Network


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

Hotspot


Dive into the research topics where Jianhai Du is active.

Publication


Featured researches published by Jianhai Du.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Pyruvate kinase and aspartate-glutamate carrier distributions reveal key metabolic links between neurons and glia in retina

Ken Lindsay; Jianhai Du; Stephanie R. Sloat; Laura Contreras; Jonathan D. Linton; Sally J. Turner; Martin Sadilek; Jorgina Satrústegui; James B. Hurley

Significance Aerobic glycolysis is a metabolic adaptation that helps cells in a tumor meet high anabolic demands. The M2 isoform of pyruvate kinase (PKM2) is associated with aerobic glycolysis in cancer cells. Aerobic glycolysis also accounts for most of the Glc metabolized in retinas. We find that photoreceptors (PRs) in retinas, like cancer cells in tumors, express PKM2. We also found very little expression of pyruvate kinase (PK) in Müller glia. We present metabolic flux analyses that show a metabolic relationship between PRs and Müller cells (MCs) that is different from the relationship between some neurons and astrocytes in brain. To compensate for PK deficiency and aspartate/glutamate carrier 1 deficiencies, MCs can fuel their mitochondria with lactate and aspartate produced by PRs. Symbiotic relationships between neurons and glia must adapt to structures, functions, and metabolic roles of the tissues they are in. We show here that Müller glia in retinas have specific enzyme deficiencies that can enhance their ability to synthesize Gln. The metabolic cost of these deficiencies is that they impair the Müller cell’s ability to metabolize Glc. We show here that the cells can compensate for this deficiency by using metabolites produced by neurons. Müller glia are deficient for pyruvate kinase (PK) and for aspartate/glutamate carrier 1 (AGC1), a key component of the malate-aspartate shuttle. In contrast, photoreceptor neurons express AGC1 and the M2 isoform of pyruvate kinase, which is commonly associated with aerobic glycolysis in tumors, proliferating cells, and some other cell types. Our findings reveal a previously unidentified type of metabolic relationship between neurons and glia. Müller glia compensate for their unique metabolic adaptations by using lactate and aspartate from neurons as surrogates for their missing PK and AGC1.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Cytosolic reducing power preserves glutamate in retina

Jianhai Du; Whitney M. Cleghorn; Laura Contreras; Jonathan D. Linton; Guy C.-K. Chan; Andrei O. Chertov; Takeyori Saheki; Viren Govindaraju; Martin Sadilek; Jorgina Satrústegui; James B. Hurley

Significance This report shows that the reducing power in the environment influences oxidation of glutamate in a neuronal tissue. Glutamate is a neurotransmitter, and it is especially important as a metabolite because it is required for synthesis of glutathione, other amino acids, and proteins. Glutamate also is a key intermediate in glutamine-dependent anaplerosis, now considered to be a principal source of citric acid cycle intermediates in cancer cells. Our analyses also show that the reducing power in the environmental can influence glutamate oxidation in cancer cells. Glutamate in neurons is an important excitatory neurotransmitter, but it also is a key metabolite. We investigated how glutamate in a neural tissue is protected from catabolism. Flux analysis using 13C-labeled fuels revealed that retinas use activities of the malate aspartate shuttle to protect >98% of their glutamate from oxidation in mitochondria. Isolation of glutamate from the oxidative pathway relies on cytosolic NADH/NAD+, which is influenced by extracellular glucose, lactate, and pyruvate.


Journal of Clinical Investigation | 2016

Reprogramming metabolism by targeting sirtuin 6 attenuates retinal degeneration

Lijuan Zhang; Jianhai Du; Sally Justus; Chun-Wei Hsu; Luis Bonet-Ponce; Wen-Hsuan Wu; Yi-Ting Tsai; Wei-Pu Wu; Yading Jia; Jimmy Duong; Vinit B. Mahajan; Chyuan-Sheng Lin; Shuang Wang; James B. Hurley; Stephen H. Tsang

Retinitis pigmentosa (RP) encompasses a diverse group of Mendelian disorders leading to progressive degeneration of rods and then cones. For reasons that remain unclear, diseased RP photoreceptors begin to deteriorate, eventually leading to cell death and, consequently, loss of vision. Here, we have hypothesized that RP associated with mutations in phosphodiesterase-6 (PDE6) provokes a metabolic aberration in rod cells that promotes the pathological consequences of elevated cGMP and Ca2+, which are induced by the Pde6 mutation. Inhibition of sirtuin 6 (SIRT6), a histone deacetylase repressor of glycolytic flux, reprogrammed rods into perpetual glycolysis, thereby driving the accumulation of biosynthetic intermediates, improving outer segment (OS) length, enhancing photoreceptor survival, and preserving vision. In mouse retinae lacking Sirt6, effectors of glycolytic flux were dramatically increased, leading to upregulation of key intermediates in glycolysis, TCA cycle, and glutaminolysis. Both transgenic and AAV2/8 gene therapy-mediated ablation of Sirt6 in rods provided electrophysiological and anatomic rescue of both rod and cone photoreceptors in a preclinical model of RP. Due to the extensive network of downstream effectors of Sirt6, this study motivates further research into the role that these pathways play in retinal degeneration. Because reprogramming metabolism by enhancing glycolysis is not gene specific, this strategy may be applicable to a wide range of neurodegenerative disorders.


eLife | 2017

Biochemical adaptations of the retina and retinal pigment epithelium support a metabolic ecosystem in the vertebrate eye

Mark A Kanow; Michelle Giarmarco; Connor Jankowski; Kristine Tsantilas; Abbi L. Engel; Jianhai Du; Jonathan D. Linton; Christopher C. Farnsworth; Stephanie R. Sloat; Austin M. Rountree; Ian R. Sweet; Ken Lindsay; Edward Parker; Susan E. Brockerhoff; Martin Sadilek; Jennifer R. Chao; James B. Hurley

Here we report multiple lines of evidence for a comprehensive model of energy metabolism in the vertebrate eye. Metabolic flux, locations of key enzymes, and our finding that glucose enters mouse and zebrafish retinas mostly through photoreceptors support a conceptually new model for retinal metabolism. In this model, glucose from the choroidal blood passes through the retinal pigment epithelium to the retina where photoreceptors convert it to lactate. Photoreceptors then export the lactate as fuel for the retinal pigment epithelium and for neighboring Müller glial cells. We used human retinal epithelial cells to show that lactate can suppress consumption of glucose by the retinal pigment epithelium. Suppression of glucose consumption in the retinal pigment epithelium can increase the amount of glucose that reaches the retina. This framework for understanding metabolic relationships in the vertebrate retina provides new insights into the underlying causes of retinal disease and age-related vision loss.


Journal of Biological Chemistry | 2017

Human retinal pigment epithelial cells prefer proline as a nutrient and transport metabolic intermediates to the retinal side

Jennifer R. Chao; Kaitlen Knight; Abbi L. Engel; Connor Jankowski; Yekai Wang; Megan A. Manson; Haiwei Gu; Danijel Djukovic; Daniel Raftery; James B. Hurley; Jianhai Du

Metabolite transport is a major function of the retinal pigment epithelium (RPE) to support the neural retina. RPE dysfunction plays a significant role in retinal degenerative diseases. We have used mass spectrometry with 13C tracers to systematically study nutrient consumption and metabolite transport in cultured human fetal RPE. LC/MS-MS detected 120 metabolites in the medium from either the apical or basal side. Surprisingly, more proline is consumed than any other nutrient, including glucose, taurine, lipids, vitamins, or other amino acids. Besides being oxidized through the Krebs cycle, proline is used to make citrate via reductive carboxylation. Citrate, made either from 13C proline or from 13C glucose, is preferentially exported to the apical side and is taken up by the retina. In conclusion, RPE cells consume multiple nutrients, including glucose and taurine, but prefer proline, and they actively synthesize and export metabolic intermediates to the apical side to nourish the outer retina.


Molecular Neurobiology | 2018

Disruption of De Novo Serine Synthesis in Müller Cells Induced Mitochondrial Dysfunction and Aggravated Oxidative Damage

Ting Zhang; Mark C. Gillies; Michele C. Madigan; Weiyong Shen; Jianhai Du; Ulrike Grünert; Fanfan Zhou; Michelle Yam; Ling Zhu

De novo serine synthesis plays important roles in normal mitochondrial function and cellular anti-oxidative capacity. It is reported to be mainly activated in glial cells of the central nervous system, but its role in retinal Müller glia remains unclear. In this study, we inhibited de novo serine synthesis using CBR-5884, a specific inhibitor of phosphoglycerate dehydrogenase (PHGDH, a rate limiting enzyme in de novo serine metabolism) in MIO-M1 cells (immortalized human Müller cells) and huPMCs (human primary Müller cells) under mild oxidative stress. Alamar blue and LDH (lactate dehydrogenase) assays showed significantly reduced metabolic activities and increased cellular damage of Müller cells, when exposed to CBR-5884 accompanied by mild oxidative stress; however, CBR-5884 alone had little effect. The increased cellular damage was partially reversed by supplementation with exogenous serine/glycine. HSP72 (an oxidative stress marker) and reactive oxygen species (ROS) levels were significantly increased; glutathione and NADPH/NADP+ levels were pronouncedly reduced under PHGDH inhibition accompanied by oxidative stress. JC-1 staining and Seahorse respiration experiments showed that inhibition of de novo serine synthesis in Müller cells can also increase mitochondrial stress and decrease mitochondrial ATP production. qPCR and Western blot demonstrated an increased expression of HSP60 (a key mitochondrial stress-related gene), and this was further validated in human retinal explants. Our study suggests that de novo serine synthesis is important for Müller cell survival, particularly when they are exposed to mild oxidative stress, possibly by maintaining mitochondrial function and generating glutathione and NADPH to counteract ROS.


Archive | 2018

How Excessive cGMP Impacts Metabolic Proteins in Retinas at the Onset of Degeneration

Jianhai Du; Jie An; Jonathan D. Linton; Yekai Wang; James B. Hurley

Aryl-hydrocarbon receptor interacting protein-like 1 (AIPL1) is essential to stabilize cGMP phosphodiesterase 6 (PDE6) in rod photoreceptors. Mutation of AIPL1 leads to loss of PDE6, accumulation of intracellular cGMP, and rapid degeneration of rods. To understand the metabolic basis for the photoreceptor degeneration caused by excessive cGMP, we performed proteomics and phosphoproteomics analyses on retinas from AIPL1-/- mice at the onset of rod cell death. AIPL1-/- retinas have about 18 times less than normal PDE6a and no detectable PDE6b. We identified twelve other proteins and thirty-nine phosphorylated proteins related to cell metabolism that are significantly altered preceding the massive degeneration of rods. They include transporters, kinases, phosphatases, transferases, and proteins involved in mitochondrial bioenergetics and metabolism of glucose, lipids, amino acids, nucleotides, and RNA. In AIPLI-/- retinas mTOR and proteins involved in mitochondrial energy production and lipid synthesis are more dephosphorylated, but glycolysis proteins and proteins involved in leucine catabolism are more phosphorylated than in normal retinas. Our findings indicate that elevating cGMP rewires cellular metabolism prior to photoreceptor degeneration and that targeting metabolism may be a productive strategy to prevent or slow retinal degeneration.


Neurobiology of Aging | 2018

Metabolic signature of the aging eye in mice

Yekai Wang; Allison Grenell; Fanyi Zhong; Michelle Yam; Allison Hauer; Elizabeth Gregor; Siyan Zhu; Daniel Lohner; Jiangjiang Zhu; Jianhai Du

Aging is a major risk factor for age-related ocular diseases including age-related macular degeneration in the retina and retinal pigment epithelium (RPE), cataracts in the lens, glaucoma in the optic nerve, and dry eye syndrome in the cornea. We used targeted metabolomics to analyze metabolites from young (6xa0weeks) and old (73xa0weeks) eyes in C57 BL6/J mice. Old mice had diminished electroretinogram responses and decreased number of photoreceptors in their retinas. Among the 297 detected metabolites, 45-114 metabolites are significantly altered in aged eye tissues, mostly in the neuronal tissues (retina and optic nerve) and less in cornea, RPE/choroid, and lens. We noted that changes of metabolites in mitochondrial metabolism and glucose metabolism are common features in the aged retina, RPE/choroid, and optic nerve. The aging retina, cornea, and optic nerve also share similar changes in Nicotinamide adenine dinucleotide (NAD), 1-methylnicotinamides, 3-methylhistidine, and other methylated metabolites. Metabolites in taurine metabolism are strikingly influenced by aging in the cornea and lens. In conclusion, the aging eye has both common and tissue-specific metabolic signatures. These changes may be attributed to dysregulated mitochondrial metabolism, reprogrammed glucose metabolism and impaired methylation in the aging eye. Our findings provide biochemical insights into the mechanisms of age-related ocular changes.


Experimental Eye Research | 2018

Deletion of GLUT1 in mouse lens epithelium leads to cataract formation

Aditi Swarup; Brent A. Bell; Jianhai Du; John Y.S. Han; Jamie Soto; E. Dale Abel; Arturo Bravo-Nuevo; Paul G. FitzGerald; Neal S. Peachey; Nancy J. Philp

ABSTRACT The primary energy substrate of the lens is glucose and uptake of glucose from the aqueous humor is dependent on glucose transporters. GLUT1, the facilitated glucose transporter encoded by Slc2a1 is expressed in the epithelium of bovine, human and rat lenses. In the current study, we examined the expression of GLUT1 in the mouse lens and determined its role in maintaining lens transparency by studying effects of postnatal deletion of Slc2a1. In situ hybridization and immunofluorescence labeling were used to determine the expression and subcellular distribution of GLUT1 in the lens. Slc2a1 was knocked out of the lens epithelium by crossing transgenic mice expressing Cre recombinase under control of the GFAP promoter with Slc2a1loxP/loxP mice to generate Slc2a1loxP/loxP;GFAP‐Cre+/0 (Lens&Dgr;Glut1) mice. Lens&Dgr;Glut1 mice developed visible lens opacities by around 3 months of age, which corresponded temporally with the total loss of detectable GLUT1 expression in the lens. Spectral domain optical coherence tomography (SD‐OCT) imaging was used to monitor the formation of cataracts over time. SD‐OCT imaging revealed that small nuclear cataracts were first apparent in the lenses of Lens&Dgr;Glut1 mice beginning at about 2.7 months of age. Longitudinal SD‐OCT imaging of Lens&Dgr;Glut1 mice revealed disruption of mature secondary fiber cells after 3 months of age. Histological sections of eyes from Lens&Dgr;Glut1 mice confirmed the disruption of the secondary fiber cells. The structural changes were most pronounced in fiber cells that had lost their organelles. In contrast, the histology of the lens epithelium in these mice appeared normal. Lactate and ATP were measured in lenses from Lens&Dgr;Glut1 and control mice at 2 and 3 months of age. At 2 months of age, when GLUT1 was still detectable in the lens epithelium, albeit at low levels, the amount of lactate and ATP were not significantly different from controls. However, in lenses isolated from 3‐month‐old Lens&Dgr;Glut1 mice, when GLUT1 was no longer detectable, levels of lactate and ATP were 50% lower than controls. Our findings demonstrate that in vivo, the transparency of mature lens fiber cells was dependent on glycolysis for ATP and the loss of GLUT1 transporters led to cataract formation. In contrast, lens epithelium and cortical fiber cells have mitochondria and could utilize other substrates to support their anabolic and catabolic needs. HIGHLIGHTSGLUT1 is the primary glucose transporter expressed in the mouse lens epithelium.Deletion of GLUT1 from the lens epithelium leads to formation of cataracts in mouse lens at the age of approximately 3 months.Cataract formation is accompanied by a decrease in ATP and lactate levels at 3 months.Optical Coherence Tomography provides a useful tool for imaging cataract formation.


Experimental Eye Research | 2018

Impact of euthanasia, dissection and postmortem delay on metabolic profile in mouse retina and RPE/choroid

Siyan Zhu; Michelle Yam; Yekai Wang; Jonathan D. Linton; Allison Grenell; James B. Hurley; Jianhai Du

ABSTRACT Metabolomics studies in the retina and retinal pigment epithelium (RPE) in animal models or postmortem donors are essential to understanding the retinal metabolism and to revealing the underlying mechanisms of retinal degenerative diseases. We have studied how different methods of euthanasia (CO2 or cervical dislocation) different isolation procedures and postmortem delay affect metabolites in mouse retina and RPE/choroid using LC MS/MS and GC MS. Compared with cervical dislocation, CO2 exposure for 5min dramatically degrades ATP and GTP into purine metabolites in the retina while raising intermediates in glucose metabolism and amino acids in the RPE/choroid. Isolation in cold buffer containing glucose has the least change in metabolites. Postmortem delay time‐dependently and differentially impacts metabolites in the retina and RPE/choroid. In the postmortem retina, 18% of metabolites were changed at 0.5h (h), 41% at 4h and 51% at 8h. However, only 6% of metabolites were changed in the postmortem RPE/choroid and it steadily increased to 20% at 8h. Notably, both postmortem retina and RPE/choroid tissue showed increased purine metabolites. Storage of eyes in cold nutrient‐rich medium substantially blocked the postmortem change in the retina and RPE/choroid. In conclusion, our study provides optimized methods to prepare fresh or postmortem retina and RPE/choroid tissue for metabolomics studies. HIGHLIGHTSEuthanasia with CO2 degrades ATP and GTP into purine metabolites only in retina.Euthanasia with CO2 increases amino acids and intermediates in glucose metabolism only in RPE/choroid.Glucose availability and cold temperature are critical to stabilize metabolism during dissection.80% of metabolites are stable within 1h in postmortem retina and 8h in RPE/choroid.Cold nutrient medium partially prevents the metabolite levels from changing in postmortem retina and RPE.

Collaboration


Dive into the Jianhai Du's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yekai Wang

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Daniel Raftery

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Martin Sadilek

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jorgina Satrústegui

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Laura Contreras

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Abbi L. Engel

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge