Elsevier

Free Radical Biology and Medicine

Volume 63, October 2013, Pages 421-431
Free Radical Biology and Medicine

Orginal Contribution
Signaling through the vascular endothelial growth factor receptor VEGFR-2 protects hippocampal neurons from mitochondrial dysfunction and oxidative stress

https://doi.org/10.1016/j.freeradbiomed.2013.05.036Get rights and content

Highlights

  • Nutritional stress upregulates VEGF and its receptors in mature hippocampal neurons.

  • Activated VEGFR-2 signals neuroprotection through the PI3K/Akt and ERK1/2 pathways.

  • VEGFR-2 inactivation decreased viability with mitochondrial dysfunction and Oxidative stress.

  • VEGF and VEGF-B signaling or NAC counteract the insult induced by VEGFR-2 inhibition.

Abstract

Vascular endothelial growth factor VEGF (VEGF-A or VEGF165) is a potent angiogenic factor that also signals neuroprotection through activation of its cognate receptor VEGFR-2. In this capacity, VEGF signaling can rescue neurons from the damage induced by stressful stimuli many of which elicit oxidative stress. However, the regulatory role that VEGFR-2 plays in providing neuroprotection remains elusive. Therefore, we investigated the effects of VEGFR-2 inhibition on primary cultures of mature hippocampal neurons undergoing nutritional stress. We found that neurons cultured under nutritional stress had increased expression of VEGF and its receptors, VEGFR-1, VEGFR-2, and NP-1, as well as enhanced levels of VEGFR-2 phosphorylation. These neurons also showed increased activation of the prosurvival pathways for MEK/ERK1/2 and PI3K/Akt, enhanced phosphorylation (inactivation) of the proapoptotic BAD, and higher levels of the antiapoptotic protein Bcl-xL, all of which were augmented by treatments with exogenous VEGF and blocked by VEGFR-2 inhibition. The blockade of VEGFR-2 function also elicited a cytotoxicity that was accompanied by caspase-3 activation, induction of hemeoxygenase-1 (HO-1), oxidative stress, and a collapse in the mitochondrial membrane potential (ΔΨm). Knockdown of VEGFR-2 by siRNA generated a similar pattern of redox change and mitochondrial impairment. Pretreatments with VEGF, VEGF-B, or the antioxidant N-acetylcysteine (NAC) rescued SU1498 or siRNA-treated neurons from the mitochondrial dysfunction and oxidative stress induced by VEGFR-2 inhibition in a timely fashion. These findings suggested that VEGF or VEGF-B can provide neuroprotection by signaling through an alternate VEGF receptor. Together, our findings suggest that VEGF signaling through VEGFR-2 plays a critical regulatory role in protecting stressed hippocampal neurons from the damaging effects of an oxidative insult. These findings also implicate VEGFR-1 or NP-1 as compensatory receptors that mediate neuroprotection when VEGFR-2 function is blocked.

Introduction

Oxidative stress and mitochondrial dysfunction contribute to the pathogenesis of a number of neurodegenerative disorders including Alzheimer's and Parkinsonʼs disease [1]. One approach for protecting neurons from oxidative insults is to administer neurotrophic growth factors. One such factor is VEGF (VEGF-A or VEGF165) which is a mitogen that stimulates angiogenesis and neuroprotection through autocrine or paracrine mechanisms [2], [3]. In addition, VEGF can stimulate axonal outgrowth, and rescue rat mesencephalic neurons or hippocampal cells from death induced by serum withdrawal, ischemia, hypoxia, and glutamate-induced toxicity [4], [5], [6] and elicit neuroprotection via angiogenesis and neurogenesis [7], [8], [9]. Delineating this neuroprotective mechanism is complex since VEGF can undergo cell-surface interactions with different cognate tyrosine kinase receptors such as VEGFR-1, VEGFR-2, and the non-tyrosine kinase members of the neuropilin family of class 3 semaphorin receptors Neuropilin-1 and -2 (NP-1, NP-2) receptors [10]. While VEGF mediates most biological effects through VEGFR-2, it can interact with NP-1 as a coreceptor that enhances VEGF signaling. Ligand binding to VEGFR-1 and VEGFR-2 results in receptor dimerization followed by autophosphorylation and activation of downstream signaling cascades [3]. In neurons, NP-1 is a cell-surface receptor for both VEGF and class 3 semaphorins and plays a functional role in axonal pathfinding, retraction, and collapse during development. While the function of VEGFR-1 remains unclear, it is implicated as a decoy receptor that sequesters VEGF from activating VEGFR-2. VEGF-B, which is a ligand for VEGFR-1 but not VEGFR-2, is poorly angiogenic in the brain but can protect against mitochondrial membrane permeabilization [11], proapoptotic gene expression [12], and is coexpressed with genes encoding mitochondrial proteins [13]. Thus, VEGF-B may protect against mitochondrial dysfunction independent of angiogenesis.

Oxidative insults are implicated as causative factors of neuronal damage in several different neurological disorders and increasing evidence from in vitro and in vivo studies shows that VEGF signaling protects neurons from insults which are known to induce oxidative stress [14], [15]. In this context, VEGF overexpression has been shown to delay the onset of neuronal death in an animal model of amyotrophic lateral sclerosis (ALS) where oxidative stress is a contributing factor [16]. VEGF has been shown to mediate neuroprotection under stress conditions through the downstream activation of the phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinase MEK/ERK1/2 pathways [6], [17], [18], [19] and by suppressing caspase activation [20]. While several of these studies show that VEGF activates VEGFR-2 to mediate survival, the protective capacity of VEGFR-2 signaling remains unclear. Therefore, the aim of these studies is to determine the impact of VEGFR-2 signaling on the survival of mature hippocampal neurons by blocking its activation using pharmacological and gene silencing methods.

In previous studies we used a neuronal model of serum-deprived SK-N-SH cells to show that VEGF signaling through VEGFR-2 prevents a caspase-dependent cell death [21]. Therefore, we addressed whether VEGF signaling through VEGFR-2 protected rat hippocampal neurons from the harmful effects of nutritional stress. Depriving cultured neurons from vital nutrients provide a viable model to identify the molecular basis of neuronal insults that would occur under pathological conditions in vivo. Our findings show that a blockade of VEGFR-2 function in hippocampal neurons leads to a rapid loss in viability that is manifested by induction of markers of oxidative stress, mitochondrial dysfunction, and a loss in the activation of prosurvival pathways. Notably, the inclusion of exogenous VEGF or VEGF-B mediates a time-dependent rescue from this response, suggesting that a molecular switch to an alternate receptor can provide neuroprotection when VEGFR-2 activity is blocked. Our findings establish a link between VEGFR-2 signaling and mitochondrial function in differentiated rat neurons and provide insight on oxidative stress-related pathways that mediate neuronal damage and how exogenous VEGF or VEGF-B may counteract these events.

Section snippets

Materials

Recombinant human vascular endothelial growth factor 165 (VEGF165) and VEGF-B were obtained from PeproTech Inc (Rocky Hill, NJ). The inhibitors of VEGFR-2 (SU1498) and PI3K/Akt (Wortmannin) were obtained from EMD Biosciences Inc. (San Diego, CA). The inhibitor of MEK1/2 (U0126) was obtained from Promega Corporation (Madison, WI). The antioxidant N-acetylcysteine (NAC) was purchased from Sigma-Aldrich (St. Louis, MO).

Primary cell culture and treatments

All animal studies were performed in accordance with the National Institutes of

VEGFR-2 signals survival in mature hippocampal neurons

We showed previously that VEGFR-2 elicits neuroprotection in serum-starved neuroblastoma cells through autocrine and paracrine mechanisms and a blockade of receptor function leads to a loss in cell viability [21], [25]. Whether VEGFR-2 plays a similar role in primary cultures of mature hippocampal neurons remains unclear. In these studies, we hypothesized that VEGF signaling through VEGFR-2 alone plays a significant role in protecting mature hippocampal neurons from oxidative stress induced by

Discussion

In this study we examined the effects of VEGFR-2 inhibition on the survival of nutritionally deprived hippocampal neurons. We found that neuronal viability was reduced only slightly in NB cultured neurons when compared to those cultured in NB supplemented with B27 and survival under both conditions was enhanced by the inclusion of exogenous VEGF and inhibited by VEGFR-2 inhibition. We attribute survival without B27 to an autocrine signaling by endogenous VEGF through VEGFR-2 based on the

Conclusions

In conclusion, we show that a blockade of VEGFR-2 function by pharmacological inhibition or gene silencing triggers oxidative stress and mitochondrial dysfunction in mature hippocampal neurons. These events are manifested by an induction of HO-1, caspase-3 cleavage with a parallel loss in both cell viability and the activation of prosurvival Akt and ERK1/2. These findings also implicate activated BAD and decreased Bcl-xL levels as participants in the mitochondrial dysfunction and oxidative

Acknowledgments

This investigation was funded by a grant from the National Center for Research Resources [RR003037], a component of the National Institutes of Health (NIH), and a NINDS/NIH grant [SC1NS066033]. Its contents are solely the responsibility of the authors and do not necessarily represent the official views of NCRR or NIH.

References (54)

  • Y. Yuan et al.

    The homeostasis of iron and suppression of HO-1 involved in the protective effects of nimodipine on neurodegeneration induced by aluminum overloading in mice

    Eur. J. Pharmacol

    (2008)
  • T. Falk et al.

    Vascular endothelial growth factor B (VEGF-B) is up-regulated and exogenous VEGF-B is neuroprotective in a culture model of Parkinsonʼs disease

    Mol. Neurodegener

    (2009)
  • M. Thirunavukkarasu et al.

    VEGFR1 (Flt-1+/-) gene knockout leads to the disruption of VEGF-mediated signaling through the nitric oxide/heme oxygenase pathway in ischemic preconditioned myocardium

    Free Radic. Biol. Med.

    (2007)
  • N. Arbel et al.

    Mediation of the antiapoptotic activity of Bcl-xL protein upon interaction with VDAC1 protein

    J. Biol. Chem

    (2012)
  • M.T. Lin et al.

    Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases

    Nature

    (2006)
  • C. Ruiz de Almodovar et al.

    Role and therapeutic potential of VEGF in the nervous system

    Physiol. Rev.

    (2009)
  • H. Matsuzaki et al.

    Vascular endothelial growth factor rescues hippocampal neurons from glutamate-induced toxicity: signal transduction cascades

    FASEB J

    (2001)
  • K.L. Jin et al.

    Vascular endothelial growth factor: direct neuroprotective effect in in vitro ischemia

    Proc. Natl. Acad. Sci. USA

    (2000)
  • A. Wick et al.

    Neuroprotection by hypoxic preconditioning requires sequential activation of vascular endothelial growth factor receptor and Akt

    J. Neurosci.

    (2002)
  • K. Jin et al.

    Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo

    Proc. Natl. Acad. Sci. USA

    (2002)
  • L. Cao et al.

    VEGF links hippocampal activity with neurogenesis, learning and memory

    Nature Genet.

    (2004)
  • Y. Sun et al.

    VEGF-induced neuroprotection, neurogenesis, and angiogenesis after focal cerebral ischemia

    J. Clin. Invest.

    (2003)
  • J. Dhondt et al.

    Neuronal FLT1 receptor and its selective ligand VEGF-B protect against retrograde degeneration of sensory neurons

    FASEB J

    (2011)
  • Y. Li et al.

    VEGF-B inhibits apoptosis via VEGFR-1-mediated suppression of the expression of BH3-only protein genes in mice and rats

    J. Clin. Invest.

    (2008)
  • C.E. Hagberg et al.

    Vascular endothelial growth factor B controls endothelial fatty acid uptake

    Nature

    (2010)
  • I. Zachary

    Neuroprotective role of vascular endothelial growth factor: signalling mechanisms, biological function, and therapeutic potential

    Neuro-Signals

    (2005)
  • E. Storkebaum et al.

    VEGF: a critical player in neurodegeneration

    J. Clin. Invest.

    (2004)
  • Cited by (26)

    • Chronic VEGFR-3 signaling preserves dendritic arborization and sensitization under stress

      2021, Brain, Behavior, and Immunity
      Citation Excerpt :

      These “vasculogenic” factors have been shown to drive adult neurogenesis and display neuroprotective and pro-survival properties in retinal ganglionic injury, stroke, and motor neuron injury models (Mauceri et al., 2020; Froger et al., 2020; Harde, et al., 2019). For example, signaling of VEGFR-2 (Hao and Rockwell, 2013), the most well-characterized angiogenic pathway, regulates neuronal branching and neuroprotection signaling via the canonical Akt and ERK signaling pathways (Hu et al., 2005). While the “lymphangiogenic” factors, VEGF-C and VEGF-D, have been less well characterized in this context, convincing data demonstrate that VEGF-C and/or VEGFR-3 signaling drive enhanced neurogenesis in young NSCs and serve as a critical dendritic arborization maintenance factors in mouse hippocampal neurons (Han et al., 2015; Mauceri et al., 2011).

    • Neuroprotective effects of glial mediators in interactions between retinal neurons and Müller cells: RGC-Müller cell communication

      2021, Experimental Eye Research
      Citation Excerpt :

      Bcl-xL, the predominant anti-apoptotic protein in the retina (Levin et al., 1997), is involved in resistance to hypoxic injury and apoptotic cell death (Dong et al., 2004). Given that the anti-apoptotic action of VEGF has been linked to an increased Bcl-2 and Bcl-xL expression in neuronal cells (Sánchez et al., 2010; Hao and Rockwell, 2013), it is conceivable that hypoxia-induced increasing VEGF levels (Fig. 2d, g, Fig. 3b) contribute to upregulation of Bcl-2 and Bcl-xL (Fig. 4a and b). Fine-tuning of Bcl-2/Bad or Bcl-xL/Bad expression ratios seems to be important for the viability status of RGCs.

    • Intracavernous Injection of Autologous Platelet-Rich Plasma Ameliorates Hyperlipidemia-Associated Erectile Dysfunction in a Rat Model

      2021, Sexual Medicine
      Citation Excerpt :

      BDNF-induced mitophagy plays a protective role against endothelial cell damage in hyperglycemic conditions.29 VEGF promotes signaling through an alternate VEGF receptor-2 in the setting of mitochondrial dysfunction and oxidative stress.30 From this previous study, it is logical to conclude that the beneficial histological and functional results of PRP treatment are derived from the PRP-mediated delivery of IGF-1, BDNF, and VEGF.

    • PI3K/Akt signaling pathway and Hsp70 activate in hippocampus of rats with chronic manganese sulfate exposure

      2018, Journal of Trace Elements in Medicine and Biology
      Citation Excerpt :

      At the same time, decreased anti-oxidant enzyme activity prevents excessive free radicals in the body to be eliminated in time, increases lipid peroxidation, and eventually results in mitochondrial dysfunction and disruption of energy production. The PI3K/Akt signaling pathway is commonly activated after oxidative stress injury, and it plays an important role in protecting against nerve cell death [43]. A previous study found that Foxo3a-siRNA significantly inhibited phospho-Foxo3a expression, resulting in the suppression of peroxiredeoxin III protein expression, and FoxO3a can protect nerve cells from oxidative stress by regulating antioxidant enzymes [44].

    View all citing articles on Scopus
    View full text