Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell Tissue Res ; 380(3): 469-486, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31989253

RESUMEN

Macrophages promote vasculogenesis during retinal neovascularization (RNV) by increasing the recruitment and differentiation of bone marrow-derived cells (BMCs). Different subtypes of macrophages (M1 and M2 macrophages) are associated with RNV. However, the mechanism underlying the regulation of BMCs by different macrophage subtypes during RNV remains unclear. In the present study, we investigated the role and mechanism of action of different macrophage subtypes that regulate BMCs during the development of RNV. The retinal avascular area and neovascularization (NV) tuft area in M2 macrophage group in vivo were the largest compared to those in the control phosphate buffer saline (PBS), unpolarized-M0, and M1 macrophage groups. The number of recruited green fluorescent protein (GFP)-positive BMCs and the degree of differentiation of BMCs into CD31-positive endothelial cells (ECs) and alpha-smooth muscle actin (α-SMA)-positive smooth muscle cells (SMCs) were higher in the M2 macrophage group than in the other groups. M2-conditional medium (M2-CM) affected the in vitro migration and activation of bone marrow mesenchymal stem cells (BMSCs, a subset of BMCs) more than M1-CM. The expression of stromal cell-derived factor-1 (SDF-1) and vascular endothelial growth factor (VEGF) in M2 macrophages and BMSCs cultured with M2-CM was also higher than that in M1 macrophages and BMSCs cultured with M1-CM. Migration of BMSCs was reduced after inhibiting the SDF-1 signaling pathway. Our results indicate that M2 macrophages may express significantly higher levels of SDF-1 and VEGF than M1 macrophages, thus regulating the recruitment and differentiation of BMCs and further aggravating vasculogenesis during RNV.


Asunto(s)
Quimiocina CXCL12/metabolismo , Macrófagos/citología , Células Madre Mesenquimatosas/citología , Neovascularización Retiniana/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Ratones , Ratones Endogámicos C57BL
2.
Exp Eye Res ; 190: 107887, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31801685

RESUMEN

Our previous work reported that minocycline induced inhibition of microglial activation aggravated visual injury in an oxygen induced retinopathy animal model. We hypothesized that minocycline might have aggravated injury to the photoreceptor. Some patients who use minocycline to treat acne complain of visual impairment; however, no studies have addressed minocycline toxicity to photoreceptors. Here, we identified mechanistic effect of minocycline on photoreceptor apoptosis. The results of Cell Counting Kit-8 and Ki67 staining demonstrated that minocycline inhibited the proliferation of 661W cells, and flow cytometry and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) demonstrated that minocycline promoted cell apoptosis. Additionally, minocycline administration activated signaling associated with endoplasmic reticulum stress, the pancreatic ER kinase-like ER kinase (PERK)-eukaryotic translation initiation factor 2α (eIF2α)-CCAAT/enhancer-binding protein homologous protein (CHOP) cascade, which represented the key mechanism underlying the initiation of apoptosis. Moreover, we observed downregulated nuclear factor erythroid 2-related factor 2 (Nrf2) after administration of minocycline for 12 h (12 hours) and Nrf2 transferred from nuclear to cytoplasm after 6 h, indicating that Nrf2 in nuclear may alleviated the pro-apoptotic effect of minocycline on photoreceptor cells. Upregulating Nrf2 inhibited apoptosis in minocycline treated 661W cells. These represent the first data demonstrating minocycline toxicity to photoreceptors via its pro-apoptotic effects through the regulation of ER stress pathways.


Asunto(s)
Antibacterianos/toxicidad , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Minociclina/toxicidad , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Animales , Western Blotting , Línea Celular , Proliferación Celular/efectos de los fármacos , Electrorretinografía , Estrés del Retículo Endoplásmico/fisiología , Factor 2 Eucariótico de Iniciación/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Etiquetado Corte-Fin in Situ , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , eIF-2 Quinasa/metabolismo
3.
Exp Eye Res ; 193: 107991, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32142723

RESUMEN

Choroidal neovascularization (CNV) is an acknowledged pathogenic mechanism of various ocular diseases, and in situ cells and mobilized bone marrow-derived cells (BMCs) are thought to participate in this process. We aimed to evaluate the roles of integrin α5 in BMCs and vascular endothelial cells (VECs) in the CNV process mediated by SDF-1/CXCR4 signaling. Adult wild-type mice were engrafted with whole BMCs obtained from GFP transgenic mice and then laser injured to induce CNV. BMCs and RF/6A cells were cultured to discover the mechanism of CNV in vitro. BMCs were mobilized to CNV areas, which expressed elevated SDF-1 and CXCR4. When SDF-1 was intravitreally injected, the number of BMCs was profoundly increased. In the SDF-1-treated group, the levels of integrin α5 expressed on BMCs and VECs were significantly higher than those on the cells in the control group. SDF-1 significantly increased the expression and positive ratio of integrin α5, which was involved in the recruitment and differentiation of BMCs into BMC-derived VECs, and these effects were suppressed by the CXCR4 inhibitor AMD3100. The PI3K/AKT pathway rather than the ERK pathway mediated SDF-1/CXCR4 induction of integrin α5. Integrin α5 suppression efficiently prevented the production of TGF-ß and bFGF but not VEGF. Inhibiting the SDF-1/CXCR4-PI3K/AKT-integrin α5 axis reduced CNV severity. Integrin α5 participates in BMC recruitment and differentiation in SDF-1/CXCR4-induced CNV and inhibition of this pathway may be a new approach to inhibit CNV.


Asunto(s)
Células de la Médula Ósea/citología , Neovascularización Coroidal/genética , Regulación de la Expresión Génica , Integrina alfa5beta1/genética , Animales , Western Blotting , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Integrina alfa5beta1/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN/genética , Transducción de Señal
4.
Angiogenesis ; 21(3): 635-652, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29675549

RESUMEN

Ocular neovascularization is a comprehensive process involved in retinal vascular development and several blinding diseases such as age-related macular degeneration and retinopathy of prematurity, with vascular endothelial growth factor (VEGF) regarded as the master regulator. However, the qualified effect of anti-VEGF therapy reveals that the underlying mechanisms are still not clearly identified. To initialize angiogenesis, endothelial cells undergo a phenotype switching to generate highly migratory and invasive cells. This process shares certain similar characters observed in endothelial-mesenchymal transition (EndMT). Here, we found that SNAI1, an EndMT transcription factor, was expressed by endothelial cells in both physiological and pathological ocular neovascularization. SNAI1 overexpression triggered cell morphological change and enhanced cell motility, while loss of SNAI1 attenuated migration, invasion and sprouting. RNA sequence analysis further revealed that SNAI1 knockdown decreased the expression of genes related to cytoskeleton rearrangement and ECM remodeling. Moreover, intravitreal injection of small interfering RNA of SNAI1 suppressed new vessel formation in developing retina as well as mice model of choroidal neovascularization and oxygen-induced retinopathy. Therefore, we propose that the EndMT transcription factor SNAI1 promotes the early phase of ocular neovascularization and may provide a potential therapeutic target.


Asunto(s)
Neovascularización Patológica/fisiopatología , Retina/fisiopatología , Neovascularización Retiniana/fisiopatología , Vasos Retinianos/fisiopatología , Factores de Transcripción de la Familia Snail/metabolismo , Animales , Movimiento Celular/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Masculino , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Retina/metabolismo , Retina/patología , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Análisis de Secuencia de ARN , Factores de Transcripción de la Familia Snail/genética
5.
Biochem Biophys Res Commun ; 483(1): 488-494, 2017 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-28007595

RESUMEN

Although it has been suggested that Dll3, one of the Notch ligands, promotes the proliferation and inhibits the apoptosis of cancer cells, the role of Dll3 in cancers remains unclear. In this study, we found that in the murine Lewis lung carcinoma (LLC) cells, the level of Dll3 mRNA changed upon tumor microenvironment (TME) stimulation, namely, decreased under hypoxia or stimulated with tumor necrosis factor (TNF)-α. Dll3 was also expressed at higher level in human lung carcinoma tissues than in the para-carcinoma tissues. Overexpression of Dll3 in LLC cells promoted cell proliferation and reduced apoptosis in vitro, and enhanced tumor growth when inoculated in vivo in mice. The Dll3-mediated proliferation could be due to increased Akt phosphorylation in LLC cells, because an Akt inhibitor counteracted Dll3-induced proliferation. Moreover, Dll3 overexpression promoted PI3K/Akt signaling through inhibiting Notch signaling.


Asunto(s)
Carcinoma Pulmonar de Lewis/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Animales , Carcinoma Pulmonar de Lewis/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Hipoxia de la Célula , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares/patología , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
ScientificWorldJournal ; 2014: 483526, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24688390

RESUMEN

High soils salinity is a main factor affecting agricultural production. Studying the function of salt-tolerance-related genes is essential to enhance crop tolerance to stress. Rab7 is a small GTP-binding protein that is distributed widely among eukaryotes. Endocytic trafficking mediated by Rab7 plays an important role in animal and yeast cells, but the current understanding of Rab7 in plants is still very limited. Herein, we isolated a vesicle trafficking gene, OsRab7, from rice. Transgenic rice over-expressing OsRab7 exhibited enhanced seedling growth and increased proline content under salt-treated conditions. Moreover, an increased number of vesicles was observed in the root tip of OsRab7 transgenic rice. The OsRab7 over-expression plants showed enhanced tolerance to salt stress, suggesting that vacuolar trafficking is important for salt tolerance in plants.


Asunto(s)
Oryza/fisiología , Tolerancia a la Sal/genética , Proteínas de Unión al GTP rab/genética , Regulación de la Expresión Génica de las Plantas , Oryza/genética , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente , Prolina/metabolismo , Plantones/genética , Plantones/crecimiento & desarrollo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
7.
Adv Healthc Mater ; : e2303527, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38411334

RESUMEN

Pathological angiogenesis with subsequent disturbed microvascular remodeling is a major cause of irreversible blindness in a number of ischemic retinal diseases. The current anti-vascular endothelial growth factor therapy can effectively inhibit angiogenesis, but it also brings significant side effects. The emergence of stem cell derived extracellular vesicles provides a new underlining strategy for ischemic retinopathy. Apoptotic vesicles (apoVs) are extracted from stem cells from human exfoliated deciduous teeth (SHED). SHED-apoVs are delivered into the eyeballs of oxygen-induced retinopathy (a most common model of angiogenic retinal dieseases) mice through intravitreal injection. The retinal neovascularization and nonperfusion area, vascular structure, and density changes are observed during the neovascularization phase (P17) and vascular remodeling phase (P21), and visual function is measured. The expression of extracellular acidification rate and lactic acid testing are used to detect endothelial cells (ECs) glycolytic activity. Furthermore, lentivirus and neutralizing antibody are used to block PD1-PDL1 axis, investigating the effects of SHED-apoVs on glycolysis and angiogenic activities. This work shows that SHED-apoVs are taken up by ECs and modulate the ECs glycolysis, leading to the decrease of abnormal neovessels and vascular remodeling. Furthermore, it is found that, at the molecular level, apoVs-carried PD1 interacts with PDL1 on hypoxic ECs to regulate the angiogenic activation. SHED-apoVs inhibit pathological angiogenesis and promote vascular remodeling in ischemic retinopathy partially by modulating ECs glycolysis through PD1/PDL1 axis. This study provides a new potential strategy for the clinical treatment of pathological retinal neovascularization.

8.
Antioxidants (Basel) ; 12(11)2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-38001832

RESUMEN

The outer blood-retina barrier (oBRB), comprises tightly connected retinal pigment epithelium (RPE) cells, Bruch's membrane, and choroid blood vessels, and is essential for retinal health and normal visual function. Disruption of the RPE barrier and its dysfunction can lead to retinal disorders such as age-related macular degeneration (AMD). In the present study, we investigated the essential role of choroid endothelial cells (ECs) in the RPE barrier formation process and its dysfunction. We discovered that ECs promoted RPE barrier formation through angiocrine signaling. Through blocking or activating endothelial Notch signaling and conducting experiments in vitro and in vivo, we confirmed that endothelial Notch signaling regulated the expression of heparin-binding epidermal growth factor (HBEGF) and consequently impacted the expression and activity of matrix metalloproteinases (MMP)-9 in RPE cells. This modulation influenced the RPE extracellular matrix deposition, tight junctions and RPE barrier function. In in vivo experiments, the intravitreal administration of recombinant HBEGF (r-HBEGF) alleviated the RPE barrier disruption induced by subretinal injection (SI) or laser treatment and also rescued RPE barrier disruption in endothelial Notch-deficient mice. Our results showed that the endothelial Notch signaling drove HBEGF expression through angiocrine signaling and effectively improved RPE barrier function by regulating the MMP-9 expression in RPE cells. It suggests that the modulation of Notch signaling in the choroidal endothelium may offer a novel therapeutic strategy for retinal degenerative diseases.

9.
J Pers Med ; 13(1)2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36675807

RESUMEN

Pathological retinal neovascularization (RNV) is the main character of ischemic ocular diseases, which causes severe visual impairments. Though retinal microglia are well acknowledged to play important roles in both physiological and pathological angiogenesis, the molecular mechanisms by which microglia communicates with endothelial cells (EC) remain unknown. In this study, using single-cell RNA sequencing, we revealed that the pro-inflammatory secreted protein Spp1 was the most upregulated gene in microglia in the mouse model of oxygen-induced retinopathy (OIR). Bioinformatic analysis showed that the expression of Spp1 in microglia was respectively regulated via nuclear factor-kappa B (NF-κB) and hypoxia-inducible factor 1α (HIF-1α) pathways, which was further confirmed through in vitro assays using BV2 microglia cell line. To mimic microglia-EC communication, the bEnd.3 endothelial cell line was cultured with conditional medium (CM) from BV2. We found that adding recombinant Spp1 to bEnd.3 as well as treating with hypoxic BV2 CM significantly enhanced EC proliferation and migration, while Spp1 neutralizing blocked those CM-induced effects. Moreover, RNA sequencing of BV2 CM-treated bEnd.3 revealed a significant downregulation of Kit, one of the type III tyrosine kinase receptors that plays a critical role in cell growth and activation. We further revealed that Spp1 increased phosphorylation and expression level of Akt/mTOR signaling cascade, which might account for its pro-angiogenic effects. Finally, we showed that intravitreal injection of Spp1 neutralizing antibody attenuated pathological RNV and improved visual function. Taken together, our work suggests that Spp1 mediates microglia-EC communication in RNV via activating endothelial Kit/Akt/mTOR signaling and is a potential target to treat ischemic ocular diseases.

10.
Cell Death Dis ; 14(6): 380, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37369647

RESUMEN

Microglia were considered as immune cells in inflammation until their angiogenic role was widely understood. Although the pro-inflammatory role of microglia in retinal angiogenesis has been explored, little is known about its role in pro-angiogenesis and the microglia-endothelia interaction. Here, we report that galectin-3 (Gal3) released by activated microglia functions as a communicator between microglia and endothelia and competitively binds to Jag1, thus inhibiting the Notch signaling pathway and enhancing endothelial angiogenic metabolism to promote angiogenesis. These results suggest that Gal3 may be a novel and effective target in the treatment of retinal angiogenesis.


Asunto(s)
Microglía , Neovascularización Patológica , Galectina 3/genética , Galectina 3/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Neovascularización Patológica/metabolismo , Transducción de Señal
11.
Life (Basel) ; 12(10)2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36295111

RESUMEN

Microglia is the resident immune cell in the retina, playing the role of immune surveillance in a traditional concept. With the heated focus on the mechanisms of microglia in pathological conditions, more and more functions of microglia have been discovered. Although the regulating role of microglia has been explored in ischemic retinopathy, little is known about its mechanisms in the different stages of the pathological process. Here, we removed microglia in the oxygen-induced retinopathy model by PLX5622 and revealed that the removal of activated microglia reduced pathological angiogenesis in the early stage after ischemic insult and alleviated the over-apoptosis of photoreceptors in the vessel remodeling phase. Our results indicated that microglia might play distinguished functions in the angiogenic and remodeling stages, and that the inhibition of microglia might be a promising target in the future treatment of ischemic retinopathy.

12.
Mol Med Rep ; 23(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33760200

RESUMEN

Proliferative vitreoretinopathy (PVR) is a disease leading to the formation of contractile preretinal membranes (PRMs) and is one of the leading causes of blindness. Connective tissue growth factor (CTGF) has been identified as a possible key determinant of progressive tissue fibrosis and excessive scarring. Therefore, the present study investigated the role and mechanism of action of CTGF in PVR. Immunohistochemical staining was performed to detect the expression of CTGF, fibronectin and collagen type III in PRMs from patients with PVR. The effects and mechanisms of recombinant human CTGF and its upstream regulator, TGF­ß1, on epithelial­mesenchymal transition (EMT) and the synthesis of extracellular matrix (ECM) by retinal pigment epithelium (RPE) cells were investigated using reverse transcription­quantitative PCR, western blotting and a [3H]proline incorporation assay. The data indicated that CTGF, fibronectin and collagen type III were highly expressed in PRMs. In vitro, CTGF significantly decreased the expression of the epithelial markers ZO­1 and E­cadherin and increased that of the mesenchymal markers fibronectin, N­cadherin and α­smooth muscle actin in a concentration­dependent manner. Furthermore, the expression of the ECM protein collagen type III was upregulated by CTGF. However, the trends in expression for the above­mentioned markers were reversed after knocking down CTGF. The incorporation of [3H]proline into RPE cells was also increased by CTGF. In addition, 8­Bromoadenosine cAMP inhibited CTGF­stimulated collagen synthesis and transient transfection of RPE cells with a CTGF antisense oligonucleotide inhibited TGF­ß1­induced collagen synthesis. The phosphorylation of PI3K and AKT in RPE cells was promoted by CTGF and TGF­ß1 and the latter promoted the expression of CTGF. The results of the present study indicated that CTGF may promote EMT and ECM synthesis in PVR via the PI3K/AKT signaling pathway and suggested that targeting CTGF signaling may have a therapeutic or preventative effect on PVR.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Transición Epitelial-Mesenquimal/genética , Pigmentos Retinianos/genética , Factor de Crecimiento Transformador beta1/genética , Vitreorretinopatía Proliferativa/genética , Western Blotting , Movimiento Celular/genética , Matriz Extracelular/genética , Fibronectinas/genética , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Epitelio Pigmentado de la Retina/crecimiento & desarrollo , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal/genética , Vitreorretinopatía Proliferativa/patología
13.
Mol Ther Nucleic Acids ; 25: 554-566, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34589277

RESUMEN

After angiogenesis-activated embryonic and early postnatal vascularization, endothelial cells (ECs) in most tissues enter a quiescent state necessary for proper tissue perfusion and EC functions. Notch signaling is essential for maintaining EC quiescence, but the mechanisms of action remain elusive. Here, we show that microRNA-218 (miR-218) is a downstream effector of Notch in quiescent ECs. Notch activation upregulated, while Notch blockade downregulated, miR-218 and its host gene Slit2, likely via transactivation of the Slit2 promoter. Overexpressing miR-218 in human umbilical vein ECs (HUVECs) significantly repressed cell proliferation and sprouting in vitro. Transcriptomics showed that miR-218 overexpression attenuated the MYC proto-oncogene, bHLH transcription factor (MYC, also known as c-myc) signature. MYC overexpression rescued miR-218-mediated proliferation and sprouting defects in HUVECs. MYC was repressed by miR-218 via multiple mechanisms, including reduction of MYC mRNA, repression of MYC translation by targeting heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), and promoting MYC degradation by targeting EYA3. Inhibition of miR-218 partially reversed Notch-induced repression of HUVEC proliferation and sprouting. In vivo, intravitreal injection of miR-218 reduced retinal EC proliferation accompanied by MYC repression, attenuated pathological choroidal neovascularization, and rescued retinal EC hyper-sprouting induced by Notch blockade. In summary, miR-218 mediates the effect of Notch activation of EC quiescence via MYC and is a potential treatment for angiogenesis-related diseases.

14.
Cell Signal ; 75: 109768, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32896607

RESUMEN

Subcapsular cataracts are common phenotype of diabetic cataracts, and abnormal lens epithelial cells (LECs) under the lens capsules have been considered to involve in the pathogenesis. Our previous studies have shown that the epithelial to mesenchymal transition (EMT), which is responsible for the LECs to lose their original polarity and tight junctions, occurs in a diabetic cataract mouse model. Autophagy is known to function in the EMT process in multiple tissues. However, the relationship between autophagy and EMT process in LECs has not yet been fully demonstrated. We found that high glucose retreatment reducing expression level of E-cadherin, an epithelial marker, but increasing that of α-smooth muscle actin (α-SMA), a mesenchymal marker, by Western blot and immunoflurence staining assays, and increased the cell migration by Transwell assay in human lens epithelial cell line HLE-B3. High glucose retreatment also led to impairment of autophagy, representing by downregulation of Beclin, LC3II/LC3I, and reducing the number of autophagosomes. Activation of autophagy by rapamycin could prevent high glucose-induced EMT. In addition, the levels of p62 and Snail were increased in high glucose-treated HLE-B3 cells, and their interactions were demonstrated by co-immunoprecipitation and immunoflurence staining, but all these changes were attenuated by application of rapamycin. These findings delineated a novel autophagy-mediated mechanism, p62 might mediate Snail underlying high glucose-induced EMT in LECs, suggesting a potential therapeutic approach for diabetic cataract by regulating autophagy.


Asunto(s)
Autofagia , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Cristalino/metabolismo , Línea Celular , Células Epiteliales/patología , Glucosa/metabolismo , Humanos , Cristalino/patología , Proteínas de Unión al ARN/metabolismo
15.
Sci Rep ; 6: 28617, 2016 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-27339903

RESUMEN

Macrophages have been recognized as an important inflammatory component in choroidal neovascularization (CNV). However, it is unclear how these cells are activated and polarized, how they affect angiogenesis and what the underlining mechanisms are during CNV. Notch signaling has been implicated in macrophage activation. Previously we have shown that inducible disruption of RBP-J, the critical transcription factor of Notch signaling, in adult mice results in enhanced CNV, but it is unclear what is the role of macrophage-specific Notch signaling in the development of CNV. In the current study, by using the myeloid specific RBP-J knockout mouse model combined with the laser-induced CNV model, we show that disruption of Notch signaling in macrophages displayed attenuated CNV growth, reduced macrophage infiltration and activation, and alleviated angiogenic response after laser induction. The inhibition of CNV occurred with reduced expression of VEGF and TNF-α in infiltrating inflammatory macrophages in myeloid specific RBP-J knockout mice. These changes might result in direct inhibition of EC lumen formation, as shown in an in vitro study. Therefore, clinical intervention of Notch signaling in CNV needs to pinpoint myeloid lineage to avoid the counteractive effects of global inhibition.


Asunto(s)
Neovascularización Coroidal/metabolismo , Macrófagos/metabolismo , Células Mieloides/metabolismo , Receptores Notch/metabolismo , Animales , Movimiento Celular/fisiología , Modelos Animales de Enfermedad , Activación de Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
J Am Heart Assoc ; 5(2)2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26857067

RESUMEN

BACKGROUND: Endothelial cells (ECs) form blood vessels through angiogenesis that is regulated by coordination of vascular endothelial growth factor (VEGF), Notch, transforming growth factor ß, and other signals, but the detailed molecular mechanisms remain unclear. METHODS AND RESULTS: Small RNA sequencing initially identified miR-342-5p as a novel downstream molecule of Notch signaling in ECs. Reporter assay, quantitative reverse transcription polymerase chain reaction and Western blot analysis indicated that miR-342-5p targeted endoglin and modulated transforming growth factor ß signaling by repressing SMAD1/5 phosphorylation in ECs. Transfection of miR-342-5p inhibited EC proliferation and lumen formation and reduced angiogenesis in vitro and in vivo, as assayed by using a fibrin beads-based sprouting assay, mouse aortic ring culture, and intravitreal injection of miR-342-5p agomir in P3 pups. Moreover, miR-342-5p promoted the migration of ECs, accompanied by reduced endothelial markers and increased mesenchymal markers, indicative of increased endothelial-mesenchymal transition. Transfection of endoglin at least partially reversed endothelial-mesenchymal transition induced by miR-342-5p. The expression of miR-342-5p was upregulated by transforming growth factor ß, and inhibition of miR-342-5p attenuated the inhibitory effects of transforming growth factor ß on lumen formation and sprouting by ECs. In addition, VEGF repressed miR-342-5p expression, and transfection of miR-342-5p repressed VEGFR2 and VEGFR3 expression and VEGF-triggered Akt phosphorylation in ECs. miR-342-5p repressed angiogenesis in a laser-induced choroidal neovascularization model in mice, highlighting its clinical potential. CONCLUSIONS: miR-342-5p acts as a multifunctional angiogenic repressor mediating the effects and interaction among angiogenic pathways.


Asunto(s)
Neovascularización Coroidal/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , MicroARNs/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Receptor Notch1/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Regiones no Traducidas 3' , Animales , Sitios de Unión , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neovascularización Coroidal/genética , Neovascularización Coroidal/patología , Neovascularización Coroidal/prevención & control , Modelos Animales de Enfermedad , Endoglina/genética , Endoglina/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células HeLa , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Transgénicos , MicroARNs/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Receptor Notch1/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA