Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 33
Filtrer
1.
Invest Ophthalmol Vis Sci ; 64(15): 15, 2023 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-38095908

RÉSUMÉ

Purpose: The underlying mechanism of congenital cataracts caused by deficiency or mutation of junctional adhesion molecule C (JAM-C) gene remains unclear. Our study aims to elucidate the abnormal developmental process in Jamc-/- lenses and reveal the genes related to lens development that JAM-C may regulate. Methods: Jamc knockout (Jamc-/-) mouse embryos and pups were generated for in vivo studies. Four key developmental stages from embryonic day (E) 12.5 to postnatal day (P) 0.5 were selected for the following experiments. Hematoxylin and eosin staining was used for histological analysis. The 5-bromo-2'-deoxyuridine (BrdU) incorporation assay and TUNEL staining were performed to label lens epithelial cell (LEC) proliferation and apoptosis, respectively. Immunofluorescence and Western blot were used to analyze the markers of lens epithelium, cell cycle exit, and lens fiber differentiation. Results: JAM-C was expressed throughout the process of lens development. Deletion of Jamc resulted in decreased lens size and disorganized lens fibers, which arose from E16.5 and aggravated gradually. The LECs of Jamc-/- lenses showed decreased quantity and proliferation, accompanied with reduction of key transcription factor, FOXE3. The fibers in Jamc-/- lenses were disorganized. Moreover, Jamc-deficient lens fibers showed significantly altered distribution patterns of Cx46 and Cx50. The marker of fiber homeostasis, γ-crystallin, was also decreased in the inner cortex and core fibers of Jamc-/- lenses. Conclusions: Deletion of JAM-C exhibits malfunction of LEC proliferation and fiber maturation during murine lens development, which may be related to the downregulation of FOXE3 expression and abnormal localization patterns of Cx46 and Cx50.


Sujet(s)
Molécule d'adhérence jonctionnelle C , Cristallin , Animaux , Souris , Différenciation cellulaire/physiologie , Prolifération cellulaire , Cellules épithéliales/métabolisme , Épithélium , Molécule d'adhérence jonctionnelle C/métabolisme , Cristallin/métabolisme , Souris knockout
2.
Signal Transduct Target Ther ; 8(1): 305, 2023 08 18.
Article de Anglais | MEDLINE | ID: mdl-37591843

RÉSUMÉ

Although VEGF-B was discovered as a VEGF-A homolog a long time ago, the angiogenic effect of VEGF-B remains poorly understood with limited and diverse findings from different groups. Notwithstanding, drugs that inhibit VEGF-B together with other VEGF family members are being used to treat patients with various neovascular diseases. It is therefore critical to have a better understanding of the angiogenic effect of VEGF-B and the underlying mechanisms. Using comprehensive in vitro and in vivo methods and models, we reveal here for the first time an unexpected and surprising function of VEGF-B as an endogenous inhibitor of angiogenesis by inhibiting the FGF2/FGFR1 pathway when the latter is abundantly expressed. Mechanistically, we unveil that VEGF-B binds to FGFR1, induces FGFR1/VEGFR1 complex formation, and suppresses FGF2-induced Erk activation, and inhibits FGF2-driven angiogenesis and tumor growth. Our work uncovers a previously unrecognized novel function of VEGF-B in tethering the FGF2/FGFR1 pathway. Given the anti-angiogenic nature of VEGF-B under conditions of high FGF2/FGFR1 levels, caution is warranted when modulating VEGF-B activity to treat neovascular diseases.


Sujet(s)
Facteur de croissance fibroblastique de type 2 , Facteur de croissance endothéliale vasculaire de type B , Humains , Facteur de croissance fibroblastique de type 2/génétique , Immunothérapie , Récepteur FGFR1/génétique
3.
Adv Mater ; 35(31): e2301166, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-37235720

RÉSUMÉ

Metal-halide perovskites possess great potential for electrochemical water splitting that has not been realized due to their intolerance to water. Here, methylammonium lead halide perovskites (MAPbX3 ) are used to electrocatalyze water oxidation in aqueous electrolytes by creating MAPbX3 @AlPO-5 host-guest composites. Due to the protective feature of the zeolite matrix, halide perovskite nanocrystals (NCs) confined in aluminophosphate AlPO-5 zeolites achieve an excellent stability in water. The resultant electrocatalyst undergoes dynamic surface restructuring during the oxygen evolution reaction (OER) with the formation of an edge-sharing α-PbO2 active layer. The existence of charge-transfer interactions at the MAPbX3 /α-PbO2 interface significantly modulates the surface electron density of the α-PbO2 and optimizes the adsorption free energy of oxygen-containing intermediate species. Furthermore, the soft-lattice nature of halide perovskites enables more facile triggering of lattice-oxygen oxidation of nanostructured α-PbO2 , exhibiting pH-dependent OER activity and non-concerted proton-electron transfer for MAPbX3 @AlPO-5 composite. As a result, the developed MAPbBr3 @AlPO-5 composite manifests an ultralow overpotential of 233 mV at 10 mA cm-2 in 1 m KOH. These findings offer facile access to halide perovskite applied to water electrolysis with enhanced intrinsic activity, providing a new paradigm for designing high-efficiency OER electrocatalysts.

4.
Comput Struct Biotechnol J ; 21: 2405-2418, 2023.
Article de Anglais | MEDLINE | ID: mdl-37066124

RÉSUMÉ

Platelet-derived growth factor-D (PDGF-D) is abundantly expressed in ocular diseases. Yet, it remains unknown whether and how PDGF-D affects ocular cells or cell-cell interactions in the eye. In this study, using single-cell RNA sequencing (scRNA-seq) and a mouse model of PDGF-D overexpression in retinal pigment epithelial (RPE) cells, we found that PDGF-D overexpression markedly upregulated the key immunoproteasome genes, leading to increased antigen processing/presentation capacity of RPE cells. Also, more than 6.5-fold ligand-receptor pairs were found in the PDGF-D overexpressing RPE-choroid tissues, suggesting markedly increased cell-cell interactions. Moreover, in the PDGF-D-overexpressing tissues, a unique cell population with a transcriptomic profile of both stromal cells and antigen-presenting RPE cells was detected, suggesting PDGF-D-induced epithelial-mesenchymal transition of RPE cells. Importantly, administration of ONX-0914, an immunoproteasome inhibitor, suppressed choroidal neovascularization (CNV) in a mouse CNV model in vivo. Together, we show that overexpression of PDGF-D increased pro-angiogenic immunoproteasome activities, and inhibiting immunoproteasome pathway may have therapeutic value for the treatment of neovascular diseases.

5.
J Cell Commun Signal ; 17(1): 151-165, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36284029

RÉSUMÉ

Although mitogen-inducible gene 6 (MIG6) is highly expressed in vascular endothelial cells, it remains unknown whether MIG6 affects vascular permeability. Here, we show for the first time a critical role of MIG6 in limiting vascular permeability. We unveil that genetic deletion of Mig6 in mice markedly increased VEGFA-induced vascular permeability, and MIG6 knockdown impaired endothelial barrier function. Mechanistically, we reveal that MIG6 inhibits VEGFR2 phosphorylation by binding to the VEGFR2 kinase domain 2, and MIG6 knockdown increases the downstream signaling of VEGFR2 by enhancing phosphorylation of PLCγ1 and eNOS. Moreover, MIG6 knockdown disrupted the balance between RAC1 and RHOA GTPase activation, leading to endothelial cell barrier breakdown and the elevation of vascular permeability. Our findings demonstrate an essential role of MIG6 in maintaining endothelial cell barrier integrity and point to potential therapeutic implications of MIG6 in the treatment of diseases involving vascular permeability. Xing et al. (2022) investigated the critical role of MIG6 in vascular permeability. MIG6 deficiency promotes VEGFA-induced vascular permeability via activation of PLCγ1-Ca2+-eNOS signaling and perturbation of the balance in RAC1/RHOA activation, resulting in endothelial barrier disruption.

7.
Water Res ; 213: 118186, 2022 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-35183014

RÉSUMÉ

Timely and simultaneously detecting BOD and nitrite concentrations is of great significance for curbing of water pollution and adjusting wastewater treatment strategies. However, existing BOD and nitrite biosensors cannot perform synchronous detection due to their single electroactivity and differences in detection time. This study reported a novel dual-function electrochemical biosensor (DFEB) that could perform fast, simultaneous detection of nitrite and dissolved BOD. DFEB conducted a potential-step chronoamperometry on the mixed-bacteria bioelectrode with bidirectional electron transfer ability to obtain response signals. DFEB accurately measured dissolved BOD in the range of 5 ∼ 100 mg BOD L-1 and nitrite in the range of 0.05 ∼ 16 mg NO2--N L-1 within 20 min and maintain stable performance over 200 tests. DFEB performed well in artificial wastewater, aquatic wastewater, anaerobic tank effluent and anammox effluent, with relative errors < 15.7% and 16.8% in detecting nitrite and dissolved BOD, respectively. Our study provided a feasible way to develop multifunctional biosensors for detecting pollutants with different redox properties in wastewater.

8.
Front Cell Dev Biol ; 9: 631413, 2021.
Article de Anglais | MEDLINE | ID: mdl-34222224

RÉSUMÉ

Retinal pigment epithelium (RPE), a postmitotic monolayer located between the neuroretina and choroid, supports the retina and is closely associated with vision loss diseases such as age-related macular degeneration (AMD) upon dysfunction. Although environmental stresses are known to play critical roles in AMD pathogenesis and the roles of other stresses have been well investigated, glucose deprivation, which can arise from choriocapillary flow voids, has yet to be fully explored. In this study, we examined the involvement of VEGFR2 in glucose deprivation-mediated cell death and the underlying mechanisms. We found that VEGFR2 levels are a determinant for RPE cell death, a critical factor for dry AMD, under glucose deprivation. RNA sequencing analysis showed that upon VEGFR2 knockdown under glucose starvation, endoplasmic reticulum (ER) stress and unfolded protein response (UPR) are reduced. Consistently, VEGFR2 overexpression increased ER stress under the same condition. Although VEGFR2 was less expressed compared to EGFR1 and c-Met in RPE cells, it could elicit a higher level of ER stress induced by glucose starvation. Finally, downregulated VEGFR2 attenuated the oxidative stress and inflammatory factor expression, two downstream targets of ER stress. Our study, for the first time, has demonstrated a novel role of VEGFR2 in RPE cells under glucose deprivation, thus providing valuable insights into the mechanisms of AMD pathogenesis and suggesting that VEGFR2 might be a potential therapeutic target for AMD prevention, which may impede its progression.

9.
Front Cell Dev Biol ; 9: 634242, 2021.
Article de Anglais | MEDLINE | ID: mdl-33693003

RÉSUMÉ

The mitogen-inducible gene 6 (MIG6) is an adaptor protein widely expressed in vascular endothelial cells. However, it remains unknown thus far whether it plays a role in angiogenesis. Here, using comprehensive in vitro and in vivo model systems, we unveil a potent anti-angiogenic effect of MIG6 in retinal development and neovascularization and the underlying molecular and cellular mechanisms. Loss of function assays using genetic deletion of Mig6 or siRNA knockdown increased angiogenesis in vivo and in vitro, while MIG6 overexpression suppressed pathological angiogenesis. Moreover, we identified the cellular target of MIG6 by revealing its direct inhibitory effect on vascular endothelial cells (ECs). Mechanistically, we found that the anti-angiogenic effect of MIG6 is fulfilled by binding to SHC1 and inhibiting its phosphorylation. Indeed, SHC1 knockdown markedly diminished the effect of MIG6 on ECs. Thus, our findings show that MIG6 is a potent endogenous inhibitor of angiogenesis that may have therapeutic value in anti-angiogenic therapy.

10.
Chemosphere ; 239: 124715, 2020 Jan.
Article de Anglais | MEDLINE | ID: mdl-31499311

RÉSUMÉ

Electrodeposition is an economical and efficient way to prepare Ti/SnO2-Sb electrode for electrochemical oxidizing pollutants in wastewater. The solvent used for electrodeposition has a great effect on electrode performance. The conventional Ti/SnO2-Sb electrode electrodeposited using aqueous solvent has poor electrochemical activity and short service life. In this study, a Ti/SnO2-Sb electrode was prepared via electrodeposition using a deep eutectic solvent (DES). This new Ti/SnO2-Sb-DES electrode performed a rate constant of 0.571 h-1 for methylene blue decolorization and long accelerated service life of 12.9 h (100 mA cm-2; 0.5 M H2SO4), which were 1.7 times and 3.2 times as high as that of the electrode prepared in aqueous solvent, respectively. The enhanced properties were related to the 1.3 times increased electrochemically active surface area of Ti/SnO2-Sb-DES electrode which had a rough, multilayer and uniform surface structure packed with nano-sized coating particles. In conclusion, this study developed a facile, green and efficient pathway to prepare Ti/SnO2-Sb electrode with high performance.


Sujet(s)
Antimoine/composition chimique , Techniques électrochimiques/méthodes , Galvanoplastie/méthodes , Solvants/composition chimique , Polluants chimiques de l'eau/composition chimique , Purification de l'eau/méthodes , Électrodes/normes , Bleu de méthylène/composition chimique , Oxydoréduction , Composés de l'étain/composition chimique , Titane/composition chimique , Eaux usées/composition chimique
11.
Pharmacol Res ; 146: 104277, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31112749

RÉSUMÉ

Neovascular diseases, such as many cancers and ocular disorders, are life threatening and devastating. Although anti-vascular endothelial growth factor A (VEGF-A) therapy is available, many patients are not responsive and drug resistance can develop. To try to overcome these problems, combination therapy targeting VEGF-A and platelet-derived growth factor B (PDGF-B) was tested. However, one obvious drawback was that the other VEGF and PDGF family members were not inhibited and therefore could compensate. Indeed, this was, at least to some extent, demonstrated by the disappointing outcomes. To this end, we designed novel multi-targeted inhibitors that can block most of the VEGF and PDGF family members simultaneously by making a fusion protein containing the ligand-binding domains of vascular endothelial growth factor receptor 1 (VEGFR1), vascular endothelial growth factor receptor 2 (VEGFR2) and platelet-derived growth factor receptor beta (PDGFRß), which can therefore act as a decoy blocker for most of the VEGF and PDGF family members. Indeed, in cultured cells, the novel inhibitors suppressed the migration and proliferation of both vascular endothelial cells and smooth muscle cells, and abolished VEGFR2 and PDGFRß activation. Importantly, in a choroidal neovascularization model in vivo, the novel inhibitor inhibited ocular neovascularization more efficiently than the mono-inhibitors against VEGFR or PDGFR alone respectively. Mechanistically, a genome-wide microarray analysis unveiled that the novel inhibitor regulated unique sets of genes that were not regulated by the mono-inhibitors, further demonstrating the functional uniqueness and superiority of the novel inhibitor. Together, we show that the multi-targeted inhibitors that can block VEGFR1, VEGFR2 and PDGFRß simultaneously suppress pathological angiogenesis more efficiently than monotherapy, and may therefore have promising therapeutic value for the treatment of neovascular diseases.


Sujet(s)
Inhibiteurs de l'angiogenèse/usage thérapeutique , Oeil/effets des médicaments et des substances chimiques , Néovascularisation pathologique/traitement médicamenteux , Récepteur au PDGF bêta/antagonistes et inhibiteurs , Protéines de fusion recombinantes/usage thérapeutique , Récepteur-1 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Inhibiteurs de l'angiogenèse/pharmacologie , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Oeil/vascularisation , Oeil/métabolisme , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/physiologie , Humains , Souris de lignée C57BL , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/physiologie , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Récepteur au PDGF bêta/métabolisme , Protéines de fusion recombinantes/pharmacologie , Transcriptome/effets des médicaments et des substances chimiques , Récepteur-1 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme
12.
Proc Natl Acad Sci U S A ; 114(40): 10737-10742, 2017 10 03.
Article de Anglais | MEDLINE | ID: mdl-28923916

RÉSUMÉ

Ocular neovascularization is a devastating pathology of numerous ocular diseases and is a major cause of blindness. Caveolin-1 (Cav-1) plays important roles in the vascular system. However, little is known regarding its function and mechanisms in ocular neovascularization. Here, using comprehensive model systems and a cell permeable peptide of Cav-1, cavtratin, we show that Cav-1 is a critical player in ocular neovascularization. The genetic deletion of Cav-1 exacerbated and cavtratin administration inhibited choroidal and retinal neovascularization. Importantly, combined administration of cavtratin and anti-VEGF-A inhibited neovascularization more effectively than monotherapy, suggesting the existence of other pathways inhibited by cavtratin in addition to VEGF-A. Indeed, we found that cavtratin suppressed multiple critical components of pathological angiogenesis, including inflammation, permeability, PDGF-B and endothelial nitric oxide synthase expression (eNOS). Mechanistically, we show that cavtratin inhibits CNV and the survival and migration of microglia and macrophages via JNK. Together, our data demonstrate the unique advantages of cavtratin in antiangiogenic therapy to treat neovascular diseases.


Sujet(s)
Inhibiteurs de l'angiogenèse/pharmacologie , Anticorps monoclonaux/pharmacologie , Cavéoline-1/physiologie , Néovascularisation choroïdienne/prévention et contrôle , MAP Kinase Kinase 4/métabolisme , Fragments peptidiques/pharmacologie , Néovascularisation rétinienne/prévention et contrôle , Animaux , Cavéoline-1/pharmacologie , Néovascularisation choroïdienne/métabolisme , Néovascularisation choroïdienne/anatomopathologie , Association de médicaments , Humains , Souris knockout , Néovascularisation rétinienne/métabolisme , Néovascularisation rétinienne/anatomopathologie , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs
13.
Thromb Haemost ; 117(4): 750-757, 2017 04 03.
Article de Anglais | MEDLINE | ID: mdl-28203682

RÉSUMÉ

Junctional adhesion molecule-C (JAM-C) has been shown to play critical roles during development and in immune responses. However, its role in adult eyes under oxidative stress remains poorly understood. Here, we report that JAM-C is abundantly expressed in adult mouse retinae and choroids in vivo and in cultured retinal pigment epithelium (RPE) and photoreceptor cells in vitro. Importantly, both JAM-C expression and its membrane localisation are downregulated by H2O2-induced oxidative stress. Under H2O2-induced oxidative stress, JAM-C is critically required for the survival of human RPE cells. Indeed, loss of JAM-C by siRNA knockdown decreased RPE cell survival. Mechanistically, we show that JAM-C is required to maintain VEGFR2 expression in RPE cells, and VEGFR2 plays an important role in keeping the RPE cells viable since overexpression of VEGFR2 partially restored impaired RPE survival caused by JAM-C knockdown and increased RPE survival. We further show that JAM-C regulates VEGFR2 expression and, in turn, modulates p38 phosphorylation. Together, our data demonstrate that JAM-C plays an important role in maintaining VEGR2 expression to promote RPE cell survival under oxidative stress. Given the vital importance of RPE in the eye, approaches that can modulate JAM-C expression may have therapeutic values in treating diseases with impaired RPE survival.


Sujet(s)
Molécules d'adhérence cellulaire/métabolisme , Cellules épithéliales/métabolisme , Immunoglobulines/métabolisme , Stress oxydatif , Épithélium pigmentaire de la rétine/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Animaux , Molécules d'adhérence cellulaire/génétique , Lignée cellulaire , Survie cellulaire , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/anatomopathologie , Femelle , Humains , Peroxyde d'hydrogène/toxicité , Immunoglobulines/génétique , Souris de lignée C57BL , Stress oxydatif/effets des médicaments et des substances chimiques , Phosphorylation , Interférence par ARN , Épithélium pigmentaire de la rétine/effets des médicaments et des substances chimiques , Épithélium pigmentaire de la rétine/anatomopathologie , Transduction du signal , Transfection , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique , p38 Mitogen-Activated Protein Kinases/métabolisme
14.
J Int Med Res ; 44(6): 1403-1413, 2016 Dec.
Article de Anglais | MEDLINE | ID: mdl-27885039

RÉSUMÉ

Objective To quantify T helper (Th)17 cells and determine interleukin (IL)-17A levels in peripheral blood mononuclear cell (PBMC) culture and vitreous fluid from patients with type 2 diabetes mellitus (T2DM) with diabetic retinopathy (DR). Methods Th17 cell frequency and IL-17A concentrations in PBMCs from 60 patients with T2DM with DR, 30 without DR and 30 sex- and age-matched healthy individuals were measured by flow cytometry and enzyme-linked immunosorbent assay (ELISA), respectively. IL-17A levels in vitreous fluid from 31 eyes with proliferative DR and diabetic macular oedema (DR group) and 32 eyes with an epiretinal membrane and macular hole (control group) that underwent vitrectomy were also examined by ELISA. Results Compared with the control group, the proportion of Th17 cells and IL-17A concentrations in PBMCs were significantly increased in patients without DR but decreased in those with DR. IL-17A concentrations and Th17 cell frequency in PBMCs tended to decrease with DR severity and were negatively correlated with body mass index, T2DM duration and glycated haemoglobin. Additionally, vitreous fluid IL-17A levels were significantly elevated in patients with DR compared with those of the control group. Conclusions We conclude that disturbances in Th17 cells and IL-17A levels are possibly associated with DR.


Sujet(s)
Diabète de type 2/immunologie , Rétinopathie diabétique/immunologie , Interleukine-17/génétique , Agranulocytes/immunologie , Cellules Th17/immunologie , Corps vitré/immunologie , Sujet âgé , Indice de masse corporelle , Études cas-témoins , Diabète de type 2/génétique , Diabète de type 2/anatomopathologie , Diabète de type 2/chirurgie , Rétinopathie diabétique/génétique , Rétinopathie diabétique/anatomopathologie , Rétinopathie diabétique/chirurgie , Membrane épirétinienne/génétique , Membrane épirétinienne/immunologie , Membrane épirétinienne/anatomopathologie , Membrane épirétinienne/chirurgie , Femelle , Expression des gènes , Hémoglobine glyquée/génétique , Hémoglobine glyquée/immunologie , Humains , Interleukine-17/immunologie , Agranulocytes/anatomopathologie , Oedème maculaire/génétique , Oedème maculaire/immunologie , Oedème maculaire/anatomopathologie , Oedème maculaire/chirurgie , Mâle , Adulte d'âge moyen , Culture de cellules primaires , Perforations de la rétine/génétique , Perforations de la rétine/immunologie , Perforations de la rétine/anatomopathologie , Perforations de la rétine/chirurgie , Cellules Th17/anatomopathologie , Vitrectomie , Corps vitré/métabolisme , Corps vitré/anatomopathologie , Corps vitré/chirurgie
15.
Oncotarget ; 7(47): 77902-77915, 2016 Nov 22.
Article de Anglais | MEDLINE | ID: mdl-27788490

RÉSUMÉ

Anti-VEGF-A therapy has proven to be effective for many neovascular diseases. However, drug resistance to anti-VEGF-A treatment can develop. Also, not all patients with neovascular diseases are responsive to anti-VEGF-A treatment. The mechanisms underlying these important issues remain unclear. In this study, using different model systems, we found that inhibition of VEGF-A directly upregulated PDGF-CC and its receptors in multiple cell types in pathological angiogenesis in vitro and in vivo. Importantly, we further revealed that combinatorial targeting of VEGF-A and PDGF-CC suppressed pathological angiogenesis more efficiently than monotherapy. Given the potent angiogenic activity of PDGF-CC, our findings suggest that the development of resistance to anti-VEGF-A treatment may be caused by the compensatory upregulation of PDGF-CC, and combined inhibition of VEGF-A and PDGF-CC may have therapeutic advantages in treating neovascular diseases.


Sujet(s)
Néovascularisation choroïdienne/traitement médicamenteux , Néovascularisation choroïdienne/métabolisme , Lymphokines/métabolisme , Facteur de croissance dérivé des plaquettes/métabolisme , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Animaux , Anticorps neutralisants/immunologie , Anticorps neutralisants/pharmacologie , Cellules cultivées , Néovascularisation choroïdienne/anatomopathologie , Résistance aux substances , Femelle , Humains , Lymphokines/biosynthèse , Souris , Souris de lignée C3H , Souris de lignée C57BL , Souris de lignée DBA , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie , Facteur de croissance dérivé des plaquettes/biosynthèse , Cellules RAW 264.7 , Épithélium pigmentaire de la rétine/cytologie , Épithélium pigmentaire de la rétine/effets des médicaments et des substances chimiques , Épithélium pigmentaire de la rétine/métabolisme , Transduction du signal , Régulation positive , Facteur de croissance endothéliale vasculaire de type A/immunologie , Facteur de croissance endothéliale vasculaire de type A/métabolisme
16.
Sci Rep ; 6: 26059, 2016 05 18.
Article de Anglais | MEDLINE | ID: mdl-27189805

RÉSUMÉ

Vascular endothelial growth factor B (VEGF-B) was discovered a long time ago. However, its role in hyperglycemia- and VEGF-A inhibition-induced retinal apoptosis remains unknown thus far. Yet, drugs that can block VEGF-B are being used to treat patients with diabetic retinopathy and other ocular neovascular diseases. It is therefore urgent to have a better understanding of the function of VEGF-B in these pathologies. Here, we report that both streptozotocin (STZ)-induced diabetes in rats and Macugen intravitreal injection in mice leads to retinal apoptosis in retinal ganglion cell and outer nuclear layers respectively. Importantly, VEGF-B treatment by intravitreal injection markedly reduced retinal apoptosis in both models. We further reveal that VEGF-B and its receptors, vascular endothelial growth factor 1 (VEGFR1) and neuropilin 1 (NP1), are abundantly expressed in rat retinae and choroids and are upregulated by high glucose with concomitant activation of Akt and Erk. These data highlight an important function of VEGF-B in protecting retinal cells from apoptosis induced by hyperglycemia and VEGF-A inhibition. VEGF-B may therefore have a therapeutic potential in treating various retinal degenerative diseases, and modulation of VEGF-B activity in the eye needs careful consideration.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Rétinopathies/traitement médicamenteux , Cellules ganglionnaires rétiniennes/physiologie , Facteur de croissance endothéliale vasculaire de type B/administration et posologie , Animaux , Aptamères nucléotidiques/toxicité , Diabète expérimental/complications , Souris de lignée C57BL , Rats , Rétinopathies/physiopathologie , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Résultat thérapeutique
17.
Proc Natl Acad Sci U S A ; 111(41): 14806-11, 2014 Oct 14.
Article de Anglais | MEDLINE | ID: mdl-25267616

RÉSUMÉ

Blood vessel degeneration is critically involved in nearly all types of degenerative diseases. Therefore strategies to enhance blood vessel protection and survival are highly needed. In this study, using different animal models and cultured cells, we show that PDGF-CC is a potent vascular protective and survival factor. PDGF-CC deficiency by genetic deletion exacerbated blood vessel regression/degeneration in various animal models. Importantly, treatment with PDGF-CC protein not only increased the survival of retinal blood vessels in a model of oxygen-induced blood vessel regression but also markedly rescued retinal and blood vessel degeneration in a disease model of retinitis pigmentosa. Mechanistically, we revealed that heme oxygenase-1 (HMOX1) activity is critically required for the vascular protective/survival effect of PDGF-CC, because blockade of HMOX1 completely abolished the protective effect of PDGF-CC in vitro and in vivo. We further found that both PDGF receptors, PDGFR-ß and PDGFR-α, are required for the vasoprotective effect of PDGF-CC. Thus our data show that PDGF-CC plays a pivotal role in maintaining blood vessel survival and may be of therapeutic value in treating various types of degenerative diseases.


Sujet(s)
Heme oxygenase-1/métabolisme , Lymphokines/métabolisme , Facteur de croissance dérivé des plaquettes/métabolisme , Dégénérescence de la rétine/enzymologie , Dégénérescence de la rétine/prévention et contrôle , Vaisseaux rétiniens/enzymologie , Vaisseaux rétiniens/anatomopathologie , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/anatomopathologie , Lymphokines/pharmacologie , Souris , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Oxygène , Facteur de croissance dérivé des plaquettes/pharmacologie , Récepteur au PDGF alpha/métabolisme , Récepteur au PDGF bêta/métabolisme , Dégénérescence de la rétine/anatomopathologie , Vaisseaux rétiniens/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques
18.
Cytokine ; 70(2): 179-84, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25138017

RÉSUMÉ

Interleukin-28A (IL-28A), a member of type III interferons (IFN-λs), promotes antiviral, antitumor and immune responses. However, its ability to regulate autoimmune diseases is poorly understood. In this study, we examined the effect of IL-28A on retinal antigen-induced experimental autoimmune uveoretinitis (EAU), a mouse model of human T-cell-mediated autoimmune eye disease. We found that administration of IL-28A enhanced EAU scores and autoimmune response parameters including delayed-type hypersensitivity (DTH), Ag-specific T cell proliferation and the production of Ag-specific IL-17 and IFN-γ in the priming phase. The effect of IL-28A was abrogated by administration of a neutralizing antibody against IL-28A. Our results suggest that IL-28A is capable of exacerbating a T-cell-mediated autoimmune disease. Thus, targeting IL-28A may provide a new therapeutic approach to T cell-mediated autoimmune diseases such as uveitis.


Sujet(s)
Maladies auto-immunes/immunologie , Auto-immunité , Interleukines/métabolisme , Rétine/immunologie , Rétine/anatomopathologie , Animaux , Anticorps/immunologie , Antigènes/immunologie , Maladies auto-immunes/anatomopathologie , Modèles animaux de maladie humaine , Relation dose-réponse (immunologie) , Humains , Hypersensibilité retardée/immunologie , Hypersensibilité retardée/anatomopathologie , Interféron gamma/biosynthèse , Interleukine-17/biosynthèse , Interleukines/génétique , Interleukines/immunologie , Noeuds lymphatiques/métabolisme , Souris , ARN messager/génétique , ARN messager/métabolisme
19.
Eur J Pharmacol ; 740: 619-26, 2014 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-24973692

RÉSUMÉ

Histone deacetylases (HDACs) regulate gene transcription by modifying the acetylation level of histone and nonhistone proteins. In this study, we examined the effect of largazole, an inhibitor of class I HDACs, on inflammatory corneal angiogenesis. In a mouse model of alkali-induced corneal neovascularization (CNV), topical application of largazole to the injured corneas attenuated CNV. In addition, in vivo treatment with largazole down-regulated the expression of the pro-angiogenic factors VEGF, b-FGF, TGFß1 and EGF but up-regulated the expression of the anti-angiogenic factors Thrombospondin-1 (Tsp-1), Tsp-2 and ADAMTS-1 in the injured corneas. Furthermore, largazole inhibited the expression of pro-angiogenic factors, migration, proliferation and tube formation by human microvascular endothelial cells (HEMC-1) in vitro. These data indicate that largazole has therapeutic potential for angiogenesis-associated diseases.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Néovascularisation cornéenne/traitement médicamenteux , Depsipeptides/usage thérapeutique , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Thiazoles/usage thérapeutique , Protéines ADAM/génétique , Animaux , Lignée cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Néovascularisation cornéenne/génétique , Néovascularisation cornéenne/métabolisme , Depsipeptides/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Cellules endothéliales/physiologie , Facteur de croissance épidermique/génétique , Femelle , Facteurs de croissance fibroblastique/génétique , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Souris de lignée BALB C , ARN messager/métabolisme , Thiazoles/pharmacologie , Thrombospondine-1/génétique , Thrombospondines/génétique , Facteur de croissance transformant bêta-1/génétique , Facteur de croissance endothéliale vasculaire de type A/génétique
20.
Arterioscler Thromb Vasc Biol ; 33(2): 330-8, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23241411

RÉSUMÉ

OBJECTIVE: Activation of the immune system via toll-like receptors (TLRs) is implicated in atherosclerosis, microvascular complications, and angiogenesis. However, the involvement of TLRs in inflammation-associated angiogenesis in ischemic neural tissue has not been investigated. The goal of this study is to determine the role of TLR4 signaling in oxygen-induced neovascularization in retina, a neural tissue. METHODS AND RESULTS: In oxygen-induced retinopathy model, we found that retinal neovascularization was significantly attenuated in TLR4(-/-) mice. The further study revealed that the absence of TLR4 led to downregulation of proinflammatory factors in association with the attenuated activation of glia in the ischemic retina, which was also associated with reduced expression of high-mobility group box-1, an endogenous ligand for TLR4. The application of high-mobility group box-1 to the ischemic retina promoted the production of proinflammatory factors in wild-type but not TLR4(-/-) mice. High-mobility group box-1 treatment in vitro also significantly promoted the production of proinflammatory factors in retinal glial cells from wild-type mice, but much less from TLR4(-/-) mice. CONCLUSIONS: Our results suggest that the release of high-mobility group box-1 in ischemic neural tissue initiates TLR4-dependent responses that contribute to neovascularization. These findings represented a previously unrecognized effect of TLR4 on angiogenesis in association with the activation of glia in ischemic neural tissue.


Sujet(s)
Ischémie/immunologie , Néovascularisation rétinienne/immunologie , Vaisseaux rétiniens/immunologie , Récepteur de type Toll-4/métabolisme , Protéines angiogéniques/génétique , Protéines angiogéniques/métabolisme , Animaux , Cellules cultivées , Modèles animaux de maladie humaine , Femelle , Régulation de l'expression des gènes , Protéine HMGB1/administration et posologie , Protéine HMGB1/métabolisme , Médiateurs de l'inflammation/métabolisme , Injections intravitréennes , Ischémie/génétique , Ischémie/métabolisme , Ischémie/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Névroglie/immunologie , Névroglie/métabolisme , Protéines recombinantes/administration et posologie , Néovascularisation rétinienne/génétique , Néovascularisation rétinienne/métabolisme , Néovascularisation rétinienne/anatomopathologie , Vaisseaux rétiniens/effets des médicaments et des substances chimiques , Vaisseaux rétiniens/métabolisme , Vaisseaux rétiniens/anatomopathologie , Transduction du signal , Récepteur de type Toll-4/déficit , Récepteur de type Toll-4/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE