Network


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

Hotspot


Dive into the research topics where Ken Lindsay is active.

Publication


Featured researches published by Ken Lindsay.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Flow of energy in the outer retina in darkness and in light

Jonathan D. Linton; Lars C. Holzhausen; Norbert Babai; Hongman Song; Kiyoharu Miyagishima; George W. Stearns; Ken Lindsay; J. Wei; Andrei O. Chertov; Theo A. Peters; Romeo Caffé; Helma Pluk; Mathias W. Seeliger; Naoyuki Tanimoto; Kimberly K. Fong; Laura Bolton; Denise L. T. Kuok; Ian R. Sweet; Theodore M. Bartoletti; Roxana A. Radu; Gabriel H. Travis; Willam N. Zagotta; Ellen Townes-Anderson; Ed Parker; Catharina E.E.M. Van der Zee; Alapakkam P. Sampath; Maxim Sokolov; Wallace B. Thoreson; James B. Hurley

Structural features of neurons create challenges for effective production and distribution of essential metabolic energy. We investigated how metabolic energy is distributed between cellular compartments in photoreceptors. In avascular retinas, aerobic production of energy occurs only in mitochondria that are located centrally within the photoreceptor. Our findings indicate that metabolic energy flows from these central mitochondria as phosphocreatine toward the photoreceptor’s synaptic terminal in darkness. In light, it flows in the opposite direction as ATP toward the outer segment. Consistent with this model, inhibition of creatine kinase in avascular retinas blocks synaptic transmission without influencing outer segment activity. Our findings also reveal how vascularization of neuronal tissue can influence the strategies neurons use for energy management. In vascularized retinas, mitochondria in the synaptic terminals of photoreceptors make neurotransmission less dependent on creatine kinase. Thus, vasculature of the tissue and the intracellular distribution of mitochondria can play key roles in setting the strategy for energy distribution in neurons.


Journal of Neuroscience Research | 2015

Glucose, lactate, and shuttling of metabolites in vertebrate retinas

James B. Hurley; Ken Lindsay; Jianhai Du

The vertebrate retina has specific functions and structures that give it a unique set of constraints on the way in which it can produce and use metabolic energy. The retinas response to illumination influences its energy requirements, and the retinas laminated structure influences the extent to which neurons and glia can access metabolic fuels. There are fundamental differences between energy metabolism in retina and that in brain. The retina relies on aerobic glycolysis much more than the brain does, and morphological differences between retina and brain limit the types of metabolic relationships that are possible between neurons and glia. This Mini‐Review summarizes the unique metabolic features of the retina with a focus on the role of lactate shuttling.


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.


Journal of Biological Chemistry | 2013

Inhibition of mitochondrial pyruvate transport by Zaprinast causes massive accumulation of aspartate at the expense of glutamate in the retina

Jianhai Du; Whitney M. Cleghorn; Laura Contreras; Ken Lindsay; Austin M. Rountree; Andrei O. Chertov; Sally J. Turner; Ayse Sahaboglu; Jonathan D. Linton; Martin Sadilek; Jorgina Satrústegui; Ian R. Sweet; François Paquet-Durand; James B. Hurley

Background: Pyruvate transport into mitochondria is a key step in energy metabolism. Zaprinast is a well known phosphodiesterase inhibitor. Results: Zaprinast has a strong influence on pyruvate transport into mitochondria. Conclusion: Inhibition of the mitochondrial pyruvate carrier by Zaprinast causes accumulation of aspartate at the expense of glutamate. Significance: Maintenance of normal amino acid levels in the retina relies on pyruvate transport into mitochondria. Transport of pyruvate into mitochondria by the mitochondrial pyruvate carrier is crucial for complete oxidation of glucose and for biosynthesis of amino acids and lipids. Zaprinast is a well known phosphodiesterase inhibitor and lead compound for sildenafil. We found Zaprinast alters the metabolomic profile of mitochondrial intermediates and amino acids in retina and brain. This metabolic effect of Zaprinast does not depend on inhibition of phosphodiesterase activity. By providing 13C-labeled glucose and glutamine as fuels, we found that the metabolic profile of the Zaprinast effect is nearly identical to that of inhibitors of the mitochondrial pyruvate carrier. Both stimulate oxidation of glutamate and massive accumulation of aspartate. Moreover, Zaprinast inhibits pyruvate-driven O2 consumption in brain mitochondria and blocks mitochondrial pyruvate carrier in liver mitochondria. Inactivation of the aspartate glutamate carrier in retina does not attenuate the metabolic effect of Zaprinast. Our results show that Zaprinast is a potent inhibitor of mitochondrial pyruvate carrier activity, and this action causes aspartate to accumulate at the expense of glutamate. Our findings show that Zaprinast is a specific mitochondrial pyruvate carrier (MPC) inhibitor and may help to elucidate the roles of MPC in amino acid metabolism and hypoglycemia.


Journal of Biological Chemistry | 2016

Phototransduction Influences Metabolic Flux and Nucleotide Metabolism in Mouse Retina.

Jianhai Du; Austin M. Rountree; Whitney M. Cleghorn; Laura Contreras; Ken Lindsay; Martin Sadilek; Haiwei Gu; Danijel Djukovic; Daniel Raftery; Jorgina Satrústegui; Mark A Kanow; Lawrence Chan; Stephen H. Tsang; Ian R. Sweet; James B. Hurley

Production of energy in a cell must keep pace with demand. Photoreceptors use ATP to maintain ion gradients in darkness, whereas in light they use it to support phototransduction. Matching production with consumption can be accomplished by coupling production directly to consumption. Alternatively, production can be set by a signal that anticipates demand. In this report we investigate the hypothesis that signaling through phototransduction controls production of energy in mouse retinas. We found that respiration in mouse retinas is not coupled tightly to ATP consumption. By analyzing metabolic flux in mouse retinas, we also found that phototransduction slows metabolic flux through glycolysis and through intermediates of the citric acid cycle. We also evaluated the relative contributions of regulation of the activities of α-ketoglutarate dehydrogenase and the aspartate-glutamate carrier 1. In addition, a comprehensive analysis of the retinal metabolome showed that phototransduction also influences steady-state concentrations of 5′-GMP, ribose-5-phosphate, ketone bodies, and purines.


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.


Archive | 2014

Energy Metabolism in the Vertebrate Retina

James B. Hurley; Andrei O. Chertov; Ken Lindsay; Michelle Giamarco; Whitney M. Cleghorn; Jianhai Du; Susan E. Brockerhoff

The fundamental need for efficient and well-controlled energy metabolism in living organisms is ubiquitous. However, vertebrate retinas have many fascinating adaptations that give them the ability to regulate energy production and anabolic activity to meet a unique set of metabolic demands.


Cell Death and Disease | 2018

Pyruvate kinase M2 regulates photoreceptor structure, function, and viability.

Ammaji Rajala; Yuhong Wang; Richard S. Brush; Kristine Tsantilas; Connor Jankowski; Ken Lindsay; Jonathan D. Linton; James B. Hurley; Robert E. Anderson; Raju V. S. Rajala

Pyruvate kinase M2 (PKM2) is a glycolytic enzyme that is expressed in cancer cells. Its role in tumor metabolism is not definitively established, but investigators have suggested that regulation of PKM2 activity can cause accumulation of glycolytic intermediates and increase flux through the pentose phosphate pathway. Recent evidence suggests that PKM2 also may have non-metabolic functions, including as a transcriptional co-activator in gene regulation. We reported previously that PKM2 is abundant in photoreceptor cells in mouse retinas. In the present study, we conditionally deleted PKM2 (rod-cre PKM2-KO) in rod photoreceptors and found that the absence of PKM2 causes increased expression of PKM1 in rods. Analysis of metabolic flux from U-13C glucose shows that rod-cre PKM2-KO retinas accumulate glycolytic intermediates, consistent with an overall reduction in the amount of pyruvate kinase activity. Rod-cre PKM2-KO mice also have an increased NADPH availability could favor lipid synthesis, but we found no difference in phospholipid synthesis between rod-cre PKM2 KO and PKM2-positive controls. As rod-cre PKM2-KO mice aged, we observed a significant loss of rod function, reduced thickness of the photoreceptor outer segment layer, and reduced expression of photoreceptor proteins, including PDE6β. The rod-cre PKM2-KO retinas showed greater TUNEL staining than wild-type retinas, indicating a slow retinal degeneration. In vitro analysis showed that PKM2 can regulate transcriptional activity from the PDE6β promoter in vitro. Our findings indicate that both the metabolic and transcriptional regulatory functions of PKM2 may contribute to photoreceptor structure, function, and viability.


Investigative Ophthalmology & Visual Science | 2016

Confocal imaging reveals glucose uptake by photoreceptors in vivo

Michelle Giarmarco; Mark A Kanow; Ken Lindsay; Jianhai Du; James B. Hurley


Investigative Ophthalmology & Visual Science | 2013

Unique expression and regulation of glycolytic enzyme PKM2 in Photoreceptor cells and the role of enzymatic activity modulating metabolism of the retina

Ken Lindsay; Jonathon Linton; James B. Hurley

Collaboration


Dive into the Ken Lindsay's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianhai Du

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian R. Sweet

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

Mark A Kanow

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jorgina Satrústegui

Spanish National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge