Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
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.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Mol Vis ; 8: 359-66, 2002 Sep 25.
Article in English | MEDLINE | ID: mdl-12355063

ABSTRACT

PURPOSE: To determine how deamidation and partial loss of the N- and C-terminal extensions alter the heat stability of betaB1-crystallin. METHODS: Human lens betaB1, a deamidated betaB1, Q204E, and alphaA-crystallins were expressed. Truncated betaB1 was generated by proteolytic removal of part of its terminal extensions. The aggregation and precipitation of these proteins due to heating was monitored by circular dichroism and light scattering. The effect of heat on the stability of both monomers and oligomers was investigated. The flexibility of the extensions in wild type and deamidated betaB1 was assessed by 1H NMR spectroscopy. RESULTS: With heat, deamidated betaB1 precipitated more readily than wild type betaB1. Similar effects were obtained for either monomers or oligomers. Flexibility of the N-terminal extension in deamidated betaB1 was significantly reduced compared to the wild type protein. Truncation of the extensions further increased the rate of heat-induced precipitation of deamidated betaB1. The presence of the molecular chaperone, alphaA-crystallin, prevented precipitation of modified betaB1s. More alphaA was needed to chaperone the truncated and deamidated betaB1 than deamidated betaB1 or truncated betaB1. CONCLUSIONS: Deamidation and truncation of betaB1 led to destabilization of the protein and decreased stability to heat. Decreased stability of lens crystallins may contribute to their insolubilization and cataract formation.


Subject(s)
Hot Temperature , Molecular Chaperones/chemistry , beta-Crystallin B Chain/chemistry , Circular Dichroism , Deamination , Humans , Light , Magnetic Resonance Spectroscopy , Molecular Chaperones/metabolism , Protein Denaturation , Scattering, Radiation , alpha-Crystallin A Chain/metabolism , beta-Crystallin B Chain/metabolism
12.
J Bone Miner Res ; 17(1): 39-47, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11771668

ABSTRACT

Mice were generated by pronuclear injection of a type II collagen transgene harboring an Arg789Cys (R789C) mutation that has been found in patients with spondyloepiphyseal dysplasia (SED). Expression was directed to cartilage by the murine Col2a1 promoter to examine the consequences of mutations involving the Y-position of the collagen helix Gly-X-Y triplet on skeletogenesis. The transgenic mice had very short limbs, short trunk, short snout, and cleft palate; they died at birth. Their growth plates were disorganized and collagen fibrils were sparse in cartilage matrix. When the transgene was expressed in RCS cells, there was no evidence that R789C-bearing collagen chains were incorporated into stable collagen molecules. Molecular modeling of the mutation raised the possibility that it destabilizes the collagen triple helix. Together our results suggest that Y-position mutations, such as R789C, can act in a dominant negative manner to destabilize collagen molecules during assembly, reducing their availability to form fibrils, the deficiency of which profoundly disturbs the template functions of cartilage during skeletogenesis.


Subject(s)
Collagen Type II/genetics , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Point Mutation , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Bone Development/genetics , Cartilage/abnormalities , Collagen Type II/biosynthesis , Collagen Type II/chemistry , Disease Models, Animal , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/metabolism , Exostoses, Multiple Hereditary/pathology , Growth Plate/abnormalities , Humans , Mice , Mice, Transgenic , Models, Molecular , Osteochondrodysplasias/metabolism , Phenotype , Protein Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...