Abstract

In order to meet the energetic demands of cell-to-cell signaling, increases in local neuronal signaling are matched by a coordinated increase in local blood flow, termed neurovascular coupling. Multiple different signals from neurons, astrocytes, and pericytes contribute to this control of blood flow. Previously, several groups demonstrated that inhibition/ablation of glutamate transporters attenuates the neurovascular response. However, it was not determined if glutamate transporter activation was sufficient to increase blood flow. Here, we used multiphoton imaging to monitor the diameter of fluorescently labeled cortical arterioles in anesthetized C57/B6J mice. We delivered vehicle, glutamate transporter substrates, or a combination of a glutamate transporter substrate with various pharmacologic agents via a glass micropipette while simultaneously visualizing changes in arteriole diameter. We developed a novel image analysis method to automate the measurement of arteriole diameter in these time-lapse analyses. Using this workflow, we first conducted pilot experiments in which we focally applied L-glutamate, D-aspartate, or L-threo-hydroxyaspartate (L-THA) and measured arteriole responses as proof of concept. We subsequently applied the selective glutamate transport substrate L-THA (applied at concentrations that do not activate glutamate receptors). We found that L-THA evoked a significantly larger dilation than that observed with focal saline application. This response was blocked by co-application of the potent glutamate transport inhibitor, L-(2S,3S)-3-[3-[4-(trifluoromethyl)-benzoylamino]benzyloxy]-aspartate (TFB-TBOA). Conversely, we were unable to demonstrate a reduction of this effect through co-application of a cocktail of glutamate and GABA receptor antagonists. These studies provide the first direct evidence that activation of glutamate transport is sufficient to increase arteriole diameter. We explored potential downstream mechanisms mediating this transporter-mediated dilation by using a Ca2+ chelator or inhibitors of reversed-mode Na+/Ca2+ exchange, nitric oxide synthetase, or cyclo-oxygenase. The estimated effects and confidence intervals suggested some form of inhibition for a number of these inhibitors. Limitations to our study design prevented definitive conclusions with respect to these downstream inhibitors; these limitations are discussed along with possible next steps. Understanding the mechanisms that control blood flow are important because changes in blood flow/energy supply are implicated in several neurodegenerative disorders and are used as a surrogate measure of neuronal activity in widely used techniques such as functional magnetic resonance imaging (fMRI).

Highlights

  • The brain matches increases in local metabolic demand caused by neuronal activity with corresponding increases in local blood flow, in a process termed neurovascular coupling (NVC)

  • We provide direct evidence that activation of glutamate transport is sufficient to cause an increase in arteriole diameter

  • The bulk of glutamate transport is mediated by two Na+dependent glutamate transporters, called GLT-1 and GLAST that are enriched in astrocytes (Rothstein et al, 1994; Danbolt et al, 2016)

Read more

Summary

Introduction

The adult human brain represents about 2% of total body weight, but consumes up to 20% of the basal metabolic rate (for reviews, see Shulman et al, 2004; Hertz et al, 2007; Harris et al, 2012; Stobart and Anderson, 2013; Weber and Barros, 2015). The brain matches increases in local metabolic demand caused by neuronal activity with corresponding increases in local blood flow, in a process termed neurovascular coupling (NVC). This process supports the energetic costs of cellular signaling, including costs associated with vesicular transmitter packaging/release and membrane repolarization after activation of ligand-/voltage-gated ion channels (Howarth et al, 2012). Neurovascular coupling provides the signal for functional/BOLD magnetic resonance imaging (fMRI) studies. It underlies some versions of 2-deoxyglucose-based positron emission tomography (PET) imaging, and near-infrared spectroscopy (Raichle and Mintun, 2006; Hillman, 2014)

Objectives
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.