Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 151(3)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38240393

RESUMEN

The spheroidal shape of the eye lens is crucial for precise light focusing onto the retina. This shape is determined by concentrically aligned, convexly elongated lens fiber cells along the anterior and posterior axis of the lens. Upon differentiation at the lens equator, the fiber cells increase in height as their apical and basal tips migrate towards the anterior and posterior poles, respectively. The forces driving this elongation and migration remain unclear. We found that, in the mouse lens, membrane protrusions or lamellipodia are observed only in the maturing fibers undergoing cell curve conversion, indicating that lamellipodium formation is not the primary driver of earlier fiber migration. We demonstrated that elevated levels of fibroblast growth factor (FGF) suppressed the extension of Rac-dependent protrusions, suggesting changes in the activity of FGF controlling Rac activity, switching to lamellipodium-driven migration. Inhibitors of ROCK, myosin and actin reduced the height of both early and later fibers, indicating that elongation of these fibers relies on actomyosin contractility. Consistent with this, active RhoA was detected throughout these fibers. Given that FGF promotes fiber elongation, we propose that it does so through regulation of Rho activity.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Cristalino , Ratones , Animales , Cristalino/metabolismo , Epitelio/metabolismo , Actinas/metabolismo , Diferenciación Celular/fisiología
2.
Exp Eye Res ; 219: 109070, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35413282

RESUMEN

Epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) principally contributes to the pathogenesis of fibrotic cataract. Sprouty (Spry) and Spred proteins are receptor tyrosine kinase (RTK) antagonists that can regulate RTK-mediated signaling pathways, such as the MAPK/ERK1/2-signaling pathway. The present study examines the ability of Spry and Spred to inhibit TGFß-induced EMT in LECs. LECs explanted from postnatal-day-21 Wistar rats were transduced with adenoviral vectors coding for Spry1, Spry2 or Spred2, and subsequently treated with or without TGFß2. Immunofluorescent labeling of explants for the epithelial membrane marker ß-catenin, and the mesenchymal marker alpha-smooth muscle actin (α-sma), were used to characterize the progression of EMT. Western blotting was used to quantify levels of α-sma and ERK1/2-signaling. Overexpression of Spry or Spred in LECs was sufficient to suppress EMT in response to TGFß, including a block to cell elongation, ß-catenin delocalization and α-sma accumulation. Spry and Spred were also shown to significantly block ERK1/2 phosphorylation for up to 18 h of TGFß treatment but did not impair the earlier activation of ERK1/2 at 20 min. These findings suggest that Spry and Spred may not directly impact ERK1/2-signaling activated by the serine/threonine kinase TGFß receptor, but may selectively target later ERK1/2-signaling driven by downstream RTK-mediated signaling. Taken together, our data establish Spry and Spred antagonists as potent negative regulators of TGFß-induced EMT that can regulate ERK1/2-signaling in a temporal manner. A greater understanding of how Spry and Spred regulate the complex signaling interactions that underlie TGFß-induced EMT will be essential to facilitate the development of novel therapeutics for different pathologies driven by EMT, including fibrotic forms of cataract.


Asunto(s)
Catarata , Cristalino , Animales , Catarata/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Cristalino/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , beta Catenina/metabolismo
3.
Dev Biol ; 467(1-2): 1-13, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32858001

RESUMEN

Fibroblast growth factor receptor (FGFR) signaling patterns multiple tissues in both vertebrates and invertebrates, largely through the activation of intracellular kinases. Recent studies have demonstrated that the phosphatase, PTEN negatively regulates FGFR signaling, such that the loss of PTEN can compensate for reduced FGFR signaling to rescue aspects of normal development. In the developing mouse lens, FGFR signaling promotes cell survival and fiber cell differentiation, and the loss of Pten largely compensates for the loss of Fgfr2 during lens development. To explore this regulatory relationship further, we focused on the phenotypic consequences of Pten loss on lens development and fiber cell differentiation in the absence of all FGFR signaling, both in vivo and in lens epithelial explants. Pten deletion partially rescues primary fiber cell elongation and γ-crystallin accumulation in FGFR-deficient lenses in vivo but fails to rescue cell survival or proliferation. However, in lens epithelial explants, where cells survive without FGFR signaling, Pten deletion rescues vitreous humor-induced lens fiber cell differentiation in the combined absence of Fgfr1, Fgfr2 and Fgfr3. This represents the first evidence that vitreous-initiated signaling cascades, independent of FGFR signaling, can drive mammalian lens fiber cell differentiation, when freed from repression by PTEN.


Asunto(s)
Proliferación Celular , Células Epiteliales/metabolismo , Cristalino/embriología , Fosfohidrolasa PTEN/deficiencia , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Animales , Supervivencia Celular , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética
4.
Exp Eye Res ; 210: 108709, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34339681

RESUMEN

Lens homeostasis and transparency are dependent on the function and intercellular communication of its epithelia. While the lens epithelium is uniquely equipped with functional repair systems to withstand reactive oxygen species (ROS)-mediated oxidative insult, ROS are not necessarily detrimental to lens cells. Lens aging, and the onset of pathogenesis leading to cataract share an underlying theme; a progressive breakdown of oxidative stress repair systems driving a pro-oxidant shift in the intracellular environment, with cumulative ROS-induced damage to lens cell biomolecules leading to cellular dysfunction and pathology. Here we provide an overview of our current understanding of the sources and essential functions of lens ROS, antioxidative defenses, and changes in the major regulatory systems that serve to maintain the finely tuned balance of oxidative signaling vs. oxidative stress in lens cells. Age-related breakdown of these redox homeostasis systems in the lens leads to the onset of cataractogenesis. We propose eight candidate hallmarks that represent common denominators of aging and cataractogenesis in the mammalian lens: oxidative stress, altered cell signaling, loss of proteostasis, mitochondrial dysfunction, dysregulated ion homeostasis, cell senescence, genomic instability and intrinsic apoptotic cell death.


Asunto(s)
Envejecimiento/fisiología , Biomarcadores/metabolismo , Catarata/metabolismo , Cristalino/metabolismo , Animales , Apoptosis , Senescencia Celular , Homeostasis , Humanos , Oxidación-Reducción , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
5.
Exp Eye Res ; 206: 108546, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33773977

RESUMEN

Transforming growth factor beta (TGFß) and bone morphogenetic protein (BMP) signaling play opposing roles in epithelial-mesenchymal transition (EMT) of lens epithelial cells, a cellular process integral to the pathogenesis of fibrotic cataract. We previously showed that BMP-7-induced Smad1/5 signaling blocks TGFß-induced Smad2/3-signaling and EMT in rat lens epithelial cell explants. To further explore the antagonistic role of BMPs on TGFß-signaling, we tested the capability of BMP-4 or newly described BMP agonists, ventromorphins, in blocking TGFß-induced lens EMT. Primary rat lens epithelial explants were treated with exogenous TGFß2 alone, or in combination with BMP-4 or ventromorphins. Treatment with TGFß2 induced lens epithelial cells to undergo EMT and transdifferentiate into myofibroblastic cells with upregulated α-SMA and nuclear translocation of Smad2/3 immunofluorescence. BMP-4 was able to suppress this EMT without blocking TGFß2-nuclear translocation of Smad2/3. In contrast, the BMP agonists, ventromorphins, were unable to block TGFß2-induced EMT, despite a transient and early ability to significantly reduce TGFß2-induced nuclear translocation of Smad2/3. This intriguing disparity highlights new complexities in the responsiveness of the lens to differing BMP-related signaling. Further research is required to better understand the antagonistic relationship between TGFß and BMPs in lens EMT leading to cataract.


Asunto(s)
Proteína Morfogenética Ósea 4/agonistas , Catarata/tratamiento farmacológico , Cristalino/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 4/metabolismo , Catarata/metabolismo , Catarata/patología , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Cristalino/metabolismo , Cristalino/patología , Masculino , Ratas , Ratas Wistar , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
6.
Exp Eye Res ; 191: 107917, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31923414

RESUMEN

The transparent and refractive properties of the ocular lens are dependent on its precise cellular structure, supported by the regulation of lens cellular processes of proliferation and differentiation that are essential throughout life. The ERK/MAPK-signalling pathway plays a crucial role in regulating lens cell proliferation and differentiation, and in turn is regulated by inhibitory molecules including the Spred family of proteins to modulate and attenuate the impact of growth factor stimulation. Given Spreds are strongly and distinctly expressed in lens, along with their established inhibitory role in a range of different tissues, we investigated the role these antagonists play in regulating lens cell proliferation and differentiation, and their contribution to lens structure and growth. Using established mice lines deficient for either or both Spred 1 and Spred 2, we demonstrate their role in regulating lens development by negatively regulating ERK1/2 activity. Mice deficient for both Spred 1 and Spred 2 have impaired lens and eye development, displaying irregular lens epithelial and fibre cell activity as a result of increased levels of phosphorylated ERK1/2. While Spred 1 and Spred 2 do not appear to be necessary for induction and early stages of lens morphogenesis (prior to E11.5), nor for the formation of the primary fibre cells, they are required for the continuous embryonic growth and differentiation of the lens.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Ojo/embriología , Cristalino/embriología , Morfogénesis/fisiología , Proteínas Represoras/fisiología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Femenino , Técnicas de Genotipaje , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Reacción en Cadena de la Polimerasa
7.
Development ; 143(2): 318-28, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26657765

RESUMEN

Lens epithelial cells differentiate into lens fibers (LFs) in response to a fibroblast growth factor (FGF) gradient. This cell fate decision requires the transcription factor Prox1, which has been hypothesized to promote cell cycle exit in differentiating LF cells. However, we find that conditional deletion of Prox1 from mouse lenses results in a failure in LF differentiation despite maintenance of normal cell cycle exit. Instead, RNA-seq demonstrated that Prox1 functions as a global regulator of LF cell gene expression. Intriguingly, Prox1 also controls the expression of fibroblast growth factor receptors (FGFRs) and can bind to their promoters, correlating with decreased downstream signaling through MAPK and AKT in Prox1 mutant lenses. Further, culturing rat lens explants in FGF increased their expression of Prox1, and this was attenuated by the addition of inhibitors of MAPK. Together, these results describe a novel feedback loop required for lens differentiation and morphogenesis, whereby Prox1 and FGFR signaling interact to mediate LF differentiation in response to FGF.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Cristalino/citología , Cristalino/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Factores de Crecimiento de Fibroblastos/farmacología , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Receptores de Factores de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/genética
8.
Exp Eye Res ; 185: 107693, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31201806

RESUMEN

The ocular lens is exposed to numerous growth factors that influence its behavior in diverse ways. While many of these, such as FGF and EGF promote normal cell behavior, TGFß is unique in that it can also induce lens cell pathology, namely, the epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) leading to fibrotic cataract formation. The present study explores how EGF impacts on TGFß-induced EMT in the lens. LECs in explants prepared from 21-day-old Wistar rats were treated with either 200 pg/ml TGFß2, 5 ng/ml EGF, or a combination of these, with or without a 2-h pre-treatment of an EGFR inhibitor (PD153035), MEK inhibitor (U0126) or Smad3 inhibitor (SIS3). Co-treatment of LECs with TGFß2 and EGF, compared with TGFß2 alone, resulted in a more pronounced elongation and transdifferentiation of the LECs into myofibroblastic cells, with higher protein levels of mesenchymal cell markers (α-SMA and tropomyosin). Combining EGF with a less potent lower dose of TGFß2 (50 pg/ml) induced LECs to undergo EMT equivalent to treatment with a higher dose of TGFß2 (200 pg/ml) within 5 days of culture. EGF alone, nor the lower dose of TGFß2, were able to induce EMT in LECs within 5 days. Co-treatment of LECs with EGF and TGFß2 induced a temporal shift in the phosphorylation levels of Smad2/3, ERK1/2 and EGFR and changed the expression patterns of downstream EMT target genes, compared to treatment of LECs with either growth factor alone. Inhibition of EGFR-signaling with PD153035 blocked the EMT response induced by co-treatment with EGF and TGFß2. Taken together, our data demonstrate that EGF can potentiate TGFß2 activity to enhance EMT in LECs, further highlighting the importance of EGFR-signaling in cataract formation. By directly blocking EGFR signaling, the activity of both EGF and TGFß2 can be simultaneously reduced, thereby serving as a potential target for cataract prevention.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Cristalino/citología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta2/farmacología , Actinas/metabolismo , Animales , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Sinergismo Farmacológico , Electroforesis en Gel de Poliacrilamida , Células Epiteliales/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Sistema de Señalización de MAP Quinasas/fisiología , Fosforilación , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Tropomiosina/metabolismo
9.
Exp Eye Res ; 178: 108-121, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30290164

RESUMEN

Epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) plays a critical role in the pathogenesis of fibrotic cataract. Transforming growth factor-beta (TGFß) is a potent inducer of this fibrotic process in lens. Recent studies in cancer progression have shown that in addition to activating the canonical Smad signaling pathway, TGFß can also transactivate the epidermal growth factor receptor (EGFR) to enhance invasive cell migration. The present study aims to elucidate the involvement of EGFR-signaling in TGFß-induced EMT in LECs. Treatment with TGFß2 induced transdifferentiation of LECs into myofibroblastic cells, typical of an EMT. TGFß2 induced the phosphorylation of the EGFR and upregulation of Egfr and Hb-egf gene expression. Pharmacologic inhibition of EGFR-signaling using PD153035 inhibited TGFß-induced EMT, including the upregulation of mesenchymal markers and downregulation of epithelial markers. Crosstalk between TGFß2-induced EGFR and ERK1/2 was evident, with both pathways impacting on Smad2/3-signaling. Our finding that TGFß2 transactivates downstream EGFR-signaling reveals a previously unknown mechanism in the pathogenesis of cataract. Understanding the complex interplay between divergent canonical and non-canonical signaling pathways, as well as downstream target genes involved in TGFß-induced EMT, will enable the development of more effective targeted therapies in the pharmacological treatment of cataract.


Asunto(s)
Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Cristalino/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta2/farmacología , Actinas/metabolismo , Animales , Western Blotting , Movimiento Celular , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Fosforilación , Quinazolinas/farmacología , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , beta Catenina/metabolismo
10.
Exp Eye Res ; 185: 107692, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31189078

RESUMEN

Many of the small molecule-based inhibitors of NADPH oxidase activity are largely inadequate to substantiate broad claims, often exhibiting a lack of Nox-isoform-specificity, and sometimes only acting as scavengers of reactive oxygen species (ROS). In the present study, we use a newly developed highly selective Nox4 inhibitor, GLX7013114, to modulate TGFß-induced lens epithelial to mesenchymal transition (EMT). Rat lens epithelial explants were pre-treated with 0.3  µM of GLX7013114, and then treated with 200 pg/ml of TGF-ß2 to induce lens EMT. ROS production was visualized microscopically using the superoxide fluorogenic probe, dihydroethidium (DHE). The EMT process was documented using phase-contrast microscopy, and molecular EMT markers were immunolabeled. qPCR was also performed to observe changes in EMT-associated genes. TGFß-induced ROS was evident at 8 h of culture and its intensity was found to be significantly reduced when GLX7013114 was applied, comparable to ROS levels measured in untreated explants. Using phase-contrast microscopy to follow TGFß-induced EMT over 5 days in the presence of the inhibitor, lens epithelial cells in explants became myofibroblastic by day 2 and underwent progressive apoptosis to reveal a bare lens capsule by day 5. Explants treated with TGFß and GLX7013114 had some increased cell survival; however, these differences were not significant. For the first time, Nox4 inhibition by GLX7013114 was shown to reduce the TGFß-induced gene expression of α-smooth muscle actin (αSMA), collagen 1a and fibronectin. GLX7013114, given that it appears to block aspects of TGFß-induced EMT, including ROS production, may be a new useful Nox4-selective inhibitor for further studies.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Cristalino/citología , NADPH Oxidasa 4/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/farmacología , Actinas/metabolismo , Animales , Colágeno Tipo I/metabolismo , Células Epiteliales/metabolismo , Fibronectinas/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica , Microscopía de Contraste de Fase , NADPH Oxidasas/antagonistas & inhibidores , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Exp Eye Res ; 178: 160-175, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30290165

RESUMEN

Spred, like Sprouty (Spry) and also Sef proteins, have been identified as important regulators of receptor tyrosine kinase (RTK)-mediated MAPK/ERK-signaling in various developmental systems, controlling cellular processes such as proliferation, migration and differentiation. Spreds are widely expressed during early embryogenesis, and in the eye lens, become more localised in the lens epithelium with later development, overlapping with other antagonists including Spry. Given the synexpression of Spreds and Spry in lens, in order to gain a better understanding of their specific roles in regulating growth factor mediated-signaling and cell behavior, we established and characterised lines of transgenic mice overexpressing Spred1 or Spred2, specifically in the lens. This overexpression of Spreds resulted in a small lens phenotype during ocular morphogenesis, retarding its growth by compromising epithelial cell proliferation and fiber differentiation. These in situ findings were shown to be dependent on the ability of Spreds to suppress MAPK-signaling, in particular FGF-induced ERK1/2-signaling in lens cells. This was validated in vitro using lens epithelial explants, that highlighted the overlapping role of Spreds with Spry2, but not Spry1. This study provides insights into the putative function of Spreds and Spry in situ, some overlapping and some distinct, and their importance in regulating lens cell proliferation and fiber differentiation contributing to lens and eye growth.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Epiteliales/citología , Cristalino/crecimiento & desarrollo , Proteínas Represoras/fisiología , Proteínas Adaptadoras Transductoras de Señales , Adenoviridae/genética , Animales , Western Blotting , Factores de Crecimiento de Fibroblastos/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Regulación del Desarrollo de la Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Transfección
12.
Dev Dyn ; 247(3): 340-345, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28480986

RESUMEN

Epithelial-mesenchymal transition (EMT) produces myofibroblasts that contribute to the formation of fibrotic tissue with an impairment of tissue homeostasis and functionality. The crystalline lens of the eye is a unique transparent and isolated tissue. The lens vesicle becomes isolated from the surface ectoderm, its cells are all contained as they line the inner surface of the lens capsule. Clinically the formation of fibrotic tissue by the lens epithelial cells causes a type of cataract or opacification and contraction of the lens capsule postcataract surgery. Production of EMT in the intact animal lens by using specific gene transfer to the lens or experimental lens injury has been shown to be a powerful tool to investigate EMT processes. It is not easy to uncover whether the origin of the myofibroblast is epithelial cell-derived or from other cell lineages in fibrotic tissues. However, myofibroblasts that appear in the crystalline lens pathology are totally derived from the lens epithelial cells for the reasons mentioned above. Here, we report on different animal models of lens EMT, using either transgenic approaches or injury to study the biological aspects of EMT. Developmental Dynamics 247:340-345, 2018. © 2017 The Authors Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Asunto(s)
Transición Epitelial-Mesenquimal , Animales , Catarata/patología , Modelos Animales de Enfermedad , Fibrosis/patología , Cristalino/patología , Ratones , Miofibroblastos/patología
13.
Exp Eye Res ; 170: 148-159, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29501879

RESUMEN

Sprouty (Spry) and Spred proteins have been identified as closely related negative regulators of the receptor tyrosine kinase (RTK)-mediated MAPK pathway, inhibiting cellular proliferation, migration and differentiation in many systems. As the different members of this antagonist family are strongly expressed in the lens epithelium in overlapping patterns, in this study we used lens epithelial explants to examine the impact of these different antagonists on the morphologic and molecular changes associated with fibroblast growth factor (FGF)-induced lens fiber differentiation. Cells in lens epithelial explants were transfected using different approaches to overexpress the different Spry (Spry1, Spry2) and Spred (Spred1, Spred2, Spred3) members, and we compared their ability to undergo FGF-induced fiber differentiation. In cells overexpressing any of the antagonists, the propensity for FGF-induced cell elongation was significantly reduced, indicative of a block to lens fiber differentiation. Of these antagonists, Spry1 and Spred2 appeared to be the most potent among their respective family members, demonstrating the greatest block in FGF-induced fiber differentiation based on the percentage of cells that failed to elongate. Consistent with the reported activity of Spry and Spred, we show that overexpression of Spry2 was able to suppress FGF-induced ERK1/2 phosphorylation in lens cells, as well as the ERK1/2-dependent fiber-specific marker Prox1, but not the accumulation of ß-crystallins. Taken together, Spry and Spred proteins that are predominantly expressed in the lens epithelium in situ, appear to have overlapping effects on negatively regulating ERK1/2-signaling associated with FGF-induced lens epithelial cell elongation leading to fiber differentiation. This highlights the important regulatory role for these RTK antagonists in establishing and maintaining the distinct architecture and polarity of the lens.


Asunto(s)
Diferenciación Celular/fisiología , Cristalino/citología , Proteínas del Tejido Nervioso/fisiología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Represoras/fisiología , Adenoviridae/genética , Animales , Animales Recién Nacidos , Western Blotting , Electroforesis en Gel de Poliacrilamida , Factores de Crecimiento de Fibroblastos/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas Fluorescentes Verdes/metabolismo , Cristalino/metabolismo , Proteínas de la Membrana/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfogénesis , Fosfoproteínas/fisiología , Fosforilación , Ratas , Ratas Wistar , Transfección
14.
Exp Eye Res ; 170: 58-66, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29458060

RESUMEN

The development and growth of the vertebrate ocular lens is dependent on the regulated proliferation of an anterior monolayer of epithelial cells, and their subsequent differentiation into elongate fiber cells. The growth factor rich ocular media that bathes the lens mediates these cellular processes, and their respective intracellular signaling pathways are in turn regulated to ensure that the proper lens architecture is maintained. Recent studies have proposed that Cysteine Rich Motor Neuron 1 (Crim1), a transmembrane protein involved in organogenesis of many tissues, might influence cell adhesion, polarity and proliferation in the lens by regulating integrin-signaling. Here, we characterise the lens and eyes of the Crim1KST264 mutant mice, and show that the loss of Crim1 function in the ocular tissues results in inappropriate differentiation of the lens epithelium into fiber cells. Furthermore, restoration of Crim1 levels in just the lens tissue of Crim1KST264 mice is sufficient to ameliorate most of the dysgenesis observed in the mutant animals. Based on our findings, we propose that tight regulation of Crim1 activity is required for maintenance of the lens epithelium, and its depletion leads to ectopic differentiation into fiber cells, dramatically altering lens structure and ultimately leading to microphthalmia and aphakia.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/fisiología , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Cristalino/embriología , Actinas/metabolismo , Animales , Diferenciación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Desarrollo Embrionario , Epitelio/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Cristalino/citología , Cristalino/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta2/metabolismo , beta-Cristalinas/metabolismo
15.
Growth Factors ; 35(4-5): 131-143, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28969468

RESUMEN

The eye lens grows by systematic proliferation of its epithelial cells and their differentiation into fibre cells. The anterior aqueous humour regulates lens epithelial cell proliferation whereas posteriorly, the vitreous stimulates lens fibre differentiation. Vitreous-derived members of the fibroblast growth factor (FGF) family induce fibre differentiation, with added support for FGFs as putative regulators of aqueous-induced lens cell proliferation. To further characterize this, given FGFs' known affinity for proteoglycans, we compared the effect of proteoglycan sulphation in growth factor- and aqueous-induced lens cell proliferation. Disruption of proteoglycan sulphation in lens cells specifically impacted on aqueous- and FGF-induced MAPK/ERK1/2-signalling, but not on that induced by other mitogens such as PDGF; however, cell proliferation was reduced in all treatment groups, regardless of the mitogen. Overall, by disrupting proteoglycan activity, we further highlight the significant role of FGFs in aqueous-induced ERK1/2 phosphorylation leading to lens cell proliferation.


Asunto(s)
Proliferación Celular , Células Epiteliales/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Cristalino/citología , Sistema de Señalización de MAP Quinasas , Animales , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Cristalino/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteoglicanos/metabolismo , Ratas , Ratas Wistar
16.
Exp Eye Res ; 159: 98-113, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28365272

RESUMEN

Transforming Growth Factor Beta (TGFß) potently induces lens epithelial to mesenchymal transition (EMT). The resultant mesenchymal cells resemble those found in plaques of human forms of subcapsular cataract. Smad signaling has long been implicated as the sole driving force of TGFß-mediated activity. Rat lens epithelial explants were used to examine the role of the Smad-independent signaling, namely the MAPK/ERK1/2 signaling pathway, in the initiation and progression of TGFß-induced EMT. Phase contrast microscopy was used to observe the morphological changes associated with TGFß-induced EMT in this model, including cell elongation, cell membrane blebbing, cell loss as indicated by the area of bare capsule and capsular wrinkling. The levels of Smad2, Smad2/3 and ERK1/2 phosphorylation measured using western blotting confirmed that the addition of UO126 was sufficient in blocking all TGFß-induced ERK1/2 activation, as well as reducing Smad signaling at 18 h. Immunofluorescent labeling and further western blotting confirmed that TGFß-induced EMT was associated with an increase in α-smooth muscle actin (α-SMA) and a reduction of E-cadherin at cell borders. Pre-treatment with UO126 was effective at blocking the TGFß-induced EMT, as evidenced by a reduction of α-SMA expression and protein labeling, E-cadherin labeling at cell borders, and a reduction of cell loss, cell elongation and capsular wrinkling. Post-treatment with UO126 at 2 and 6 h after TGFß addition was also effective at blocking EMT while post-treatment with UO126 at 24 and 48 h was not sufficient in hampering TGFß-induced EMT. Our data implicates ERK1/2 signaling in the initiation but not the progression of TGFß-induced EMT in rat lens epithelial cells. The tight regulation of intracellular signaling pathways such as ERK1/2 are required for the maintenance of lens epithelial cell integrity and hence tissue transparency. A greater understanding of the molecular mechanisms that drive the induction and progression of EMT in the lens will provide the basis for potential therapeutics for human cataract.


Asunto(s)
Catarata/metabolismo , Transición Epitelial-Mesenquimal/genética , Cristalino/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Factor de Crecimiento Transformador beta/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Catarata/genética , Catarata/patología , Proliferación Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Cristalino/efectos de los fármacos , Cristalino/patología , Microscopía de Contraste de Fase , Fosforilación , ARN/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
17.
Optom Vis Sci ; 94(2): 270-276, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27801692

RESUMEN

PURPOSE: To report the histopathological features of anterior subcapsular cataract associated with atopic dermatitis. CASE REPORT: A 29-year-old man with atopic dermatitis presented with bilateral anterior subcapsular cataract. After routine cataract surgery, the anterior subcapsular cataractous tissue was obtained as an anterior capsulorhexis flap and prepared as a wholemount for histological analysis. The wholemount consisted of a well-demarcated central grayish-white plaque surrounded by transparent capsule, corroborating the slit-lamp biomicroscopic appearance. Higher magnification of the plaque revealed a fibrous and amorphous mass, most likely extracellular matrix owing to the presence of irregularly arranged bundled strands of fibrils, typical of collagen. Lens epithelial cells at the plaque were densely packed and myofibroblast-like and immunoreactive for alpha-smooth muscle actin. In contrast, lens epithelial cells more distant from the plaque retained their regular cuboidal arrangement and regular spacing, and were not labeled for alpha-smooth muscle actin, similar to lens epithelial cells obtained from a non-cataractous case. CONCLUSIONS: The presence of alpha-smooth muscle actin-reactive elongated cells at the plaque suggests that the cuboidal lens epithelial cells making up the anterior subcapsular cataract have transdifferentiated into spindle-shaped myofibroblastic cells that produce and deposit aberrant extracellular matrix. This transdifferentiation process, more commonly known as an epithelial-mesenchymal transition, contributes to a fibrotic response leading to the development of human anterior subcapsular cataract.


Asunto(s)
Catarata/patología , Dermatitis Atópica/complicaciones , Adulto , Catarata/etiología , Extracción de Catarata , Transdiferenciación Celular , Células Epiteliales/patología , Humanos , Cápsula del Cristalino/patología , Cristalino/patología , Masculino
18.
Dev Biol ; 406(2): 129-46, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26375880

RESUMEN

Sprouty proteins function as negative regulators of the receptor tyrosine kinase (RTK)-mediated Ras/Raf/MAPK pathway in many varied physiological and developmental processes, inhibiting growth factor-induced cellular proliferation, migration and differentiation. Like other negative regulators, Sprouty proteins are expressed in various organs during development, including the eye; ubiquitously expressed in the optic vesicle, lens pit, optic cup and lens vesicle. Given the synexpression of different antagonists (e.g, Sprouty, Sef, Spred) in the developing lens, to gain a better understanding of their specific role, in particular, their ability to regulate ocular growth factor signaling in lens cells, we characterized transgenic mice overexpressing Sprouty1 or Sprouty2 in the eye. Overexpression of Sprouty in the lens resulted in reduced lens and eye size during ocular morphogenesis, influenced by changes to the lens epithelium, aberrant fiber cell differentiation and compromised de novo maintenance of the lens capsule. Here we demonstrate an important inhibitory role for Sprouty in the regulation of lens cell proliferation and fiber differentiation in situ, potentially through its ability to modulate FGF- (and even EGF-) mediated MAPK/ERK1/2 signaling in lens cells. Whilst growth factor regulation of lens cell proliferation and fiber differentiation are required for orchestrating lens morphogenesis and growth, in turn, antagonists such as Sprouty are just as important for regulating the intracellular signaling pathways driving lens cellular processes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Cristalino/embriología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Morfogénesis/genética , Fenotipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Western Blotting , Bromodesoxiuridina , Diferenciación Celular/genética , Proliferación Celular/genética , Técnica del Anticuerpo Fluorescente , Técnicas Histológicas , Cristalino/citología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Transgénicos , Morfogénesis/fisiología
19.
Development ; 139(24): 4601-12, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23136392

RESUMEN

Most growth factor receptor tyrosine kinases (RTKs) signal through similar intracellular pathways, but they often have divergent biological effects. Therefore, elucidating the mechanism of channeling the intracellular effect of RTK stimulation to facilitate specific biological responses represents a fundamental biological challenge. Lens epithelial cells express numerous RTKs with the ability to initiate the phosphorylation (activation) of Erk1/2 and PI3-K/Akt signaling. However, only Fgfr stimulation leads to lens fiber cell differentiation in the developing mammalian embryo. Additionally, within the lens, only Fgfrs activate the signal transduction molecule Frs2α. Loss of Frs2α in the lens significantly increases apoptosis and decreases phosphorylation of both Erk1/2 and Akt. Also, Frs2α deficiency decreases the expression of several proteins characteristic of lens fiber cell differentiation, including Prox1, p57(KIP2), aquaporin 0 and ß-crystallins. Although not normally expressed in the lens, the RTK TrkC phosphorylates Frs2α in response to binding the ligand NT3. Transgenic lens epithelial cells expressing both TrkC and NT3 exhibit several features characteristic of lens fiber cells. These include elongation, increased Erk1/2 and Akt phosphorylation, and the expression of ß-crystallins. All these characteristics of NT3-TrkC transgenic lens epithelial cells depend on Frs2α. Therefore, tyrosine phosphorylation of Frs2α mediates Fgfr-dependent lens cell survival and provides a mechanistic basis for the unique fiber-differentiating capacity of Fgfs on mammalian lens epithelial cells.


Asunto(s)
Diferenciación Celular/genética , Factores de Crecimiento de Fibroblastos/fisiología , Cristalino/embriología , Proteínas de la Membrana/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Sinergismo Farmacológico , Embrión de Mamíferos , Factores de Crecimiento de Fibroblastos/farmacología , Cristalino/crecimiento & desarrollo , Cristalino/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Morfogénesis/fisiología , Ratas , Ratas Wistar , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Receptor trkC/genética , Receptor trkC/metabolismo , Receptor trkC/fisiología , Regulación hacia Arriba/genética
20.
Exp Eye Res ; 132: 9-16, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25576668

RESUMEN

An eclectic range of ocular growth factors with differing actions are present within the aqueous and vitreous humors that bathe the lens. Growth factors that exert their actions via receptor tyrosine kinases (RTKs), such as FGF, play a normal regulatory role in lens; whereas other factors, such as TGFß, can lead to an epithelial to mesenchymal transition (EMT) that underlies several forms of cataract. The respective downstream intracellular signaling pathways of these factors are in turn tightly regulated. One level of negative regulation is thought to be through RTK-antagonists, namely, Sprouty (Spry), Sef and Spred that are all expressed in the lens. In this study, we tested these different negative regulators and compared their ability to block TGFß-induced EMT in rat lens epithelial cells. Spred expression within the rodent eye was confirmed using RT-PCR, western blotting and immunofluorescence. Rat lens epithelial explants were used to examine the morphological changes associated with TGFß-induced EMT over 3 days of culture, as well as α-smooth muscle actin (α-sma) immunolabeling. Cells in lens epithelial explants were transfected with either a reporter (EGFP) vector (pLXSG), or with plasmids also coding for different RTK-antagonists (i.e. pLSXG-Spry1, pLSXG-Spry2, pLXSG-Sef, pLSXG-Spred1, pLSXG-Spred2, pLSXG-Spred3), before treating with TGFß for up to 3 days. The percentages of transfected cells that underwent TGFß-induced morphological changes consistent with an EMT were determined using cell counts and validated with a paired two-tailed t-test. Explants transfected with pLXSG demonstrated a distinct transition in cell morphology after TGFß treatment, with ∼60% of the cells undergoing fibrotic-like cell elongation. This percentage was significantly reduced in cells overexpressing the different antagonists, indicative of a block in lens EMT. Of the antagonists tested under these in vitro conditions, Spred1 was the most potent demonstrating the greatest block in TGFß-induced fibrotic cell elongation/EMT. Through the overexpression of RTK-antagonists in lens epithelial cells we have established a novel role for Spry, Spred and Sef as negative regulators of TGFß-induced EMT. Further investigations may help us develop a better understanding of the molecular mechanisms involved in maintaining the integrity of the normal lens epithelium, with these antagonists serving as putative therapeutic agents for prevention of EMT, and hence cataractogenesis.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Cristalino/efectos de los fármacos , Proteínas de la Membrana/fisiología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/farmacología , Animales , Western Blotting , Catarata/metabolismo , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/fisiología , Cristalino/fisiología , Proteínas de la Membrana/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA