Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lucy Evans is active.

Publication


Featured researches published by Lucy Evans.


Nature Medicine | 2016

Retinal lipid and glucose metabolism dictates angiogenesis through the lipid sensor Ffar1

Jean-Sebastien Joyal; Ye Sun; Marin L. Gantner; Zhuo Shao; Lucy Evans; Nicholas Saba; Thomas Fredrick; Samuel Burnim; Jin Sung Kim; Gauri Patel; Aimee M. Juan; Christian G. Hurst; Colman J. Hatton; Zhenghao Cui; Kerry A. Pierce; Patrick Bherer; Edith Aguilar; Michael B. Powner; Kristis Vevis; Michel Boisvert; Zhongjie Fu; Emile Levy; Marcus Fruttiger; Alan Packard; Flavio Rezende; Bruno Maranda; Przemyslaw Sapieha; Jing Chen; Martin Friedlander; Clary B. Clish

Tissues with high metabolic rates often use lipids, as well as glucose, for energy, conferring a survival advantage during feast and famine. Current dogma suggests that high-energy–consuming photoreceptors depend on glucose. Here we show that the retina also uses fatty acid β-oxidation for energy. Moreover, we identify a lipid sensor, free fatty acid receptor 1 (Ffar1), that curbs glucose uptake when fatty acids are available. Very-low-density lipoprotein receptor (Vldlr), which is present in photoreceptors and is expressed in other tissues with a high metabolic rate, facilitates the uptake of triglyceride-derived fatty acid. In the retinas of Vldlr−/− mice with low fatty acid uptake but high circulating lipid levels, we found that Ffar1 suppresses expression of the glucose transporter Glut1. Impaired glucose entry into photoreceptors results in a dual (lipid and glucose) fuel shortage and a reduction in the levels of the Krebs cycle intermediate α-ketoglutarate (α-KG). Low α-KG levels promotes stabilization of hypoxia-induced factor 1a (Hif1a) and secretion of vascular endothelial growth factor A (Vegfa) by starved Vldlr−/− photoreceptors, leading to neovascularization. The aberrant vessels in the Vldlr−/− retinas, which invade normally avascular photoreceptors, are reminiscent of the vascular defects in retinal angiomatous proliferation, a subset of neovascular age-related macular degeneration (AMD), which is associated with high vitreous VEGFA levels in humans. Dysregulated lipid and glucose photoreceptor energy metabolism may therefore be a driving force in macular telangiectasia, neovascular AMD and other retinal diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Endothelial microRNA-150 is an intrinsic suppressor of pathologic ocular neovascularization

Chi-Hsiu Liu; Ye Sun; Jie Li; Yan Gong; Katherine Tian; Lucy Evans; Peyton Morss; Thomas Fredrick; Nicholas Saba; Jing Chen

Significance Pathologic vascular growth causes vision impairment in several vascular eye diseases. Specifically targeting molecular signatures distinguishing pathologic neovascularization from normal vessels will allow targeted treatment options. This study demonstrates that miR-150 was specifically enriched in normal retinal vessels and down-regulated in pathologic neovessels in a mouse model of proliferative retinopathy. MiR-150 suppressed pathologic ocular neovascularization in mice with decreased expression of angiogenic target genes and inhibited endothelial cell function in vitro. Loss of miR-150 also promoted vascular sprouting in ex vivo aortic and choroidal assays and laser-induced choroidal neovascularization in mice. These data suggest that endothelial miR-150 is an endogenous suppressor of ocular neovascularization and a drug target for vascular eye diseases. Pathologic ocular neovascularization commonly causes blindness. It is critical to identify the factors altered in pathologically proliferating versus normally quiescent vessels to develop effective targeted therapeutics. MicroRNAs regulate both physiological and pathological angiogenesis through modulating expression of gene targets at the posttranscriptional level. However, it is not completely understood if specific microRNAs are altered in pathologic ocular blood vessels, influencing vascular eye diseases. Here we investigated the potential role of a specific microRNA, miR-150, in regulating ocular neovascularization. We found that miR-150 was highly expressed in normal quiescent retinal blood vessels and significantly suppressed in pathologic neovessels in a mouse model of oxygen-induced proliferative retinopathy. MiR-150 substantially decreased endothelial cell function including cell proliferation, migration, and tubular formation and specifically suppressed the expression of multiple angiogenic regulators, CXCR4, DLL4, and FZD4, in endothelial cells. Intravitreal injection of miR-150 mimic significantly decreased pathologic retinal neovascularization in vivo in both wild-type and miR-150 knockout mice. Loss of miR-150 significantly promoted angiogenesis in aortic rings and choroidal explants ex vivo and laser-induced choroidal neovascularization in vivo. In conclusion, miR-150 is specifically enriched in quiescent normal vessels and functions as an endothelium-specific endogenous inhibitor of pathologic ocular neovascularization.


The American Journal of Clinical Nutrition | 2015

Dietary ω-3 polyunsaturated fatty acids decrease retinal neovascularization by adipose–endoplasmic reticulum stress reduction to increase adiponectin

Zhongjie Fu; Chatarina Löfqvist; Zhuo Shao; Ye Sun; Jean-Sebastien Joyal; Christian G. Hurst; Ricky Zhenghao Cui; Lucy Evans; Katherine Tian; John Paul SanGiovanni; Jing Chen; David Ley; Ingrid Hansen Pupp; Ann Hellström; Lois E. H. Smith

BACKGROUND Retinopathy of prematurity (ROP) is a vision-threatening disease in premature infants. Serum adiponectin (APN) concentrations positively correlate with postnatal growth and gestational age, important risk factors for ROP development. Dietary ω-3 (n-3) long-chain polyunsaturated fatty acids (ω-3 LCPUFAs) suppress ROP and oxygen-induced retinopathy (OIR) in a mouse model of human ROP, but the mechanism is not fully understood. OBJECTIVE We examined the role of APN in ROP development and whether circulating APN concentrations are increased by dietary ω-3 LCPUFAs to mediate the protective effect in ROP. DESIGN Serum APN concentrations were correlated with ROP development and serum ω-3 LCPUFA concentrations in preterm infants. Mouse OIR was then used to determine whether ω-3 LCPUFA supplementation increases serum APN concentrations, which then suppress retinopathy. RESULTS We found that in preterm infants, low serum APN concentrations positively correlate with ROP, and serum APN concentrations positively correlate with serum ω-3 LCPUFA concentrations. In mouse OIR, serum total APN and bioactive high-molecular-weight APN concentrations are increased by ω-3 LCPUFA feed. White adipose tissue, where APN is produced and assembled in the endoplasmic reticulum, is the major source of serum APN. In mouse OIR, adipose endoplasmic reticulum stress is increased, and APN production is suppressed. ω-3 LCPUFA feed in mice increases APN production by reducing adipose endoplasmic reticulum stress markers. Dietary ω-3 LCPUFA suppression of neovascularization is reduced from 70% to 10% with APN deficiency. APN receptors localize in the retina, particularly to pathologic neovessels. CONCLUSION Our findings suggest that increasing APN by ω-3 LCPUFA supplementation in total parental nutrition for preterm infants may suppress ROP.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Cytochrome P450 2C8 ω3-Long-Chain Polyunsaturated Fatty Acid Metabolites Increase Mouse Retinal Pathologic Neovascularization—Brief Report

Zhuo Shao; Zhongjie Fu; Andreas Stahl; Jean-Sebastien Joyal; Colman J. Hatton; Aimee Juan; Christian G. Hurst; Lucy Evans; Zhenghao Cui; Dorothy T. Pei; Yan Gong; Dan Xu; Katherine Tian; Hannah H Bogardus; Matthew L. Edin; Fred B. Lih; Przemyslaw Sapieha; Jing Chen; Dipak Panigrahy; Ann Hellström; Darryl C. Zeldin; Lois E. H. Smith

Objective— Regulation of angiogenesis is critical for many diseases. Specifically, pathological retinal neovascularization, a major cause of blindness, is suppressed with dietary &ohgr;3-long-chain polyunsaturated fatty acids (&ohgr;3LCPUFAs) through antiangiogenic metabolites of cyclooxygenase and lipoxygenase. Cytochrome P450 epoxygenases (CYP2C8) also metabolize LCPUFAs, producing bioactive epoxides, which are inactivated by soluble epoxide hydrolase (sEH) to transdihydrodiols. The effect of these enzymes and their metabolites on neovascularization is unknown. Approach and Results— The mouse model of oxygen-induced retinopathy was used to investigate retinal neovascularization. We found that CYP2C (localized in wild-type monocytes/macrophages) is upregulated in oxygen-induced retinopathy, whereas sEH is suppressed, resulting in an increased retinal epoxide:diol ratio. With a &ohgr;3LCPUFA-enriched diet, retinal neovascularization increases in Tie2-driven human-CYP2C8–overexpressing mice (Tie2-CYP2C8-Tg), associated with increased plasma 19,20-epoxydocosapentaenoic acid and retinal epoxide:diol ratio. 19,20-Epoxydocosapentaenoic acids and the epoxide:diol ratio are decreased with overexpression of sEH (Tie2-sEH-Tg). Overexpression of CYP2C8 or sEH in mice does not change normal retinal vascular development compared with their wild-type littermate controls. The proangiogenic role in retina of CYP2C8 with both &ohgr;3LCPUFA and &ohgr;6LCPUFA and antiangiogenic role of sEH in &ohgr;3LCPUFA metabolism were corroborated in aortic ring assays. Conclusions— Our results suggest that CYP2C &ohgr;3LCPUFA metabolites promote retinal pathological angiogenesis. CYP2C8 is part of a novel lipid metabolic pathway influencing retinal neovascularization.


PLOS ONE | 2013

Choroid Sprouting Assay: An Ex Vivo Model of Microvascular Angiogenesis

Zhuo Shao; Mollie Friedlander; Christian G. Hurst; Zhenghao Cui; Dorothy T. Pei; Lucy Evans; Aimee M. Juan; Houda Tahir; François Duhamel; Jing Chen; Przemyslaw Sapieha; Sylvain Chemtob; Jean-Sebastien Joyal; Lois E. H. Smith

Angiogenesis of the microvasculature is central to the etiology of many diseases including proliferative retinopathy, age-related macular degeneration and cancer. A mouse model of microvascular angiogenesis would be very valuable and enable access to a wide range of genetically manipulated tissues that closely approximate small blood vessel growth in vivo. Vascular endothelial cells cultured in vitro are widely used, however, isolating pure vascular murine endothelial cells is technically challenging. A microvascular mouse explant model that is robust, quantitative and can be reproduced without difficulty would overcome these limitations. Here we characterized and optimized for reproducibility an organotypic microvascular angiogenesis mouse and rat model from the choroid, a microvascular bed in the posterior of eye. The choroidal tissues from C57BL/6J and 129S6/SvEvTac mice and Sprague Dawley rats were isolated and incubated in Matrigel. Vascular sprouting was comparable between choroid samples obtained from different animals of the same genetic background. The sprouting area, normalized to controls, was highly reproducible between independent experiments. We developed a semi-automated macro in ImageJ software to allow for more efficient quantification of sprouting area. Isolated choroid explants responded to manipulation of the external environment while maintaining the local interactions of endothelial cells with neighboring cells, including pericytes and macrophages as evidenced by immunohistochemistry and fluorescence-activated cell sorting (FACS) analysis. This reproducible ex vivo angiogenesis assay can be used to evaluate angiogenic potential of pharmacologic compounds on microvessels and can take advantage of genetically manipulated mouse tissue for microvascular disease research.


Angiogenesis | 2013

Neuronal sirtuin1 mediates retinal vascular regeneration in oxygen-induced ischemic retinopathy.

Jing Chen; Shaday Michan; Aimee M. Juan; Christian G. Hurst; Colman J. Hatton; Dorothy T. Pei; Jean-Sebastien Joyal; Lucy Evans; Zhenghao Cui; Andreas Stahl; Przemyslaw Sapieha; David A. Sinclair; Lois E. H. Smith

Regeneration of blood vessels in ischemic neuronal tissue is critical to reduce tissue damage in diseases. In proliferative retinopathy, initial vessel loss leads to retinal ischemia, which can induce either regrowth of vessels to restore normal metabolism and minimize damage, or progress to hypoxia-induced sight-threatening pathologic vaso-proliferation. It is not well understood how retinal neurons mediate regeneration of vascular growth in response to ischemic insults. In this study we aim to investigate the potential role of Sirtuin 1 (Sirt1), a metabolically-regulated protein deacetylase, in mediating the response of ischemic neurons to regulate vascular regrowth in a mouse model of oxygen-induced ischemic retinopathy (OIR). We found that Sirt1 is highly induced in the avascular ischemic retina in OIR. Conditional depletion of neuronal Sirt1 leads to significantly decreased retinal vascular regeneration into the avascular zone and increased hypoxia-induced pathologic vascular growth. This effect is likely independent of PGC-1α, a known Sirt1 target, as absence of PGC-1α in knockout mice does not impact vascular growth in retinopathy. We found that neuronal Sirt1 controls vascular regrowth in part through modulating deacetylation and stability of hypoxia-induced factor 1α and 2α, and thereby modulating expression of angiogenic factors. These results indicate that ischemic neurons induce Sirt1 to promote revascularization into ischemic neuronal areas, suggesting a novel role of neuronal Sirt1 in mediating vascular regeneration in ischemic conditions, with potential implications beyond retinopathy.


PLOS ONE | 2015

Optimization of an Image-Guided Laser-Induced Choroidal Neovascularization Model in Mice

Yan Gong; Jie Li; Ye Sun; Zhongjie Fu; Chi-Hsiu Liu; Lucy Evans; Katherine Tian; Nicholas Saba; Thomas Fredrick; Peyton Morss; Jing Chen; Lois E. H. Smith

The mouse model of laser-induced choroidal neovascularization (CNV) has been used in studies of the exudative form of age-related macular degeneration using both the conventional slit lamp and a new image-guided laser system. A standardized protocol is needed for consistent results using this model, which has been lacking. We optimized details of laser-induced CNV using the image-guided laser photocoagulation system. Four lesions with similar size were consistently applied per eye at approximately double the disc diameter away from the optic nerve, using different laser power levels, and mice of various ages and genders. After 7 days, the mice were sacrificed and retinal pigment epithelium/choroid/sclera was flat-mounted, stained with Isolectin B4, and imaged. Quantification of the area of the laser-induced lesions was performed using an established and constant threshold. Exclusion criteria are described that were necessary for reliable data analysis of the laser-induced CNV lesions. The CNV lesion area was proportional to the laser power levels. Mice at 12-16 weeks of age developed more severe CNV than those at 6-8 weeks of age, and the gender difference was only significant in mice at 12-16 weeks of age, but not in those at 6-8 weeks of age. Dietary intake of omega-3 long-chain polyunsaturated fatty acid reduced laser-induced CNV in mice. Taken together, laser-induced CNV lesions can be easily and consistently applied using the image-guided laser platform. Mice at 6-8 weeks of age are ideal for the laser-induced CNV model.


Science Signaling | 2015

SOCS3 in retinal neurons and glial cells suppresses VEGF signaling to prevent pathological neovascular growth

Ye Sun; Meihua Ju; Zhiqiang Lin; Thomas Fredrick; Lucy Evans; Katherine Tian; Nicholas Saba; Peyton Morss; William T. Pu; Jing Chen; Andreas Stahl; Jean-Sebastien Joyal; Lois E. H. Smith

Enhancing SOCS3 activity in neurons and glial cells in the retina may prevent abnormal blood vessel formation in proliferative retinopathies. Halting blood vessel formation in the eye Diabetics and preterm infants are susceptible to vision loss or blindness because of abnormal blood vessel formation in the eye, a process called retinal neovascularization. Using a model of this disease process, Sun et al. found that mice that lacked a protein called SOCS3 in the neurons and glial cells of the eye had greater retinal neovascularization than did control mice. Their results suggest that SOCS3 prevents neurons and glial cells from releasing too much VEGF (a cytokine that promotes blood vessel formation) by inhibiting the transcription factor STAT3, which can trigger the production of VEGF. Neurons and glial cells in the retina contribute to neovascularization, or the formation of abnormal new blood vessels, in proliferative retinopathy, a condition that can lead to vision loss or blindness. We identified a mechanism by which suppressor of cytokine signaling 3 (SOCS3) in neurons and glial cells prevents neovascularization. We found that Socs3 expression was increased in the retinal ganglion cell and inner nuclear layers after oxygen-induced retinopathy. Mice with Socs3 deficiency in neuronal and glial cells had substantially reduced vaso-obliterated retinal areas and increased pathological retinal neovascularization in response to oxygen-induced retinopathy, suggesting that loss of neuronal/glial SOCS3 increased both retinal vascular regrowth and pathological neovascularization. Furthermore, retinal expression of Vegfa (which encodes vascular endothelial growth factor A) was higher in these mice than in Socs3 flox/flox controls, indicating that neuronal and glial SOCS3 suppressed Vegfa expression during pathological conditions. Lack of neuronal and glial SOCS3 resulted in greater phosphorylation and activation of STAT3, which led to increased expression of its gene target Vegfa, and increased endothelial cell proliferation. In summary, SOCS3 in neurons and glial cells inhibited the STAT3-mediated secretion of VEGF from these cells, which suppresses endothelial cell activation, resulting in decreased endothelial cell proliferation and angiogenesis. These results suggest that neuronal and glial cell SOCS3 limits pathological retinal angiogenesis by suppressing VEGF signaling.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Nuclear receptor RORα regulates pathologic retinal angiogenesis by modulating SOCS3-dependent inflammation

Ye Sun; Chi-Hsiu Liu; John Paul SanGiovanni; Lucy Evans; Katherine Tian; Bing Zhang; Andreas Stahl; William T. Pu; Theodore M. Kamenecka; Laura A. Solt; Jing Chen

Significance Pathologic retinal neovascularization commonly causes blindness. Retinoic-acid-receptor–related orphan receptor alpha (RORα), a lipid-sensing nuclear receptor, is genetically associated with the risk of developing neovascular eye disease in humans. We demonstrate that RORα expression was highly increased in a mouse model of oxygen-induced proliferative retinopathy with pathologic neovessels. Both genetic deficiency and pharmacologic inhibition of RORα suppressed pathologic retinal neovascularization in mice with dampened inflammation. RORα transcriptionally regulated suppressors of cytokine signaling 3 (SOCS3), a negative mediator of macrophage function and inflammation. Suppression of SOCS3 attenuated the protective effects of RORα inhibition in retinopathy. Our data demonstrate an important role of RORα in controlling pathologic retinal neovascularization and suggest that RORα may represent a druggable target for treating ocular neovascularization. Pathologic ocular angiogenesis is a leading cause of blindness, influenced by both dysregulated lipid metabolism and inflammation. Retinoic-acid-receptor–related orphan receptor alpha (RORα) is a lipid-sensing nuclear receptor with diverse biologic function including regulation of lipid metabolism and inflammation; however, its role in pathologic retinal angiogenesis remains poorly understood. Using a mouse model of oxygen-induced proliferative retinopathy, we showed that RORα expression was significantly increased and genetic deficiency of RORα substantially suppressed pathologic retinal neovascularization. Loss of RORα led to decreased levels of proinflammatory cytokines and increased levels of antiinflammatory cytokines in retinopathy. RORα directly suppressed the gene transcription of suppressors of cytokine signaling 3 (SOCS3), a critical negative regulator of inflammation. Inhibition of SOCS3 abolished the antiinflammatory and vasoprotective effects of RORα deficiency in vitro and in vivo. Moreover, treatment with a RORα inverse agonist SR1001 effectively protected against pathologic neovascularization in both oxygen-induced retinopathy and another angiogenic model of very-low–density lipoprotein receptor (Vldlr)-deficient (Vldlr−/−) mice with spontaneous subretinal neovascularization, whereas a RORα agonist worsened oxygen-induced retinopathy. Our data demonstrate that RORα is a novel regulator of pathologic retinal neovascularization, and RORα inhibition may represent a new way to treat ocular neovascularization.


PLOS ONE | 2014

Sirtuin1 Over-Expression Does Not Impact Retinal Vascular and Neuronal Degeneration in a Mouse Model of Oxygen-Induced Retinopathy

Shaday Michan; Aimee M. Juan; Christian G. Hurst; Zhenghao Cui; Lucy Evans; Colman J. Hatton; Dorothy T. Pei; Meihua Ju; David A. Sinclair; Lois E. H. Smith; Jing Chen

Proliferative retinopathy is a leading cause of blindness, including retinopathy of prematurity (ROP) in children and diabetic retinopathy in adults. Retinopathy is characterized by an initial phase of vessel loss, leading to tissue ischemia and hypoxia, followed by sight threatening pathologic neovascularization in the second phase. Previously we found that Sirtuin1 (Sirt1), a metabolically dependent protein deacetylase, regulates vascular regeneration in a mouse model of oxygen-induced proliferative retinopathy (OIR), as neuronal depletion of Sirt1 in retina worsens retinopathy. In this study we assessed whether over-expression of Sirtuin1 in retinal neurons and vessels achieved by crossing Sirt1 over-expressing flox mice with Nestin-Cre mice or Tie2-Cre mice, respectively, may protect against retinopathy. We found that over-expression of Sirt1 in Nestin expressing retinal neurons does not impact vaso-obliteration or pathologic neovascularization in OIR, nor does it influence neuronal degeneration in OIR. Similarly, increased expression of Sirt1 in Tie2 expressing vascular endothelial cells and monocytes/macrophages does not protect retinal vessels in OIR. In addition to the genetic approaches, dietary supplement with Sirt1 activators, resveratrol or SRT1720, were fed to wild type mice with OIR. Neither treatment showed significant vaso-protective effects in retinopathy. Together these results indicate that although endogenous Sirt1 is important as a stress-induced protector in retinopathy, over-expression of Sirt1 or treatment with small molecule activators at the examined doses do not provide additional protection against retinopathy in mice. Further studies are needed to examine in depth whether increasing levels of Sirt1 may serve as a potential therapeutic approach to treat or prevent retinopathy.

Collaboration


Dive into the Lucy Evans's collaboration.

Top Co-Authors

Avatar

Jing Chen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lois E. H. Smith

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Katherine Tian

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhenghao Cui

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Ye Sun

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nicholas Saba

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Thomas Fredrick

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Zhongjie Fu

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge