Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cells ; 13(2)2024 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-38247805

RESUMEN

Low-dose aspirin (LDA) is efficacious in preventing preeclampsia, but its mechanism of action is unclear. Conflicting evidence suggests that it may inhibit placental trophoblast release of soluble fms-like tyrosine kinase-1 (sFlt1), a key mediator of preeclampsia. We examined whether, and at what concentrations, aspirin and its principal metabolite, salicylic acid, modulate sFlt1 release and/or expression in trophoblasts. Human trophoblast lines BeWo and HTR-8/SVneo were cultured; BeWo cells were also treated with 1% oxygen vs. normoxia to mimic hypoxia in preeclamptic placentas. Cells were treated with aspirin or salicylic acid vs. vehicle for 24 h at concentrations relevant to LDA and at higher concentrations. Protein concentrations (ELISA) and mRNA expression (RT-PCR) of sFlt1 were determined. Under normoxia, LDA-relevant concentrations of aspirin (10-50 µmol/L) or salicylic acid (20-100 µmol/L) had no significant effect on sFlt1 protein release or mRNA expression in BeWo cells. However, inhibition was observed at higher concentrations (1 mmol/L for aspirin and ≥200 µmol/L for salicylic acid). Hypoxia enhanced sFlt1 protein release and mRNA expression in BeWo cells, but these responses were not significantly affected by either aspirin or salicylic acid at LDA concentrations. Similarly, neither drug altered sFlt1 protein secretion or mRNA expression in normoxic HTR-8/SVneo cells at LDA concentrations. We suggest that direct modulation of trophoblast release or expression of sFlt1 is unlikely to be a mechanism underlying the clinical efficacy of LDA in preeclampsia.


Asunto(s)
Aspirina , Preeclampsia , Trofoblastos , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Femenino , Humanos , Embarazo , Aspirina/farmacología , Hipoxia , Placenta , Preeclampsia/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras , ARN Mensajero/genética , Ácido Salicílico/farmacología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
2.
FASEB Bioadv ; 3(1): 23-35, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33521587

RESUMEN

Preeclampsia remains a challenge without an effective therapy. Evidence supports targetability of soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng), which are released excessively from the placenta under ischemic and hypoxic stresses. We compared four trophoblast cell lines, BeWo, Jar, Jeg-3, and HTR-8/SVneo, in order to identify a suitable model for drug screening. Cultured trophoblasts were exposed to 1% oxygen vs. normoxia for 24-48 hr; human umbilical vein and aortic endothelial cells were included for comparison. Supernatant sFlt-1 and sEng concentrations were measured by ELISA, and sFlt-1 mRNA expression determined by RT-PCR. Cellular responses to experimental therapeutics were explored. All four trophoblast lines secreted sEng, which did not increase by hypoxia. BeWo, Jar, and Jeg-3 exhibited significantly enhanced expression of sFlt-1 i13 and e15a mRNA in response to hypoxia; however, only BeWo released a detectable level of sFlt-1 protein, which was doubled by hypoxia. In contrast, hypoxia decreased sFlt-1 mRNA expression and protein release in HTR-8/SVneo, similarly to endothelial cells. The cellular mechanism involved HIFα. BeWo responded to representative agents similarly to human primary placental tissues in the literature. These data support that the BeWo-hypoxia model mimics a key pathogenic mechanism of preeclampsia and has potential value for translational drug discovery.

3.
Artículo en Inglés | MEDLINE | ID: mdl-33504507

RESUMEN

INTRODUCTION: Pre-eclampsia (PE) is increased ~4-fold by maternal diabetes. Elevated plasma antiangiogenic factors, soluble fms-like tyrosine kinase (sFLT-1) and soluble endoglin (sENG), precede PE onset. We investigated whether diabetes-related stresses, modified lipoproteins and elevated glucose enhance trophoblast sFLT-1 and sENG release and/or alter placental barrier function and whether oxidized low-density lipoprotein (Ox-LDL) is in placental tissue. RESEARCH DESIGN AND METHODS: HTR8/SVneo cells were exposed to 'heavily-oxidized, glycated' LDL (HOG-LDL) versus native LDL (N-LDL) (10-200 mg protein/L) for 24 hours ±pretreatment with glucose (30 mmol/L, 72 hours). Concentrations of sFLT-1 and sENG in supernatants (by ELISA) and expressions of sFLT-1-I13 and sFLT-1-E15A isoforms, endoglin (ENG) and matrix metalloproteinase-14 (MMP-14; by RT-PCR) were quantified. For barrier studies, JAR cells were cultured in Transwell plates (12-14 days), then exposed to LDL. Transepithelial electrical resistance (TEER) was measured after 6, 12 and 24 hours. In placental sections from women with and without type 1 diabetes, immunostaining of apolipoprotein B100 (ApoB, a marker of LDL), Ox-LDL and lipoxidation product 4-hydroxynonenal was performed. RESULTS: HOG-LDL (50 mg/L) increased sFLT-1 (2.7-fold, p<0.01) and sENG (6.4-fold, p<0.001) in supernatants versus N-LDL. HOG-LDL increased expression of sFLT-1-I13 (twofold, p<0.05), sFLT-1-E15A (1.9-fold, p<0.05), ENG (1.6-fold, p<0.01) and MMP-14 (1.8-fold, p<0.05) versus N-LDL. High glucose did not by itself alter sFLT-1 or sENG concentrations, but potentiated effects of HOG-LDL on sFLT-1 by 1.5-fold (p<0.05) and on sENG by 1.8-fold (p<0.01). HOG-LDL (200 mg/L) induced trophoblast barrier impairment, decreasing TEER at 6 hours (p<0.01), 12 hours (p<0.01) and 24 hours (p<0.05) versus N-LDL. Immunostaining of term placental samples from women both with and without diabetes revealed presence of intravillous modified lipoproteins. CONCLUSION: These findings may explain, in part, the high risk for PE in women with diabetes. The trophoblast culture model has potential for evaluating novel therapies targeting barrier dysfunction.


Asunto(s)
Diabetes Mellitus , Preeclampsia , Femenino , Humanos , Lipoproteínas , Placenta , Embarazo , Trofoblastos , Receptor 1 de Factores de Crecimiento Endotelial Vascular
4.
Prog Retin Eye Res ; 69: 159-172, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30352305

RESUMEN

The retina is an immune privileged tissue, which is protected from external and internal insults by its blood-retina barriers and immune suppressive microenvironment. Apart from the avoidance and tolerance strategies, the retina is also protected by its own defense system, i.e., microglia and the complement system. The immune privilege and defense mechanisms work together to maintain retinal homeostasis. During aging, the retina is at an increased risk of developing various degenerative diseases such as age-related macular degeneration, diabetic retinopathy, and glaucomatous retinopathy. Previously, we have shown that aging induces a para-inflammatory response in the retina. In this review, we explore the impact of aging on retinal immune regulation and the connection between homeostatic control of retinal immune privilege and para-inflammation under aging conditions and present a view that may explain why aging puts the retina at risk of developing degenerative diseases.


Asunto(s)
Envejecimiento , Privilegio Inmunológico/fisiología , Retina , Enfermedades de la Retina , Envejecimiento/inmunología , Envejecimiento/fisiología , Animales , Barrera Hematorretinal/fisiología , Células Epiteliales/fisiología , Humanos , Inflamación/inmunología , Inflamación/fisiopatología , Microglía/fisiología , Neuronas/fisiología , Retina/inmunología , Retina/fisiología , Enfermedades de la Retina/inmunología , Enfermedades de la Retina/fisiopatología , Epitelio Pigmentado de la Retina/fisiología
5.
Aging (Albany NY) ; 10(6): 1380-1389, 2018 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-29905533

RESUMEN

Age-related para-inflammation in the retina-choroidal interface is featured by low-levels of complement activation and subretinal macrophage accumulation. This study aimed to understand how complement expression in macrophages is regulated by retinal pigment epithelium (RPE). Bone marrow-derived macrophages (BMDMs) and RPE cells were cultured from 8-10 weeks old C57BL/6J mice. The BMDMs were co-cultured with normal RPE, or oxidized photoreceptor outer segment (oxPOS) or TNF-α pre-treated RPE, or apoptotic RPE, or RPE-choroid eyecups. Macrophages were then isolated and processed for real-time RT-PCR. The expression of complement inhibitor C1INH in BMDMs was significantly upregulated by RPE and RPE-choroid eyecups. The eyecups also upregulated CFH, CD59a, and Crry in BMDMs. oxPOS pre-treated RPE upregulated C1qb but down-regulated C3 expression in BMDMs. TNF-α pre-treated RPE enhanced C1INH and CFB expression. When BMDMs were treated with apoptotic RPE, the expression of C1qb, CFH, and CD59a was reduced, whereas the expression of C3, CFB and C1INH was increased. Our results suggest that RPE can modulate macrophages complement expression at the retina-choroidal interface even under aging or oxidative conditions. However, during inflammation, they may promote the alternative pathway of complement activation through down-regulating CFH and CD59a and upregulating CFB and C3.


Asunto(s)
Proteína Inhibidora del Complemento C1/metabolismo , Células Epiteliales/metabolismo , Macrófagos/metabolismo , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Epitelio Pigmentado de la Retina/citología , Animales , Bovinos , Células Cultivadas , Técnicas de Cocultivo , Ratones , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
6.
Am J Pathol ; 188(4): 1007-1020, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29452101

RESUMEN

The suppressor of cytokine signaling protein 3 (SOCS3) critically controls immune cell activation, although its role in macrophage polarization and function remains controversial. Using experimental autoimmune uveoretinitis (EAU) as a model, we show that inflammation-mediated retinal degeneration is exaggerated and retinal angiogenesis is accelerated in mice with SOCS3 deficiency in myeloid cells (LysMCre/+SOCS3fl/fl). At the acute stage of EAU, the population of infiltrating neutrophils was increased and the population of macrophages decreased in LysMCre/+SOCS3fl/fl mice compared with that in wild-type (WT) mice. Real-time RT-PCR showed that the expression of tumor necrosis factor-α, IL-1ß, interferon-γ, granulocyte-macrophage colony-stimulating factor, and arginase-1 was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina in contrast to the WT EAU retina. The percentage of arginase-1+ infiltrating cells was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina than that in the WT EAU retina. In addition, bone marrow-derived macrophages and neutrophils from the LysMCre/+SOCS3fl/fl mice express significantly higher levels of chemokine (C-C motif) ligand 2 and arginase-1 compared with those from WT mice. Inhibition of arginase using an l-arginine analog amino-2-borono-6-hexanoic suppressed inflammation-induced retinal angiogenesis without affecting the severity of inflammation. Our results suggest that SOCS3 critically controls the phenotype and function of macrophages and neutrophils under inflammatory conditions and loss of SOCS3 promotes the angiogenic phenotype of the cells through up-regulation of arginase-1.


Asunto(s)
Arginasa/genética , Enfermedades Autoinmunes/genética , Células Mieloides/metabolismo , Neovascularización Patológica/metabolismo , Degeneración Retiniana/patología , Proteína 3 Supresora de la Señalización de Citocinas/deficiencia , Regulación hacia Arriba/genética , Enfermedades de la Úvea/genética , Animales , Arginasa/antagonistas & inhibidores , Arginasa/metabolismo , Células de la Médula Ósea/metabolismo , Inflamación/patología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
7.
Invest Ophthalmol Vis Sci ; 55(11): 7572-82, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25342612

RESUMEN

PURPOSE: To investigate the roles of the CCL2-CCR2 and CX3CL1-CX3CR1 pathways in experimental autoimmune uveoretinitis (EAU)-mediated retinal tissue damage and angiogenesis. METHODS: The C57BL/6J wild-type (WT) and CCL2(-/-)CX3CR1(gfp/gfp) (double knockout [DKO]) mice were immunized with IRBP1₋20. Retinal inflammation and tissue damage were evaluated clinically and histologically at different days postimmunization (p.i.). Retinal neovascular membranes were evaluated by confocal microscopy of retinal flat mounts, and immune cell infiltration by flow cytometry. RESULTS: At day 25 p.i., DKO mice had lower clinical and histological scores and fewer CD45(high)CD11b(+) infiltrating cells compared with WT mice. The F4/80(+) macrophages constitute 40% and 21% and CD11b(+)Gr-1(+)Ly6G(+) neutrophils constitute 10% and 22% of retinal infiltrating cells in WT and DKO mice, respectively. At the late stages of EAU (day 60-90 p.i.), DKO and WT mice had similar levels of inflammatory score. However, less structural damage and reduced angiogenesis were detected in DKO mice. Neutrophils were rarely detected in the inflamed retina in both WT and DKO mice. Macrophages and myeloid-derived suppressor cells (MDSCs) accounted for 8% and 3% in DKO EAU retina, and 19% and 10% in WT EAU retina; 71% of infiltrating cells were T/B-lymphocytes in DKO EAU retina and 50% in WT EAU retina. CONCLUSIONS: Experimental autoimmune uveoretinitis-mediated retinal tissue damage and angiogenesis is reduced in CCL2(-/-)CX3CR1(gfp/gfp) mice. Retinal inflammation is dominated by neutrophils at the acute stage and lymphocytes at the chronic stage in these mice. Our results suggest that CCR2(+) and CX3CR1(+) monocytes are both involved in tissue damage and angiogenesis in EAU.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Quimiocinas/metabolismo , Retina/patología , Retinitis/inmunología , Uveítis/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Quimiocinas/genética , Quimiocinas/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/inmunología , Retina/metabolismo , Retinitis/genética , Retinitis/patología , Uveítis/genética , Uveítis/patología
8.
Exp Eye Res ; 112: 93-101, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23644095

RESUMEN

Complement activation is involved in a variety of retinal diseases. We have shown previously that a number of complement components and regulators can be produced locally in the eye, and that retinal pigment epithelial (RPE) cells are the major source of complement expression at the retina-choroidal interface. The expression of complement components by RPE cells is regulated by inflammatory cytokines. Under aging or inflammatory conditions, microglia and macrophages accumulate in the subretinal space, where they are in close contact with RPE cells. In this study, we investigated the effect of activated macrophages on complement expression by RPE cells. Mouse RPE cells were treated with the supernatants from un-activated bone marrow-derived macrophages (BM-DMs), the classically activated BM-DMs (M1) and different types of the alternatively activated BM-DMs (M2a by IL-4, M2b by immune complex and lipopolysaccharide (LPS), M2c by IL-10). The expression of inflammatory cytokines and complement genes by RPE cells were determined by real-time RT-PCR. The protein expression of CFB, C3, C1INH, and C1r was examined by Western blot. Our results show that un-stimulated RPE cells express a variety of complement-related genes, and that the expression levels of complement regulators, including C1r, factor H (CFH), DAF1, CD59, C1INH, Crry, and C4BP genes are significantly higher than those of complement component genes (C2, C4, CFB, C3, and C5). Macrophage supernatants increased inflammatory cytokine (IL-1ß, IL-6, iNOS), chemokine (CCL2) and complement expression in RPE cells. The supernatants from M0, M2a and M2c macrophages mildly up-regulated (2-3.5-fold) CFB, CFH and C3 gene expression in RPE cells, whereas the supernatants from M1 and M2b macrophages massively increased (10-30-fold) CFB and C3 gene expression in RPE cells. The expression of other genes, including C1r, C2, C4, CFH, Masp1, C1INH, and C4BP in RPE cells was also increased by the supernatants of M1 and M2b macrophages; however, the increment levels were significantly lower than CFB and C3 genes. M1 and M2b macrophage supernatants enhanced CFB (Bb fragment) protein expression and C3 secretion by RPE cells. M1 macrophages may affect complement expression in RPE cells through the STAT1 pathway. Our results suggest that under inflammatory conditions, activated macrophages could promote the alternative pathway of complement activation in the retina via induction of RPE cell CFB and C3 expression.


Asunto(s)
Proteínas del Sistema Complemento/genética , Regulación de la Expresión Génica/fisiología , Activación de Macrófagos/fisiología , Macrófagos/fisiología , Epitelio Pigmentado de la Retina/metabolismo , Animales , Western Blotting , Células Cultivadas , Activación de Complemento/fisiología , Complemento C3/metabolismo , Factor B del Complemento/metabolismo , Vía Alternativa del Complemento/fisiología , Proteínas del Sistema Complemento/metabolismo , Medios de Cultivo Condicionados/farmacología , Citocinas/farmacología , Ensayo de Inmunoadsorción Enzimática , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT1/metabolismo
9.
PLoS One ; 8(4): e61381, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637822

RESUMEN

Previous studies have shown that CCL2/CX3CR1 deficient mice on C57BL/6N background (with rd8 mutation) have an early onset (6 weeks) of spontaneous retinal degeneration. In this study, we generated CCL2(-/-)CX3CR1(GFP/GFP) mice on the C57BL/6J background. Retinal degeneration was not detected in CCL2(-/-)CX3CR1(GFP/GFP) mice younger than 6 months. Patches of whitish/yellowish fundus lesions were observed in 17∼60% of 12-month, and 30∼100% of 18-month CCL2(-/-)CX3CR1(GFP/GFP) mice. Fluorescein angiography revealed no choroidal neovascularisation in these mice. Patches of retinal pigment epithelium (RPE) and photoreceptor damage were detected in 30% and 50% of 12- and 18-month CCL2(-/-)CX3CR1(GFP/GFP) mice respectively, but not in wild-type mice. All CCL2(-/-)CX3CR1(GFP/GFP) mice exposed to extra-light (∼800lux, 6 h/day, 6 months) developed patches of retinal atrophy, and only 20-25% of WT mice which underwent the same light treatment developed atrophic lesions. In addition, synaptophysin expression was detected in the outer nucler layer (ONL) of area related to photoreceptor loss in CCL2(-/-)CX3CR1(GFP/GFP) mice. Markedly increased rhodopsin but reduced cone arrestin expression was observed in retinal outer layers in aged CCL2(-/-)CX3CR1(GFP/GFP) mice. GABA expression was reduced in the inner retina of aged CCL2(-/-)CX3CR1(GFP/GFP) mice. Significantly increased Müller glial and microglial activation was observed in CCL2(-/-)CX3CR1(GFP/GFP) mice compared to age-matched WT mice. Macrophages from CCL2(-/-)CX3CR1(GFP/GFP) mice were less phagocytic, but expressed higher levels of iNOS, IL-1ß, IL-12 and TNF-α under hypoxia conditions. Our results suggest that the deletions of CCL2 and CX3CR1 predispose mice to age- and light-mediated retinal damage. The CCL2/CX3CR1 deficient mouse may thus serve as a model for age-related atrophic degeneration of the RPE, including the dry type of macular degeneration, geographic atrophy.


Asunto(s)
Quimiocina CCL2/deficiencia , Degeneración Macular/patología , Receptores de Citocinas/deficiencia , Receptores del VIH/deficiencia , Retina/fisiología , Degeneración Retiniana/patología , Animales , Receptor 1 de Quimiocinas CX3C , Quimiocina CCL2/genética , Proteínas Fluorescentes Verdes/metabolismo , Luz , Macrófagos/fisiología , Degeneración Macular/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/fisiología , Receptores de Citocinas/genética , Receptores del VIH/genética , Retina/patología
10.
Glia ; 60(5): 833-42, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22362506

RESUMEN

Previous studies have shown that following whole-body irradiation bone marrow (BM)-derived cells can migrate into the central nervous system, including the retina, to give rise to microglia-like cells. The detailed mechanism, however, remains elusive. We show in this study that a single-dose whole-body γ-ray irradiation (8 Gy) induced subclinical damage (i.e., DNA damage) in the neuronal retina, which is accompanied by a low-grade chronic inflammation, para-inflammation, characterized by upregulated expression of chemokines (CCL2, CXCL12, and CX3CL1) and complement components (C4 and CFH), and microglial activation. The upregulation of chemokines CCL2 and CXCL12 and complement C4 lasted for more than 160 days, whereas the expression of CX3CL1 and CFH was upregulated for 2 weeks. Both resident microglia and BM-derived phagocytes displayed mild activation in the neuronal retina following irradiation. When BM cells from CX3CR1(gfp/+) mice or CX3CR1(gfp/gfp) mice were transplanted to wild-type C57BL/6 mice, more than 90% of resident CD11b(+) cells were replaced by donor-derived GFP(+) cells after 6 months. However, when transplanting CX3CR1(gfp/+) BM cells into CCL2-deficient mice, only 20% of retinal CD11b(+) cells were replaced by donor-derived cells at 6 month. Our results suggest that the neuronal retina suffers from a chronic stress following whole-body irradiation, and a para-inflammatory response is initiated, presumably to rectify the insults and maintain homeostasis. The recruitment of BM-derived myeloid cells is a part of the para-inflammatory response and is CCL2 but not CX3CL1 dependent.


Asunto(s)
Células de la Médula Ósea/metabolismo , Quimiocina CCL2/fisiología , Mediadores de Inflamación/fisiología , Células Mieloides/metabolismo , Retina/metabolismo , Irradiación Corporal Total/efectos adversos , Animales , Células de la Médula Ósea/patología , Células de la Médula Ósea/efectos de la radiación , Quimiocina CCL2/efectos de la radiación , Quimiocina CXCL1/fisiología , Mediadores de Inflamación/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/patología , Células Mieloides/efectos de la radiación , Retina/patología , Retina/efectos de la radiación
11.
Am J Pathol ; 180(1): 235-45, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22067906

RESUMEN

Neovascular retinal disease is a leading cause of blindness orchestrated by inflammatory responses. Although noninfectious uveoretinitis is mediated by CD4(+) T cells, in the persistent phase of disease, angiogenic responses are observed, along with degeneration of the retina. Full clinical manifestation relies on myeloid-derived cells, which are phenotypically distinct from, but potentially sharing common effector responses to age-related macular degeneration. To interrogate inflammation-mediated angiogenesis, we investigated experimental autoimmune uveoretinitis, an animal model for human uveitis. After the initial acute phase of severe inflammation, the retina sustains a persistent low-grade inflammation with tissue-infiltrating leukocytes for over 4 months. During this persistent phase, angiogenesis is observed as retinal neovascular membranes that arise from inflamed venules and postcapillary venules, increase in size as the disease progresses, and are associated with infiltrating arginase-1(+) macrophages. In the absence of thrombospondin-1, retinal neovascular membranes are markedly increased and are associated with arginase-1(-) CD68(+) macrophages, whereas deletion of the chemokine receptor CCR2 resulted in reduced retinal neovascular membranes in association with a predominant neutrophil infiltrate. CCR2 is important for macrophage recruitment to the retina in experimental autoimmune uveoretinitis and promotes chronicity in the form of a persistent angiogenesis response, which in turn is regulated by constitutive expression of angiogenic inhibitors like thrombospondin-1. This model offers a new platform to dissect the molecular and cellular pathology of inflammation-induced ocular angiogenesis.


Asunto(s)
Receptores CCR2/fisiología , Neovascularización Retiniana/etiología , Retinitis/etiología , Trombospondina 1/fisiología , Uveítis/etiología , Animales , Citocinas/biosíntesis , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/deficiencia , Receptores CCR2/metabolismo , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Retinitis/metabolismo , Retinitis/patología , Trombospondina 1/deficiencia , Uveítis/metabolismo , Uveítis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA