Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Immun Ageing ; 20(1): 53, 2023 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-37838654

RESUMO

BACKGROUND: Increased age is a risk factor for the development and progression of retinal diseases including age-related macular degeneration (AMD). Understanding the changes that occur in the eye due to aging is important in enhancing our understanding of AMD pathogenesis and the development of novel AMD therapies. Microglia, the resident brain and retinal immune cells are associated with both maintaining homeostasis and protection of neurons and loss of microglia homeostasis could be a significant player in age related neurodegeneration. One important characteristic of retinal aging is the migration of microglia from the inner to outer retina where they reside in the subretinal space (SRS) in contact with the retinal pigment epithelial (RPE) cells. The role of aged subretinal microglia is unknown. Here, we depleted microglia in aged C57/BL6 mice fed for 6 weeks with a chow containing PLX5622, a small molecule inhibitor of colony-stimulating factor-1 receptor (Csf1r) required for microglial survival. RESULTS: The subretinal P2RY12 + microglia in aged mice displayed a highly amoeboid and activated morphology and were filled with autofluorescence droplets reminiscent of lipofuscin. TEM indicates that subretinal microglia actively phagocytize shed photoreceptor outer segments, one of the main functions of retinal pigmented epithelial cells. PLX5622 treatment depleted up to 90% of the retinal microglia and was associated with significant loss in visual function. Mice on the microglia depletion diet showed reduced contrast sensitivity and significantly lower electroretinogram for the c-wave, a measurement of RPE functionality, compared to age-matched controls. The loss of c-wave coincided with a loss of RPE cells and increased RPE swelling in the absence of microglia. CONCLUSIONS: We conclude that microglia preserve visual function in aged mice and support RPE cell function, by phagocytosing shed photoreceptor outer segments and lipids, therefore compensating for the known age-related decline of RPE phagocytosis.

4.
Metabolites ; 13(2)2023 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-36837806

RESUMO

Redox homeostasis is a delicate balancing act of maintaining appropriate levels of antioxidant defense mechanisms and reactive oxidizing oxygen and nitrogen species. Any disruption of this balance leads to oxidative stress, which is a key pathogenic factor in several ocular diseases. In this review, we present the current evidence for oxidative stress and mitochondrial dysfunction in conditions affecting both the anterior segment (e.g., dry eye disease, keratoconus, cataract) and posterior segment (age-related macular degeneration, proliferative vitreoretinopathy, diabetic retinopathy, glaucoma) of the human eye. We posit that further development of therapeutic interventions to promote pro-regenerative responses and maintenance of the redox balance may delay or prevent the progression of these major ocular pathologies. Continued efforts in this field will not only yield a better understanding of the molecular mechanisms underlying the pathogenesis of ocular diseases but also enable the identification of novel druggable redox targets and antioxidant therapies.

5.
Metabolites ; 13(2)2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36837832

RESUMO

Epithelial-mesenchymal transition (EMT) is a dedifferentiation program in which polarized, differentiated epithelial cells lose their cell-cell adhesions and transform into matrix-producing mesenchymal cells. EMT of retinal pigment epithelial (RPE) cells plays a crucial role in many retinal diseases, including age-related macular degeneration, proliferative vitreoretinopathy, and diabetic retinopathy. This dynamic process requires complex metabolic reprogramming to accommodate the demands of this dramatic cellular transformation. Both transforming growth factor-beta 2 (TGFß2) and tumor necrosis factor-alpha (TNFα) have the capacity to induce EMT in RPE cells; however, little is known about their impact on the RPE metabolome. Untargeted metabolomics using high-resolution mass spectrometry was performed to reveal the metabolomic signatures of cellular and secreted metabolites of primary human fetal RPE cells treated with either TGFß2 or TNFα for 5 days. A total of 638 metabolites were detected in both samples; 188 were annotated as primary metabolites. Metabolomics profiling showed distinct metabolomic signatures associated with TGFß2 and TNFα treatment. Enrichment pathway network analysis revealed alterations in the pentose phosphate pathway, galactose metabolism, nucleotide and pyrimidine metabolism, purine metabolism, and arginine and proline metabolism in TNFα-treated cells compared to untreated control cells, whereas TGFß2 treatment induced perturbations in fatty acid biosynthesis metabolism, the linoleic acid pathway, and the Notch signaling pathway. These results provide a broad metabolic understanding of the bioenergetic rewiring processes governing TGFß2- and TNFα-dependent induction of EMT. Elucidating the contributions of TGFß2 and TNFα and their mechanistic differences in promoting EMT of RPE will enable the identification of novel biomarkers for diagnosis, management, and tailored drug development for retinal fibrotic diseases.

6.
J Vis Exp ; (192)2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36804905

RESUMO

Metabolic dysfunction of retinal pigment epithelial cells (RPE) is a key pathogenic driver of retinal diseases such as age-related macular degeneration (AMD) and proliferative vitreoretinopathy (PVR). Since RPE are highly metabolically-active cells, alterations in their metabolic status reflect changes in their health and function. High-resolution respirometry allows for real-time kinetic analysis of the two major bioenergetic pathways, glycolysis and mitochondrial oxidative phosphorylation (OXPHOS), through quantification of the extracellular acidification rate (ECAR) and oxygen consumption rate (OCR), respectively. The following is an optimized protocol for conducting high-resolution respirometry on primary human retinal pigment epithelial cells (H-RPE). This protocol provides a detailed description of the steps involved in producing bioenergetic profiles of RPE to define their basal and maximal OXPHOS and glycolytic capacities. Exposing H-RPE to different drug injections targeting the mitochondrial and glycolytic machinery results in defined bioenergetic profiles, from which key metabolic parameters can be calculated. This protocol highlights the enhanced response of BAM15 as an uncoupling agent compared to carbonyl cyanide p-trifluoromethoxyphenylhydrazone (FCCP) to induce the maximal respiration capacity in RPE. This protocol can be utilized to study the bioenergetic status of RPE under different disease conditions and test the efficacy of novel drugs in restoring the basal metabolic status of RPE.


Assuntos
Metabolismo Energético , Glicólise , Humanos , Cinética , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/metabolismo , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/farmacologia , Células Epiteliais/metabolismo , Pigmentos da Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo
7.
Front Mol Neurosci ; 15: 896786, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35813071

RESUMO

The retinal pigment epithelium (RPE) acts as a metabolic gatekeeper between photoreceptors and the choroidal vasculature to maintain retinal function. RPE dysfunction is a key feature of age-related macular degeneration (AMD), the leading cause of blindness in developed countries. Inflammation is a key pathogenic mechanism in AMD and tumor necrosis factor-alpha (TNFα) has been implicated as a pro-inflammatory cytokine involved in AMD. While mitochondrial dysfunction has been implicated in AMD pathogenesis, the interplay between inflammation and cellular metabolism remains elusive. The present study explores how the pro-inflammatory cytokine, TNFα, impacts mitochondrial morphology and metabolic function in RPE. Matured human primary RPE (H-RPE) were treated with TNFα (10 ng/ml) for up to 5 days. TNFα-induced upregulation of IL-6 secretion and inflammatory genes (IL-6, IL-8, MCP-1) was accompanied by increased oxidative phosphorylation (OXPHOS) and reduced glycolysis, leading to an increase in cellular adenosine triphosphate (ATP) content. Transmission electron microscopy (TEM) revealed defects in mitochondrial morphology with engorged mitochondria and loss of cristae integrity following TNFα treatment. Pre-treatment with the anti-inflammatory drug, 80 µM dimethyl fumarate (DMFu), blocked TNFα-induced inflammatory activation of RPE (IL-6, IL-8, MCP-1, CFH, CFB, C3) and normalized their bioenergetic profile to control levels by regulating PFKFB3 and PKM2 gene expression. Furthermore, DMFu prevented TNFα-induced mitochondrial dysfunction and morphological anomalies. Thus, our results indicate that DMFu serves as a novel therapeutic avenue for combating inflammatory activation and metabolic dysfunction of RPE in AMD.

8.
Cells ; 10(10)2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34685584

RESUMO

Bone morphogenetic proteins (BMPs) are a diverse class of growth factors that belong to the transforming growth factor-beta (TGFß) superfamily. Although originally discovered to possess osteogenic properties, BMPs have since been identified as critical regulators of many biological processes, including cell-fate determination, cell proliferation, differentiation and morphogenesis, throughout the body. In the ocular lens, BMPs are important in orchestrating fundamental developmental processes such as induction of lens morphogenesis, and specialized differentiation of its fiber cells. Moreover, BMPs have been reported to facilitate regeneration of the lens, as well as abrogate pathological processes such as TGFß-induced epithelial-mesenchymal transition (EMT) and apoptosis. In this review, we summarize recent insights in this topic and discuss the complexities of BMP-signaling including the role of individual BMP ligands, receptors, extracellular antagonists and cross-talk between canonical and non-canonical BMP-signaling cascades in the lens. By understanding the molecular mechanisms underlying BMP activity, we can advance their potential therapeutic role in cataract prevention and lens regeneration.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Cristalino/metabolismo , Cristalino/patologia , Proteínas Morfogenéticas Ósseas/metabolismo , Catarata/metabolismo , Proliferação de Células/fisiologia , Humanos , Fator de Crescimento Transformador beta/metabolismo
9.
Exp Eye Res ; 210: 108709, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34339681

RESUMO

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.


Assuntos
Envelhecimento/fisiologia , Biomarcadores/metabolismo , Catarata/metabolismo , Cristalino/metabolismo , Animais , Apoptose , Senescência Celular , Homeostase , Humanos , Oxirredução , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
10.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946753

RESUMO

PGC-1α, a key orchestrator of mitochondrial metabolism, plays a crucial role in governing the energetically demanding needs of retinal pigment epithelial cells (RPE). We previously showed that silencing PGC-1α induced RPE to undergo an epithelial-mesenchymal-transition (EMT). Here, we show that induction of EMT in RPE using transforming growth factor-beta 2 (TGFß2) suppressed PGC-1α expression. Correspondingly, TGFß2 induced defects in mitochondrial network integrity with increased sphericity and fragmentation. TGFß2 reduced expression of genes regulating mitochondrial dynamics, reduced citrate synthase activity and intracellular ATP content. High-resolution respirometry showed that TGFß2 reduced mitochondrial OXPHOS levels consistent with reduced expression of NDUFB5. The reduced mitochondrial respiration was associated with a compensatory increase in glycolytic reserve, glucose uptake and gene expression of glycolytic enzymes (PFKFB3, PKM2, LDHA). Treatment with ZLN005, a selective small molecule activator of PGC-1α, blocked TGFß2-induced upregulation of mesenchymal genes (αSMA, Snai1, CTGF, COL1A1) and TGFß2-induced migration using the scratch wound assay. Our data show that EMT is accompanied by mitochondrial dysfunction and a metabolic shift towards reduced OXPHOS and increased glycolysis that may be driven by PGC-1α suppression. ZLN005 effectively blocks EMT in RPE and thus serves as a novel therapeutic avenue for treatment of subretinal fibrosis.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/antagonistas & inibidores , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Fator de Crescimento Transformador beta2/farmacologia , Benzimidazóis/farmacologia , Linhagem Celular , Células Cultivadas , Metabolismo Energético/efeitos dos fármacos , Transição Epitelial-Mesenquimal/fisiologia , Fibrose , Expressão Gênica/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Glicólise/genética , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Epitélio Pigmentado da Retina/citologia
11.
Exp Eye Res ; 206: 108546, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33773977

RESUMO

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.


Assuntos
Proteína Morfogenética Óssea 4/agonistas , Catarata/tratamento farmacológico , Cristalino/efeitos dos fármacos , Animais , Proteína Morfogenética Óssea 4/metabolismo , Catarata/metabolismo , Catarata/patologia , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Cristalino/metabolismo , Cristalino/patologia , Masculino , Ratos , Ratos Wistar , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
12.
Int J Mol Sci ; 21(12)2020 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-32560057

RESUMO

Epithelial-mesenchymal transition (EMT) and endothelial-mesenchymal transition (EndMT) are physiological processes required for normal embryogenesis. However, these processes can be hijacked in pathological conditions to facilitate tissue fibrosis and cancer metastasis. In the eye, EMT and EndMT play key roles in the pathogenesis of subretinal fibrosis, the end-stage of age-related macular degeneration (AMD) that leads to profound and permanent vision loss. Predominant in subretinal fibrotic lesions are matrix-producing mesenchymal cells believed to originate from the retinal pigment epithelium (RPE) and/or choroidal endothelial cells (CECs) through EMT and EndMT, respectively. Recent evidence suggests that EMT of RPE may also be implicated during the early stages of AMD. Transforming growth factor-beta (TGFß) is a key cytokine orchestrating both EMT and EndMT. Investigations in the molecular mechanisms underpinning EMT and EndMT in AMD have implicated a myriad of contributing factors including signaling pathways, extracellular matrix remodelling, oxidative stress, inflammation, autophagy, metabolism and mitochondrial dysfunction. Questions arise as to differences in the mesenchymal cells derived from these two processes and their distinct mechanistic contributions to the pathogenesis of AMD. Detailed discussion on the AMD microenvironment highlights the synergistic interactions between RPE and CECs that may augment the EMT and EndMT processes in vivo. Understanding the differential regulatory networks of EMT and EndMT and their contributions to both the dry and wet forms of AMD can aid the development of therapeutic strategies targeting both RPE and CECs to potentially reverse the aberrant cellular transdifferentiation processes, regenerate the retina and thus restore vision.


Assuntos
Células Endoteliais/patologia , Degeneração Macular/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Humanos , Degeneração Macular/patologia , Estresse Oxidativo , Epitélio Pigmentado da Retina/metabolismo
13.
Sci Rep ; 9(1): 8079, 2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31147562

RESUMO

We previously demonstrated that inhibition of epidermal growth factor receptor (EGFR) slowed corneal epithelial migration. Here we examine the effect of EGF on transforming growth factor-beta receptor II (TGF-ßRII) in a corneal wound-healing model and primary human corneal epithelial cells (pHCE). Corneal debridement wounds were made and allowed to heal ± Tyrphostin AG1478 (EGFR inhibitor), and assayed for EGFR activation and EGFR and TGF-ßRII localization. Primary HCE were treated with EGF ± U0126 (MEK inhibitor) and assayed for TGF-ßRII expression. EGFR activation was maximal 15 minutes after wounding and localized in the migrating epithelial cells. TGF-ßRII localization was also observed in the migrating epithelium and was reduced when EGFR was blocked. When pHCE were treated with EGF for 6 hours, the cells produced enhanced levels of TGF-ßRII, which was blocked by U0126. Downstream signaling pathways of MEK (p38MAPK and ERK1/2MAPK) were then examined, and TGF-ß1 and EGF were found to have differential effects on the phosphorylation of p38 and ERK1/2, with TGF-ß1 upregulating p-p38 but not pERK1/2 and EGF upregulating pERK1/2 but not p-p38. Taken together, these data indicate that EGF stimulates TGF-ßRII through ERK1/2 and EGFR signaling, suggesting interplay between EGF- and TGF-ß-signaling pathways during corneal wound repair.


Assuntos
Lesões da Córnea/patologia , Fator de Crescimento Epidérmico/metabolismo , Epitélio Corneano/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Cicatrização/fisiologia , Animais , Butadienos/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Epitélio Corneano/citologia , Epitélio Corneano/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Nitrilas/farmacologia , Cultura Primária de Células , Quinazolinas/farmacologia , Ratos , Fator de Crescimento Transformador beta1/metabolismo , Tirfostinas/farmacologia , Cicatrização/efeitos dos fármacos
14.
Exp Eye Res ; 185: 107693, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31201806

RESUMO

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.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Cristalino/citologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta2/farmacologia , Actinas/metabolismo , Animais , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Sinergismo Farmacológico , Eletroforese em Gel de Poliacrilamida , Células Epiteliais/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Sistema de Sinalização das MAP Quinases/fisiologia , Fosforilação , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Tropomiosina/metabolismo
15.
Life Sci Alliance ; 2(3)2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31101737

RESUMO

The retinal pigment epithelium (RPE) supports visual processing and photoreceptor homeostasis via energetically demanding cellular functions. Here, we describe the consequences of repressing peroxisome proliferator-activated receptor γ coactivator-1 α (PGC-1α), a master regulator of mitochondrial function and biogenesis, on RPE epithelial integrity. The sustained silencing of PGC-1α in differentiating human RPE cells affected mitochondria/autophagy function, redox state, and impaired energy sensor activity ultimately inducing epithelial to mesenchymal transition (EMT). Adult conditional knockout of PGC-1 coactivators in mice resulted in rapid RPE dysfunction and transdifferentiation associated with severe photoreceptor degeneration. RPE anomalies were characteristic of autophagic defect and mesenchymal transition comparable with the ones observed in age-related macular degeneration. These findings demonstrate that PGC-1α is required to maintain the functional and phenotypic status of RPE by supporting the cells' oxidative metabolism and autophagy-mediated repression of EMT.

17.
Exp Eye Res ; 178: 108-121, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30290164

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Cristalino/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta2/farmacologia , Actinas/metabolismo , Animais , Western Blotting , Movimento Celular , Eletroforese em Gel de Poliacrilamida , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Fosforilação , Quinazolinas/farmacologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , beta Catenina/metabolismo
18.
Curr Eye Res ; 43(8): 986-997, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29652528

RESUMO

PURPOSE: This study aims to highlight some of the genes that are differentially regulated by ERK1/2 signaling in TGFß-induced EMT in lens, and their potential contribution to this pathological process. MATERIALS AND METHODS: Rat lens epithelial explants were cultured with or without TGFß over a 3-day-culture period to induce EMT, in the presence or absence of UO126 (ERK1/2 signaling inhibitor), both prior to TGFß-treatment, or 24 or 48 hours after TGFß treatment. Smad2/3-nuclear immunolabeling was used to indicate active TGFß signaling, and quantitative RT-PCR was used to analyze changes in the different treatment groups in expression of the following representative genes: TGFß signaling (Smad7, Smurf1, and Rnf111), epithelial markers (Pax6, Cdh1, Zeb1, and Zeb2), cell survival/death regulators (Bcl2, Bax, and Bad) and lens mesenchymal markers (Mmp9, Fn1, and Col1a1), over the 3 days of culture. RESULTS: ERK1/2 was found to regulate the expression of Smurf1, Smad7, Rnf11, Cdh1, Pax6, Zeb1, Bcl2, Bax, and Bad genes in lens cells. TGFß signaling was evident by nuclear localization of Smad2/3 and this was effectively blocked by pre-treatment with UO126, but not by post-treatment with this ERK1/2 signaling inhibitor. TGFß induced the expression of its signaling partners (Smad7, Smurf1, and Rnf111), as well as lens mesenchymal genes (Mmp9, Fn1, and Col1a1), consistent with its role in inducing an EMT. These TGFß-responsive signaling genes, as well as the mesenchymal markers, were all positively regulated by ERK1/2-activity. The expression levels of the lens epithelial genes we examined, and genes that were associated with cell death/survival, were not directly impacted by TGFß. CONCLUSIONS: TGFß-mediated ERK1/2 signaling positively modulates the expression of mesenchymal genes in lens epithelial explants undergoing EMT, in addition to regulating TGFß-mediated regulatory genes. Independent of TGFß, ERK1/2 activity can also regulate the expression of endogenous lens epithelial genes, highlighting its potential key role in regulation of both normal and pathological lens cellular processes.


Assuntos
Catarata/genética , Transição Epitelial-Mesenquimal/genética , Expressão Gênica/efeitos dos fármacos , Cristalino/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fator de Crescimento Transformador beta2/farmacologia , Animais , Butadienos/farmacologia , Catarata/metabolismo , Catarata/patologia , Células Cultivadas , Modelos Animais de Doenças , Cristalino/patologia , Nitrilas/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
19.
Prog Retin Eye Res ; 60: 44-65, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28807717

RESUMO

Wound healing is one of the most complex biological processes to occur in life. Repair of tissue following injury involves dynamic interactions between multiple cell types, growth factors, inflammatory mediators and components of the extracellular matrix (ECM). Aberrant and uncontrolled wound healing leads to a non-functional mass of fibrotic tissue. In the eye, fibrotic disease disrupts the normally transparent ocular tissues resulting in irreversible loss of vision. A common feature in fibrotic eye disease is the transdifferentiation of cells into myofibroblasts that can occur through a process known as epithelial-mesenchymal transition (EMT). Myofibroblasts rapidly produce excessive amounts of ECM and exert tractional forces across the ECM, resulting in the distortion of tissue architecture. Transforming growth factor-beta (TGFß) plays a major role in myofibroblast transdifferentiation and has been implicated in numerous fibrotic eye diseases including corneal opacification, pterygium, anterior subcapsular cataract, posterior capsular opacification, proliferative vitreoretinopathy, fibrovascular membrane formation associated with proliferative diabetic retinopathy, submacular fibrosis, glaucoma and orbital fibrosis. This review serves to introduce the pathological functions of the myofibroblast in fibrotic eye disease. We also highlight recent developments in elucidating the multiple signaling pathways involved in fibrogenesis that may be exploited in the development of novel anti-fibrotic therapies to reduce ocular morbidity due to scarring.


Assuntos
Transdiferenciação Celular/fisiologia , Oftalmopatias/fisiopatologia , Fibrose/fisiopatologia , Miofibroblastos/fisiologia , Cicatrização/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Humanos , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
20.
J Neuromuscul Dis ; 4(2): 159-164, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28582869

RESUMO

BACKGROUND: The L25 mouse line was generated by random genomic insertion of a lens-specific transgene. Inbreeding of L25 hemizygotes revealed an unanticipated spastic phenotype in the hind limbs. OBJECTIVE: The goals were to characterize the motor phenotype in the L25 mice and to map the transgene insert site within the mouse genome. METHODS: Six pairs of L25+/- mice were repeatedly mated. Beginning at weaning, all progeny were inspected for body weight and motor signs twice weekly until they displayed predefined ethical criteria for termination. The transgene insert site was determined by whole genome sequencing. Western blotting was used to compare the expression levels of beta-IV spectrin protein in the brain. RESULTS: Matings of hemizygous L25+/- × L25+/- mice yielded 20% (29/148) affected weanlings, identified by an abnormal retraction of the hind limbs when lifted by the tail, and a fine tremor. Affected mice were less mobile and grew more slowly than wild-type littermates. All affected mice required termination due to >15% loss of body weight (50% survival age 92 days). At the endpoint, mice showed varying degrees of spastic paresis or spastic paralysis localised to the hind limbs. Motor endplates remained fully innervated. Genome sequencing confirmed that the transgene was inserted in the locus of ßIV spectrin of L25 mice. Western blotting indicated that this random insertion had greatly reduced the expression of ßIV spectrin protein in the affected L25 mice. CONCLUSIONS: The results confirm the importance of ßIV spectrin for maintaining central motor pathway control of the hind limbs, and provide a developmental time course for the phenotype.


Assuntos
Espasticidade Muscular/metabolismo , Mutagênese Insercional , Espectrina/metabolismo , Animais , Peso Corporal/fisiologia , Encéfalo/metabolismo , Feminino , Expressão Gênica , Membro Posterior , Masculino , Camundongos Transgênicos , Placa Motora/metabolismo , Placa Motora/patologia , Espasticidade Muscular/patologia , Paresia/metabolismo , Paresia/patologia , Fenótipo , Espectrina/genética , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA