Short- and long-term changes in extracellular glutamate and acetylcholine concentrations in the rat hippocampus following hypoxia

https://doi.org/10.1016/j.neuint.2012.03.009Get rights and content

Abstract

Hypoxia at birth is a major source of brain damage and it is associated with serious neurological sequelae in survivors. Alterations in the extracellular turnover of glutamate (Glu) and acetylcholine (ACh), two neurotransmitters that are essential for normal hippocampal function and learning and memory processes, may contribute to some of the neurological effects of perinatal hypoxia. We set out to determine the immediate and long-lasting effects of hypoxia on the turnover of these neurotransmitters by using microdialysis to measure the extracellular concentration of Glu and ACh in hippocampus, when hypoxia was induced in rats at postnatal day (PD) 7, and again at PD30. In PD7 rats, hypoxia induced an increase in extracellular Glu concentrations that lasted for up to 2.5 h and a decrease in extracellular ACh concentrations over this period. By contrast, perinatal hypoxia attenuated Glu release in asphyxiated rats, inducing a decrease in basal Glu levels when these animals reached PD30. Unlike Glu, the basal ACh levels in these animals were greater than in controls at PD30, although ACh release was stimulated less strongly than in control animals. These results provide the first evidence of the initial and long term consequences of the hypoxia on Glu and ACh turnover in the brain, demonstrating that hypoxia produces significant alterations in hippocampal neurochemistry and physiology.

Highlights

► In the hippocampus, extracellular Glu levels increase during hypoxia in PD7 rats. ► In the hippocampus, extracellular Ach levels decrease during hypoxia in PD7 rats. ► Neonatal hypoxia induces long term alterations in extracellular Glu and ACh levels in PD30 rats.

Introduction

During the postnatal period, hypoxia due to birth complications is a major cause of brain damage (Pasupathy et al., 2009) and this perinatal hypoxia is an important public health issue in many countries. Depending on the severity, hypoxia can lead to perinatal asphyxia syndrome and hypoxic-ischemic encephalopathy and it is associated with a significant number of newborn deaths. Moreover, survivors of brain injury due to oxygen deprivation have a considerable risk of developing neurological complications such as schizophrenia, learning disabilities, seizures, cerebral palsy and mental retardation (Nelson and Grether, 1999, McNeil et al., 2000, Mitchell, 2009).

Hypoxia is produced by a decrease in oxygen availability to the entire organism, resulting in changes in blood flow to the brain, heart and adrenal glandules (Williams et al., 1993, Guerri et al., 2008, Momen et al., 2009). Respiratory and metabolic acidosis also have been described in hypoxic conditions due to the overproduction of lactic acid by interruption of the Krebs cycle and secondary hyponatremia (Na+ plasma concentrations below 135 mmol/L: Moritz and Ayus, 2010, Duan et al., 2011). This immediate systemic response is accompanied by a series of alterations in the brain that affect neuronal function and neurotransmitter turnover. Extracellular acidosis may interfere with the normal function of certain K+ channels, such as acid-sensitive K+-channels of the tandem P-domain K+-channel family (TASKs) and neuronal voltage-gated K+ (kV) channels (Xiong et al., 2007, Trapp et al., 2008, Ortiz et al., 2009). Alterations in intracellular Na2+ homeostasis (Sheldon et al., 2004) and changes in the normal oscillation of mitochondrial NADPH (Mironov and Richter, 2001, Ratan et al., 2007) have also been demonstrated during hypoxia. Together, these observations suggest that hypoxia induces significant alterations in the normal ionic environment in the brain (Jiang et al., 1992, Muller and Somjen, 2000) and that it affects ATP production. Increases in [Ca2+]i and Ca2+-dependent neuronal damage have been described in hypoxic conditions (Oka et al., 2003, Pandit and Buckler, 2009), suggesting that Ca2+-dependent vesicle fusion, a pre-requisite for neurotransmitter release, may be affected. These observations suggest that hypoxia may induce changes in the extracellular concentration of neurotransmitters in the brain.

Glutamate (Glu) and acetylcholine (ACh) are two excitatory neurotransmitters that participate in the activation of hippocampal neuronal pathways involved in learning and memory. Significantly, the levels of both neurotransmitters are altered in children exposed to hypoxia during the neonatal period (Sun et al., 2002). In neurons, Glu is mainly produced from α-ketoglutarate, an intermediate in the Krebs cycle, and it accumulates in intracellular synaptic vesicles from which it is released in response to Ca2+-induced depolarization. The postsynaptic effects of Glu are mediated by ionotropic receptors, named according to their selective agonist: N-Methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA), and kainate (KA). These receptors are cation channels and their opening is enhanced by Glu binding to specific sites within the receptor structure, resulting in the production of excitatory postsynaptic potentials (Hassel and Digledine, 2006, Rambhadran et al., 2010, Terhag et al., 2010). In addition to the ionotropic receptors, Glu can also activate eight distinct metabotropic receptors (mGlur1 to mGluR8). These receptors share a common structural design of seven transmembrane domains, typical of G protein-coupled receptors. mGluRs are associated with a wide array of diverse cytoplasmic signaling enzymes, including phospholipase C and adenylate cyclase (Hassel and Digledine, 2006, Traynelis et al., 2010). Glu is released in the hippocampus, acting in the trisynaptic circuit between the dentate gyrus and the CA fields.

ACh is the product of the acetylation of choline by acetyl coenzyme A (acetyl-CoA) that is stored in cytoplasmic vesicles, from which it is released in response to an influx of Ca2+. ACh is considered a regulatory neurotransmitter in the brain, where it is involved in memory storage and retrieval, long-term potentiation and attention pathways (Terry, 2006, Buchanan et al., 2010, Bailey et al., 2010). ACh exerts its postsynaptic effects through two types of receptors:nicotinic cation-activated receptors and muscarinic G protein–coupled receptors (Taylor and Brown, 2006, Albuquerque et al., 2009). Importantly, the metabolism of both Glu and ACh metabolism is dependent on oxygen supply.

Increases in extracellular Glu concentrations have been described in animal models involving hypoxia, such as hypoxia–ischemia induced by experimental vascular manipulation (Park et al., 2010), or in brain slices (Dos-Anjos et al., 2009) and cell cultures (Lehmann et al., 2009, Sivakumar et al., 2010). Similarly, the availability of ACh in neuronal tissue during hypoxia has been indirectly explored (Chathu et al., 2008). However, few studies have directly assessed the temporal profile of Glu and ACh in a pure in vivo model of hypoxia, uncomplicated by cerebral ischemia. The neonatal seizure model (Jensen et al., 1995) is a useful model to actively monitor the extracellular concentration of neurotransmitters in conscious animals during and after the induction of hypoxia. In the present study, we have investigated the immediate and long-term effects of neonatal oxygen deprivation on extracellular levels of Glu and ACh in the hippocampus of freely moving rats. We observed a large increase in extracellular Glu levels during hypoxia in rats on postnatal day (PD) 7, which lasted for up to 2 h after the induction of hypoxia. Upon reaching PD30, these animals exhibited lower basal Glu concentrations than control animals. Extracellular ACh concentrations decreased in PD7 rats during hypoxia, although they surpassed the control levels by PD30.

Section snippets

Animals

Pregnant Wistar rats (n = 32) were housed in independent cages, with 12 × 12 light cycle and ad libitum access to food and water. At birth, only males were selected to form litters of eight animals. The pups remained with their mother until PD7. All efforts were made to minimize animal suffering, and to reduce the number of animals used.

Surgery at PD7

PD7 rats were implanted with a microdialysis probe (CMA/7, 7 mm shaft length and 2mm membrane length, mean recovery 32 ± 5.6%) in the dorsal hippocampus under ether

Results

Exposure to hypoxic conditions (8% O2/92% N2) resulted in an increased rate of respiration, immobility and a characteristic head clonus, all of which were observed for up to3 h hours after hypoxia induction. Only rats that exhibited all of these characteristics were included in the study.

The enzymatic method for Glu determination was sensitive and produced a linear response in the concentration range used (0.75–25 μM, r2 = 0.997: Fig. 2, insert). Accordingly, it was deemed to be suitable to measure

Discussion

We have quantified the concentration of extracellular Glu and ACh before, during and after PD7 rats were exposed to hypoxia and we investigated the long term effects of hypoxia by measuring the concentrations of these neurotransmitters in PD 30 rats exposed to perinatal hypoxia. Perinatal hypoxia is a common cause of neonatal seizures and encephalopathy, and it can result in permanent brain damage, although the precise mechanisms underlying these effects remain unknown. To explore the roles of

Acknowledgements

This study was supported by PROMEP (EXB-086) and CONACyT-CB (105807)

References (69)

  • H. Luna-Munguia et al.

    Effects of high frecuency electrical stimulation and R-verapamil on seizure susceptibility and glutamate and GABA release in a model of phenytoin-resistant seizures

    Neuropharmacology

    (2011)
  • T. McNeil et al.

    Obstetric complications and congenital malformation in schizophrenia

    Brain Res. Rev.

    (2000)
  • R. Otoya et al.

    Acute and long-lasting effects of neonatal hypoxia on (+)-3-[125I]MK-801 binding to NMDA brain receptors

    Exp. Neurol.

    (1997)
  • J. Pandit et al.

    Differential effects of halothane and sevoflurane onhypoxia-induced intracellular calcium transients of neonatal rat carotid body type I cells

    Br. J. Anaesth.

    (2009)
  • E. Park et al.

    Correlation between extracellular glutamate release and neuronal cell death in an eleven vessel occlusion model in rat

    Brain Res.

    (2010)
  • A. Rambhadran et al.

    Subunit arrangement in N-methyl-d-aspartate (NMDA) receptors

    J. Biol. Chem.

    (2010)
  • B. Row et al.

    Impaired spatial working memory and altered choline acetyltransferase (CHAT) immunoreactivity and nicotinic receptor binding in rats exposed to intermittent hypoxia during sleep

    Behav. Brain Res.

    (2007)
  • A. Rubaj et al.

    The epileptigenic effect of seizures induced by hypoxia. The role of NMDA and AMPA/KA antagonist

    Pharmacol. Biochem. Behav.

    (2003)
  • C. Williams et al.

    Pathophysiology of perinatal asphyxia

    Clin. Perinatol.

    (1993)
  • N. Woolf et al.

    Cholinergic systems in the rat brain. I. Projections to the limbic telencephalon

    Brain Res. Bull.

    (1984)
  • E. Albuquerque et al.

    Mammalian nicotinic acetylcholine receptors: from structure to function

    Physiol. Rev.

    (2009)
  • Bailey, C., De Biasi, M., Fletcher, P., Lambe, E., 2010. The nicotinic acetylcholine receptor α5 subunit plays a key...
  • A. Bekker et al.

    Physostigmine reverses cognitive dysfunction caused by moderate hypoxia in adult mice

    Anesth. Analg.

    (2007)
  • R. Burke et al.

    Quantitative morphological analysis of striatal cholinergic neurons in perinatal asphyxia

    Ann. Neurol.

    (1990)
  • G. Ceresoli-Borroni et al.

    Neonatal asphyxia in rats: acute effects on cerebral kynurenine metabolism

    Pediatr. Res.

    (2001)
  • E. Chleide et al.

    Hypoxia-induced decrease of brain acetylcholine release detected by microdialysis

    Neuroreport

    (1990)
  • M. Dallas et al.

    Hypoxia suppresses glutamate transport in astrocytes

    J. Neurosci.

    (2007)
  • B. Duan et al.

    Extracellular spermine exacerbates ischemic neuronal injury through sensitization of ASIC1A channels to extracellular acidosis

    J. Neurosci.

    (2011)
  • F. Galeffi et al.

    Simultaneous monitoring of tissue PO2 and NADH fluorescence during synaptic stimulation and spreading depression reveals a transient dissociation between oxygen utilization and mitochondrial redox state in rat hippocampal slices

    J. Cereb. Blood Flow Metab.

    (2011)
  • G. Gibson et al.

    Impaired synthesis of acetylcholine by mild hypoxic hypoxia or nitrous oxide

    J. Neurochem.

    (1981)
  • G. Gibson et al.

    Decreases in amino acids and acetylcholine metabolism during hypoxia

    J. Neurochem.

    (1981)
  • R. Guerri et al.

    Effects of hypoxia in the chemobaroreflex interaction

    Rev. Fed. Arg. Cardiol.

    (2008)
  • B. Hassel et al.

    Glutamate

  • G. Hilton et al.

    Glutamate-mediated excitotoxicity in neonatal hippocampal neurons is mediated by mGluR-induced release of Ca2+ from intracellular stores and is prevented by estradiol

    Eur. J. Neurosci.

    (2006)
  • Cited by (13)

    • Pharmacological inhibition of poly (ADP-ribose) polymerase by olaparib, prevents acute lung injury associated cognitive deficits potentially through suppression of inflammatory response

      2020, European Journal of Pharmacology
      Citation Excerpt :

      Additionally, we have observed no significant adverse effect on motor function (motor coordination/spontaneous motor activity), upon ALI induction (Sahu et al., 2018). Further, an elevation in serum pro-inflammatory markers (TNF-α and IL-1β) has been reported to be co-related with cognitive defects (Huber et al., 2001; Lopez-Perez et al., 2012; Lv et al., 2010). It appears that olaparib, when given 30 min before each hit, is capable of preventing the induction of the systemic inflammation, as reflected by reduced expression of the aforementioned inflammatory cytokines at both the mRNA and the protein levels.

    • Two hit induced acute lung injury impairs cognitive function in mice: A potential model to study cross talk between lung and brain

      2018, Brain, Behavior, and Immunity
      Citation Excerpt :

      Perhaps, the inflammatory mediators produced and persisted during ALI/ARDS, can exert their detrimental effects by inducing some dangerous cross talk between lung and brain (Stevens and Puybasset, 2011). Indeed, increase in systemic pro-inflammatory factors has been linked to the compromised cognitive function (Farkas et al., 2007; Hopkins, 2007; Huber et al., 2001; Lopez-Perez et al., 2012; Lv et al., 2010). Despite recent advances in understanding the pathophysiology, treatment and long-term outcomes of ALI/ARDS, the incidence and prevalence of the condition and its associated cognitive impairment, remain uncertain which in turn reflects the heterogeneity of the syndrome (Dowdy et al., 2006; Hopkins et al., 2004; Hopkins et al., 2005; Stevens and Puybasset, 2011; Gonzalvo et al., 2007; Lopez-Aguilar et al., 2013).

    • Do sedation and analgesia contribute to long-term cognitive dysfunction in critical care survivors?

      2018, Medicina Intensiva
      Citation Excerpt :

      Hypoxemia events, dyspnea and air hunger, and even the MV itself are important components to consider. Experimental studies have shown that systemic PaO2 oscillations cause mild brain injury64; specifically, brain structures such as the hippocampus are extremely vulnerable to hypoxemia.65 Furthermore, dyspnea and air hunger cause activation of the afferent pathways through chemo or baroreceptors, reflected by an increase in the signal in magnetic resonance in brain areas such as the limbic system which are related to psychological, emotional and memory disorders.66

    • A novel method for simultaneous glutamate and extracellular activity measurement in brain slices with high temporal resolution

      2015, Talanta
      Citation Excerpt :

      Enzyme assays present a new alternative methodology. These consist of converting Glu and other neurotransmitters to a derivate, which is mainly based on the generation of hydrogen peroxide (H2O2) as a product of the reaction by different series of specific enzymes for each neurotransmitter, that can be detected by electrochemical [18], fluorescence [19] and luminescence procedures [20]. One of the most common brain preparations used for in vitro studies is the brain slice, which retains many of the aspects of the in vivo brain region and permits the study of the physiology and pharmacology of the brain in more detail and with the use of fewer animals than in vivo models.

    • Simultaneous recording of hippocampal oxygen and glucose in real time using constant potential amperometry in the freely-moving rat

      2013, Journal of Neuroscience Methods
      Citation Excerpt :

      However, there has been less attention has been afforded to the hippocampus (Freund et al., 1989; Hu and Wilson, 1997) despite its pivotal roles in declarative memory (Scoville and Milner, 1957; Cohen et al., 1999; Eacott and Easton, 2011), spatial navigation (O’Keefe and Nadel, 1978; Morris et al., 1982; D’Hooge and De Deyn, 2001) and its links to various neurodegenerative/psychiatric disorders (Heckers and Konradi, 2010; Marlatt and Lucassen, 2010; Bast, 2011; Dhikav and Anand, 2011; Bonilha et al., 2012). Therefore, there is a need for sensor data recorded in real-time to supplement the large amount of electrophysiological data (Bliss and Lomo, 1973; Martin et al., 2000; Morris et al., 2003; Colgin and Moser, 2010), microdialysis data (McNay et al., 2001; Gold, 2003; De Bundel et al., 2009; López-Pérez et al., 2012) and molecular data from tissue samples (Gooney et al., 2002; Minichiello, 2009; Barry and Commins, 2011) already published from work in the hippocampus. Some behavioural work with hippocampal sensors has been performed; it has been recently shown that oxygen sensors can be used to differentiate between the activity of the dorsal and ventral regions of the hippocampus in a spatial memory task (McHugh et al., 2011).

    View all citing articles on Scopus
    View full text