription, cell growth arrest, caspase activation and apoptosis. Conclusion Artificial protein farnesylation impaired the transcriptional activity of p53. This could be prevented by Farnesyl transferase inhibition. These data highlight the fact that the artificial prenylation of proteins provides a novel system for controlling the function of a transactivating factor.
Collapse
Affiliation(s)
- Bettina Couderc
- INSERM U563, Department Innovations thérapeutiques et Oncologie moléculaire, Institut Claudius Regaud and Faculté des Sciences Pharmaceutiques, Toulouse, France
| | - Marie Penary
- INSERM U563, Department Innovations thérapeutiques et Oncologie moléculaire, Institut Claudius Regaud and Faculté des Sciences Pharmaceutiques, Toulouse, France
| | - Mustapha Tohfe
- INSERM U563, Department Innovations thérapeutiques et Oncologie moléculaire, Institut Claudius Regaud and Faculté des Sciences Pharmaceutiques, Toulouse, France
| | - Anne Pradines
- INSERM U563, Department Innovations thérapeutiques et Oncologie moléculaire, Institut Claudius Regaud and Faculté des Sciences Pharmaceutiques, Toulouse, France
| | - Antoine Casteignau
- INSERM U563, Department Innovations thérapeutiques et Oncologie moléculaire, Institut Claudius Regaud and Faculté des Sciences Pharmaceutiques, Toulouse, France
| | - Danièle Berg
- INSERM U563, Department Innovations thérapeutiques et Oncologie moléculaire, Institut Claudius Regaud and Faculté des Sciences Pharmaceutiques, Toulouse, France
| | - Gilles Favre
- INSERM U563, Department Innovations thérapeutiques et Oncologie moléculaire, Institut Claudius Regaud and Faculté des Sciences Pharmaceutiques, Toulouse, France
| |
Collapse
|
12
|
Malecaze F, Lubsen NH, Serre B, Decha A, Duboue M, Penary M, Berg D, Arnaud JD, Titeux M, Kremer EJ, Couderc B. Lens cell targetting for gene therapy of prevention of posterior capsule opacification. Gene Ther 2006; 13:1422-9. [PMID: 16724094 DOI: 10.1038/sj.gt.3302790] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Posterior capsule opacification is the main complication of cataract surgery. Using adenovirus-mediated gene transfer, we recently reported that it was feasible to prevent PCO by overexpressing pro-apoptotic molecules such as pro-caspase 3 or Bax in the residual lens epithelial cells post-cataract surgery. However, this approach is feasible only if gene transfer can be restricted to the residual cells responsible for PCO. Initially, we tested an adenovirus (human serotype 5, HAd5), a lentivirus (HIV) and an oncoretrovirus (MLV) vector for the their in vivo transduction efficiency of rabbit lens cells. We found that HAd5 vectors were the most efficient (>90% of the cells could be transduced). Six potential lens-specific promoters were then cloned into HAd5 vectors and assayed for their ability to target expression to a specific population of cells, using in vitro, ex vivo and in vivo rabbit tissues and human lens capsular bags. We found that the LEP503, MIP and Filensin promoters induced strong lens-specific expression of a reporter gene, in human lens cells. Following this ex vivo assay, we showed in a rabbit PCO model that gene transfer could be spatially restricted to the capsular bag by confining the vector with Matrigel. Our combined approach using a lens-specific promoter and a biocompatible gel should render feasible a novel therapeutic strategy for PCO that targets the remaining lens cells.
Collapse
Affiliation(s)
- F Malecaze
- INSERM U563, Department of Ophtalmologie et Pathologie des épithéliums, UPS, Faculté de Médecine Toulouse Rangueil, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Chen Q, Liang D, Yang T, Leone G, Overbeek PA. Distinct capacities of individual E2Fs to induce cell cycle re-entry in postmitotic lens fiber cells of transgenic mice. Dev Neurosci 2005; 26:435-45. [PMID: 15855772 DOI: 10.1159/000082285] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Accepted: 08/30/2004] [Indexed: 11/19/2022] Open
Abstract
PURPOSE Inactivation of the retinoblastoma gene in human retinoblasts or mouse lens fiber cells causes inappropriate cell cycle entry, presumably as a consequence of elevated activity of the E2F transcription factors. Although E2Fs are known to be critical regulators of the cell cycle, it is still unclear whether family members E2F3a, E2F4 or E2F5 are individually capable of inducing cell cycle entry in vivo. In this study, we designed experiments to test whether lens-specific expression of these E2F family members would induce postmitotic fiber cells to re-enter the cell cycle. METHODS Transgenic mice were generated by microinjection of constructs that contained E2F cDNAs (E2F3a, E2F4 or E2F5) linked to the mouse lens-specific alphaA-crystallin promoter. The mice were characterized by histology, in situ hybridization, immunohistochemistry, BrdU incorporation, TUNEL assay and Western blots. RESULTS E2F3a expression was sufficient to induce cell cycle entry in lens fiber cells. Cell cycle re-entry was accompanied by apoptotic cell death resulting in microphthalmia. E2F4 expression stimulated a modest level of cell cycle re-entry, but the transgenic lenses remained normal in size and did not show significant apoptosis. Transgenic mice expressing E2F5 did not show lens defects. In both the E2F3a and E2F4 transgenic lenses, cyclin A2 and cyclin B1 expression were upregulated. Phosphorylated histone H3, a marker for mitosis, was detected in the E2F3a fiber cells. Western blots showed that both p53 and p73alpha were upregulated in the E2F3a lenses. However, expression of p21, a well-known p53 target gene, was not activated, suggesting that p73alpha might be responsible for inducing apoptosis and blocking unregulated proliferation in lens cells overexpressing E2F3a. CONCLUSIONS E2F3a and E2F4, but not E2F5, function to induce cell cycle entry, although E2F4 has more modest activity. E2F3a may induce cell death primarily through activation of p73alpha.
Collapse
Affiliation(s)
- Qin Chen
- College of Optometry, University of Houston, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
14
|
Cvekl A, Yang Y, Chauhan BK, Cveklova K. Regulation of gene expression by Pax6 in ocular cells: a case of tissue-preferred expression of crystallins in lens. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2005; 48:829-44. [PMID: 15558475 PMCID: PMC2080872 DOI: 10.1387/ijdb.041866ac] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Lens development is an excellent model for genetic and biochemical studies of embryonic induction, cell cycle regulation, cellular differentiation and signal transduction. Differentiation of lens is characterized by lens-preferred expression and accumulation of water-soluble proteins, crystallins. Crystallins are required for light transparency, refraction and maintenance of lens integrity. Here, we review mechanisms of lens-preferred expression of crystallin genes by employing synergism between developmentally regulated DNA-binding transcription factors: Pax6, c-Maf, MafA/L-Maf, MafB, NRL, Sox2, Sox1, RARbeta/RXRbeta, RORalpha, Prox1, Six3, gammaFBP-B and HSF2. These factors are differentially expressed in lens precursor cells, lens epithelium and primary and secondary lens fibers. They exert their function in combination with ubiquitously expressed factors (e.g. AP-1, CREB, pRb, TFIID and USF) and co-activators/chromatin remodeling proteins (e.g. ASC-2 and CBP/p300). A special function belongs to Pax6, a paired domain and homeodomain-containing protein, which is essential for lens formation. Pax6 is expressed in lens progenitor cells before the onset of crystallin expression and it serves as an important regulatory factor required for expression of c-Maf, MafA/L-Maf, Six3, Prox1 and retinoic acid signaling both in lens precursor cells and the developing lens. The roles of these factors are illustrated by promoter studies of mouse alphaA-, alphaB-, gammaF- and guinea pig zeta-crystallins. Pax6 forms functional complexes with a number of transcription factors including the retinoblastoma protein, pRb, MafA, Mitf and Sox2. We present novel data showing that pRb antagonizes Pax6-mediated activation of the alphaA-crystallin promoter likely by inhibiting binding of Pax6 to DNA.
Collapse
Affiliation(s)
- Ales Cvekl
- The Department of Ophthalmology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | | | |
Collapse
|
15
|
Chen Q, Liang D, Fromm LD, Overbeek PA. Inhibition of Lens Fiber Cell Morphogenesis by Expression of a Mutant SV40 Large T Antigen That Binds CREB-binding Protein/p300 but Not pRb. J Biol Chem 2004; 279:17667-73. [PMID: 14742445 DOI: 10.1074/jbc.m311678200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Simian virus (SV) 40 large T antigen can both induce tumors and inhibit cellular differentiation. It is not clear whether these cellular changes are synonymous, sequential, or distinct responses to the protein. T antigen is known to bind to p53, to the retinoblastoma (Rb) family of tumor suppressor proteins, and to other cellular proteins such as p300 family members. To test whether SV40 large T antigen inhibits cellular differentiation in vivo in the absence of cell cycle induction, we generated transgenic mice that express in the lens a mutant version of the early region of SV40. This mutant, which we term E107KDelta, has a deletion that eliminates synthesis of small t antigen and a point mutation (E107K) that results in loss of the ability to bind to Rb family members. At embryonic day 15.5 (E15.5), the transgenic lenses show dramatic defects in lens fiber cell differentiation. The fiber cells become post-mitotic, but do not elongate properly. The cells show a dramatic reduction in expression of their beta- and gamma-crystallins. Because CBP and p300 are co-activators for crystallin gene expression, we assayed for interactions between E107KDelta and CBP/p300. Our studies demonstrate that cellular differentiation can be inhibited by SV40 large T antigen in the absence of pRb inactivation, and that interaction of large T antigen with CBP/p300 may be enhanced by a mutation that eliminates the binding to pRb.
Collapse
Affiliation(s)
- Qin Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
16
|
Abstract
Human adenoviruses (Ads) have the ability to transform primary cells, and certain Ads, the subgenus A adenoviruses such as Ad12, induce tumours in immunocompetent rodents. The oncogenic phenotype of the subgenus A adenoviruses is determined by the viral E1A oncogene. In order to generate tumours, Ad12-transformed cells must evade the cellular immune system of the host. Ad12 E1A gene products mediate transcriptional repression of several genes in the major histocompatibility complex (MHC) involved in antigen processing and presentation, resulting in evasion of cytotoxic T lymphocyte (CTL) killing of transformed cells. In this review, the molecular mechanisms of E1A-mediated transcriptional repression of MHC gene expression are described. In addition, evasion of natural killer (NK) cell killing by Ad-transformed cells is also considered.
Collapse
Affiliation(s)
- G E Blair
- School of Biochemistry and Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | | |
Collapse
|
17
|
Bhat SP. Crystallins, genes and cataract. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2003; 60:205-62. [PMID: 12790344 DOI: 10.1007/978-3-0348-8012-1_7] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Far from being a physical entity, assembled of inanimate structural proteins, the ocular lens epitomizes the biological ingenuity that sustains an essential and near-perfect physical system of immaculate optics. Crystallins (alpha, beta, and gamma) provide transparency by dint of their high concentration, but it is debatable whether proteins that provide transparency are any different, biologically or structurally, from those that are present in non-transparent structures or tissues. It is becoming increasingly clear that crystallins may have a plethora of metabolic and regulatory functions, both within the lens as well as outside of it. Alpha-crystallins are members of a small heat shock family of proteins and beta/gamma-crystallins belong to the family of epidermis-specific differentiation proteins. Crystallin gene expression has been studied from the perspective of the lens specificity of their promoters. Mutations in alpha-, beta-, and gamma-crystallins are linked with the phenotype of the loss of transparency. Understanding catalytic, non-structural properties of crystallins may be critical for understanding the malfunction in molecular cascades that lead to cataractogenesis and its eventual therapeutic amelioration.
Collapse
Affiliation(s)
- Suraj P Bhat
- Jules Stein Eye Institute and Brain Research Institute, Geffen School of Medicine at UCLA, Los Angeles, CA 90077-7000, USA.
| |
Collapse
|
18
|
Chen Q, Dowhan DH, Liang D, Moore DD, Overbeek PA. CREB-binding protein/p300 co-activation of crystallin gene expression. J Biol Chem 2002; 277:24081-9. [PMID: 11943779 DOI: 10.1074/jbc.m201821200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although some of the transcription factors that are required for expression of crystallins during lens development have been identified, the molecular interactions that contribute to enhanced crystallin expression are not yet well defined. In this study, we designed experiments to test whether the co-activators CREB-binding protein (CBP) and/or p300 interact with c-Maf, Prox-1, or Sox-1 to enhance transcription of crystallin genes. Promoter regions from the mouse alphaA-, betaB2-, and gammaF-crystallin genes were linked to a luciferase reporter. Expression of c-Maf transactivated each of these promoters. Of particular interest, co-expression of CBP or p300 with c-Maf was found to synergistically co-activate each promoter. CBP and p300 were less effective or ineffective at co-activation with Prox-1 or Sox-1. Co-immunoprecipitation and mammalian two-hybrid experiments revealed that CBP and p300 bind to c-Maf and Prox-1 but not to Sox-1. The co-activation of c-Maf by CBP/p300 requires histone acetyltransferase activity. Our results suggest that c-Maf recruits CBP and/or p300 to crystallin promoters leading to up-regulation of crystallin gene expression through localized histone acetylation and consequent chromatin re-modeling. In a promoter-specific fashion, co-activation can be modulated by Prox-1 and/or Sox-1. This modulation may help to specify the endogenous levels of crystallin gene expression.
Collapse
Affiliation(s)
- Qin Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|