Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci Res ; 100(2): 638-652, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34822722

RESUMO

Glaucoma is one of the leading causes of irreversible blindness and can result from abnormalities in anterior segment structures required for aqueous humor outflow, including the trabecular meshwork (TM) and Schlemm's canal (SC). Transcription factors such as AP-2ß play critical roles in anterior segment development. Here, we show that the Mgp-Cre knock-in (Mgp-Cre.KI) mouse can be used to target the embryonic periocular mesenchyme giving rise to the TM and SC. Fate mapping of male and female mice indicates that AP-2ß loss causes a decrease in iridocorneal angle cells derived from Mgp-Cre.KI-expressing populations compared to controls. Moreover, histological analyses revealed peripheral iridocorneal adhesions in AP-2ß mutants that were accompanied by a decrease in expression of TM and SC markers, as observed using immunohistochemistry. In addition, rebound tonometry showed significantly higher intraocular pressure (IOP) that was correlated with a progressive significant loss of retinal ganglion cells, reduced retinal thickness, and reduced retinal function, as measured using an electroretinogram, in AP-2ß mutants compared with controls, reflecting pathology described in late-stage glaucoma patients. Importantly, elevated IOP in AP-2ß mutants was significantly reduced by treatment with latanoprost, a prostaglandin analog that increases unconventional outflow. These findings demonstrate that AP-2ß is critical for TM and SC development, and that these mutant mice can serve as a model for understanding and treating progressive human primary angle-closure glaucoma.


Assuntos
Glaucoma , Malha Trabecular , Fator de Transcrição AP-2 , Animais , Humor Aquoso/metabolismo , Feminino , Glaucoma/genética , Glaucoma/metabolismo , Humanos , Pressão Intraocular , Masculino , Camundongos , Malha Trabecular/metabolismo , Malha Trabecular/patologia , Fator de Transcrição AP-2/genética
2.
Int J Mol Sci ; 22(16)2021 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-34445433

RESUMO

The cornea is an anterior eye structure specialized for vision. The corneal endothelium and stroma are derived from the periocular mesenchyme (POM), which originates from neural crest cells (NCCs), while the stratified corneal epithelium develops from the surface ectoderm. Activating protein-2ß (AP-2ß) is highly expressed in the POM and important for anterior segment development. Using a mouse model in which AP-2ß is conditionally deleted in the NCCs (AP-2ß NCC KO), we investigated resulting corneal epithelial abnormalities. Through PAS and IHC staining, we observed structural and phenotypic changes to the epithelium associated with AP-2ß deletion. In addition to failure of the mutant epithelium to stratify, we also observed that Keratin-12, a marker of the differentiated epithelium, was absent, and Keratin-15, a limbal and conjunctival marker, was expanded across the central epithelium. Transcription factors PAX6 and P63 were not observed to be differentially expressed between WT and mutant. However, growth factor BMP4 was suppressed in the mutant epithelium. Given the non-NCC origin of the epithelium, we hypothesize that the abnormalities in the AP-2ß NCC KO mouse result from changes to regulatory signaling from the POM-derived stroma. Our findings suggest that stromal pathways such as Wnt/ß-Catenin signaling may regulate BMP4 expression, which influences cell fate and stratification.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Regulação para Baixo , Epitélio Corneano/anormalidades , Deleção de Genes , Fator de Transcrição AP-2/genética , Animais , Proteína Morfogenética Óssea 4/genética , Diferenciação Celular , Epitélio Corneano/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Queratina-12/metabolismo , Queratina-15/metabolismo , Masculino , Camundongos , Crista Neural/metabolismo , Fenótipo , Fator de Transcrição AP-2/metabolismo , Via de Sinalização Wnt
3.
Int J Mol Sci ; 22(21)2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34769418

RESUMO

Fibrotic cataracts have been attributed to transforming growth factor-beta (TGF-ß)-induced epithelial-to-mesenchymal transition (EMT). Using mouse knockout (KO) models, our laboratory has identified MMP9 as a crucial protein in the TGF-ß-induced EMT process. In this study, we further revealed an absence of alpha-smooth muscle actin (αSMA) and filamentous-actin (F-actin) stress fibers in MMP9KO mouse lens epithelial cell explants (LECs). Expression analysis using NanoString revealed no marked differences in αSMA (ACTA2) and beta-actin (ß-actin) (ACTB) mRNA between the lenses of TGF-ß-overexpressing (TGF-ßtg) mice and TGF-ßtg mice on a MMP9KO background. We subsequently conducted a protein array that revealed differential regulation of proteins known to be involved in actin polymerization and cell migration in TGF-ß-treated MMP9KO mouse LECs when compared to untreated controls. Immunofluorescence analyses using rat LECs and the novel MMP9-specific inhibitor, JNJ0966, revealed similar differential regulation of cortactin, FAK, LIMK1 and MLC2 as observed in the array. Finally, a reduction in the nuclear localization of MRTF-A, a master regulator of cytoskeletal remodeling during EMT, was observed in rat LECs co-treated with JNJ0966 and TGF-ß. In conclusion, MMP9 deficiency results in differential regulation of proteins involved in actin polymerization and cell migration, and this in turn prevents TGF-ß-induced EMT in the lens.


Assuntos
Actinas/metabolismo , Cristalino/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteoma/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Movimento Celular/fisiologia , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Cristalino/patologia , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Polimerização , Transcriptoma
4.
Exp Eye Res ; 201: 108231, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33039457

RESUMO

The cornea is a highly specialized transparent tissue located at the anterior most surface of the eye. It consists of three main layers, the outer stratified squamous epithelium, the inner endothelium, and the intermediate stroma. Formation of these layers during development involves a complex interaction between ectodermal-derived structures, such as the overlying head ectoderm with the periocular mesenchyme (POM), the latter of which is comprised of neural crest cells (NCC) and mesoderm-derived progenitor cells. Regulation of corneal epithelial development, including both epithelial cell fate and stratification, has been shown to depend on numerous bi-directional mesenchymal-epithelial signaling pathways. In this review we pay particular attention to the genes and signaling pathways that involve the POM.


Assuntos
Córnea/diagnóstico por imagem , Crista Neural/crescimento & desenvolvimento , Animais , Diferenciação Celular , Córnea/metabolismo , Humanos , Crista Neural/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo
5.
Exp Eye Res ; 195: 108042, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32353428

RESUMO

Previously, we have shown that Tfap2b, the gene encoding transcription factor AP-2ß, is needed for normal mouse eye development. Specifically, targeted loss of Tfap2b in neural crest cells (NCCs)1 and their derivatives, particularly the periocular mesenchyme (POM), resulted in anterior segment defects affecting the cornea and angle tissue. These defects were further associated with an increase in intraocular pressure (IOP). The present study investigates the underlying changes in embryonic and postnatal POM cell development and differentiation caused by loss of AP-2ß in the NCCs, particularly in the structures that control aqueous outflow, using Wnt1Cre+/-; Tfap2b-/lox; tdTomatolox/+ mice (AP-2ß neural crest cell knockout or AP-2ß NCC KO). Toluidine blue-stained sections and ultrathin sections stained with uranyl acetate and lead citrate were used to assess morphology and ultrastructure, respectively. Immunohistochemistry of KO and control eyes was performed at embryonic day (E) 15.5, E18.5, postnatal day (P) 1, P7 and P14 using phospho-histone H3 (PH3), α-smooth muscle actin (α-SMA), myocilin and endomucin antibodies, as well as a TUNEL assay. Conditional deletion of AP-2ß in the NCC-derived POM resulted in defects that appeared during both embryogenesis and postnatal stages. Fate mapping of the knockout cells in the mutants revealed that the POM migrated appropriately into the eye during embryogenesis. However, during postnatal stages a significant reduction in POM proliferation in the angle region was observed in the mutants compared to controls. This was accompanied by a lack of expression of appropriate trabecular meshwork and Schlemm's canal markers. This is the first study to show that AP-2ß is required for development and differentiation of the trabecular meshwork and Schlemm's canal. Together, these defects likely contributed to the elevated intraocular pressure (IOP) previously reported in the AP-2ß NCC KO mice.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Pressão Intraocular/fisiologia , RNA/genética , Malha Trabecular/crescimento & desenvolvimento , Fator de Transcrição AP-2/genética , Animais , Células Cultivadas , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Modelos Animais , RNA/metabolismo , Malha Trabecular/metabolismo , Fator de Transcrição AP-2/metabolismo
6.
J Neurosci Res ; 97(1): 7-15, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29660784

RESUMO

Development of the eye is closely associated with neural crest cell migration and specification. Eye development is extremely complex, as it requires the working of a combination of local factors, receptors, inductors, and signaling interactions between tissues such as the optic cup and periocular mesenchyme (POM). The POM is comprised of neural crest-derived mesenchymal progenitor cells that give rise to numerous important ocular structures including those tissues that form the optic cup and anterior segment of the eye. A number of genes are involved in the migration and specification of the POM such as PITX2, PITX3, FOXC1, FOXE3, PAX6, LMX1B, GPR48, TFAP2A, and TFAP2B. In this review, we will discuss the relevance of these genes in the development of the POM and how mutations and defects result in rare ocular diseases.


Assuntos
Anormalidades do Olho/genética , Oftalmopatias/genética , Crista Neural/anormalidades , Crista Neural/metabolismo , Doenças Raras/genética , Segmento Anterior do Olho/anormalidades , Oftalmopatias/patologia , Humanos , Mutação , Segmento Posterior do Olho/anormalidades , Doenças Raras/patologia , Fatores de Transcrição
7.
Int J Mol Sci ; 20(9)2019 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-31035577

RESUMO

Cataracts are the leading cause of blindness worldwide. Although surgery is a successful method to restore vision loss due to cataracts, post-surgical complications can occur, such as secondary cataracts, also known as posterior capsular opacification (PCO). PCO arises when lens epithelial cells (LEC) are left behind in the capsular bag following surgery and are induced to undergo epithelial to mesenchymal transition (EMT). Following EMT, LEC morphology and phenotype are altered leading to a loss of transparency and vision. Transforming growth factor (TGF)-ß-induced signaling through both canonical, TGF-ß/Smad, and non-canonical, ß-catenin/Wnt and Rho/ROCK/MRTF-A, pathways have been shown to be involved in lens EMT, and thus PCO. However, the interactions between these signaling pathways in the lens have not been thoroughly explored. In the current study we use rat LEC explants as an ex vivo model, to examine the interplay between three TGF-ß-mediated pathways using α-smooth muscle actin (α-SMA) as a molecular marker for EMT. We show that Smad3 inhibition via SIS3 prevents nuclear translocation of ß-catenin and MRTF-A, and α-SMA expression, suggesting a key role of Smad3 in regulation of MRTF-A and ß-catenin nuclear transport in LECs. Further, we demonstrate that inhibition of ß-catenin/CBP interaction by ICG-001 decreased the amount of phosphorylated Smad3 upon TGF-ß stimulation in addition to significantly decreasing the expression levels of TGF-ß receptors, TBRII and TBRI. Overall, our findings demonstrate interdependence between the canonical and non-canonical TGF-ß-mediated signaling pathways controlling EMT in the lens.


Assuntos
Transição Epitelial-Mesenquimal , Cristalino/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , beta Catenina/metabolismo , Animais , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Ligação Proteica , Transporte Proteico , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/genética , Fator de Crescimento Transformador beta/farmacologia , beta Catenina/genética
8.
Nephrol Dial Transplant ; 32(1): 50-61, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27190383

RESUMO

Background: For patients using peritoneal dialysis (PD), the peritoneal membrane can develop fibrosis and angiogenesis, leading to ultrafiltration failure, chronic hypervolemia and increased risk of technique failure and mortality. Matrix metalloproteinases (MMPs), and specifically the gelatinases (MMP2 and MMP9), may be involved in peritoneal membrane injury. Methods: From stable PD patients, mesothelial cells were assayed for MMP gene expression. MMP9 was overexpressed in mouse peritoneum by adenovirus, and MMP9 -/- mice were subjected to transforming growth factor ß (TGF-ß)-induced peritoneal fibrosis. Results: MMP9 mRNA expression correlated with peritoneal membrane solute transport properties. Overexpression of MMP9 in the mouse peritoneum induced submesothelial thickening and angiogenesis. MMP9 induced mesothelial cell transition to a myofibroblast phenotype measured by increased alpha smooth muscle actin and decreased E-cadherin expression. Angiogenesis was markedly reduced in MMP9 -/- mice treated with an adenovirus expressing active TGF-ß compared with wild-type mice. TGF-ß-mediated E-cadherin cleavage was MMP9 dependent, and E-cadherin cleavage led to ß-catenin-mediated signaling. A ß-catenin inhibitor blocked the angiogenic response induced by AdMMP9. Conclusions: Our data suggest that MMP9 is involved in peritoneal membrane injury possibly through cleavage of E-cadherin and induction of ß-catenin signaling. MMP9 is a potential biomarker for peritoneal membrane injury and is a therapeutic target to protect the peritoneal membrane in PD patients.


Assuntos
Caderinas/metabolismo , Soluções para Hemodiálise/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Patológica/etiologia , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/etiologia , beta Catenina/metabolismo , Animais , Transporte Biológico , Caderinas/genética , Humanos , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fibrose Peritoneal/metabolismo , Fibrose Peritoneal/patologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , beta Catenina/genética
9.
Mol Med ; 22: 713-723, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27704140

RESUMO

Transforming growth factor (TGF)-ß-induced epithelial-mesenchymal transition (EMT) leads to the formation of ocular fibrotic pathologies, such as anterior subcapsular cataract and posterior capsule opacification. Remodeling of the actin cytoskeleton, mediated by the Rho family of GTPases, plays a key role in EMT, however, how actin dynamics affect downstream markers of EMT has not been fully determined. Our previous work suggests that myocardin related transcription factor A (MRTF-A), an actin-binding protein, might be an important mediator of TGFß-induced EMT in lens epithelial cells. The aim of the current study was to determine the requirement of RhoA/ROCK signaling in mediating TGFß-induced nuclear accumulation of MRTF-A, and ultimate expression of α-smooth muscle actin (αSMA), a marker of a contractile, myofibroblast phenotype. Using rat lens epithelial explants, we demonstrate that ROCK inhibition using Y-27632 prevents TGFß-induced nuclear accumulation of MRTF-A, E-cadherin/ß-catenin complex disassembly, and αSMA expression. Using a novel inhibitor specifically targeting MRTF-A signaling, CCG-203971, we further demonstrate the requirement of MRTF-A nuclear localization and activity in the induction of αSMA expression. Overall, our findings suggest that TGFß-induced cytoskeletal reorganization through RhoA/ROCK/MRTF-A signaling is critical to EMT of lens epithelial cells.

10.
Am J Pathol ; 184(7): 2001-12, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24814605

RESUMO

Epithelial-mesenchymal transition (EMT) is associated with fibrotic diseases in the lens, such as anterior subcapsular cataract (ASC) formation. Often mediated by transforming growth factor (TGF)-ß, EMT in the lens involves the transformation of lens epithelial cells into a multilayering of myofibroblasts, which manifest as plaques beneath the lens capsule. TGF-ß-induced EMT and ASC have been associated with the up-regulation of two matrix metalloproteinases (MMPs): MMP-2 and MMP-9. The current study used MMP-2 and MMP-9 knockout (KO) mice to further determine their unique roles in TGF-ß-induced ASC formation. Adenoviral injection of active TGF-ß1 into the anterior chamber of all wild-type and MMP-2 KO mice led to the formation of distinct ASC plaques that were positive for α-smooth muscle actin, a marker of EMT. In contrast, only a small proportion of the MMP-9 KO eyes injected with adenovirus-expressing TGF-ß1 exhibited ASC plaques. Isolated lens epithelial explants from wild-type and MMP-2 KO mice that were treated with TGF-ß exhibited features indicative of EMT, whereas those from MMP-9 KO mice did not acquire a mesenchymal phenotype. MMP-9 KO mice were further bred onto a TGF-ß1 transgenic mouse line that exhibits severe ASC formation, but shows a resistance to ASC formation in the absence of MMP-9. These findings suggest that MMP-9 expression is more critical than MMP-2 in mediating TGF-ß-induced ASC formation.


Assuntos
Catarata/genética , Transição Epitelial-Mesenquimal , Cápsula do Cristalino/patologia , Metaloproteinase 9 da Matriz/genética , Fator de Crescimento Transformador beta1/farmacologia , Actinas/metabolismo , Animais , Caderinas/metabolismo , Catarata/induzido quimicamente , Metaloproteinase 2 da Matriz/genética , Camundongos Knockout , Camundongos Transgênicos
11.
Dev Dyn ; 243(10): 1298-309, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24753151

RESUMO

BACKGROUND: Transcription factors are critical in regulating lens development. The AP-2 family of transcription factors functions in differentiation, cell growth and apoptosis, and in lens and eye development. AP-2α, in particular, is important in early lens development, and when conditionally deleted at the placode stage defective separation of the lens vesicle from the surface ectoderm results. AP-2α's role during later stages of lens development is unknown. To address this, the MLR10-Cre transgene was used to delete AP-2α from the lens epithelium beginning at embryonic day (E) 10.5. RESULTS: The loss of AP-2α after lens vesicle separation resulted in morphological defects beginning at E18.5. By P4, a small highly vacuolated lens with a multilayered epithelium was evident in the MLR10-AP-2α mutants. Epithelial cells appeared elongated and expressed fiber cell specific ßB1 and γ-crystallins. Epithelial cell polarity and lens cell adhesion was disrupted and accompanied by the misexpression of ZO-1, N-Cadherin, and ß-catenin. Cell death was observed in the mutant lens epithelium between postnatal day (P) 14 and P30, and correlated with altered arrangements of cells within the epithelium. CONCLUSIONS: Our findings demonstrate that AP-2α continues to be required after lens vesicle separation to maintain a normal lens epithelial cell phenotype and overall lens integrity and to ensure correct fiber cell differentiation.


Assuntos
Cristalino/fisiologia , Fator de Transcrição AP-2/fisiologia , Animais , Catarata/genética , Adesão Celular/genética , Diferenciação Celular/genética , Polaridade Celular/genética , Embrião de Mamíferos , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Epitélio/metabolismo , Epitélio/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Cristalino/embriologia , Camundongos , Camundongos Transgênicos , Fenótipo
12.
Mol Vis ; 19: 1017-28, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23687438

RESUMO

PURPOSE: Transforming growth factor beta (TGFß) is a known inducer of epithelial to mesenchymal transition (EMT), and studies in other systems have shown that nuclear localization of the myocardin-related transcription factor (MRTF) is downstream of TGFß. In the following study, we investigated whether nuclear translocation of MRTF-A or MRTF-B is involved in TGFß-induced EMT of lens epithelial cells (LECs). We further investigated the relationship between matrix metalloproteinase-2 and -9 (MMP-2/9) and MRTF in the EMT of LECs. METHODS: Rat lens explant cultures were used as the model system. Explants were treated with TGFß, an MMP-2/9 inhibitor, or actin binding drugs and immunostained for alpha smooth muscle actin (αSMA), MRTF-A, and MRTF-B. Cytoplasmic and nuclear intensities of cells were measured using ImageJ. Production of αSMA was measured using western blot analysis and ImageJ. RESULTS: Untreated explant cells exhibited little αSMA expression, and MRTF-A and B were found to reside primarily in the cytosol. However, when stimulated with TGFß, a significantly greater number of cells exhibited nuclear expression of MRTF-A, accompanied by an increase in αSMA expression. However, MRTF-B remained in the cytoplasm following TGFß treatment. Cotreatment with an MMP-2/9 inhibitor and TGFß resulted in reduced MRTF-A nuclear localization and αSMA expression compared to cells treated with TGFß alone. CONCLUSIONS: Our results are the first to demonstrate the expression of MRTF-A in LECs and that its nuclear translocation can be stimulated by TGFß. Our data further suggest that MMP-2 and -9 are involved in the translocation of MRTF-A in LECs during TGFß-induced EMT.


Assuntos
Núcleo Celular/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Cristalino/citologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Actinas/metabolismo , Animais , Núcleo Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Wistar
13.
Mol Vis ; 19: 684-95, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23559862

RESUMO

PURPOSE: Extracellular matrix remodeling is thought to have profound effects on tissue architecture and associated function. We have shown previously that overexpression of transforming growth factor beta (TGFß), which stimulates matrix accumulation, results in altered morphology, cataract, and ocular hypertension in rodents. We have further shown that TGFß-induced cataracts can be mitigated through inhibition of the matrix metalloproteinases (MMP) MMP-2 and MMP-9. We therefore sought to determine whether loss of MMP expression also altered TGFß-induced changes in intraocular pressure (IOP). METHODS: To carry out this study, TGFß1 transgenic mice were bred onto a MMP-9 null background. IOP measurements were made at 1- to 2-, 2- to 3-, and 3- to 4-month time points using a TonoLab rebound tonometer. Histological and immunofluorescence findings were obtained at the same time points. RESULTS: Our results demonstrate that lens-specific expression of TGFß1 in mice results in altered morphology of the anterior segment and an accompanying significant increase in IOP. TGFß1 transgenic mice bred onto the MMP-9 null background exhibited a further increase in IOP. Interestingly, the MMP-9-deficient animals (without the TGFß transgene), which exhibited normal angle morphology, had increased IOP levels compared to their wild-type littermates. CONCLUSION: These results indicate that TGFß and MMP-9 likely act independently in regulating IOP. Additionally, MMP-9 plays an important role in maintaining IOP, and further investigation into the mechanisms of MMP-9 activity in the anterior angle may give clues to how extracellular matrix remodeling participates in ocular hypertension and glaucoma.


Assuntos
Pressão Intraocular/fisiologia , Metaloproteinase 9 da Matriz/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Actinas/metabolismo , Animais , Caderinas/metabolismo , Adesão Celular , Colágeno Tipo IV/metabolismo , Córnea/metabolismo , Iris/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Coloração e Rotulagem , Fatores de Tempo
14.
Dev Dyn ; 241(4): 814-29, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22411557

RESUMO

BACKGROUND: We have previously shown that the transcription factor AP-2α (Tcfap2a) is expressed in postmitotic developing amacrine cells in the mouse retina. Although retina-specific deletion of Tcfap2a did not affect retinogenesis, two other family members, AP-2ß and AP-2γ, showed expression patterns similar to AP-2α. RESULTS: Here we show that, in addition to their highly overlapping expression patterns in amacrine cells, AP-2α and AP-2ß are also co-expressed in developing horizontal cells. AP-2γ expression is restricted to amacrine cells, in a subset that is partially distinct from the AP-2α/ß-immunopositive population. To address possible redundant roles for AP-2α and AP-2ß during retinogenesis, Tcfap2a/b-deficient retinas were examined. These double mutants showed a striking loss of horizontal cells and an altered staining pattern in amacrine cells that were not detected upon deletion of either family member alone. CONCLUSIONS: These studies have uncovered critical roles for AP-2 activity in retinogenesis, delineating the overlapping expression patterns of Tcfap2a, Tcfap2b, and Tcfap2c in the neural retina, and revealing a redundant requirement for Tcfap2a and Tcfap2b in horizontal and amacrine cell development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Retina/embriologia , Fator de Transcrição AP-2/genética , Células Amácrinas/citologia , Células Amácrinas/fisiologia , Animais , Camundongos , Organogênese/fisiologia , Retina/fisiologia
15.
Biomolecules ; 13(12)2023 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-38136638

RESUMO

Fibrotic cataracts, posterior capsular opacification (PCO), and anterior subcapsular cataracts (ASC) are mainly attributed to the transforming growth factor-ß (TGFß)-induced epithelial-to-mesenchymal transition (EMT) of lens epithelial cells (LECs). Previous investigations from our laboratory have shown the novel role of non-canonical TGFß signaling in the progression of EMT in LECs. In this study, we have identified YAP as a critical signaling molecule involved in lens fibrosis. The observed increase in nuclear YAP in capsules of human ASC patients points toward the involvement of YAP in lens fibrosis. In addition, the immunohistochemical (IHC) analyses on ocular sections from mice that overexpress TGFß in the lens (TGFßtg) showed a co-expression of YAP and α-SMA in the fibrotic plaques when compared to wild-type littermate lenses, which do not. The incubation of rat lens explants with verteporfin, a YAP inhibitor, prevented a TGFß-induced fiber-like phenotype, α-SMA, and fibronectin expression, as well as delocalization of E-cadherin and ß-catenin. Finally, LECs co-incubated with TGFß and YAP inhibitor did not exhibit an induction in matrix metalloproteinase 2 compared to those LECs treated with TGFß alone. In conclusion, these data demonstrate that YAP is required for TGFß-mediated lens EMT and fibrosis.


Assuntos
Opacificação da Cápsula , Cristalino , Humanos , Ratos , Animais , Camundongos , Metaloproteinase 2 da Matriz/metabolismo , Proteínas de Sinalização YAP , Cristalino/metabolismo , Células Epiteliais/metabolismo , Opacificação da Cápsula/patologia , Fator de Crescimento Transformador beta/metabolismo , Fibrose
16.
Hum Mol Genet ; 19(9): 1791-804, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20150232

RESUMO

Appropriate development of the retina and optic nerve requires that the forebrain-derived optic neuroepithelium undergoes a precisely coordinated sequence of patterning and morphogenetic events, processes which are highly influenced by signals from adjacent tissues. Our previous work has suggested that transcription factor activating protein-2 alpha (AP-2alpha; Tcfap2a) has a non-cell autonomous role in optic cup (OC) development; however, it remained unclear how OC abnormalities in AP-2alpha knockout (KO) mice arise at the morphological and molecular level. In this study, we show that patterning and morphogenetic defects in the AP-2alpha KO optic neuroepithelium begin at the optic vesicle stage. During subsequent OC formation, ectopic neural retina and optic stalk-like tissue replaced regions of retinal pigment epithelium. AP-2alpha KO eyes also displayed coloboma in the ventral retina, and a rare phenotype in which the optic stalk completely failed to extend, causing the OCs to be drawn inward to the midline. We detected evidence of increased sonic hedgehog signaling in the AP-2alpha KO forebrain neuroepithelium, which likely contributed to multiple aspects of the ocular phenotype, including expansion of PAX2-positive optic stalk-like tissue into the OC. Our data suggest that loss of AP-2alpha in multiple tissues in the craniofacial region leads to severe OC and optic stalk abnormalities by disturbing the tissue-tissue interactions required for ocular development. In view of recent data showing that mutations in human TFAP2A result in similar eye defects, the current findings demonstrate that AP-2alpha KO mice provide a valuable model for human ocular disease.


Assuntos
Modelos Animais de Doenças , Anormalidades do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Morfogênese/genética , Nervo Óptico/embriologia , Retina/embriologia , Fator de Transcrição AP-2/genética , Animais , Primers do DNA/genética , Anormalidades do Olho/genética , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Hedgehog/metabolismo , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Morfogênese/fisiologia , Reação em Cadeia da Polimerase , Prosencéfalo/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fator de Transcrição AP-2/metabolismo
17.
Front Cell Dev Biol ; 10: 886053, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35656546

RESUMO

Injury to the ocular lens perturbs cell-cell and cell-capsule/basement membrane interactions leading to a myriad of interconnected signaling events. These events include cell-adhesion and growth factor-mediated signaling pathways that can ultimately result in the induction and progression of epithelial-mesenchymal transition (EMT) of lens epithelial cells and fibrosis. Since the lens is avascular, consisting of a single layer of epithelial cells on its anterior surface and encased in a matrix rich capsule, it is one of the most simple and desired systems to investigate injury-induced signaling pathways that contribute to EMT and fibrosis. In this review, we will discuss the role of key cell-adhesion and mechanotransduction related signaling pathways that regulate EMT and fibrosis in the lens.

18.
Curr Eye Res ; 46(10): 1509-1515, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33689532

RESUMO

Purpose: Our lab has shown that conditionally disrupting the transcription factor activating protein 2ß (Tfap2b) gene, responsible for the activating protein-2ß (AP-2ß) transcription factor, exclusively in cranial neural crest cells (AP-2ß NCC KO), leads to anterior segment dysgenesis and a closed angle phenotype. The purpose of the current study is to determine if there is a progressive loss of retinal ganglion cells (RGCs) in the mutant over time and whether this loss was associated with macroglial activity changes and elevated intraocular pressure (IOP).Methods: Using the Cre-loxP system, we generated a conditional knockout of Tfap2b exclusively in cranial NCC (AP-2ß NCC KO). Immunohistochemistry was performed using anti-Brn3a, anti-GFAP and anti-Vimentin antibodies. IOP was measured using a tonometer and the data was analyzed using GraphPad Prism software. Brn3a and DAPI positive cells were counted using Image-J and statistical analysis was performed with GraphPad Prism software.Results: Our findings revealed that while no statistical difference in Brn3a expression was observed between wild-type and mutant mice at postnatal day (P) 4 or P10, at P40 (p < .01) and P42 (p < .0001) Brn3a expression was significantly reduced in the mutant retina at the region of the ONH. There was also increased expression of glial fibrillary acidic protein (GFAP) by Müller cells in the AP-2ß NCC KO mice at P35 and P40, indicating the presence of neuroinflammation. Moreover, increased IOP was observed starting at P35 and continuing at P40 and P42 (p < .0001 for all three ages examined).Conclusions: Together, these findings suggest that the retinal damage observed in the KO mouse becomes apparent by P40 after increased IOP was observed at P35 and progressed over time. The AP-2ß NCC KO mouse may therefore be a novel experimental model for glaucoma.


Assuntos
Glaucoma/diagnóstico , Crista Neural/metabolismo , Doenças Retinianas/diagnóstico , Células Ganglionares da Retina/patologia , Fator de Transcrição AP-2/genética , Animais , Progressão da Doença , Eletroforese , Glaucoma/genética , Glaucoma/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Pressão Intraocular/fisiologia , Camundongos , Camundongos Knockout , Microglia/patologia , Reação em Cadeia da Polimerase , Doenças Retinianas/genética , Doenças Retinianas/metabolismo , Tonometria Ocular , Fator de Transcrição Brn-3A/metabolismo , Vimentina/metabolismo
19.
Dev Biol ; 326(2): 403-17, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18996109

RESUMO

The classical cadherins are known to have both adhesive and signaling functions. It has also been proposed that localized regulation of cadherin activity may be important in cell assortment during development. In the context of eye development, it has been suggested that cadherins are important for separation of the invaginated lens vesicle from the surface ectoderm. To test this hypothesis, we conditionally deleted N-cadherin or E-cadherin from the presumptive lens ectoderm of the mouse. Conditional deletion of either cadherin alone did not produce a lens vesicle separation defect. However, these conditional mutants did exhibit common structural deficits, including microphthalmia, severe iris hyperplasia, persistent vacuolization within the fibre cell region, and eventual lens epithelial cell deterioration. To assess the co-operative roles of E-cadherin and N-cadherin within the developing lens, double conditional knockout embryos were generated. These mice displayed distinct defects in lens vesicle separation and persistent expression of another classical cadherin, P-cadherin, within the cells of the persistent lens stalk. Double mutant lenses also exhibited severe defects in lens epithelial cell adhesion and survival. Finally, the severity of the lens phenotype was shown to be sensitive to the number of wild-type E- and N-cadherin alleles. These data suggest that the co-operative expression of both E- and N-cadherin during lens development is essential for normal cell sorting and subsequent lens vesicle separation.


Assuntos
Caderinas/metabolismo , Sobrevivência Celular , Células Epiteliais , Cristalino/embriologia , Cristalino/crescimento & desenvolvimento , Morfogênese , Animais , Biomarcadores/metabolismo , Caderinas/genética , Proteínas Cdh1 , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Hiperplasia/patologia , Iris/patologia , Cristalino/anormalidades , Cristalino/anatomia & histologia , Camundongos , Camundongos Knockout , Microftalmia/genética , Fenótipo
20.
Mol Cell Biol ; 27(21): 7497-510, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17724084

RESUMO

Activating protein 2alpha (AP-2alpha) is known to be expressed in the retina, and AP-2alpha-null mice exhibit defects in the developing optic cup, including patterning of the neural retina (NR) and a replacement of the dorsal retinal pigmented epithelium (RPE) with NR. In this study, we analyzed the temporal and spatial retinal expression patterns of AP-2alpha and created a conditional deletion of AP-2alpha in the developing retina. AP-2alpha exhibited a distinct expression pattern in the developing inner nuclear layer of the retina, and colocalization studies indicated that AP-2alpha was exclusively expressed in postmitotic amacrine cell populations. Targeted deletion of AP-2alpha in the developing retina did not result in observable retinal defects. Further examination of AP-2alpha-null mutants revealed that the severity of the RPE defect was variable and, although defects in retinal lamination occur at later embryonic stages, earlier stages showed normal lamination and expression of markers for amacrine and ganglion cells. Together, these data demonstrate that, whereas AP-2alpha alone does not play an intrinsic role in retinogenesis, it has non-cell-autonomous effects on optic cup development. Additional expression analyses showed that multiple AP-2 proteins are present in the developing retina, which will be important to future studies.


Assuntos
Deleção de Genes , Retina/citologia , Retina/embriologia , Fator de Transcrição AP-2/deficiência , Fator de Transcrição AP-2/metabolismo , Células Amácrinas/citologia , Animais , Calbindina 2 , Contagem de Células , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Gânglios Sensitivos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Integrases/metabolismo , Camundongos , Mitose , Especificidade de Órgãos , Organogênese , Transporte Proteico , Retina/enzimologia , Proteína G de Ligação ao Cálcio S100/metabolismo , Sintaxina 1/metabolismo , Fator de Transcrição AP-2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA