Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
iScience ; 27(3): 109213, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38439953

RESUMEN

Patients with Graves' disease (GD) can develop Graves' ophthalmopathy (GO), but the underlying pathological mechanisms driving this development remain unclear. In our study, which included patients with GD and GO, we utilized single-cell RNA sequencing (scRNA-seq) and multiplatform analyses to investigate CD169+ classical monocytes, which secrete proinflammatory cytokines and are expanded through activated interferon signaling. We found that CD169+ clas_mono was clinically significant in predicting GO progression and prognosis, and differentiated into CD169+ macrophages that promote inflammation, adipogenesis, and fibrosis. Our murine model of early-stage GO showed that CD169+ classical monocytes accumulated in orbital tissue via the Cxcl12-Cxcr4 axis. Further studies are needed to investigate whether targeting circulating monocytes and the Cxcl12-Cxcr4 axis could alleviate GO progression.

2.
Commun Biol ; 6(1): 1048, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37848613

RESUMEN

Behect's disease is a chronic vasculitis characterized by complex multi-organ immune aberrations. However, a comprehensive understanding of the gene-regulatory profile of peripheral autoimmunity and the diverse immune responses across distinct cell types in Behcet's disease (BD) is still lacking. Here, we present a multi-omic single-cell study of 424,817 cells in BD patients and non-BD individuals. This study maps chromatin accessibility and gene expression in the same biological samples, unraveling vast cellular heterogeneity. We identify widespread cell-type-specific, disease-associated active and pro-inflammatory immunity in both transcript and epigenomic aspects. Notably, integrative multi-omic analysis reveals putative TF regulators that might contribute to chromatin accessibility and gene expression in BD. Moreover, we predicted gene-regulatory networks within nominated TF activators, including AP-1, NF-kB, and ETS transcript factor families, which may regulate cellular interaction and govern inflammation. Our study illustrates the epigenetic and transcriptional landscape in BD peripheral blood and expands understanding of potential epigenomic immunopathology in this disease.


Asunto(s)
Síndrome de Behçet , Vasculitis , Humanos , Síndrome de Behçet/genética , Transcriptoma , Cromatina/genética , Perfilación de la Expresión Génica
3.
Signal Transduct Target Ther ; 8(1): 305, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37591843

RESUMEN

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.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos , Factor B de Crecimiento Endotelial Vascular , Humanos , Factor 2 de Crecimiento de Fibroblastos/genética , Inmunoterapia , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
4.
iScience ; 26(5): 106729, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37216113

RESUMEN

Gingiva-derived mesenchymal stem cells (GMSCs) have shown astonishing efficacy in the treatment of various autoimmune diseases. However, the mechanisms underlying these immunosuppressive properties remain poorly understood. Here, we generated a lymph node single-cell transcriptomic atlas of GMSC-treated experimental autoimmune uveitis mice. GMSC exerted profound rescue effects on T cells, B cells, dendritic cells, and monocytes. GMSCs rescued the proportion of T helper 17 (Th17) cells and increased the proportion of regulatory T cells. In addition to globally altered transcriptional factors (Fosb and Jund), we observed cell type-dependent gene regulation (e.g., Il17a and Rac1 in Th17 cells), highlighting the GMSCs' cell type-dependent immunomodulatory capacity. GMSCs strongly influenced the phenotypes of Th17 cells, suppressing the formation of the highly inflammatory CCR6-CCR2+ phenotype and enhancing the production of interleukin (IL) -10 in the CCR6+CCR2+ phenotype. Integration of the glucocorticoid-treated transcriptome suggests a more specific immunosuppressive effect of GMSCs on lymphocytes.

5.
Comput Struct Biotechnol J ; 21: 2405-2418, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37066124

RESUMEN

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.

6.
Angiogenesis ; 25(4): 517-533, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35859222

RESUMEN

The critical factors regulating stem cell endothelial commitment and renewal remain not well understood. Here, using loss- and gain-of-function assays together with bioinformatic analysis and multiple model systems, we show that PDGFD is an essential factor that switches on endothelial commitment of embryonic stem cells (ESCs). PDGFD genetic deletion or knockdown inhibits ESC differentiation into EC lineage and increases ESC self-renewal, and PDGFD overexpression activates ESC differentiation towards ECs. RNA sequencing reveals a critical requirement of PDGFD for the expression of vascular-differentiation related genes in ESCs. Importantly, PDGFD genetic deletion or knockdown increases ESC self-renewal and decreases blood vessel densities in both embryonic and neonatal mice and in teratomas. Mechanistically, we reveal that PDGFD fulfills this function via the MAPK/ERK pathway. Our findings provide new insight of PDGFD as a novel regulator of ESC fate determination, and suggest therapeutic implications of modulating PDGFD activity in stem cell therapy.


Asunto(s)
Células Madre Embrionarias , Modelos Biológicos , Animales , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones
7.
Biochem Pharmacol ; 202: 115116, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35671791

RESUMEN

Cyclosporine A (CsA) is a widely known immunosuppressive agent that is clinically important in autoimmune diseases owing to its selective suppression of T lymphocytes. Although it has long been recognized to inhibit T cell responses by blocking calcineurin, the potential targets and specific downstream mechanisms remain elusive. Herein, we built a comprehensive single-cell transcriptomic landscape of immune cells in the blank, untreated experimental autoimmune uveitis (EAU), and CsA-treated EAU mice. CsA reversed EAU-associated changes in cell type composition, genomic expression, cell trajectory, and cell-cell communication. We found that CsA reverses the proportion change of disease-related immune cells; regulates several crucial pathogenic factors (eg. IL1r1, CD48, and Bhlhe40) in T helper 17 cells (Th17), the transcription factor Bhlhe40 was also rescued in T helper 1 cells (Th1); and may differentiate Tregs into a state of enhanced immunosuppression. In addition, we revealed the rescued impact of CsA on all immune cell types, especially on plasma B cells differentiation and immunoglobulin secretion. Furthermore, comparisons with glucocorticoids showed that CsA might have a more premium rescue effect involved in attenuating the pathogenicity of autoreactive T cells. Our work provides a comprehensive single-cell transcriptional atlas of immune cells under CsA therapy, providing advanced insights into the mechanisms underlying CsA and a reference for developing new therapeutic strategies for autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Uveítis , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Ciclosporina/farmacología , Ciclosporina/uso terapéutico , Modelos Animales de Enfermedad , Ratones , Análisis de la Célula Individual , Células Th17
9.
J Immunol ; 207(3): 837-848, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34282004

RESUMEN

Dendritic cells (DCs) are critical for pathogen recognition and Ag processing/presentation. Human monocyte-derived DCs (moDCs) have been extensively used in experimental studies and DC-based immunotherapy approaches. However, the extent of human moDC and peripheral DCs heterogeneity and their interrelationship remain elusive. In this study, we performed single-cell RNA sequencing of human moDCs and blood DCs. We identified seven subtypes within moDCs: five corresponded to type 2 conventional DCs (cDC2s), and the other two were CLEC10A+CD127+ cells with no resemblance to any peripheral DC subpopulations characterized to date. Moreover, we defined five similar subtypes in human cDC2s, revealed the potential differentiation trajectory among them, and unveiled the transcriptomic differences between moDCs and cDC2s. We further studied the transcriptomic changes of each moDC subtype during maturation, demonstrating SLAMF7 and IL15RA as maturation markers and CLEC10A and SIGLEC10 as markers for immature DCs. These findings will enable more accurate functional/developmental analyses of human cDC2s and moDCs.


Asunto(s)
Células Dendríticas/fisiología , Monocitos/fisiología , Análisis de la Célula Individual/métodos , Adulto , Diferenciación Celular/genética , Células Cultivadas , Citocinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Lectinas/genética , Lectinas Tipo C/genética , Masculino , Receptores de Superficie Celular/genética , Receptores de Interleucina-15/genética , Análisis de Secuencia de ARN , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Células Th2/inmunología , Adulto Joven
10.
Front Cell Dev Biol ; 9: 686886, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34150781

RESUMEN

Platelet-derived growth factor-D (PDGF-D) is highly expressed in immune cells. However, the potential role of PDGF-D in immune system remains thus far unclear. Here, we reveal a novel function of PDGF-D in activating both classical and alternative complement pathways that markedly increase chemokine and cytokine responses to promote macrophage polarization. Pharmacological targeting of the complement C3a receptor using SB290157 alleviated PDGF-D-induced neuroinflammation by blocking macrophage polarization and inhibited pathological choroidal neovascularization. Our study thus suggests that therapeutic strategies targeting both PDGF-D and the complement system may open up new possibilities for the treatment of neovascular diseases.

11.
Front Cell Dev Biol ; 9: 634242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33693003

RESUMEN

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.

12.
J Ocul Pharmacol Ther ; 35(6): 331-340, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31343394

RESUMEN

Purpose: Lysophosphatidic acid (LPA) is a growth factor-like phospholipid that has been recognized as a profibrotic mediator in numerous tissues, yet, whether it plays a role in subconjunctival fibrosis remains to be investigated. Therefore, this study was designed to examine the effect of LPA1-3 signaling inhibitor, Ki16425 on the conversion of human Tenon's fibroblasts (HTFs) into myofibroblasts. Methods: Primary cultured HTFs were incubated with transforming growth factor-ß1 (TGF-ß1) alone or combined with Ki16425, the cell proliferation and migration were measured by Cell Counting Kit-8 and the scratch wound assay, respectively. HTFs contractility was evaluated with 3-dimensional (3D) Collagen Contraction assay. The mRNA and protein levels of α-smooth muscle actin (α-SMA), Snail and the phosphorylation levels of Smad2/3, p38MAPK, and ERK1/2 were determined by real-time quantitative polymerase chain reaction (RT-qPCR), western blot, and immunofluorescence staining. Results: Ki16425 significantly prevent the proliferation and migration of Tenon's fibroblasts (HTFs) in a dose-dependent manner. Furthermore, Ki16425 blocked HTFs myofibroblast differentiation via downregulation of mRNA and protein expression of α-SMA. 3D collagen gel contraction assay demonstrated that Ki16425 effectively inhibits myofibroblast contraction induced by TGF-ß1. Mechanistically, we revealed that Ki16425 reduces Smad2/3 but not p38MAPK or ERK1/2 phosphorylation by TGF-ß1. By using an LPA1-specific inhibitor, AM095, we confirmed that LPA1 signaling but not LPA2 or LPA3 is involved in TGF-ß1 induced HTFs activation. Conclusions: Our results show that inhibition of LPA1 signaling presents potent antifibrotic effect in HTFs, which may serve as a promising intervention strategy for preventing subconjunctival fibrosis caused by glaucoma filtration surgery.


Asunto(s)
Fibroblastos/citología , Isoxazoles/farmacología , Lisofosfolípidos/metabolismo , Miofibroblastos/citología , Propionatos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Glaucoma/cirugía , Humanos , Isoxazoles/administración & dosificación , Propionatos/administración & dosificación , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
13.
Pharmacol Res ; 146: 104277, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31112749

RESUMEN

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.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Ojo/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ojo/irrigación sanguínea , Ojo/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transcriptoma/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Pharmacol Res ; 143: 33-39, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30851357

RESUMEN

Oxidative stress, due to insufficiency of antioxidants or over-production of oxidants, can lead to severe cell and tissue damage. Oxidative stress occurs constantly and has been shown to be involved in innumerable diseases, such as degenerative, cardiovascular, neurological, and metabolic disorders, cancer, and aging, thus highlighting the vital need of antioxidant defense mechanisms. Vascular endothelial growth factor B (VEGF-B) was discovered a long time ago, and is abundantly expressed in most types of cells and tissues. VEGF-B remained functionally mysterious for many years and later on has been shown to be minimally angiogenic. Recently, VEGF-B is reported to be a potent antioxidant by boosting the expression of key antioxidant enzymes. Thus, one major role of VEGF-B lies in safeguarding tissues and cells from oxidative stress-induced damage. VEGF-B may therefore have promising therapeutic utilities in treating oxidative stress-related diseases. In this review, we discuss the current knowledge on the newly discovered antioxidant function of VEGF-B and the related molecular mechanisms, particularly, in relationship to some oxidative stress-related diseases, such as retinitis pigmentosa, age-related macular degeneration, diabetic retinopathy, glaucoma, amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease.


Asunto(s)
Antioxidantes/uso terapéutico , Oftalmopatías/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Factor B de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Antioxidantes/farmacología , Oftalmopatías/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Factor B de Crecimiento Endotelial Vascular/metabolismo , Factor B de Crecimiento Endotelial Vascular/farmacología
15.
Mol Med Rep ; 19(1): 468-476, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30483803

RESUMEN

Subconjunctival fibrosis represents the primary cause of postoperative failure of trabeculectomy, and at present there is a lack of effective intervention strategies. The present study aimed to investigate the effect of the mitogen­activated protein kinase kinase (MEK) inhibitor U0126 on human tenon fibroblast (HTF) myofibrosis transdifferentiation, and to illuminate the underlying molecular mechanisms involved. It was demonstrated that U0126 significantly inhibited the proliferation, migration and collagen contraction of HTFs stimulated with TGF­ß1. In addition, U0126 largely attenuated the TGF­ß1­induced conversion of HTFs into myofibroblasts, as indicated by a downregulation of the mRNA and protein expression of α­smooth muscle actin and zinc finger protein SNAI1, and by ameliorating the 3D­collagen contraction response. Mechanistically, U0126 suppressed the TGF­ß1­stimulated phosphorylation of mothers against decapentaplegic homolog 2/3, P38 mitogen­activated protein kinase and extracellular signal­regulated kinase 1/2, indicating that U0126 may inhibit HTF activation through the canonical and non­canonical signaling pathways of TGF­ß1. Therefore, U0126 exhibits a potent anti­fibrotic effect among HTFs, and the inhibition of MEK signaling may serve as an alternative intervention strategy for the treatment of trabeculectomy­associated fibrosis.


Asunto(s)
Transdiferenciación Celular/fisiología , Fibroblastos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Miofibroblastos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Actinas/metabolismo , Butadienos/farmacología , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Humanos , Miofibroblastos/efectos de los fármacos , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Transcripción de la Familia Snail/metabolismo
16.
Proc Natl Acad Sci U S A ; 115(41): 10351-10356, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30249667

RESUMEN

VEGF-B was discovered a long time ago. However, unlike VEGF-A, whose function has been extensively studied, the function of VEGF-B and the mechanisms involved still remain poorly understood. Notwithstanding, drugs that inhibit VEGF-B and other VEGF family members have been used to treat patients with neovascular diseases. It is therefore critical to have a better understanding of VEGF-B function and the underlying mechanisms. Here, using comprehensive methods and models, we have identified VEGF-B as a potent antioxidant. Loss of Vegf-b by gene deletion leads to retinal degeneration in mice, and treatment with VEGF-B rescues retinal cells from death in a retinitis pigmentosa model. Mechanistically, we demonstrate that VEGF-B up-regulates numerous key antioxidative genes, particularly, Gpx1 Loss of Gpx1 activity largely diminished the antioxidative effect of VEGF-B, demonstrating that Gpx1 is at least one of the critical downstream effectors of VEGF-B. In addition, we found that the antioxidant function of VEGF-B is mediated mainly by VEGFR1. Given that oxidative stress is a crucial factor in numerous human diseases, VEGF-B may have therapeutic value for the treatment of such diseases.


Asunto(s)
Antioxidantes/metabolismo , Degeneración Retiniana/genética , Factor B de Crecimiento Endotelial Vascular/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glutatión Peroxidasa/genética , Ratones Endogámicos C57BL , Ratones Mutantes , Estrés Oxidativo , Retina/efectos de los fármacos , Retina/patología , Degeneración Retiniana/tratamiento farmacológico , Retinitis Pigmentosa/genética , Factor B de Crecimiento Endotelial Vascular/genética , Factor B de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Glutatión Peroxidasa GPX1
17.
Mol Vis ; 24: 789-800, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30636861

RESUMEN

Purpose: This study aimed to investigate the effect of nintedanib on the conversion of human Tenon's fibroblasts (HTFs) into myofibroblasts and reveal the molecular mechanisms involved. Methods: Primary cultured HTFs were incubated with transforming growth factor ß1 (TGF-ß1) alone or combined with nintedanib, and cell proliferation and migration were measured by cell counting kit-8 (CCK8) and the scratch wound assay, respectively. HTF contractility was evaluated with a 3D collagen contraction assay. The mRNA and protein levels of α smooth muscle actin (α-SMA) and Snail and the phosphorylation levels of Smad2/3, p38 mitogen-activated protein kinase (p38MAPK), and extracellular signal-regulated kinase ½ (ERK1/2) were determined by quantitative reverse transcription polymerase chain reaction (RT-PCR), western blot, and immunofluorescence staining. Results: Nintedanib inhibited the proliferation and migration of HTFs in a dose-dependent manner. Furthermore, nintedanib prevented HTF myofibroblast differentiation via downregulation of mRNA and protein expression of α-SMA and Snail. A three-dimensional (3D) collagen gel contraction assay demonstrated that nintedanib effectively inhibits myofibroblast contraction induced by TGF-ß1. Mechanistically, we revealed that nintedanib reduces the TGF-ß1-induced phosphorylation of Smad2/3, p38MAPK, and ERK1/2, suggesting that nintedanib acts through both classic and nonclassic signaling pathways of TGF-ß1 to prevent HTF activation. Conclusions: Our study provides new evidence that nintedanib has potent antifibrotic effects in HTFs and suggests that it may be used as a potential therapeutic agent for subconjunctival fibrosis.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Indoles/farmacología , Miofibroblastos/efectos de los fármacos , Actinas/antagonistas & inhibidores , Actinas/genética , Actinas/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miofibroblastos/citología , Miofibroblastos/metabolismo , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Cápsula de Tenon/citología , Cápsula de Tenon/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
PLoS One ; 12(9): e0185287, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28949992

RESUMEN

PURPOSE: To analyze the validity of the Hospital Anxiety and Depression Scale (HADS) among Chinese cataract population. METHODS: A total of 275 participants with unilateral or bilateral cataract were recruited to complete the Chinese version of HADS. The patients' demographic and ophthalmic characteristics were documented. Rasch analysis was conducted to examine the model fit statistics, the thresholds ordering of the polytomous items, targeting, person separation index and reliability, local dependency, unidimentionality, differential item functioning (DIF) and construct validity of the HADS individual and summary measures. RESULTS: Rasch analysis was performed on anxiety and depression subscales as well as HADS-Total score respectively. The items of original HADS-Anxiety, HADS-Depression and HADS-Total demonstrated evidence of misfit of the Rasch model. Removing items A7 for anxiety subscale and rescoring items D14 for depression subscale significantly improved Rasch model fit. A 12-item higher order total scale with further removal of D12 was found to fit the Rasch model. The modified items had ordered response thresholds. No uniform DIF was detected, whereas notable non-uniform DIF in high-ability group was found. The revised cut-off points were given for the modified anxiety and depression subscales. CONCLUSION: The modified version of HADS with HADS-A and HADS-D as subscale and HADS-T as a higher-order measure is a reliable and valid instrument that may be useful for assessing anxiety and depression states in Chinese cataract population.


Asunto(s)
Ansiedad/psicología , Catarata/psicología , Depresión/psicología , Anciano , China , Femenino , Humanos , Masculino
19.
Proc Natl Acad Sci U S A ; 114(40): 10737-10742, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28923916

RESUMEN

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.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Caveolina 1/fisiología , Neovascularización Coroidal/prevención & control , MAP Quinasa Quinasa 4/metabolismo , Fragmentos de Péptidos/farmacología , Neovascularización Retiniana/prevención & control , Animales , Caveolina 1/farmacología , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Quimioterapia Combinada , Humanos , Ratones Noqueados , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
20.
Curr Eye Res ; 42(8): 1124-1129, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28494163

RESUMEN

PURPOSE: 14-3-3 zeta protein plays a potential protective role in neurodegenerative disease. Given that glaucoma and neurodegenerative diseases share a similar pathogenesis, it is possible that 14-3-3 zeta may have a similar protective effect in the glaucomatous process. In the present study, we measured the expression of 14-3-3 zeta in vivo (mouse eyes) and in vitro in a transformed human trabecular meshwork (HTM) cell line, TM-1, and assessed the possible roles of this protein in dexamethasone (DEX)-treated eyes and HTM cells. METHODS: Mouse eyes were randomly treated with 0.1% dexamethasone (DEX) eye drops or phosphate-buffered solution (PBS) for 28 days. The expression and distribution of 14-3-3 zeta protein in mouse eyes were examined using immunofluorescence. TM-1 cells were treated with DEX (10-6 or 10-7 M) or PBS for 1, 4, or 7 days, and the mRNA and protein expression of 14-3-3 zeta were detected by real-time RT-PCR and Western blotting. RESULTS: 14-3-3 zeta protein was highly expressed in the mouse cornea, trabecular meshwork (TM), and ciliary body. Intraocular pressure (IOP) was significantly elevated, whereas the 14-3-3 zeta expression was significantly decreased in mouse TM after 0.1% DEX treatment for 28 days. In vitro, treatment with 10-7 M DEX mildly increased 14-3-3 zeta mRNA and protein expression (p > 0.05), whereas 10-6 M DEX significantly decreased expression of 14-3-3 zeta mRNA and protein (p < 0.05) compared to the control (Ctrl) group at the seventh day. CONCLUSIONS: DEX can increase IOP in mouse eyes and concurrently downregulate 14-3-3 zeta protein expression in mouse TM. The effects of DEX on 14-3-3 zeta expression in vitro were both dose- and time-related. Our results suggest that alterations in 14-3-3 zeta protein may be implicated in DEX-induced pathological elevated IOP.


Asunto(s)
Proteínas 14-3-3/genética , Dexametasona/toxicidad , Regulación de la Expresión Génica , Glaucoma/genética , Presión Intraocular/efectos de los fármacos , ARN/genética , Malla Trabecular/metabolismo , Proteínas 14-3-3/biosíntesis , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Glaucoma/inducido químicamente , Glaucoma/metabolismo , Glucocorticoides/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena en Tiempo Real de la Polimerasa , Malla Trabecular/efectos de los fármacos , Malla Trabecular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...