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










Publication year range
1.
Mol Biol Cell ; 35(6): ar75, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38598298

ABSTRACT

To cause vision-disrupting fibrotic secondary cataract (PCO), lens epithelial cells that survive cataract surgery must migrate to the posterior of the lens capsule and differentiate into myofibroblasts. During this process, the cells become exposed to the FGF that diffuses out of the vitreous body. In normal development, such relatively high levels of FGF induce lens epithelial cells to differentiate into lens fiber cells. It has been a mystery as to how lens cells could instead undergo a mutually exclusive cell fate, namely epithelial to myofibroblast transition, in the FGF-rich environment of the posterior capsule. We and others have reported that the ability of TGFß to induce lens cell fibrosis requires the activity of endogenous ErbBs. We show here that lens fiber-promoting levels of FGF induce desensitization of ErbB1 (EGFR) that involves its phosphorylation on threonine 669 mediated by both ERK and p38 activity. Transinhibition of ErbB1 by FGF is overcome by a time-dependent increase in ErbB1 levels induced by TGFß, the activation of which is increased after cataract surgery. Our studies provide a rationale for why TGFß upregulates ErbB1 in lens cells and further support the receptor as a therapeutic target for PCO.


Subject(s)
Cataract , Epithelial Cells , ErbB Receptors , Fibrosis , Lens, Crystalline , Transforming Growth Factor beta , Humans , Cataract/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , ErbB Receptors/metabolism , Fibroblast Growth Factors/metabolism , Lens, Crystalline/metabolism , Myofibroblasts/metabolism , Myofibroblasts/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Signal Transduction , Transforming Growth Factor beta/metabolism
2.
Mol Biol Cell ; 35(3): ar35, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38170570

ABSTRACT

Fibrosis is a major, but incompletely understood, component of many diseases. The most common vision-disrupting complication of cataract surgery involves differentiation of residual lens cells into myofibroblasts. In serum-free primary cultures of lens epithelial cells (DCDMLs), inhibitors of either ERK or of ErbB signaling prevent TGFß from upregulating both early (fibronectin) and late (αSMA) markers of myofibroblast differentiation. TGFß stimulates ERK in DCDMLs within 1.5 h. Kinase inhibitors of ErbBs, but not of several other growth factor receptors in lens cells, reduce phospho ERK to below basal levels in the absence or presence of TGFß. This effect is attributable to constitutive ErbB activity playing a major role in regulating the basal levels pERK. Additional studies support a model in which TGFß-generated reactive oxygen species serve to indirectly amplify ERK signaling downstream of tonically active ErbBs to mediate myofibroblast differentiation. ERK activity is in turn essential for expression of ErbB1 and ErbB2, major inducers of ERK signaling. By mechanistically linking TGFß, ErbB, and ERK signaling to myofibroblast differentiation, our data elucidate a new role for ErbBs in fibrosis and reveal a novel mode by which TGFß directs lens cell fate.


Subject(s)
Myofibroblasts , Signal Transduction , Humans , Epithelial Cells/metabolism , Fibrosis , Myofibroblasts/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , ErbB Receptors
3.
Invest Ophthalmol Vis Sci ; 64(10): 6, 2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37418274

ABSTRACT

Purpose: TGFß-induced epithelial-to-myofibroblast transition (EMyT) of lens cells has been linked to the most common vision-disrupting complication of cataract surgery-namely, posterior capsule opacification (PCO; secondary cataract). Although inhibitors of the ErbB family of receptor tyrosine kinases have been shown to block some PCO-associated processes in model systems, our knowledge of ErbB signaling in the lens is very limited. Here, we investigate the expression of ErbBs and their ligands in primary cultures of chick lens epithelial cells (dissociated cell-derived monolayer cultures [DCDMLs]) and how TGFß affects ErbB function. Methods: DCDMLs were analyzed by immunofluorescence microscopy and Western blotting under basal and profibrotic conditions. Results: Small-molecule ErbB kinase blockers, including the human therapeutic lapatinib, selectively inhibit TGFß-induced EMyT of DCDMLs. Lens cells constitutively express ErbB1 (EGFR), ErbB2, and ErbB4 protein on the plasma membrane and release into the medium ErbB-activating ligand. Culturing DCDMLs with TGFß increases soluble bioactive ErbB ligand and markedly alters ErbBs, reducing total and cell surface ErbB2 and ErbB4 while increasing ErbB1 expression and homodimer formation. Similar, TGFß-dependent changes in relative ErbB expression are induced when lens cells are exposed to the profibrotic substrate fibronectin. A single, 1-hour treatment with lapatinib inhibits EMyT in DCDMLs assessed 6 days later. Short-term exposure to lower doses of lapatinib is also capable of eliciting a durable response when combined with suboptimal levels of a mechanistically distinct multikinase inhibitor. Conclusions: Our findings support ErbB1 as a therapeutic target for fibrotic PCO, which could be leveraged to pharmaceutically preserve the vision of millions of patients with cataracts.


Subject(s)
Capsule Opacification , Cataract , Humans , Capsule Opacification/metabolism , Lapatinib/metabolism , Ligands , Cataract/etiology , Cataract/metabolism , Epithelial Cells/metabolism , Transforming Growth Factor beta/metabolism , Fibrosis
4.
Oncogene ; 38(8): 1200-1210, 2019 02.
Article in English | MEDLINE | ID: mdl-30242244

ABSTRACT

Certain cancers, including gastrointestinal stromal tumor (GIST) and subsets of melanoma, are caused by somatic KIT mutations that result in KIT receptor tyrosine kinase constitutive activity, which drives proliferation. The treatment of KIT-mutant GIST has been revolutionized with the advent of KIT-directed cancer therapies. KIT tyrosine kinase inhibitors (TKI) are superior to conventional chemotherapy in their ability to control advanced KIT-mutant disease. However, these therapies have a limited duration of activity due to drug-resistant secondary KIT mutations that arise (or that are selected for) during KIT TKI treatment. To overcome the problem of KIT TKI resistance, we sought to identify novel therapeutic targets in KIT-mutant GIST and melanoma cells using a human tyrosine kinome siRNA screen. From this screen, we identified lemur tyrosine kinase 3 (LMTK3) and herein describe its role as a novel KIT regulator in KIT-mutant GIST and melanoma cells. We find that LMTK3 regulated the translation rate of KIT, such that loss of LMTK3 reduced total KIT, and thus KIT downstream signaling in cancer cells. Silencing of LMTK3 decreased cell viability and increased cell death in KIT-dependent, but not KIT-independent GIST and melanoma cell lines. Notably, LMTK3 silencing reduced viability of all KIT-mutant cell lines tested, even those with drug-resistant KIT secondary mutations. Furthermore, targeting of LMTK3 with siRNA delayed KIT-dependent GIST growth in a xenograft model. Our data suggest the potential of LMTK3 as a target for treatment of patients with KIT-mutant cancer, particularly after failure of KIT TKIs.


Subject(s)
Gastrointestinal Stromal Tumors/drug therapy , Melanoma/drug therapy , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate/administration & dosage , Melanoma/genetics , Melanoma/pathology , Mice , Mutation/drug effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
5.
J Cell Sci ; 131(22)2018 11 20.
Article in English | MEDLINE | ID: mdl-30404825

ABSTRACT

Lens epithelial cells are bound to the lens extracellular matrix capsule, of which laminin is a major component. After cataract surgery, surviving lens epithelial cells are exposed to increased levels of fibronectin, and so we addressed whether fibronectin influences lens cell fate, using DCDML cells as a serum-free primary lens epithelial cell culture system. We found that culturing DCDMLs with plasma-derived fibronectin upregulated canonical TGFß signaling relative to cells plated on laminin. Fibronectin-exposed cultures also showed increased TGFß signaling-dependent differentiation into the two cell types responsible for posterior capsule opacification after cataract surgery, namely myofibroblasts and lens fiber cells. Increased TGFß activity could be identified in the conditioned medium recovered from cells grown on fibronectin. Other experiments showed that plating DCDMLs on fibronectin overcomes the need for BMP in fibroblast growth factor (FGF)-induced lens fiber cell differentiation, a requirement that is restored when endogenous TGFß signaling is inhibited. These results demonstrate how the TGFß-fibronectin axis can profoundly affect lens cell fate. This axis represents a novel target for prevention of late-onset posterior capsule opacification, a common but currently intractable complication of cataract surgery.


Subject(s)
Eye Proteins/metabolism , Fibronectins/metabolism , Lens, Crystalline/metabolism , Cell Differentiation , Humans , Signal Transduction
6.
Dev Dyn ; 247(11): 1186-1198, 2018 11.
Article in English | MEDLINE | ID: mdl-30295986

ABSTRACT

BACKGROUND: The tissue-specific transcriptional programs during normal development require tight control by distal cis-regulatory elements, such as enhancers, with specific DNA sequences recognized by transcription factors, coactivators, and chromatin remodeling enzymes. Gata3 is a sequence-specific DNA-binding transcription factor that regulates formation of multiple tissues and organs, including inner ear, lens, mammary gland, T-cells, urogenital system, and thyroid gland. In the eye, Gata3 has a highly restricted expression domain in the posterior part of the lens vesicle; however, the underlying regulatory mechanisms are unknown. RESULTS: Here we describe the identification of a novel bipartite Gata3 lens-specific enhancer located ∼18 kb upstream from its transcriptional start site. We also found that a 5-kb Gata3 promoter possesses low activity in the lens. The bipartite enhancer contains arrays of AP-1, Ets-, and Smad1/5-binding sites as well as binding sites for lens-associated DNA-binding factors. Transient transfection studies of the promoter with the bipartite enhancer showed enhanced activation by BMP4 and FGF2. CONCLUSIONS: These studies identify a novel distal enhancer of Gata3 with high activity in lens and indicate that BMP and FGF signaling can up-regulate expression of Gata3 in differentiating lens fiber cells through the identified Gata3 enhancer and promoter elements. Developmental Dynamics 247:1186-1198, 2018. © 2018 The Authors. Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Subject(s)
Enhancer Elements, Genetic , GATA3 Transcription Factor/genetics , Lens, Crystalline/embryology , Animals , Binding Sites , Bone Morphogenetic Protein 4/physiology , DNA-Binding Proteins , Fibroblast Growth Factor 2/physiology , GATA3 Transcription Factor/chemistry , GATA3 Transcription Factor/metabolism , Mice , Promoter Regions, Genetic , Transcriptional Activation
7.
Mol Biol Cell ; 28(7): 907-921, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28209733

ABSTRACT

The most common vision-disrupting complication of cataract surgery is posterior capsule opacification (PCO; secondary cataract). PCO is caused by residual lens cells undergoing one of two very different cell fates: either transdifferentiating into myofibroblasts or maturing into lens fiber cells. Although TGFß has been strongly implicated in lens cell fibrosis, the factors responsible for the latter process have not been identified. We show here for the first time that TGFß can induce purified primary lens epithelial cells within the same culture to undergo differentiation into either lens fiber cells or myofibroblasts. Marker analysis confirmed that the two cell phenotypes were mutually exclusive. Blocking the p38 kinase pathway, either with direct inhibitors of the p38 MAP kinase or a small-molecule therapeutic that also inhibits the activation of p38, prevented TGFß from inducing epithelial-myofibroblast transition and cell migration but did not prevent fiber cell differentiation. Rapamycin had the converse effect, linking MTOR signaling to induction of fiber cell differentiation by TGFß. In addition to providing novel potential therapeutic strategies for PCO, our findings extend the so-called TGFß paradox, in which TGFß can induce two disparate cell fates, to a new epithelial disease state.


Subject(s)
Lens, Crystalline/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chick Embryo , Epithelial Cells/metabolism , Epithelium/metabolism , Eye Proteins/metabolism , Humans , Myofibroblasts/metabolism , Signal Transduction
8.
J Biol Chem ; 291(8): 3947-58, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26719333

ABSTRACT

Fibroblast growth factor (FGF) signaling regulates a multitude of cellular processes, including cell proliferation, survival, migration, and differentiation. In the vertebrate lens, FGF signaling regulates fiber cell differentiation characterized by high expression of crystallin proteins. However, a direct link between FGF signaling and crystallin gene transcriptional machinery remains to be established. Previously, we have shown that the bZIP proto-oncogene c-Maf regulates expression of αA-crystallin (Cryaa) through binding to its promoter and distal enhancer, DCR1, both activated by FGF2 in cell culture. Herein, we identified and characterized a novel FGF2-responsive region in the c-Maf promoter (-272/-70, FRE). Both c-Maf and Cryaa regulatory regions contain arrays of AP-1 and Ets-binding sites. Chromatin immunoprecipitation (ChIP) assays established binding of c-Jun (an AP-1 factor) and Etv5/ERM (an Ets factor) to these regions in lens chromatin. Analysis of temporal and spatial expression of c-Jun, phospho-c-Jun, and Etv5/ERM in wild type and ERK1/2 deficient lenses supports their roles as nuclear effectors of FGF signaling in mouse embryonic lens. Collectively, these studies show that FGF signaling up-regulates expression of αA-crystallin both directly and indirectly via up-regulation of c-Maf. These molecular mechanisms are applicable for other crystallins and genes highly expressed in terminally differentiated lens fibers.


Subject(s)
Crystallins/biosynthesis , Fibroblast Growth Factor 2/metabolism , Lens, Crystalline/metabolism , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-maf/biosynthesis , Animals , Chick Embryo , Crystallins/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblast Growth Factor 2/genetics , Humans , Lens, Crystalline/cytology , MCF-7 Cells , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-maf/genetics , Response Elements/physiology , Up-Regulation/physiology
9.
Mol Biol Cell ; 26(13): 2561-72, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25947138

ABSTRACT

Fibroblast growth factors (FGFs) play a central role in two processes essential for lens transparency--fiber cell differentiation and gap junction-mediated intercellular communication (GJIC). Using serum-free primary cultures of chick lens epithelial cells (DCDMLs), we investigated how the FGF and bone morphogenetic protein (BMP) signaling pathways positively cooperate to regulate lens development and function. We found that culturing DCDMLs for 6 d with the BMP blocker noggin inhibits the canonical FGF-to-ERK pathway upstream of FRS2 activation and also prevents FGF from stimulating FRS2- and ERK-independent gene expression, indicating that BMP signaling is required at the level of FGF receptors. Other experiments revealed a second type of BMP/FGF interaction by which FGF promotes expression of BMP target genes as well as of BMP4. Together these studies reveal a novel mode of cooperation between the FGF and BMP pathways in which BMP keeps lens cells in an optimally FGF-responsive state and, reciprocally, FGF enhances BMP-mediated gene expression. This interaction provides a mechanistic explanation for why disruption of either FGF or BMP signaling in the lens leads to defects in lens development and function.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Crystallins/metabolism , Fibroblast Growth Factors/metabolism , Lens, Crystalline/metabolism , Animals , Bone Morphogenetic Proteins/pharmacology , Cattle , Cell Communication/physiology , Cell Differentiation/physiology , Cells, Cultured , Chick Embryo , Epithelial Cells/metabolism , Fibroblast Growth Factors/pharmacology , Gap Junctions/metabolism , Humans , Lens, Crystalline/growth & development , Recombinant Proteins/pharmacology , Signal Transduction
10.
J Membr Biol ; 245(7): 357-68, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22797938

ABSTRACT

A major limitation in lens gap junction research has been the lack of experimentally tractable ex vivo systems to study the formation and regulation of fiber-type gap junctions. Although immortalized lens-derived cell lines are amenable to both gene transfection and siRNA-mediated knockdown, to our knowledge none are capable of undergoing appreciable epithelial-to-fiber differentiation. Lens central epithelial explants have the converse limitation. A key advance in the field was the development of a primary embryonic chick lens cell culture system by Drs. Sue Menko and Ross Johnson. Unlike central epithelial explants, these cultures also include cells from the peripheral (preequatorial and equatorial) epithelium, which is the most physiologically relevant population for the study of fiber-type gap junction formation. We have modified the Menko/Johnson system and refer to our cultures as dissociated cell-derived monolayer cultures (DCDMLs). We culture DCDMLs without serum to mimic the avascular lens environment and on laminin, the major matrix component of the lens capsule. Here, I review the features of the DCDML system and how we have used it to study lens gap junctions and fiber cell differentiation. Our results demonstrate the power of DCDMLs to generate new findings germane to the mammalian lens and how these cultures can be exploited to conduct experiments that would be impossible, prohibitively expensive and/or difficult to interpret using transgenic animals in vivo.


Subject(s)
Gap Junctions/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Animals , Cell Differentiation/physiology , Chick Embryo , Chickens , Connexins/metabolism
12.
Mol Biol Cell ; 21(10): 1686-97, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20357001

ABSTRACT

Gap junction-mediated intercellular communication (GJIC) is essential for the proper function of many organs, including the lens. GJIC in lens epithelial cells is increased by FGF in a concentration-dependent process that has been linked to the intralenticular gradient of GJIC required for lens transparency. Unlike FGF, elevated levels of TGF-beta are associated with lens dysfunction. We show that TGF-beta1 or -2 up-regulates dye coupling in serum-free primary cultures of chick lens epithelial cells (dissociated cell-derived monolayer cultures [DCDMLs]) via a mechanism distinct from that utilized by other growth factors. Remarkably, the ability of TGF-beta and of FGF to up-regulate GJIC is abolished if DCDMLs are simultaneously exposed to both factors despite undiminished cell-cell contact. This reduction in dye coupling is attributable to an inhibition of gap junction assembly. Connexin 45.6, 43, and 56-containing gap junctions are restored, and intercellular dye coupling is increased, if the activity of p38 kinase is blocked. Our data reveal a new type of cross-talk between the FGF and TGF-beta pathways, as well as a novel role for TGF-beta and p38 kinase in the regulation of GJIC. They also provide an explanation for how pathologically increased TGF-beta signaling could contribute to cataract formation.


Subject(s)
Gap Junctions/metabolism , Lens, Crystalline/cytology , Transforming Growth Factor beta/pharmacology , Animals , Cell Communication/drug effects , Cell Communication/physiology , Chick Embryo , Connexins , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Epithelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/physiology , Gap Junctions/physiology , Lens, Crystalline/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation/drug effects
13.
Mol Biol Cell ; 20(9): 2451-63, 2009 May.
Article in English | MEDLINE | ID: mdl-19297523

ABSTRACT

For all previously well-characterized oligomeric integral membrane proteins, folding, multisubunit assembly, and recognition of conformationally immature molecules for degradation occurs at their organelle of synthesis. This cannot, however, be the case for the gap junction-forming protein connexin43 (Cx43), which when endogenously expressed undergoes multisubunit assembly into connexons only after its transport to the trans-Golgi network. We have developed two novel assays to assess Cx43 folding and assembly: acquisition of resistance of disulfide bonds to reduction by extracellularly added DTT and Triton X-114 detergent phase partitioning. We show that Cx43 synthesized at physiologically relevant levels undergoes a multistep conformational maturation process in which folding of connexin monomers within the ER is a prerequisite for multisubunit assembly in the TGN. Similar results were obtained with Cx32, disproving the widely reported contention that the site of endogenous beta connexin assembly is the ER. Exogenous overexpression of Cx43, Cx32, or Cx26 allows these events to take place within the ER, the first example of the TGN and ER as alternative sites for oligomeric assembly. Our findings also constitute the first biochemical evidence that defective connexin folding is a cause of the human disorder X-linked Charcot-Marie-Tooth disease.


Subject(s)
Connexins/metabolism , Endoplasmic Reticulum/metabolism , Protein Subunits/metabolism , Animals , Cell Line, Tumor , Charcot-Marie-Tooth Disease/metabolism , Connexin 26 , Connexin 43/chemistry , Connexin 43/metabolism , Connexins/chemistry , Disulfides/metabolism , Humans , Intracellular Space , Mice , Mutant Proteins/metabolism , Protein Conformation , Protein Folding , Rats , Gap Junction beta-1 Protein
14.
Exp Eye Res ; 88(5): 919-27, 2009 May.
Article in English | MEDLINE | ID: mdl-19103198

ABSTRACT

The cells of the lens are joined by an extensive network of gap junction intercellular channels consisting of connexins 43, 46, and 50. We have proposed, and experimentally supported, the hypothesis that fibroblast growth factor (FGF) signaling is required for upregulation of gap junction-mediated intercellular coupling (GJIC) at the lens equator. The ability of FGF to increase GJIC in cultured lens cells requires sustained activation of extracellular signal-regulated kinase (ERK). In other cell types, activation of ERK has been shown to block GJIC mediated by connexin43 (Cx43). Why ERK signaling does not block lens cell coupling is not known. Another unresolved issue in lens gap junction regulation is how connexins, synthesized before the loss of biosynthetic organelles in mature lens fiber cells, avoid degradation during formation of the organelle-free zone. We have addressed these questions using serum-free cultures (termed DCDMLs) of primary embryonic chick lens epithelial cells. We show that FGF stimulates ERK in DCDMLs via the canonical Ras/Raf1 pathway, and that the reason that neither basal nor growth factor-stimulated GJIC is blocked by activation of ERK is because it is not mediated by Cx43. In fibroblastic cells, the normally rapid rate of degradation of Cx43 after its transport to the plasma membrane is reduced by treatments that either directly (ALLN; epoxomicin) or indirectly (generation of oxidatively un/mis-folded proteins by arsenic compounds) prevent the ubiquitin/proteasome system (UPS) from acting on its normal substrates. We show here that Cx45.6 and Cx56, the chick orthologs of mammalian Cx50 and Cx46, behave similarly in DCDMLs. When organelles lyse during the maturation of fiber cells, they release into the cytosol a large amount of new proteins that have the potential to saturate the capacity, and/or compromise the function, of the UPS. This would serve to spare gap junctions from degradation during formation of the organelle-free zone, thereby preserving GJIC between mature fiber cells despite the lack of de novo connexin synthesis.


Subject(s)
Gap Junctions/physiology , Lens, Crystalline/cytology , Up-Regulation/physiology , Animals , Cell Communication/physiology , Cells, Cultured , Chick Embryo , Connexin 43/physiology , Culture Media, Serum-Free , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/physiology , Lens, Crystalline/metabolism , Phosphorylation/physiology , Plasmids , Proteasome Endopeptidase Complex/physiology , Transfection , Ubiquitin/physiology
15.
Dev Biol ; 324(2): 202-12, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18848538

ABSTRACT

It is widely accepted that vitreous humor-derived FGFs are required for the differentiation of anterior lens epithelial cells into crystallin-rich fibers. We show that BMP2, 4, and 7 can induce the expression of markers of fiber differentiation in primary lens cell cultures to an extent equivalent to FGF or medium conditioned by intact vitreous bodies (VBCM). Abolishing BMP2/4/7 signaling with noggin inhibited VBCM from upregulating fiber marker expression. Remarkably, noggin and anti-BMP antibodies also prevented purified FGF (but not unrelated stimuli) from upregulating the same fiber-specific proteins. This effect is attributable to inhibition of BMPs produced by the lens cells themselves. Although BMP signaling is required for FGF to enhance fiber differentiation, the converse is not true. Expression of noggin in the lenses of transgenic mice resulted in a postnatal block of epithelial-to-secondary fiber differentiation, with extension of the epithelial monolayer to the posterior pole of the organ. These results reveal the central importance of BMP in secondary fiber formation and show that although FGF may be necessary for this process, it is not sufficient. Differentiation of fiber cells, and thus proper vision, is dependent on cross-talk between the FGF and BMP signaling pathways.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Cell Differentiation , Fibroblast Growth Factors/metabolism , Lens, Crystalline/embryology , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins/genetics , Cells, Cultured , Chick Embryo , Culture Media, Conditioned , Epithelial Cells/metabolism , Eye Proteins/metabolism , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental , Intermediate Filament Proteins/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/drug effects , Mice , Mice, Transgenic , Vitreous Body/metabolism , delta-Crystallins/metabolism
16.
Mol Biol Cell ; 19(6): 2631-41, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18400943

ABSTRACT

Homeostasis in the lens is dependent on an extensive network of cell-to-cell gap junctional channels. Gap junction-mediated intercellular coupling (GJIC) is higher in the equatorial region of the lens than at either pole, an asymmetry believed essential for lens transparency. Primary cultures of embryonic chick lens epithelial cells up-regulate GJIC in response to purified fibroblast growth factor (FGF)1/2 or to medium conditioned by vitreous bodies, the major reservoir of factors (including FGF) for the lens equator. We show that purified bone morphogenetic protein (BMP)2, -4, and -7 also up-regulate GJIC in these cultures. BMP2, -4, or both are present in vitreous body conditioned medium, and BMP4 and -7 are endogenously expressed by lens cells. Remarkably, lens-derived BMP signaling is required for up-regulation of GJIC by purified FGF, and sufficient for up-regulation by vitreous humor. This is the first demonstration of an obligatory interaction between FGF and BMPs in postplacode lens cells, and of a role for FGF/BMP cross-talk in regulating GJIC in any cell type. Our results support a model in which the angular gradient in GJIC in the lens, and thus proper lens function, is dependent on signaling between the FGF and BMP pathways.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Communication/drug effects , Fibroblast Growth Factor 2/pharmacology , Gap Junctions/drug effects , Gap Junctions/metabolism , Lens, Crystalline/cytology , Animals , Bone Morphogenetic Protein 7/metabolism , Carrier Proteins/metabolism , Culture Media, Conditioned , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lens, Crystalline/enzymology , Signal Transduction/drug effects , Up-Regulation/drug effects , Vitreous Body/enzymology
17.
Mol Biol Cell ; 18(11): 4279-91, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17699585

ABSTRACT

ER-associated, ubiquitin-proteasome system (UPS)-mediated degradation of the wild-type (WT) gap junction protein connexin32 (Cx32) is inhibited by mild forms of cytosolic stress at a step before its dislocation into the cytosol. We show that the same conditions (a 30-min, 42 degrees C heat shock or oxidative stress induced by arsenite) also reduce the endoplasmic reticulum (ER)-associated turnover of disease-causing mutants of Cx32 and the cystic fibrosis transmembrane conductance regulator (CFTR), as well as that of WT CFTR and unassembled Ig light chain. Stress-stabilized WT Cx32 and CFTR, but not the mutant/unassembled proteins examined, could traverse the secretory pathway. Heat shock also slowed the otherwise rapid UPS-mediated turnover of the cytosolic proteins myoD and GFPu, but not the degradation of an ubiquitination-independent construct (GFP-ODC) closely related to the latter. Analysis of mutant Cx32 from cells exposed to proteasome inhibitors and/or cytosolic stress indicated that stress reduces degradation at the level of substrate polyubiquitination. These findings reveal a new link between the cytosolic stress-induced heat shock response, ER-associated degradation, and polyubiquitination. Stress-denatured proteins may titer a limiting component of the ubiquitination machinery away from pre-existing UPS substrates, thereby sparing the latter from degradation.


Subject(s)
Cytosol/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , CHO Cells , Connexins/genetics , Connexins/metabolism , Cricetinae , Cricetulus , Endoplasmic Reticulum/metabolism , Glutamic Acid/genetics , Glutamic Acid/metabolism , Hot Temperature , Mutation/genetics , Protein Transport , Substrate Specificity
18.
Mol Biol Cell ; 16(11): 5247-57, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16135529

ABSTRACT

The protein constituents of gap junctions, connexins, have a rapid basal rate of degradation even after transport to the cell surface. We have used cell surface biotinylation to label gap junction-unassembled plasma membrane pools of connexin43 (Cx43) and show that their degradation is inhibited by mild hyperthermia, oxidative stress, and proteasome inhibitors. Cytosolic stress does not perturb endocytosis of biotinylated Cx43, but instead it seems to interfere with its targeting and/or transport to the lysosome, possibly by increasing the level of unfolded protein in the cytosol. This allows more Cx43 molecules to recycle to the cell surface, where they are assembled into long-lived, functional gap junctions in otherwise gap junction assembly-inefficient cells. Cytosolic stress also slowed degradation of biotinylated Cx43 in gap junction assembly-efficient normal rat kidney fibroblasts, and reduced the rate at which gap junctions disappeared from cell interfaces under conditions that blocked transport of nascent connexin molecules to the plasma membrane. These data demonstrate that degradation from the cell surface can be down-regulated by physiologically relevant forms of stress. For connexins, this may serve to enhance or preserve gap junction-mediated intercellular communication even under conditions in which protein synthesis and/or intracellular transport are compromised.


Subject(s)
Connexin 43/metabolism , Gap Junctions/physiology , Animals , Biotinylation , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Cricetinae , Cricetulus , Cytosol/physiology , Endocytosis , Endoplasmic Reticulum/metabolism , Hot Temperature , Lysosomes/metabolism , Lysosomes/physiology , Mice , Oxidative Stress/physiology , Proteasome Endopeptidase Complex/physiology , Protein Folding , Rats , Transfection , Up-Regulation
19.
Cell Commun Adhes ; 10(4-6): 329-33, 2003.
Article in English | MEDLINE | ID: mdl-14681037

ABSTRACT

Little is known about the mechanism and regulation of connexin turnover from the plasma membrane. We have used a combination of cell surface biotinylation, immunofluorescence microscopy, and scrape-load dye transfer assays to investigate the effect of the protein synthesis inhibitor cycloheximide on connexin43 and connexin32 after their transport to the plasmalemma. The results obtained demonstrate that cycloheximide inhibits the turnover of connexins from the surface of both gap junction assembly-deficient and -efficient cells. Moreover, cell surface connexin saved from destruction by cycloheximide can assemble into long-lived, functional gap junctional plaques. These findings support the concept that downregulation of connexin degradation from the plasma membrane can serve as a mechanism to enhance gap junction-mediated intercellular communication.


Subject(s)
Cell Membrane/metabolism , Connexin 43/metabolism , Connexins/metabolism , Cycloheximide/pharmacology , Animals , Biological Transport/drug effects , CHO Cells , Cell Membrane/drug effects , Cricetinae , Cricetulus , Gap Junctions/metabolism , Microscopy, Fluorescence , Protein Synthesis Inhibitors/pharmacology , Gap Junction beta-1 Protein
20.
J Cell Biol ; 157(3): 381-94, 2002 Apr 29.
Article in English | MEDLINE | ID: mdl-11980915

ABSTRACT

Akey step in ER-associated degradation (ERAD) is dislocation of the substrate protein from the ER into the cytosol to gain access to the proteasome. Very little is known about how this process is regulated, especially in the case of polytopic proteins. Using pulse-chase analysis combined with subcellular fractionation, we show that connexins, the four transmembrane structural components of gap junctions, can be chased in an intact form from the ER membrane into the cytosol of proteasome inhibitor-treated cells. Dislocation of endogenously expressed connexin from the ER was reduced 50-80% when the cytosolic heat shock response was induced by mild oxidative or thermal stress, but not by treatments that instead upregulate the ER unfolded protein response. Cytosolic but not ER stresses slowed the normally rapid degradation of connexins, and led to a striking increase in gap junction formation and function in otherwise assembly-inefficient cell types. These treatments also inhibited the dislocation and turnover of a connexin-unrelated ERAD substrate, unassembled major histocompatibility complex class I heavy chain. Our findings demonstrate that dislocation is negatively regulated by physiologically relevant, nonlethal stress. They also reveal a previously unrecognized relationship between cytosolic stress and intercellular communication.


Subject(s)
Connexins/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Proteasome Endopeptidase Complex , Animals , CHO Cells , Cell Line , Cricetinae , Cysteine Proteinase Inhibitors/pharmacology , Gap Junctions/metabolism , Heat-Shock Response , Leupeptins/pharmacology , Oxidation-Reduction , Peptide Hydrolases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...