Network


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

Hotspot


Dive into the research topics where Zhuo Shao is active.

Publication


Featured researches published by Zhuo Shao.


Blood | 2011

Ischemic neurons prevent vascular regeneration of neural tissue by secreting semaphorin 3A

Jean-Sebastien Joyal; Nicholas Sitaras; François Binet; José Carlos Rivera; Andreas Stahl; Karine Zaniolo; Zhuo Shao; Anna Polosa; Tang Zhu; David Hamel; Mikheil Djavari; Dario Kunik; Jean-Claude Honoré; Emilie Picard; Alexandra Zabeida; Daya R. Varma; Gilles R.X. Hickson; Joseph A. Mancini; Michael Klagsbrun; Santiago Costantino; Christian M. Beauséjour; Pierre Lachapelle; Lois E. H. Smith; Sylvain Chemtob; Przemyslaw Sapieha

The failure of blood vessels to revascularize ischemic neural tissue represents a significant challenge for vascular biology. Examples include proliferative retinopathies (PRs) such as retinopathy of prematurity and proliferative diabetic retinopathy, which are the leading causes of blindness in children and working-age adults. PRs are characterized by initial microvascular degeneration, followed by a compensatory albeit pathologic hypervascularization mounted by the hypoxic retina attempting to reinstate metabolic equilibrium. Paradoxically, this secondary revascularization fails to grow into the most ischemic regions of the retina. Instead, the new vessels are misdirected toward the vitreous, suggesting that vasorepulsive forces operate in the avascular hypoxic retina. In the present study, we demonstrate that the neuronal guidance cue semaphorin 3A (Sema3A) is secreted by hypoxic neurons in the avascular retina in response to the proinflammatory cytokine IL-1β. Sema3A contributes to vascular decay and later forms a chemical barrier that repels neo-vessels toward the vitreous. Conversely, silencing Sema3A expression enhances normal vascular regeneration within the ischemic retina, thereby diminishing aberrant neovascularization and preserving neuroretinal function. Overcoming the chemical barrier (Sema3A) released by ischemic neurons accelerates the vascular regeneration of neural tissues, which restores metabolic supply and improves retinal function. Our findings may be applicable to other neurovascular ischemic conditions such as stroke.


The International Journal of Biochemistry & Cell Biology | 2010

Proliferative retinopathies: angiogenesis that blinds.

Przemyslaw Sapieha; David Hamel; Zhuo Shao; José Carlos Rivera; Karine Zaniolo; Jean Sébastien Joyal; Sylvain Chemtob

Proliferative ischemic retinopathies such as proliferative diabetic retinopathy (PDR), retinopathy of prematurity (ROP) and those stemming from retinal vein occlusion are the leading causes of blindness in the working age and pediatric populations of industrialized countries. They present major financial burdens for health care systems and account for significant loss of productivity. These pathologies are characterized by excessive pre-retinal blood vessel growth that can ultimately lead to a fibrous scar formation and culminate in retinal detachment. This abnormal and disproportionate hyper-vascularization is a compensatory mechanism to overcome an earlier phase of microvessel degeneration and reinstate metabolic equilibrium to the hypoxic retina. To date, the treatment modalities to counter these diseases largely rely on invasive and moderately efficient surgical interventions. In this review, we discuss the current views on retinal vaso-obliteration, neovascularization and available treatments and present future strategies to tackle these diseases.


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.


Investigative Ophthalmology & Visual Science | 2011

Choroidal Involution Is a Key Component of Oxygen-Induced Retinopathy

Zhuo Shao; A. Dorfman; Swathi Seshadri; Mikheil Djavari; Elsa Kermorvant-Duchemin; Florian Sennlaub; Martine Blais; Anna Polosa; Daya R. Varma; Jean-Sebastien Joyal; Pierre Lachapelle; Pierre Hardy; Nicholas Sitaras; Emilie Picard; Joseph G. Mancini; Przemyslaw Sapieha; Sylvain Chemtob

PURPOSE Retinopathy of prematurity (ROP) is a major cause of visual handicap in the pediatric population. To date, this disorder is thought to stem from deficient retinal vascularization. Intriguingly, functional electrophysiological studies in patients with mild or moderate ROP and in the oxygen-induced retinopathy (OIR) model in rats reveal central photoreceptor disruption that overlies modest retinal vessel loss; a paucity of retinal vasculature occurs predominantly at the periphery. Given that choroidal circulation is the major source of oxygen and nutrients to the photoreceptors, the authors set out to investigate whether the choroidal vasculature system may be affected in OIR. METHODS Rat models of OIR treating newborn animals with 80% or 50/10% alternated oxygen level for the first two postnatal weeks were used to mimic ROP in humans. Immunohistology staining and vascular corrosion casts were used to investigate the vessel layout of the eye. To investigate the effect of 15-deoxy-Δ12,14-PGJ(2) (15d-PGJ(2); a nonenzymatic product of prostaglandin D(2)) on endothelial cells, in vitro cell culture and ex vivo choroid explants were employed and intravitreal injections were performed in animals. RESULTS The authors herein demonstrate that deficient vascularity occurs not only in the retinal plexus but also in the choroid. This sustained, marked choroidal degeneration is specifically confined to central regions of the retina that present persistent photoreceptor loss and corresponding functional deficits. Moreover, the authors show that 15d-PGJ(2) is a prominent contributor to this choroidal decay. CONCLUSIONS The authors demonstrate for the first time pronounced, sustained choroidal vascular involution during the development of ROP. Findings also suggest that effective therapeutic strategies to counter ROP should consider choroidal preservation.


Journal of Clinical Investigation | 2014

Neuropilin-1 mediates myeloid cell chemoattraction and influences retinal neuroimmune crosstalk

Agnieszka Dejda; Gaelle Mawambo; Agustin Cerani; Khalil Miloudi; Zhuo Shao; Jean-François Daudelin; Salix Boulet; Malika Oubaha; Felix Beaudoin; Naoufal Akla; Sullivan Henriques; Catherine Ménard; Andreas Stahl; Jean-Sébastien Delisle; Flavio Rezende; Nathalie Labrecque; Przemyslaw Sapieha

Immunological activity in the CNS is largely dependent on an innate immune response and is heightened in diseases, such as diabetic retinopathy, multiple sclerosis, amyotrophic lateral sclerosis, and Alzheimers disease. The molecular dynamics governing immune cell recruitment to sites of injury and disease in the CNS during sterile inflammation remain poorly defined. Here, we identified a subset of mononuclear phagocytes (MPs) that responds to local chemotactic cues that are conserved among central neurons, vessels, and immune cells. Patients suffering from late-stage proliferative diabetic retinopathy (PDR) had elevated vitreous semaphorin 3A (SEMA3A). Using a murine model, we found that SEMA3A acts as a potent attractant for neuropilin-1-positive (NRP-1-positive) MPs. These proangiogenic MPs were selectively recruited to sites of pathological neovascularization in response to locally produced SEMA3A as well as VEGF. NRP-1-positive MPs were essential for disease progression, as NRP-1-deficient MPs failed to enter the retina in a murine model of oxygen-induced retinopathy (OIR), a proxy for PDR. OIR mice with NRP-1-deficient MPs exhibited decreased vascular degeneration and diminished pathological preretinal neovascularization. Intravitreal administration of a NRP-1-derived trap effectively mimicked the therapeutic benefits observed in mice lacking NRP-1-expressing MPs. Our findings indicate that NRP-1 is an obligate receptor for MP chemotaxis, bridging neural ischemia to an innate immune response in neovascular retinal disease.


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.


Nature Medicine | 2014

Subcellular localization of coagulation factor II receptor-like 1 in neurons governs angiogenesis

Jean Sébastien Joyal; Satra Nim; Tang Zhu; Nicholas Sitaras; José Carlos Rivera; Zhuo Shao; Przemyslaw Sapieha; David Hamel; Melanie Sanchez; Karine Zaniolo; Manon St-Louis; Johanne Ouellette; Martín Montoya-Zavala; Alexandra Zabeida; Emilie Picard; Pierre Hardy; Vikrant K. Bhosle; Daya R. Varma; Christian M. Beauséjour; Christelle Boileau; William H. Klein; Morley D. Hollenberg; Alfredo Ribeiro-da-Silva; Gregor Andelfinger; Sylvain Chemtob

Neurons have an important role in retinal vascular development. Here we show that the G protein–coupled receptor (GPCR) coagulation factor II receptor-like 1 (F2rl1, previously known as Par2) is abundant in retinal ganglion cells and is associated with new blood vessel formation during retinal development and in ischemic retinopathy. After stimulation, F2rl1 in retinal ganglion cells translocates from the plasma membrane to the cell nucleus using a microtubule-dependent shuttle that requires sorting nexin 11 (Snx11). At the nucleus, F2rl1 facilitates recruitment of the transcription factor Sp1 to trigger Vegfa expression and, in turn, neovascularization. In contrast, classical plasma membrane activation of F2rl1 leads to the expression of distinct genes, including Ang1, that are involved in vessel maturation. Mutant versions of F2rl1 that prevent nuclear relocalization but not plasma membrane activation interfere with Vegfa but not Ang1 expression. Complementary angiogenic factors are therefore regulated by the subcellular localization of a receptor (F2rl1) that governs angiogenesis. These findings may have implications for the selectivity of drug actions based on the subcellular distribution of their targets.


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.


Investigative Ophthalmology & Visual Science | 2011

Ghrelin Modulates Physiologic and Pathologic Retinal Angiogenesis through GHSR-1a

Karine Zaniolo; Przemyslaw Sapieha; Zhuo Shao; Andreas Stahl; Tang Zhu; Sophie Tremblay; Emilie Picard; Ankush Madaan; Martine Blais; Pierre Lachapelle; Joseph G. Mancini; Pierre Hardy; Lois E. H. Smith; Huy Ong; Sylvain Chemtob

PURPOSE Vascular degeneration and the ensuing abnormal vascular proliferation are central to proliferative retinopathies. Given the metabolic discordance associated with these diseases, the authors explored the role of ghrelin and its growth hormone secretagogue receptor 1a (GHSR-1a) in proliferative retinopathy. METHODS In a rat model of oxygen-induced retinopathy (OIR), the contribution of ghrelin and GHSR-1a was investigated using the stable ghrelin analogs [Dap3]-ghrelin and GHRP6 and the GSHR-1a antagonists JMV-2959 and [D-Lys3]-GHRP-6. Plasma and retinal levels of ghrelin were analyzed by ELISA, whereas retinal expression and localization of GHSR-1a were examined by immunohistochemistry and Western blot analysis. The angiogenic and vasoprotective properties of ghrelin and its receptor were further confirmed in aortic explants and in models of vaso-obliteration. RESULTS Ghrelin is produced locally in the retina, whereas GHSR-1a is abundantly expressed in retinal endothelial cells. Ghrelin levels decrease during the vaso-obliterative phase and rise during the proliferative phase of OIR. Intravitreal delivery of [Dap3]-ghrelin during OIR significantly reduces retinal vessel loss when administered during the hyperoxic phase. Conversely, during the neovascular phase, ghrelin promotes pathologic angiogenesis through the activation of GHSR-1a. These angiogenic effects were confirmed ex vivo in aortic explants. CONCLUSIONS New roles were disclosed for the ghrelin-GHSR-1a pathway in the preservation of retinal vasculature during the vaso-obliterative phase of OIR and during the angiogenic phase of OIR. These findings suggest that the ghrelin-GHSR-1a pathway can exert opposing effects on retinal vasculature, depending on the phase of retinopathy, and thus holds therapeutic potential for proliferative retinopathies.


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.

Collaboration


Dive into the Zhuo Shao's collaboration.

Top Co-Authors

Avatar

Sylvain Chemtob

Hôpital Maisonneuve-Rosemont

View shared research outputs
Top Co-Authors

Avatar

Przemyslaw Sapieha

Hôpital Maisonneuve-Rosemont

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lois E. H. Smith

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Pierre Lachapelle

McGill University Health Centre

View shared research outputs
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

Tang Zhu

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge