Chronic hypobaric hypoxia induced apoptosis in CA1 region of hippocampus: A possible role of NMDAR mediated p75NTR upregulation

https://doi.org/10.1016/j.expneurol.2008.01.030Get rights and content

Abstract

Hypobaric hypoxia has been implicated with neural degeneration and memory loss. Though there has been considerable knowledge on the role of the p75NTR in triggering apoptosis, the occurrence of a similar mechanism in hypoxic stress still remains to be explored. We, in the present study, have tried to explore the role of p75NTR in mediating apoptosis in hypobaric hypoxia. Male Sprague Dawley rats were exposed to an altitude of 7,620 m for different durations. To study the contribution of apoptosis to hypobaric hypoxia induced cell death in the hippocampus, rat brains were examined for the occurrence of apoptosis by determining the number of cells showing DNA breaks using terminal deoxynucleotidyl transferase-mediated UTP nick end labeling (TUNEL) assay and chromatin condensation using Hoechst staining along with estimation of caspase activity and expression of active Caspase 3. Expression of p75NTR was studied to determine its possible role in triggering apoptosis in hypobaric hypoxia. Exposure to hypobaric hypoxia was found to progressively increase the number of TUNEL positive and Hoechst positive cells along with increase in caspase activity, thus suggesting apoptotic mode of cell death. p75NTR was found to be upregulated on prolonged exposure to hypobaric hypoxia corresponding to the increase in the number of apoptotic cells. Further, reduced expression of p75NTR expression by antisense nucleotide administration significantly decreased apoptosis in the CA1 region of hippocampus. Blocking of NMDA receptors by MK801 interestingly decreased p75NTR expression and the number of TUNEL positive cells as compared to hypoxic animals. These findings suggest the regulation of p75NTR by NMDA receptors and its role in inducing apoptosis in hypoxia.

Introduction

Hypobaric hypoxia has been reported to cause memory impairment and cognitive dysfunctions (Kramer et al., 1993, Shukitt-Hale et al., 1998). Altered neurotransmitter synthesis, uptake and release (Benveniste et al., 1984, Rossi et al., 2000), free radical generation (Askew, 2002) and changes in gene expression and protein functions (Pellegrini-Giampietro et al., 1992, Gorter et al., 1997, Chandel et al., 1998) are typically associated with hypoxia and ischemia (Hartman et al., 2005). We have previously shown the occurrence of oxidative stress in hypobaric hypoxia (Maiti et al., 2006) that might play a key role in causing memory impairment. However limited neuroprotection has been reported on antioxidant supplementation in hypoxic and ischemic stress indicating involvement of other complex molecular mechanisms that might lead to neural degeneration in hypobaric hypoxia (Adcock et al., 2002). There also has been ample evidence on the occurrence of glutamate excitotoxicity in hypoxic stress and ischemia (Goldberg et al., 1987, Won et al., 2002). The CA1 region of the hippocampus in particular has been earlier reported to be vulnerable to hypoxic insult (Maiti et al., 2007). We have earlier reported on the altered expression of NMDA and AMPA receptor subunits in hypobaric hypoxia and the occurrence of excitotoxicity in the hippocampus (Hota et al., in press). However the events downstream of glutamate receptor activation in hypobaric hypoxia still remains an enigma. The role of neurotrophins in mediating neuronal damage in hypoxic stress has been a matter of debate in recent days. Though there has been reports on the neuroprotective role of neurotrophins and their receptors in hypoxic and ischemic stress (Narumiya et al., 1998), the findings on potentiation of neuronal injury by the neurotrophins (Behrens et al., 1999) has also drawn considerable attention.

Neurotrophins are a paradigmal family of neurotrophic factors that are crucial in the development and maintenance of the nervous system (Barde, 1989, Kaplan and Stephens, 1994, Lewin and Barde, 1996). The intracellular signaling cascades of neurotrophins are mediated by two discrete receptor subtypes. The agonist-selective high affinity trk family of neurotrophin receptors that selectively bind to different members of the neurotrophin family, with NGF binding to trkA, BDNF and neurotrophin-4/5 to trkB and neurotrophin 3 binding to trkC, mediate neuronal growth and survivability (Kaplan and Miller, 1997). The agonist-nonselective low affinity p75NTRs that form the second subtype on the other hand appear to be quite paradoxical and the signaling cascades pertaining to its activation still remain to be characterized.

The p75NTR is a 75-kDa glycoprotein with an extracellular domain containing four cysteine-rich repeats required for neurotrophin binding, a single transmembrane domain, and an intracellular domain that contains a Type II death domain and lacks catalytic activity (Dechant and Barde, 2002, Hempstead, 2002). Over expression of p75NTR induces apoptosis in neuronal cells (Frade et al., 1996, Roux et al., 1999, Friedman, 2000). In vivo over expression of the intracellular domain of p75NTR in transgenic mouse neurons leads to widespread apoptosis of peripheral and central neurons (Majdan et al., 1997). Similarly, disruption of ligand binding to p75NTR reduces apoptosis in the retina (Frade et al., 1996). Expression of p75NTR has also been reported to elicit death of hippocampal neurons (Carter and Lewin, 1997, Friedman, 2000). There also has been evidence on upregulation of these receptors after damage in several neuronal populations.

Caspase 3 activation has been shown to be central to the apoptotic pathway triggered by p75NTR over expression (Troy et al., 2002). This is further supported by the complete lack of Caspase 3 activation in the hippocampus of p75-null animals after pilocarpine treatment induced seizures (Troy et al., 2002). Though the molecular mechanisms involved in transcriptional regulation still remain to be elucidated, cell swelling and nitric oxide (NO) signaling cascades have been proposed to elicit p75NTR expression in conditions of neural damage (Peterson and Bogenmann, 2003). Similar upregulation of neuronal nitric oxide synthase (nNOS) in excitotoxic conditions therefore indicates towards a calcium mediated regulation of p75NTR expression.

In view of the above findings the present study aimed at exploring the occurrence of apoptotic neuronal death in the CA1 region of the hippocampus of rats on exposure to different durations of hypobaric hypoxia at a simulated altitude of 7620 m by Terminal deoxynucleotidyl transferase-mediated UTP nick end labeling (TUNEL), Hoechst staining, and estimation of Caspase 3 activity. The expression of p75NTR on different durations of hypoxic exposure was also studied. Regulation of p75NTR expression by NMDAR activation was explored by administering the selective NMDAR antagonist MK801 and studying its effect on p75NTR expression during hypobaric hypoxia.

Section snippets

Materials

Primary antibodies for p75, active Caspase 3 and GAPDH, blocking serum and HRP-conjugated secondary antibodies were obtained from Santa Cruz. Chemiluminescent substrate kit, Caspase 3 activity assay kit and Hoechst 33342 were obtained from Sigma Chemicals. Chemicals for western blot were procured from Biorad. TUNEL Assay kit was purchased from Chemicon. All other chemicals and reagents were supplied by Sigma Chemicals.

Animals

Experiments were conducted with male Sprague Dawley rats weighing 220 ± 15 g

DNA fragmentation

DNA fragmentation in the hippocampal region which is also an indicator of apoptosis was studied by TUNEL assay. Nearly 2% of cells in the CA1 region of control animals showed DNA fragmentation which increased to more than 10% when the animals were exposed to an altitude of 25,000 ft (7620 m) for 3 days (Fig. 1). A progressive increase in DNA fragmentation was observed with the increase in duration of exposure (20% on 7 days of exposure and 25% on 14 days of exposure) as shown in Fig. 1.

Chromatin condensation

Brain

Discussion

Hypobaric hypoxia, a condition prevalent at high altitude due to low partial pressure of oxygen, is known to cause cognitive dysfunctions and memory impairment (Shukitt-Hale et al., 1998). Occurrence of oxidative stress and neuronal degeneration has been attributed for the occurrence of memory impairment on exposure to extreme high altitude (Maiti et al., 2006). However supplementation of antioxidants alone has not been able to completely ameliorate the memory impairment at high altitude. The

Acknowledgment

The study was completely supported by Defence Research and Development Organization, Ministry of Defence, Government of India.

References (63)

  • ErnforsP. et al.

    Expression of nerve growth factor receptor mRNA is developmentally regulated and increased after axotomy in rat spinal cord motoneurons

    Neuron

    (1989)
  • GallC. et al.

    Kainic acid-induced seizures stimulate increased expression of nerve growth factor mRNA in rat hippocampus

    Mol. Brain Res.

    (1991)
  • HartmanR.E. et al.

    Characterizing learning deficits and hippocampal neuron loss following transient global cerebral ischemia in rats

    Brain Res.

    (2005)
  • HempsteadB.L.

    The many faces of p75NTR

    Curr. Opin. Neurobiol.

    (2002)
  • KaplanD.R. et al.

    Signal transduction by the neurotrophin receptors

    Curr. Opin. Cell Biol.

    (1997)
  • KokaiaZ. et al.

    Focal cerebral ischemia in rats induces expression of p75 neurotrophin receptor in resistant striatal cholinergic neurons

    Neuroscience

    (1998)
  • MaitiP. et al.

    Hypobaric hypoxia induces oxidative stress in rat brain

    Neurochem. Int.

    (2006)
  • MaitiP. et al.

    Hypobaric hypoxia damages the hippocampal pyramidal neurons in the rat brain

    Brain Res.

    (2007)
  • MukaiJ. et al.

    NADE, a p75NTR associated cell death executor, is involved in signal transduction mediated by the common neurotrophin receptor p75NTR

    J. Biol. Chem.

    (2000)
  • NarumiyaS. et al.

    Enhanced expression of full-length TrkB receptors in young rat brain with hypoxic/ischemic injury

    Brain Res.

    (1998)
  • PetersonS. et al.

    Osmotic swelling induces p75 neurotrophin receptor (p75NTR) expression via nitric oxide⁎

    J. Biol. Chem.

    (2003)
  • RiceA.C. et al.

    NMDA receptor activation during status epilepticus is required for the development of epilepsy

    Brain Res.

    (1998)
  • RouxP.P. et al.

    Neurotrophin signaling through the p75 neurotrophin receptor

    Prog. Neurobiol.

    (2002)
  • RydenM. et al.

    Differential modulation of neuron survival during development by nerve growth factor binding to the p75 neurotrophin receptor

    J. Biol. Chem.

    (1997)
  • TroyC.M. et al.

    Mechanisms of p75-mediated death of hippocampal neurons. Role of caspases

    J. Biol. Chem.

    (2002)
  • VerdiJ.M. et al.

    P75LNGFR regulates Trk signal transduction and NGF induced neuronal differentiation in MAH cells

    Neuron

    (1994)
  • YuanJ. et al.

    Diversity in mechanisms of neuronal cell death

    Neuron

    (2003)
  • ZimmermannK.C. et al.

    How cells die: apoptosis pathways

    J. Allergy Clin. Immunol.

    (2001)
  • K.H. Adcock et al.

    Neuroprotection of creatine supplementation in neonatal rats with transient cerebral hypoxia–ischemia

    Dev. Neurosci.

    (2002)
  • AloyzR.S. et al.

    p53 is essential for developmental neuron death as regulated by the TrkA and p75 neurotrophin receptors

    J. Cell Biol.

    (1998)
  • ArmstrongD.M. et al.

    Expression of choline acetyltransferase and nerve growth factor receptor within hypoglossal motoneurons following nerve injury

    J. Comp. Neurol.

    (1991)
  • Cited by (36)

    • Effects of datumetine on hippocampal NMDAR activity

      2021, Toxicology Reports
      Citation Excerpt :

      Similar to our results, exposure to hypoxia reduced BDNF expression and increase the expression of NMDAR [115]. The authors attributed the reduced expression of BDNF observed to increase in activity of GluN2B subunit of NMDAR as they have earlier reported increase activity of NMDAR in hypoxic condition [117,118]. Although the specific subunit of datumetine binding to NMDAR is not known, whether it requires the presence of glutamate at the synapse for its activity.

    • Molecular regulation of dendritic spine dynamics and their potential impact on synaptic plasticity and neurological diseases

      2015, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      Since the last two decade, our laboratory has worked with hypobaric hypoxia, an adverse environmental condition encountered at high altitude. Similar to ischemic/hypoxic damage, we have also observed that hypobaric hypoxia increases oxidative stress, including reactive oxygen (ROS) and reactive nitrogen species (RNS; Maiti et al., 2006), glutamate excitotoxicity (Hota et al., 2008a,b), increases in stress hormones especially glucocorticoids (Baitharu et al., 2013), which cause neuronal death and dendritic spine loss in cortical and hippocampal neurons leading to impaired learning and memory in rats (Maiti et al., 2008a). We have also been studying the effects of different altitudes (3000–8000 m) and duration of exposure to the degree of neuronal vulnerability, as well as synaptic damage, and we have observed that the degree of spine loss depends on the severity of altitude and duration of stay in that environment (Fig. 18; Maiti et al., 2008a).

    • Investigation of prolonged hypobaric hypoxia-induced change in rat brain using T2 relaxometry and diffusion tensor imaging at 7T

      2015, Neuroscience
      Citation Excerpt :

      The CC has also been reported to be sensitive to hypoxic damage (Hackett and Roach, 2004; Kallenberg et al., 2008; Bailey et al., 2009). Previous biochemical studies performed on the rodent brain (especially the hippocampal region) have shown oxidative stress (Barhwal et al., 2007), apoptosis (Hota et al., 2008), and neurodegeneration (Maiti et al., 2010), in response to prolonged HH exposure. Harik et al. (1995) and Barhwal et al. (2007) reported that prolonged hypoxia increases brain micro-vasculature as an adaptive response to hypoxia.

    View all citing articles on Scopus
    View full text