Network


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

Hotspot


Dive into the research topics where Michael B. Robinson is active.

Publication


Featured researches published by Michael B. Robinson.


Nature Medicine | 1999

Selective inhibition of NAALADase, which converts NAAG to glutamate, reduces ischemic brain injury

Barbara S. Slusher; James J. Vornov; Ajit G. Thomas; Patricia D. Hurn; Izumi Harukuni; Anish Bhardwaj; Richard J. Traystman; Michael B. Robinson; Paul Britton; X.-C. May Lu; Frank C. Tortella; Krystyna M. Wozniak; Marc Yudkoff; Beth M. Potter; Paul F. Jackson

We describe here a new strategy for the treatment of stroke, through the inhibition of NAALADase (N-acetylated-α-linked-acidic dipeptidase), an enzyme responsible for the hydrolysis of the neuropeptide NAAG (N-acetyl-aspartyl-glutamate) to N-acetyl-aspartate and glutamate. We demonstrate that the newly described NAALADase inhibitor 2-PMPA (2-(phosphonomethyl)pentanedioic acid) robustly protects against ischemic injury in a neuronal culture model of stroke and in rats after transient middle cerebral artery occlusion. Consistent with inhibition of NAALADase, we show that 2-PMPA increases NAAG and attenuates the ischemia-induced rise in glutamate. Both effects could contribute to neuroprotection. These data indicate that NAALADase inhibition may have use in neurological disorders in which excessive excitatory amino acid transmission is pathogenic.


Advances in pharmacology | 1996

Heterogeneity and Functional Properties of Subtypes of Sodium-Dependent Glutamate Transporters in the Mammalian Central Nervous System

Michael B. Robinson; Lisa A. Dowd

Publisher Summary In recent times, there has been a tremendous expansion in knowledge of Na + -dependent glutamate (Glu) transporters. A large family has been identified using pharmacological as well as molecular biological techniques. At present, the cloned transporters do not reconstitute all of the pharmacological subtypes observed in vivo and in vitro . Furthermore, the clones provide evidence for subtypes of transport activity that were not differentiated pharmacologically. The acidic amino acids, glutamate (Glu), and aspartate (Asp) are the predominant excitatory neurotransmitters in the mammalian central nervous system (CNS). Several types of transport activities for Glu and Asp have been differentiated in mammals. Although at least two families of these transporters are localized to the plasma membrane, other subtypes of transporters appear to be selectively localized to subcellular organelles. Only one of the families of transporters that is localized to the plasma membrane appears to be coupled to an electrochemical gradient that permits transport of the acidic amino acids against their concentration gradient. This family of active Na + -dependent transporters is the focus of this chapter. The pharmacological evidence to suggest that there are several subtypes of this transport system are presented here.


Neurochemistry International | 2012

The glutamate transporter, GLAST, participates in a macromolecular complex that supports glutamate metabolism.

Deborah E. Bauer; Joshua G. Jackson; Elizabeth N. Genda; Misty M. Montoya; Marc Yudkoff; Michael B. Robinson

GLAST is the predominant glutamate transporter in the cerebellum and contributes substantially to glutamate transport in forebrain. This astroglial glutamate transporter quickly binds and clears synaptically released glutamate and is principally responsible for ensuring that synaptic glutamate concentrations remain low. This process is associated with a significant energetic cost. Compartmentalization of GLAST with mitochondria and proteins involved in energy metabolism could provide energetic support for glutamate transport. Therefore, we performed immunoprecipitation and co-localization experiments to determine if GLAST might co-compartmentalize with proteins involved in energy metabolism. GLAST was immunoprecipitated from rat cerebellum and subunits of the Na(+)/K(+) ATPase, glycolytic enzymes, and mitochondrial proteins were detected. GLAST co-localized with mitochondria in cerebellar tissue. GLAST also co-localized with mitochondria in fine processes of astrocytes in organotypic hippocampal slice cultures. From these data, we hypothesized that mitochondria participate in a macromolecular complex with GLAST to support oxidative metabolism of transported glutamate. To determine the functional metabolic role of this complex, we measured CO(2) production from radiolabeled glutamate in cultured astrocytes and compared it to overall glutamate uptake. Within 15 min, 9% of transported glutamate was converted to CO(2). This CO(2) production was blocked by inhibitors of glutamate transport and glutamate dehydrogenase, but not by an inhibitor of glutamine synthetase. Our data support a model in which GLAST exists in a macromolecular complex that allows transported glutamate to be metabolized in mitochondria to support energy production.


Journal of Biological Chemistry | 2008

The Endoplasmic Reticulum Exit of Glutamate Transporter Is Regulated by the Inducible Mammalian Yip6b/GTRAP3-18 Protein

Alicia M. Ruggiero; Yiting Liu; Svetlana Vidensky; Susanne Maier; Elizabeth Jung; Hesso Farhan; Michael B. Robinson; Harald H. Sitte; Jeffrey D. Rothstein

GTRAP3-18 interacts with and reduces the activity of the neuronal specific Na+/K+ glutamate transporter, EAAC1 both in vitro and in vivo. GTRAP3-18 and the related isoform, JM4, are distant relatives of the Rab GTPase-interacting factor PRA1, and share a topology of four transmembrane domains and cytosolic termini. GTRAP3-18 and JM4 are resident endoplasmic reticulum (ER) proteins. The physiological role of GTRAP3-18 is poorly understood. We demonstrate for the first time that GTRAP3-18 is a regulator of ER protein trafficking. Expression of GTRAP3-18 delays the ER exit of EAAC1, as well as other members of the excitatory amino acid transporter family. GTRAP3-18 uses hydrophobic domain interactions in the ER membrane to self-associate and cytoplasmic interactions at the C terminus to regulate trafficking. The features of GTRAP3-18 activity are consistent with recent phylogenic sequence analyses suggesting GTRAP3-18 and JM4 be reclassified as mammalian isoforms of the yeast protein family Yip, Yip6b, and Yip6a, respectively.


European Journal of Neuroscience | 1996

The Glutamate Transport Inhibitor L-trans-pyrrolidine-2,4-dicarboxylate Indirectly Evokes NMDA Receptor Mediated Neurotoxicity in Rat Cortical Cultures

Rachel C. Blitzblau; Shalini Gupta; Sina Djali; Michael B. Robinson; Paul A. Rosenberg

Because of the well‐documented importance of glutamate uptake in protecting neurons against glutamate toxicity, we were interested in testing the effects of L‐trans‐pyrrolidine‐2,4‐ dicarboxylate (PDC) on rat cortical cultures. This compound is a substrate for glutamate transporters and is a potent glutamate transport inhibitor that does not interact significantly with glutamate receptors. Using a 30 min exposure, and assessing neuronal survival after 20‐24 h, PDC was neurotoxic in conventional astrocyte‐rich cortical cultures, with an EC50 in these cultures of 320 ± 157 μM. In astrocyte‐poor cultures, an EC50 for PDC of 50 ± 5 μM was determined. The neurotoxicity of PDC in both astrocyte‐rich and astrocyte‐poor cultures was blocked by the NMDA antagonist MK‐801, but not by the non‐NMDA receptor antagonist 6‐cyano‐7‐nitroquinoxaline‐2,3‐dione (CNQX). We tested the possibility that the neurotoxicity of PDC might be due to release of excitatory amino acids using several approaches. After pre‐loading cells with the non‐metabolizable analogue of glutamate, [3H]‐D‐aspartate, first we demonstrated that PDC caused significant efflux of [3H]‐D‐aspartate. This effect of PDC was dependent upon extracellular sodium. In contrast with glutamate neurotoxicity, PDC neurotoxicity was inhibited by removal of extracellular sodium. In the presence of 1 mM PDC, sodium caused neurotoxicity with an EC50 of 18 ± 7.6 mM. Tetrodotoxin had no effect on either PDC neurotoxicity or on PDC‐evoked [3H]‐D‐aspartate release. PDC‐evoked release of [3H]‐D‐aspartate was demonstrable in astrocyte cultures with no neurons present. PDC also evoked release of endogenous glutamate. Finally, the neurotoxicity of PDC was blocked by coincubation with glutamate‐pyruvate transaminase plus pyruvate to degrade extracellular glutamate. These results demonstrate the neurotoxicity of PDC, and suggest that the mechanism of this toxicity is the glutamate transporter‐dependent accumulation of glutamate in the extracellular space.


Neurochemistry International | 2016

Astroglial glutamate transporters coordinate excitatory signaling and brain energetics

Michael B. Robinson; Joshua G. Jackson

In the mammalian brain, a family of sodium-dependent transporters maintains low extracellular glutamate and shapes excitatory signaling. The bulk of this activity is mediated by the astroglial glutamate transporters GLT-1 and GLAST (also called EAAT2 and EAAT1). In this review, we will discuss evidence that these transporters co-localize with, form physical (co-immunoprecipitable) interactions with, and functionally couple to various energy-generating systems, including the Na(+)/K(+)-ATPase, the Na(+)/Ca(2+) exchanger, glycogen metabolizing enzymes, glycolytic enzymes, and mitochondria/mitochondrial proteins. This functional coupling is bi-directional with many of these systems both being regulated by glutamate transport and providing the fuel to support glutamate uptake. Given the importance of glutamate uptake to maintaining synaptic signaling and preventing excitotoxicity, it should not be surprising that some of these systems appear to redundantly support the energetic costs of glutamate uptake. Although the glutamate-glutamine cycle contributes to recycling of neurotransmitter pools of glutamate, this is an over-simplification. The ramifications of co-compartmentalization of glutamate transporters with mitochondria for glutamate metabolism are discussed. Energy consumption in the brain accounts for ∼20% of the basal metabolic rate and relies almost exclusively on glucose for the production of ATP. However, the brain does not possess substantial reserves of glucose or other fuels. To ensure adequate energetic supply, increases in neuronal activity are matched by increases in cerebral blood flow via a process known as neurovascular coupling. While the mechanisms for this coupling are not completely resolved, it is generally agreed that astrocytes, with processes that extend to synapses and endfeet that surround blood vessels, mediate at least some of the signal that causes vasodilation. Several studies have shown that either genetic deletion or pharmacologic inhibition of glutamate transport impairs neurovascular coupling. Together these studies strongly suggest that glutamate transport not only coordinates excitatory signaling, but also plays a pivotal role in regulating brain energetics.


The Journal of Neuroscience | 2011

Nuclear Factor-κB Contributes to Neuron-Dependent Induction of Glutamate Transporter-1 Expression in Astrocytes

Mausam Ghosh; Yongjie Yang; Jeffrey D. Rothstein; Michael B. Robinson

The glutamate transporter-1 [GLT-1 (excitatory amino acid transporter 2)] subtype of glutamate transporter ensures crisp excitatory signaling and limits excitotoxicity in the CNS. Astrocytic expression of GLT-1 is regulated during development, by neuronal activity, and in neurodegenerative diseases. Although neurons activate astrocytic expression of GLT-1, the mechanisms involved have not been identified. In the present study, astrocytes from transgenic mice that express enhanced green fluorescent protein (eGFP) under the control of a bacterial artificial chromosome (BAC) containing a very large region of DNA surrounding the GLT-1 gene (BAC GLT-1 eGFP mice) were used to assess the role of nuclear factor-κB (NF-κB) in neuron-dependent activation of the GLT-1 promoter. We provide evidence that neurons activate NF-κB signaling in astrocytes. Transduction of astrocytes from the BAC GLT-1 eGFP mice with dominant-negative inhibitors of NF-κB signaling completely blocked neuron-dependent activation of a NF-κB reporter construct and attenuated induction of eGFP. Exogenous expression of p65 and/or p50 NF-κB subunits induced expression of eGFP or GLT-1 and increased GLT-1-mediated transport activity. Using wild-type and mutant GLT-1 promoter reporter constructs, we found that NF-κB sites at −583 or −251 relative to the transcription start site were required for neuron-dependent reporter activation. Electrophoretic mobility shift and supershift assays reveal that p65 and p50 interact with these same sites ex vivo. Finally, chromatin immunoprecipitation showed that p65 and p50 interact with these sites in adult cortex, but not in kidney (a tissue that expresses no detectable GLT-1). Together, these studies strongly suggest that NF-κB contributes to neuron-dependent regulation of astrocytic GLT-1 transcription.


European Journal of Neuroscience | 1998

Dihydrokainate-sensitive neuronal glutamate transport is required for protection of rat cortical neurons in culture against synaptically released glutamate.

Guang Jian Wang; Hye Joo Chung; Jamie Schnuer; Elizabeth Lea; Michael B. Robinson; William K. Potthoff; Elias Aizenman; Paul A. Rosenberg

Glutamate transport in nearly pure rat cortical neurons in culture (less than 0.2% astrocytes) is potently inhibited by dihydrokainate, l‐serine‐O‐sulphate, but not by l‐α‐amino‐adipate. This system allows for a test of the hypothesis that glutamate transport is important for protecting neurons against the toxicity of endogenous synaptically released glutamate. In support of this hypothesis, a 20–24 h exposure to 1 mm dihydrokainate reduced cell survival to only 14.8 ± 9.8% in neuronal cultures (P < 0.001;n = 3), although it had no effect on neuronal survival in astrocyte‐rich cultures (P > 0.05;n = 3). Dihydrokainate also significantly caused accumulation of glutamate in the extracellular medium of cortical neuronal cultures (6.6 ± 4.9 μm, compared to 1.2 ± 0.3 μm in control, n = 14, P < 0.01). The neurotoxicity of dihydrokainate was blocked by 10 μm MK‐801, 10 μm tetrodotoxin, and an enzyme system that degrades extracellular glutamate. The latter two also abolished the accumulation of glutamate in the extracellular medium. Dihydrokainate (1 mm) inhibited the 45calcium uptake stimulated by 30 μmN‐methyl‐d‐aspartate (NMDA), but not by higher concentrations consistent with a weak antagonist action of dihydrokainate at the NMDA receptor. Whole cell recordings showed that 1 mm dihydrokainate produced ≈ 25% inhibition of 30 μm NMDA‐induced current in cortical neurons. Dihydrokainate (1 mm) alone generated a small current (17% of the current produced by 30 μm NMDA) that was blocked by 30 μm 5,7‐dichlorokynurenate and only weakly by 10 μm 6‐cyano‐7‐nitroquinoxaline‐2,3‐dione (CNQX). These results suggest that the toxicity of dihydrokainate in neuronal cultures is due to its ability to block glutamate transport in these cultures, and that dihydrokainate‐sensitive neuronal glutamate transport may be important in protecting neurons against the toxicity of synaptically released glutamate.


Science Signaling | 2015

Regulation of brain glutamate metabolism by nitric oxide and S-nitrosylation

Karthik Raju; Paschalis-Thomas Doulias; Perry Evans; Elizabeth N. Krizman; Joshua G. Jackson; Oksana Horyn; Yevgeny Daikhin; Ilana Nissim; Marc Yudkoff; Itzhak Nissim; Kim A. Sharp; Michael B. Robinson; Harry Ischiropoulos

Protein S-nitrosylation controls the brain’s use of glutamate for energy or neurotransmission. Regulating neuronal glutamate status The gasotransmitter nitric oxide (NO) is generated by nitric oxide synthase (NOS) and can affect protein function by modifying cysteine residues in a process called S-nitrosylation. Mice lacking the neuronal NOS (nNOS) isoform have a phenotype that could be explained by decreased availability of glutamate, an amino acid that is also an excitatory neurotransmitter. Glutamate is derived from and can be converted to glutamine, and oxidation of glutamate feeds into the energy-providing TCA (tricarboxylic acid) cycle. Raju et al. found that mice lacking nNOS showed decreased S-nitrosylation of proteins involved in glutamate metabolism and uptake. In extracts from these mice, conversion of glutamate to glutamine was decreased, oxidation of glutamate was decreased, and glutamate uptake was increased. Nitric oxide (NO) is a signaling intermediate during glutamatergic neurotransmission in the central nervous system (CNS). NO signaling is in part accomplished through cysteine S-nitrosylation, a posttranslational modification by which NO regulates protein function and signaling. In our investigation of the protein targets and functional impact of S-nitrosylation in the CNS under physiological conditions, we identified 269 S-nitrosocysteine residues in 136 proteins in the wild-type mouse brain. The number of sites was significantly reduced in the brains of mice lacking endothelial nitric oxide synthase (eNOS−/−) or neuronal nitric oxide synthase (nNOS−/−). In particular, nNOS−/− animals showed decreased S-nitrosylation of proteins that participate in the glutamate/glutamine cycle, a metabolic process by which synaptic glutamate is recycled or oxidized to provide energy. 15N-glutamine–based metabolomic profiling and enzymatic activity assays indicated that brain extracts from nNOS−/− mice converted less glutamate to glutamine and oxidized more glutamate than those from mice of the other genotypes. GLT1 [also known as EAAT2 (excitatory amino acid transporter 2)], a glutamate transporter in astrocytes, was S-nitrosylated at Cys373 and Cys561 in wild-type and eNOS−/− mice, but not in nNOS−/− mice. A form of rat GLT1 that could not be S-nitrosylated at the equivalent sites had increased glutamate uptake compared to wild-type GLT1 in cells exposed to an S-nitrosylating agent. Thus, NO modulates glutamatergic neurotransmission through the selective, nNOS-dependent S-nitrosylation of proteins that govern glutamate transport and metabolism.


Developmental Brain Research | 1995

EVIDENCE OF EXCITOTOXICITY IN THE BRAIN OF THE ORNITHINE CARBAMOYLTRANSFERASE DEFICIENT SPARSE FUR MOUSE

Michael B. Robinson; Keri J Hopkins; Mark L. Batshaw; BethAnn McLaughlin; Melvyn P. Heyes; Mary Lou Oster-Granite

Ornithine carbamoyltransferase deficiency (OCTD) is the most common inborn error of urea synthesis. An X-linked disorder, OCTD males commonly present with hyperammonemic coma in the newborn period. There is a high rate of mortality and morbidity, with most survivors sustaining severe brain damage and resultant developmental disabilities. Although ammonia is presumed to be the principal neurotoxin, there is evidence that other neurochemical alterations may also be involved. The OCTD sparse fur (spf/Y) mouse has proven to be a useful model of this disease with similar metabolic and neurochemical alterations to those found in the human disease. In this study, the levels of the tryptophan derived excitotoxin quinolinic acid were examined in the brains of spf/Y mice. In addition, the neuropathology was examined using both light and electron microscopic approaches. Consistent with reports in children with urea cycle disorders, the levels of tryptophan and quinolinic acid were increased two-fold in various brain regions of the spf/Y mouse. Quinolinic acid, an agonist at the N-methyl-D-aspartate (NMDA) receptors, is known to produce selective cell loss in the striatum. We found a significant loss of medium spiny neurons and increased numbers of reactive oligodendroglia and microglia in the striatum of spf/Y mice. These neurochemical and neuropathological observations are consistent with an excitotoxic influence on brain injury in OCTD. It leads us to suggest that administration of NMDA receptor antagonists may ameliorate brain damage in children with inborn errors of urea synthesis.

Collaboration


Dive into the Michael B. Robinson's collaboration.

Top Co-Authors

Avatar

Joshua G. Jackson

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Elizabeth N. Krizman

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Mark L. Batshaw

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marc Yudkoff

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Robert Ghrist

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Sina Djali

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Barbara S. Slusher

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Dana M. Correale

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Deborah E. Bauer

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge