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










Publication year range
1.
Exp Cell Res ; 330(1): 186-98, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25128817

ABSTRACT

Cyclin dependent kinase 5 (Cdk5), a proline-directed serine/threonine kinase, requires p39 for its enzymatic activity, and is implicated in cytoskeletal organization and contraction in numerous cell types. The C-terminus of p39 binds muskelin, a multi-domain scaffolding protein known to affect cytoskeletal organization, but the mechanisms by which muskelin affects cytoskeletal organization remain unclear. The present study sought to determine whether p39 might serve as an adaptor protein that links muskelin to stress fibers and to investigate the possible biological relevance of such an interaction. Double immunoprecipitation showed that muskelin, p39, and myosin II are components of a single intracellular complex, and suppressing p39 abrogated the interaction between muskelin and the myosin subunits, demonstrating that p39 is required to link muskelin to myosin II. Muskelin is colocalized with myosin regulatory light chain (MRLC) and on stress fibers. The suppression of muskelin reduced Rho-GTP, MRLC phosphorylation, disrupted stress fiber organization, and promoted cell migration, all of which closely mimic the effect of Cdk5 inhibition. Moreover, suppressing muskelin and inhibiting Cdk5 together have no additional effect, indicating that muskelin plays an important role in Cdk5-dependent signaling. p39 is necessary and sufficient for Cdk5-dependent regulation of MRLC phosphorylation, as suppression of p39, but not p35, reduces MRLC phosphorylation. Together, these results demonstrate that p39 specifically links muskelin to myosin II and consequently, to stress fibers and reveal a novel role for muskelin in regulating myosin phosphorylation and cytoskeletal organization.


Subject(s)
Actins/metabolism , Cell Adhesion Molecules/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lens, Crystalline/metabolism , Myosin Type II/metabolism , Nerve Tissue Proteins/metabolism , Stress Fibers/metabolism , Binding Sites , Cell Line , Humans , Myosin Light Chains/metabolism , Nerve Tissue Proteins/chemistry , Phosphorylation , Protein Binding , rho GTP-Binding Proteins/metabolism
2.
G3 (Bethesda) ; 3(12): 2239-55, 2013 Dec 09.
Article in English | MEDLINE | ID: mdl-24142921

ABSTRACT

MicroRNAs (miRNAs) and fibroblast growth factor (FGF) signaling regulate a wide range of cellular functions, including cell specification, proliferation, migration, differentiation, and survival. In lens, both these systems control lens fiber cell differentiation; however, a possible link between these processes remains to be examined. Herein, the functional requirement for miRNAs in differentiating lens fiber cells was demonstrated via conditional inactivation of Dicer1 in mouse (Mus musculus) lens. To dissect the miRNA-dependent pathways during lens differentiation, we used a rat (Rattus norvegicus) lens epithelial explant system, induced by FGF2 to differentiate, followed by mRNA and miRNA expression profiling. Transcriptome and miRNome analysis identified extensive FGF2-regulated cellular responses that were both independent and dependent on miRNAs. We identified 131 FGF2-regulated miRNAs. Seventy-six of these miRNAs had at least two in silico predicted and inversely regulated target mRNAs. Genes modulated by the greatest number of FGF-regulated miRNAs include DNA-binding transcription factors Nfib, Nfat5/OREBP, c-Maf, Ets1, and N-Myc. Activated FGF signaling influenced bone morphogenetic factor/transforming growth factor-ß, Notch, and Wnt signaling cascades implicated earlier in lens differentiation. Specific miRNA:mRNA interaction networks were predicted for c-Maf, N-Myc, and Nfib (DNA-binding transcription factors); Cnot6, Cpsf6, Dicer1, and Tnrc6b (RNA to miRNA processing); and Ash1l, Med1/PBP, and Kdm5b/Jarid1b/Plu1 (chromatin remodeling). Three miRNAs, including miR-143, miR-155, and miR-301a, down-regulated expression of c-Maf in the 3'-UTR luciferase reporter assays. These present studies demonstrate for the first time global impact of activated FGF signaling in lens cell culture system and predicted novel gene regulatory networks connected by multiple miRNAs that regulate lens differentiation.


Subject(s)
Cell Differentiation/genetics , Fibroblast Growth Factor 2/genetics , Lens, Crystalline/cytology , MicroRNAs/metabolism , Animals , Cell Cycle Checkpoints/genetics , Cells, Cultured , DEAD-box RNA Helicases/genetics , Female , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression Profiling , Gene Regulatory Networks , In Vitro Techniques , Lens, Crystalline/abnormalities , Lens, Crystalline/drug effects , Lens, Crystalline/physiology , Mice , Mice, Mutant Strains , Pregnancy , RNA, Messenger/metabolism , Rats , Ribonuclease III/genetics , Transcriptome
3.
J Biol Chem ; 287(48): 40779-92, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23012360

ABSTRACT

BACKGROUND: Lens degeneration in Fpr1(-/-) mice prompted us to search for functional FPR1 expression directly on lens epithelial cells. RESULTS: FPR1 is functionally expressed on human lens epithelial cells but has atypical properties compared with hematopoietic cell FPR1. CONCLUSION: Lens epithelial cell FPR1 may be involved in development and maintenance of the lens. SIGNIFICANCE: This is the first link between non-hematopoietic expression of FPR1 and an ophthalmologic phenotype. Formyl peptide receptor 1 (FPR1) is a G protein-coupled chemoattractant receptor expressed mainly on leukocytes. Surprisingly, aging Fpr1(-/-) mice develop spontaneous lens degeneration without inflammation or infection (J.-L. Gao et al., manuscript in preparation). Therefore, we hypothesized that FPR1 is functionally expressed directly on lens epithelial cells, the only cell type in the lens. Consistent with this, the human fetal lens epithelial cell line FHL 124 expressed FPR1 mRNA and was strongly FPR1 protein-positive by Western blot and FACS. Competition binding using FPR1 ligands N-formyl-Nle-Leu-Phe-Nle-Tyr-Lys (Nle = Norleucine), formylmethionylleucylphenylalanine, and peptide W revealed the same profile for FHL 124 cells, neutrophils, and FPR1-transfected HEK 293 cells. Saturation binding with fluorescein-labeled N-formyl-Nle-Leu-Phe-Nle-Tyr-Lys revealed ~2500 specific binding sites on FHL-124 cells (K(D) ~ 0.5 nm) versus ~40,000 sites on neutrophils (K(D) = 3.2 nm). Moreover, formylmethionylleucylphenylalanine induced pertussis toxin-sensitive Ca(2+) flux in FHL 124 cells, consistent with classic G(i)-mediated FPR1 signaling. FHL 124 cell FPR1 was atypical in that it resisted agonist-induced internalization. Expression of FPR1 was additionally supported by detection of the intact full-length open reading frame in sequenced cDNA from FHL 124 cells. Thus, FHL-124 cells express functional FPR1, which is consistent with a direct functional role for FPR1 in the lens, as suggested by the phenotype of Fpr1 knock-out mice.


Subject(s)
Epithelial Cells/metabolism , Lens, Crystalline/metabolism , Receptors, Formyl Peptide/genetics , Animals , Cell Line , Humans , Mice , Mice, Knockout , Receptors, Formyl Peptide/metabolism
4.
Dev Biol ; 362(2): 219-29, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22173065

ABSTRACT

Notch signaling is essential for proper lens development, however the specific requirements of individual Notch receptors have not been investigated. Here we report the lens phenotypes of Notch2 conditionally mutant mice, which exhibited severe microphthalmia, reduced pupillary openings, disrupted fiber cell morphology, eventual loss of the anterior epithelium, fiber cell dysgenesis, denucleation defects, and cataracts. Notch2 mutants also had persistent lens stalks as early as E11.5, and aberrant DNA synthesis in the fiber cell compartment by E14.5. Gene expression analyses showed that upon loss of Notch2, there were elevated levels of the cell cycle regulators Cdkn1a (p21Cip1), Ccnd2 (CyclinD2), and Trp63 (p63) that negatively regulates Wnt signaling, plus down-regulation of Cdh1 (E-Cadherin). Removal of Notch2 also resulted in an increased proportion of fiber cells, as was found in Rbpj and Jag1 conditional mutant lenses. However, Notch2 is not required for AEL proliferation, suggesting that a different receptor regulates this process. We found that Notch2 normally blocks lens progenitor cell death. Overall, we conclude that Notch2-mediated signaling regulates lens morphogenesis, apoptosis, cell cycle withdrawal, and secondary fiber cell differentiation.


Subject(s)
Apoptosis/genetics , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/physiology , Lens, Crystalline/embryology , Morphogenesis/physiology , Receptor, Notch2/metabolism , Signal Transduction/physiology , Animals , Cell Cycle/physiology , DNA Primers/genetics , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Lens, Crystalline/metabolism , Lens, Crystalline/ultrastructure , Mice , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
5.
Cell Mol Life Sci ; 68(20): 3425-36, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21442427

ABSTRACT

The non-receptor tyrosine kinase Src is a critical regulator of cytoskeletal contraction, cell adhesion, and migration. In normal cells, Src activity is stringently controlled by Csk-dependent phosphorylation of Src(Y530), and by Cullin-5-dependent ubiquitinylation, which affects active Src(pY419) exclusively, leading to its degradation by the proteosome. Previous work has shown that Src activity is also limited by Cdk5, a proline-directed kinase, which has been shown to phosphorylate Src(S75). Here we show that this phosphorylation promotes the ubiquitin-dependent degradation of Src, thus restricting the availability of active Src. We demonstrate that Src(S75) phosphorylation occurs in vivo in epithelial cells, and like ubiquitinylation, is associated only with active Src. Preventing Cdk5-dependent phosphorylation of Src(S75), by site-specific mutation of S75 or by Cdk5 inhibition or suppression, increases Src(Y419) phosphorylation and kinase activity, resulting in Src-dependent cytoskeletal changes. In transfected cells, ubiquitinylation of Src(S75A) is about 35% that of wild-type Src-V5, and its half-life is approximately 2.5-fold greater. Cdk5 suppression leads to a comparable decrease in the ubiquitinylation of endogenous Src and a similar increase in Src stability. Together, these findings demonstrate that Cdk5-dependent phosphorylation of Src(S75) is a physiologically significant mechanism of regulating intracellular Src activity.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Epithelial Cells/metabolism , Lens, Crystalline/metabolism , Protein-Tyrosine Kinases/metabolism , Ubiquitin/metabolism , Blotting, Western , CSK Tyrosine-Protein Kinase , Cells, Cultured , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Lens, Crystalline/cytology , Lens, Crystalline/drug effects , Mutation/genetics , Phosphorylation/drug effects , Protein-Tyrosine Kinases/genetics , Ubiquitination , src-Family Kinases
6.
Cell Adh Migr ; 4(3): 333-6, 2010.
Article in English | MEDLINE | ID: mdl-20190570

ABSTRACT

Cell adhesion is a fundamental property of epithelial cells required for anchoring, migration and survival. During cell migration, the formation and disruption of adhesion sites is stringently regulated by integration of multiple, sequential signals acting in distinct regions of the cell. Recent findings implicate cyclin dependent kinase 5 (Cdk5) in the signaling pathways that regulate cell adhesion and migration of a variety of cell types. Experiments with epithelial cell lines indicate that Cdk5 activity exerts its effects by limiting Src activity in regions where Rho activity is required for stress fiber contraction and by phosphorylating the talin head to stabilize nascent focal adhesions. Both pathways regulate cell migration by increasing adhesive strength.


Subject(s)
Cell Movement , Cyclin-Dependent Kinase 5/metabolism , Epithelial Cells/cytology , Epithelial Cells/enzymology , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Epithelial Cells/drug effects , Humans , Models, Biological , Myosin Light Chains/metabolism , Protein Kinase Inhibitors/pharmacology , Stress Fibers/drug effects , Stress Fibers/metabolism , Talin/metabolism
7.
Mol Cell Biol ; 29(24): 6488-99, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19822667

ABSTRACT

Cdk5 regulates adhesion and migration in a variety of cell types. We previously showed that Cdk5 is strongly activated during stress fiber formation and contraction in spreading cells. Here we determine the mechanism linking Cdk5 to stress fiber contractility and its relevance to cell migration. Immunofluorescence showed that Cdk5 colocalized with phosphorylated myosin regulatory light chain (pMRLC) on contracting stress fibers. Inhibiting Cdk5 activity by various means significantly reduced pMRLC level and cytoskeletal contraction, with loss of central stress fibers. Blocking Cdk5 activity also reduced Rho-Rho kinase (ROCK) signaling, which is the principal pathway of myosin phosphorylation under these conditions. Next, we examined the effect of Cdk5 activity on Src, a known regulator of Rho. Inhibiting Cdk5 activity increased Src activation and phosphorylation of its substrate, p190RhoGAP, an upstream inhibitor of Rho. Inhibiting both Cdk5 and Src activity completely reversed the effect of Cdk5 inhibition on Rho and prevented the loss of central stress fibers, demonstrating that Cdk5 exerts its effects on Rho-ROCK signaling by suppressing Src activity. Moreover, inhibiting either Cdk5 or ROCK activity increased cell migration to an equal extent, while inhibiting both kinases produced no additional effect, demonstrating that Cdk5-dependent regulation of ROCK activity is a physiological determinant of migration rate.


Subject(s)
Cell Movement/physiology , Cyclin-Dependent Kinase 5/metabolism , Cytoskeleton/metabolism , Epithelial Cells/physiology , Guanine Nucleotide Exchange Factors/metabolism , Repressor Proteins/metabolism , rho GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism , Cell Line , Cyclin-Dependent Kinase 5/genetics , Enzyme Activation , Enzyme Inhibitors/metabolism , Epithelial Cells/cytology , Guanine Nucleotide Exchange Factors/genetics , Humans , Kinetin/metabolism , Myosin Light Chains/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction/physiology , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , src-Family Kinases/genetics
8.
J Vis Exp ; (31)2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19773734

ABSTRACT

The anterior surface of the ocular lens is covered by a monolayer of epithelial cells, which proliferate in an annular zone underlying the ciliary body. Following division, these cells migrate posteriorly, where FGF diffusing from the retina induces them to differentiate into a posterior array of elongated lens fiber cells, which compose the bulk of the lens. Differentiation of lens epithelial cells into lens fibers can be induced in vitro by culturing explants of the central region of the anterior epithelium in the presence of FGF-2. Explants are prepared from lenses of neonatal rats by removing the lens from the eye and grasping the lens capsule on the posterior side with dissecting tweezers. The posterior capsule is then gently torn open and pressed down into the plastic bottom of a tissue culture dish. The peripheral regions of the explant are removed with a scalpel and the central area is then cultured in the presence of 100 ng/ml FGF-2 for as long as 2-3 weeks, depending on the parameters to be studied. Since epithelial cells in cultured explants differentiate in approximate synchrony over a period of days to weeks, the time course of signaling and gene expression can be determined using molecular, biochemical, and pharmacological techniques. Immunofluorescence microscopy is a powerful adjunct to these methods as it demonstrates the subcellular localization of proteins of interest and can reveal the physiological consequences of experimental manipulations of signaling pathways.


Subject(s)
Cell Differentiation/physiology , Lens, Crystalline/cytology , Tissue Culture Techniques/methods , Animals , Cells, Cultured , Epithelial Cells/cytology , Fibroblast Growth Factor 2/pharmacology , Rats
9.
Dev Biol ; 332(1): 166-76, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19481073

ABSTRACT

Previous studies of the developing lens have shown that Notch signaling regulates differentiation of lens fiber cells by maintaining a proliferating precursor pool in the anterior epithelium. However, whether Notch signaling is further required after the onset of fiber cell differentiation is not clear. This work investigates the role of Notch2 and Jagged1 (Jag1) in secondary fiber cell differentiation using rat lens epithelial explants undergoing FGF-2 dependent differentiation in vitro. FGF induced Jag1 expression and Notch2 signaling (as judged by the appearance of activated Notch2 Intracellular Domain (N2ICD)) within 12-24 h. These changes were correlated with induction of the Notch effector, Hes5, upregulation of N-cadherin (N-cad), and downregulation of E-cadherin (E-cad), a cadherin switch characteristic of fiber cell differentiation. Induction of Jag1 was efficiently blocked by U0126, a specific inhibitor of MAPK/ERK signaling, indicating a requirement for signaling through this pathway downstream of the FGF receptor. Other growth factors that activate MAPK/ERK signaling (EGF, PDGF, IGF) did not induce Jag1. Inhibition of Notch signaling using gamma secretase inhibitors DAPT and L-685,458 or anti-Jag1 antibody markedly decreased FGF-dependent expression of Jag1 demonstrating Notch-dependent lateral induction. In addition, inhibition of Notch signaling reduced expression of N-cad, and the cyclin dependent kinase inhibitor, p57Kip2, indicating a direct role for Notch signaling in secondary fiber cell differentiation. These results demonstrate that Notch-mediated lateral induction of Jag1 is an essential component of FGF-dependent lens fiber cell differentiation.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Differentiation/drug effects , Fibroblast Growth Factors/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/drug effects , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Biomarkers/metabolism , Cadherins/metabolism , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Epithelium/drug effects , Epithelium/metabolism , Insulin-Like Growth Factor I/pharmacology , Jagged-1 Protein , MAP Kinase Signaling System/drug effects , Microdissection , Models, Biological , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Wistar , Serrate-Jagged Proteins
10.
Exp Cell Res ; 314(19): 3542-50, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18838073

ABSTRACT

Previous studies have shown that Cdk5 promotes lens epithelial cell adhesion. Here we use a cell spreading assay to investigate the mechanism of this effect. As cells spread, forming matrix adhesions and stress fibers, Cdk5(Y15) phosphorylation and Cdk5 kinase activity increased. Cdk5(Y15) phosphorylation was inhibited by PP1, a Src family kinase inhibitor. To identify the PP1-sensitive kinase, we transfected cells with siRNA oligonucleotides for cSrc and related kinases. Only cSrc siRNA oligonucleotides inhibited Cdk5(Y15) phosphorylation. Cdk5(pY15) and its activator, p35, colocalized with actin in stress fibers. To examine Cdk5 function, we inhibited Cdk5 activity under conditions that also prevent phosphorylation at Y15: expression of kinase inactive mutations Cdk5(Y15F) and Cdk5(K33T), and siRNA suppression of Cdk5. Stress fiber formation was severely inhibited. To distinguish between a requirement for Cdk5 kinase activity and a possible adaptor role for Cdk5(pY15), we used two methods that inhibit kinase activity without inhibiting phosphorylation at Y15: pharmacological inhibition with olomoucine and expression of the kinase inactive mutation, Cdk5(D144N). Stress fiber organization was altered, but stress fiber formation was not blocked. These findings indicate that Cdk5(Y15) phosphorylation and Cdk5 activity have distinct functions required for stress fiber formation and organization, respectively.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Stress Fibers/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cyclin-Dependent Kinase 5/analysis , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cytoskeleton/metabolism , Epithelial Cells/metabolism , Humans , Mutation , Phosphorylation , RNA, Small Interfering/metabolism , Rabbits , Stress Fibers/ultrastructure , Transfection , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/metabolism
11.
Mol Vis ; 14: 542-9, 2008 Mar 17.
Article in English | MEDLINE | ID: mdl-18385789

ABSTRACT

PURPOSE: To investigate the effect of the Cdk5 inhibitor olomoucine on corneal debridement wound healing in vivo. METHODS: Corneal debridement wounds of 1.5 mm were made on the ocular surface of CD-1 mice. A 20 microl drop of 15 microM olomoucine in 1% DMSO was applied to the wound area immediately after wounding and again after 6 h. Control mice received identical applications of 1% DMSO. Mice were euthanized after 18 h, two weeks, and three weeks for evaluation of wound healing and restratification. Corneas were stained with Richardson's dye, photographed, and processed for histology and immunofluorescence as whole mounts or paraffin sections. The remaining wound area at 18 h was measured by image analysis. Scratch wounded cultures of human corneal-limbal epithelial cells (HCLE) were used to examine the effect of olomoucine on matrix metalloproteinase (MMP) expression in vitro. MMP-2 and MMP-9 were detected by immunofluorescence and immunoblotting. RESULTS: Olomoucine treatment significantly enhanced corneal wound closure without increasing inflammation or infiltration of polymorphonuclear leukocytes 18 h after wounding (p<0.05). The increased localization of MMP-9 within epithelial cells at the wound edge was further enhanced by olomoucine while the expression of MMP-2 was reduced. Olomoucine treatment of scratch wounded HCLE cells produced similar changes in MMP-9 and MMP-2 expression. The examination of treated corneas two and three weeks after wounding showed normal epithelial restratification with no evidence of inflammation or stromal disorganization. CONCLUSIONS: Topical application of olomoucine in 1% DMSO significantly enhances closure of small epithelial debridement wounds without increasing inflammation or impairing reepithelialization.


Subject(s)
Cornea/physiopathology , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Debridement , Enzyme Inhibitors/pharmacology , Kinetin/pharmacology , Wound Healing/drug effects , Animals , Cadherins/metabolism , Cells, Cultured , Cornea/metabolism , Cornea/pathology , Cornea/surgery , Epithelial Cells/enzymology , Humans , Limbus Corneae/drug effects , Limbus Corneae/enzymology , Limbus Corneae/injuries , Limbus Corneae/pathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred Strains , Neutrophils/pathology , Time Factors , Tissue Distribution
12.
Biochem Biophys Res Commun ; 354(4): 1034-9, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17276406

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) has been shown to regulate adhesion and migration of lens and corneal epithelial cells. To explore protein-protein interactions that may mediate these functions, we performed yeast two-hybrid screening on an embryonic rat lens library using Cdk5 and its regulators, p35 and p39 as baits. This screen identified an interaction between p39 and non-muscle myosin essential light chain (MLC(17)). GST pull-down experiments demonstrated that p39 binds directly to MLC(17) through a strong binding site in the N-terminal 109 amino acids of p39. Immunoprecipitation of proteins from Cos1 cells co-transfected with GFP-MLC(17) and HA-p39 confirmed that these proteins interact intracellularly. Immunofluorescence microscopy of co-transfected lens epithelial cells showed that GFP-MLC(17) and HA-p39 co-localize along cytoskeletal fibrils. Moreover, endogenous rat lens p39 co-immunoprecipitated with MLC(17) and myosin heavy chain II (MHC II), demonstrating that the interaction is physiological and serves to link p39 to the cytoskeleton.


Subject(s)
Myosin Light Chains/metabolism , Nerve Tissue Proteins/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Cyclin-Dependent Kinase 5/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Myosin Heavy Chains/metabolism , Rabbits , Rats , Subcellular Fractions/chemistry , Two-Hybrid System Techniques
13.
J Biol Chem ; 280(22): 21376-83, 2005 Jun 03.
Article in English | MEDLINE | ID: mdl-15797862

ABSTRACT

Previous studies implicate cyclin-dependent kinase 5 in cell adhesion and migration of epithelial cells of the cornea and lens. To explore molecular interactions underlying these functions, we performed yeast two-hybrid screening of an embryonic rat lens library for proteins that interact with cyclin-dependent kinase 5 and its regulators, p35 and p39. This screen identified a specific interaction between p39 and muskelin, an intracellular protein known to affect cytoskeletal organization in adherent cells. Immunohistochemistry detected muskelin in the developing lens and in other tissues, including brain and muscle. Glutathione S-transferase pull-down experiments and co-immunoprecipitations confirmed the specificity of the p39-muskelin interaction. Deletion analysis of p39 showed that muskelin binds to the p39 C terminus, which contains a short insertion (amino acids 329-366) absent from p35. Similar analysis of muskelin mapped the interaction with p39 to the fifth kelch repeat. Co-expression of p39 and muskelin in COS1 cells or lens epithelial cells altered the intracellular localization of muskelin, recruiting it to the cell periphery. These findings demonstrate a novel interaction between muskelin and the cyclin-dependent kinase 5 activator p39 and suggest that p39 may regulate the subcellular localization of muskelin.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Actins/chemistry , Animals , Binding Sites , Brain/metabolism , COS Cells , Cell Adhesion , Cell Adhesion Molecules , Cell Line , Cell Movement , Cyclin-Dependent Kinase 5 , Cytoskeleton/metabolism , DNA, Complementary/metabolism , Epithelial Cells/metabolism , Gene Deletion , Gene Library , Glutathione Transferase/metabolism , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Lens, Crystalline/metabolism , Microscopy, Fluorescence , Muscles/metabolism , Oligonucleotides/chemistry , Peptides/chemistry , Phosphotransferases/metabolism , Plasmids/metabolism , Protein Binding , RNA/chemistry , RNA, Messenger/metabolism , Rabbits , Rats , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Two-Hybrid System Techniques
14.
Int J Dev Biol ; 48(8-9): 857-65, 2004.
Article in English | MEDLINE | ID: mdl-15558477

ABSTRACT

Cell movements during lens development and differentiation involve dynamic regulation of cell-matrix and cell-cell adhesion. How these processes are regulated depends on the particular array of matrix components and adhesion proteins that are expressed, as well as the signaling pathways that affect them. This review examines what is known about adhesion proteins and their regulation in the lens in light of recent findings about the mechanism of cell migration. The characteristic shape and organization of the lens depends on highly regulated cell movements during development and differentiation. Epithelial cells at the equator migrate posteriorly, bringing them into contact with factors in the vitreous humor and initiating differentiation. Elongation of the differentiating fiber cells is coupled with directed migration, posteriorly along the capsule and anteriorly along the fiber cell-epithelial interface, to generate a symmetrically organized fiber cell mass with aligned suture planes. To make these movements, cells systematically create and dissolve cell-cell and cell-matrix adhesions, form connections between these adhesions and the cytoskeleton, and generate contractile force. Since errors in cell migration may lead to aberrant lens shape or misplacement of the lens sutures, precise regulation of each step is essential for the optical quality of the lens. Recent advances in cellular developmental biology have begun to shed light on the molecular mechanisms underlying cell movements and the changes in adhesion that make them possible. This review will summarize those findings and relate them to relevant studies of the lens to provide an outline of the cellular events that lead to lens morphogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Lens, Crystalline/embryology , Animals , Cell Adhesion , Cell Differentiation , Cell Movement , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Humans , Integrins/metabolism , Lens, Crystalline/metabolism , Models, Biological , Protein Isoforms , Signal Transduction , rho GTP-Binding Proteins/metabolism
15.
Mol Vis ; 10: 663-7, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15448618

ABSTRACT

PURPOSE: Constitutive expression of RNA sequences complementary to the chemokine CCL27 mRNA has been found in the normal mouse eye. This study examines the nature and location of these endogenous RNAs in ocular tissues. METHODS: Conventional RT-PCR, 5' RACE, and dideoxy DNA sequencing were used to examine the sequences of CCL27 related RNAs in the eye. Expression levels of specific RNAs were measured by real time PCR. Tissue distribution of RNA transcripts was determined by RT-PCR using RNA from microdissected tissues and by in situ hybridization with radiolabeled riboprobes. RESULTS: We detect 5 distinct splice variants derived from transcription of the CCL27 gene locus. The most abundant form codes for a non-secreted protein, PESKY, and is expressed in lens, cornea, and retina. Another variant corresponds to the mRNA of the secreted chemokine and is synthesized in the cornea, but not in retina or lens. The remaining splice variants are novel and may be eye specific, but have only short open reading frames (<50 amino acids). CCL27 transcripts are most abundantly expressed in the retina, as judged by in situ hybridization. CONCLUSIONS: PESKY and other CCL27 splice variants of unknown function are widely expressed in ocular tissues. Analysis of CCL27 transcripts from lens, retina, and cornea indicates that mRNA for the secreted chemokine, CCL27, is endogenously expressed only in the cornea and may play a role in ocular immune responses involving CD4 lymphocytes in this tissue.


Subject(s)
Alternative Splicing/genetics , Chemokines, CC/genetics , Cornea/metabolism , Gene Expression Regulation/physiology , Lens, Crystalline/metabolism , RNA, Messenger/metabolism , Retina/metabolism , Animals , Chemokine CCL27 , In Situ Hybridization , Mice , Mice, Inbred BALB C , Oligonucleotide Probes/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
16.
J Biol Chem ; 279(30): 31813-22, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15145928

ABSTRACT

Lens major intrinsic protein (MIP), exclusive to the vertebrate lens, otherwise known as MIP26 and Aquaporin 0, is abundantly expressed as a lens fiber membrane protein. Although relatively less efficient compared with other aquaporins, MIP is suggested to function as a water channel, as an adhesion molecule, and is required for lens transparency. Because MIP is specifically expressed in lens fiber cells, we investigated in this study the activation of Mip expression after triggering differentiation of rat lens epithelia explants by fibroblast growth factor (FGF)-2. Here, we show that Mip expression in the lens cells is regulated by FGF-2. Using Real time PCR we demonstrate that endogenous Mip levels in the explants were up-regulated upon FGF-2 stimulation, in a concentration-dependent manner. Up-regulation of Mip at the transcriptional level was simultaneous with the activation of the FGF down-stream signaling components, ERK1/2 and JNK. Specific inhibitors, UO126 for ERK1/2 and SP600125 for JNK, abrogated Mip expression in response to FGF-2 in the explants. This inhibition pattern was recapitulated in reporter assays for transfection of the rat lens epithelia explants, driven by the Mip promoter (-1648/+44). Our studies show that ERK1/2 and JNK signaling pathways are required for Mip expression in lens epithelia explants induced to differentiate by FGF-2.


Subject(s)
Eye Proteins/metabolism , Fibroblast Growth Factor 2/pharmacology , Lens Capsule, Crystalline/drug effects , Lens Capsule, Crystalline/metabolism , Membrane Glycoproteins/metabolism , Animals , Aquaporins , Base Sequence , Cell Differentiation/drug effects , Culture Techniques , DNA Primers/genetics , Enzyme Activation/drug effects , Epithelium/drug effects , Epithelium/growth & development , Epithelium/metabolism , Eye Proteins/genetics , Gene Expression Regulation/drug effects , JNK Mitogen-Activated Protein Kinases , Lens Capsule, Crystalline/growth & development , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
17.
Invest Ophthalmol Vis Sci ; 45(3): 863-71, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14985303

ABSTRACT

PURPOSE: Major intrinsic protein (MIP), also called aquaporin-0, is essential for lens transparency and is specifically expressed in the lens fiber cell membranes. The goal of the current study was to identify and characterize proteins that interact with MIP and to elucidate the role of these interactions in MIP functions. METHODS: The C-terminal 74-amino-acid fragment of MIP was used as bait to screen a rat lens cDNA yeast two-hybrid library. The full-length MIP was expressed as enhanced green fluorescent protein (EGFP)-tagged or myc-tagged proteins, and gammaE-crystallin was expressed as FLAG-tagged or red fluorescent protein (HcRed)-tagged proteins, respectively, in the RK13 rabbit kidney epithelial cell line. Protein-protein interactions were analyzed by coimmunoprecipitation assays and visualized by confocal fluorescence microscopy. RESULTS: gammaE-Crystallin, a water-soluble protein that is specifically expressed in lens fibers, was identified as a binding protein to the MIP C-terminal peptide. Coimmunoprecipitation assays demonstrated that gammaE-crystallin interacts specifically with full-length MIP in mammalian cells. MIP did not interact with gammaD-crystallin, another member of the highly conserved gamma-crystallin gene family. Confocal fluorescence microscopy demonstrated that MIP interacted with gammaE-crystallin in individual mammalian cells and that this interaction resulted in the recruitment of gammaE-crystallin from the cytoplasm to the plasma membrane. CONCLUSIONS: These experiments provide the first demonstration of MIP interaction with other lens proteins at the molecular level and raise the possibility of a structural role of MIP in the organization of gamma-crystallins in lens fibers.


Subject(s)
Antigens, Surface/metabolism , Cell Membrane/metabolism , Crystallins/metabolism , Eye Proteins/metabolism , Membrane Glycoproteins/metabolism , Animals , Aquaporins , Blotting, Western , Cell Line , Gene Library , Green Fluorescent Proteins , Kidney/cytology , Kidney/metabolism , Luminescent Proteins/metabolism , Microscopy, Confocal , Plasmids , Protein Binding , Protein Interaction Mapping , Rabbits , Rats , Two-Hybrid System Techniques , gamma-Crystallins
18.
Invest Ophthalmol Vis Sci ; 44(12): 5259-68, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14638725

ABSTRACT

PURPOSE: To demonstrate the interactions of PKCgamma with caveolin (Cav)-1 and connexin(Cx)43 in lipid rafts and its regulation of gap junctions. METHODS: N/N1003A lens epithelial cells, bovine primary lens epithelial cells, and stably transfected N/N1003A lens epithelial cells were used. Coimmunoprecipitation and Western blot analysis were used to detect protein expression and their interactions. Cav-1-containing lipid rafts and redistribution of Cav-1, PKCgamma, and Cx43 were analyzed by sucrose gradients and by consequent Western blot analysis. Cell surface gap junction Cx43 plaques were detected by confocal microscopy. PKCgamma activity was measured with a PKC assay kit. RESULTS: Cav-1 and -2 were found in N/N1003A and bovine primary lens epithelial cells. Cx43 was associated with Cav-1 in lipid rafts. Phorbol ester (TPA) and insulin-like growth factor (IGF)-1 recruited PKCgamma into Cav-1-containing lipid rafts and stimulated the interactions of PKCgamma with Cav-1 and Cx43. TPA and IGF-1 induced redistribution of Cav-1 and Cx43 from light-density fractions to higher density fractions on sucrose gradients. PKCgamma redistributed with Cav-1- and Cx43-containing fractions on stimulation with TPA or IGF-1. Overexpression of PKCgamma-enhanced green fluorescent protein (EGFP) increased the interaction of PKCgamma-EGFP with Cav-1 and Cx43 and decreased gap junction Cx43 plaques without exogenous growth factors. Overexpression of a loss-of-function PKCgamma mutant did not decrease gap junction Cx43 plaques or cause redistribution in lipid rafts, even though the PKCgamma mutant still interacted with Cav-1 and Cx43. CONCLUSIONS: Activation of PKCgamma by TPA or IGF-1 stimulated the interaction of PKCgamma with Cav-1 and Cx43 in lipid rafts, causing Cx43, Cav-1, and PKCgamma to redistribute within the lipid rafts, and this resulted in a decrease in gap junction plaques.


Subject(s)
Caveolins/metabolism , Epithelial Cells/metabolism , Gap Junctions/metabolism , Lens, Crystalline/metabolism , Lipid Metabolism , Protein Kinase C/metabolism , Animals , Blotting, Western , Cattle , Caveolin 1 , Cells, Cultured , Centrifugation, Density Gradient , Connexin 43/metabolism , Epithelial Cells/drug effects , Green Fluorescent Proteins , Insulin-Like Growth Factor I/pharmacology , Isoenzymes/metabolism , Lens, Crystalline/drug effects , Luminescent Proteins , Microscopy, Confocal , Mutagenesis, Site-Directed , Rabbits , Tetradecanoylphorbol Acetate/pharmacology , Transfection
19.
J Biol Chem ; 278(7): 5388-98, 2003 Feb 14.
Article in English | MEDLINE | ID: mdl-12464613

ABSTRACT

12(S)-hydroxyeicosatetraenoic acid (12(S)HETE) is a bioactive metabolite of arachidonic acid synthesized by 12-lipoxygenase. The 12-lipoxygenase blocker, baicalein, prevents epidermal growth factor (EGF)-induced activation of protein kinase C (PKC) alpha and beta in lens epithelial cells, whereas supplementation with 12(S)HETE reverses this effect, suggesting that EGF and 12(S)HETE may work together to activate PKC. This study investigates the mechanism of PKCbeta activation by EGF and 12(S)HETE. 12(S)HETE alone directed translocation of PKCbeta through the C1 rather than the C2 domain, without activating phosphoinositide 3-kinase (PI3K) or MAPK signaling or increasing intracellular calcium concentration. In the presence of baicalein, EGF triggered an asymmetric phosphorylation of the EGF receptor initiating signaling through PI3K and MAPK, but not PLCgamma. Together, 12(S)HETE and EGF synergistically increased phosphorylation of PKCbeta in the activation loop and C terminus as well as PKCbeta-specific activity. PI3K inhibitors blocked phosphorylation, but MEK1 inhibitors did not. Microvesicles containing phosphatidylinositol 3,4,5-trisphosphate mimicked the action of EGF on PKCbeta activity in the presence of 12(S)HETE. Kinase-inactive PKCbeta mutations in either activation loop or C terminus were effectively translocated by 12(S)HETE, as was PKCbeta in the presence of chelerythrine or Gö-6983. These findings indicate that unphosphorylated PKCbeta is translocated to the membrane by 12(S)HETE and phosphorylated by EGF-dependent PI3K signaling, to generate catalytically competent PKCbeta.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Epidermal Growth Factor/pharmacology , Epithelial Cells/enzymology , Protein Kinase C/metabolism , Signal Transduction/drug effects , Animals , Cell Line , Drug Synergism , Enzyme Activation/drug effects , Lens, Crystalline/cytology , Lens, Crystalline/enzymology , Rabbits
20.
Transgenic Res ; 11(4): 397-410, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12212842

ABSTRACT

Transgenic mouse technology has been very valuable for the study of lens fiber cells since they can not be propagated in cell culture. The targeting of transgenes to the lens has traditionally been done with the alphaA-crystallin promoter. However, while lens-specific, transgenic lines made with the alphaA-crystallin promoter express the transgene at levels 100-300-fold lower than endogenous alphaA-crystallin. Here we propose an alternative, the chicken betaB1-crystallin promoter (-432/+30). Transgenic mice made with this promoter have successfully expressed CAT, d/n m-calpain, Weel, and betaB2-crystallin mRNA at levels comparable to the endogenous betaB1-crystallin gene and no eye abnormalities such as cataracts, have resulted. All of the transgenic lines made with the chicken betaB1-crystallin promoter have expressed the transgene in the lens fiber cells, and the best lines express at levels close to endogenous betaB1-crystallin. While RNA expression is very high, only moderate protein expression has been achieved, implying that the high protein expression of the crystallins is partially controlled at the level of translation. Thus, the chicken betaB1-crystallin promoter directs high level RNA expression to lens fiber cells, which may be especially useful for the expression of ribozyme and anti-sense RNAs in addition to ectopic proteins.


Subject(s)
Crystallins/genetics , Lens, Crystalline/physiology , Promoter Regions, Genetic , Animals , Base Sequence , Chickens , Consensus Sequence , DNA Primers , Electrophoresis, Gel, Two-Dimensional , Humans , Lens, Crystalline/cytology , Mice , Mice, Transgenic , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Nucleic Acid , beta-Crystallin B Chain
SELECTION OF CITATIONS
SEARCH DETAIL
...