Network


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

Hotspot


Dive into the research topics where Zhenghao Cui is active.

Publication


Featured researches published by Zhenghao Cui.


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.


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 | 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.


Nature Medicine | 2016

Corrigendum: 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

Jean-Sébastien Joyal, Ye Sun, Marin L Gantner, Zhuo Shao, Lucy P 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 A Rezende, Bruno Maranda, Przemyslaw Sapieha, Jing Chen, Martin Friedlander, Clary B Clish & Lois E H Smith Nat. Med.; doi:10.1038/nm.4059; corrected 24 March 2016


PLOS ONE | 2013

Correction: 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 Tahiri; François Duhamel; Jing Chen; Przemyslaw Sapieha; Sylvain Chemtob; Jean-Sebastien Joyal; Lois E. H. Smith


Investigative Ophthalmology & Visual Science | 2014

Endothelial miR-150 regulates pathologic retinal vascular growth in a mouse model of retinopathy of prematurity

Chi-Hsiu Liu; Zhenghao Cui; Lucy Evans; Hannah H Bogardus; Christian G. Hurst; Katherine Tian; Jing Chen


Investigative Ophthalmology & Visual Science | 2014

Over-expression of sirtuins does not protect against retinal neovascularization in a mouse model of oxygen-induced retinopathy

Meihua Ju; Shaday Michan; Christian G. Hurst; Zhenghao Cui; Lucy Evans; Lois E. H. Smith; Jing Chen


Investigative Ophthalmology & Visual Science | 2013

Evaluating Choroidal Microvascular Angiogenesis by Choroid Sprouting Assay

Zhuo Shao; Mollie Friedlander; Christian G. Hurst; Zhenghao Cui; Lucy Evans; Jing Chen; Przemyslaw Sapieha; Sylvain Chemtob; Jean-Sebastien Joyal; Lois E. H. Smith

Collaboration


Dive into the Zhenghao Cui's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jing Chen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lucy Evans

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Przemyslaw Sapieha

Hôpital Maisonneuve-Rosemont

View shared research outputs
Top Co-Authors

Avatar

Aimee M. Juan

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Dorothy T. Pei

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Colman J. Hatton

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge