Elsevier

Neurobiology of Disease

Volume 85, January 2016, Pages 144-154
Neurobiology of Disease

mTOR pathway inhibition prevents neuroinflammation and neuronal death in a mouse model of cerebral palsy

https://doi.org/10.1016/j.nbd.2015.10.001Get rights and content

Highlights

  • In mice, hypoxia, ischemia and lipopolysaccharide (HIL) models cerebral palsy.

  • Rapamycin treatment prevents brain injury at 24 h and 1 month induced by HIL.

  • Rapamycin reduces neuroinflammation at 24 h and 1 month induced by HIL.

  • Rapamycin may inhibit neuronal death by inducing autophagy.

  • mTOR inhibition may provide a new preventative treatment option for cerebral palsy.

Abstract

Background and purpose

Mammalian target of rapamycin (mTOR) pathway signaling governs cellular responses to hypoxia and inflammation including induction of autophagy and cell survival. Cerebral palsy (CP) is a neurodevelopmental disorder linked to hypoxic and inflammatory brain injury however, a role for mTOR modulation in CP has not been investigated. We hypothesized that mTOR pathway inhibition would diminish inflammation and prevent neuronal death in a mouse model of CP.

Methods

Mouse pups (P6) were subjected to hypoxia–ischemia and lipopolysaccharide-induced inflammation (HIL), a model of CP causing neuronal injury within the hippocampus, periventricular white matter, and neocortex. mTOR pathway inhibition was achieved with rapamycin (an mTOR inhibitor; 5 mg/kg) or PF-4708671 (an inhibitor of the downstream p70S6kinase, S6K, 75 mg/kg) immediately following HIL, and then for 3 subsequent days. Phospho-activation of the mTOR effectors p70S6kinase and ribosomal S6 protein and expression of hypoxia inducible factor 1 (HIF-1α) were assayed. Neuronal cell death was defined with Fluoro-Jade C (FJC) and autophagy was measured using Beclin-1 and LC3II expression. Iba-1 labeled, activated microglia were quantified.

Results

Neuronal death, enhanced HIF-1α expression, and numerous Iba-1 labeled, activated microglia were evident at 24 and 48 h following HIL. Basal mTOR signaling, as evidenced by phosphorylated-S6 and -S6K levels, was unchanged by HIL. Rapamycin or PF-4,708,671 treatment significantly reduced mTOR signaling, neuronal death, HIF-1α expression, and microglial activation, coincident with enhanced expression of Beclin-1 and LC3II, markers of autophagy induction.

Conclusions

mTOR pathway inhibition prevented neuronal death and diminished neuroinflammation in this model of CP. Persistent mTOR signaling following HIL suggests a failure of autophagy induction, which may contribute to neuronal death in CP. These results suggest that mTOR signaling may be a novel therapeutic target to reduce neuronal cell death in CP.

Introduction

Cerebral palsy (CP) is among the most common neurodevelopmental disorders, affecting 2–3 out of every 1000 live births (Kirby et al., 2011). CP is characterized by a heterogeneous phenotype including impaired motor function, intellectual disability, blindness, and epilepsy in 25–50% of affected children (Bax et al., 2005, Pakula et al., 2009). Clear risk factors for CP include prematurity, low birth weight, multiple gestations, coagulation disorders, intraventricular hemorrhage, placental pathology, and especially, hypoxic–ischemic brain injury (Keogh and Badawi, 2006). Of particular relevance, inflammation resulting from maternal or fetal infection dramatically increases the risk for CP, especially if superimposed on prenatal or perinatal hypoxia–ischemia (Nelson and Grether, 1998, Wheater and Rennie, 2000). Retrospective case–control analysis illustrates that preterm infants with CP and white matter injury had increased culture-positive infections of the blood, cerebrospinal fluid, and trachea during the neonatal period (Graham et al., 2004). Moreover, the activation of inflammatory pathways during fetal life may sensitize the brain to the effects of hypoxia–ischemia (Fleiss and Gressens, 2012). Indeed, the most widely accepted animal models of CP combine hypoxia–ischemia plus lipopolysaccharide-induced inflammation (HIL) exposure during development, resulting in greater cellular injury and more substantial motor and behavioral deficits than either insult alone (Eklind et al., 2004, Shen et al., 2010, Shen et al., 2012, Girard et al., 2009, Hu et al., 2013). The patterns of neuronal injury in established HIL mouse models serve to model one subtype of CP (Shen et al., 2010, Shen et al., 2012) and include selective cell death in the hippocampus, cortex, thalamus, and the periventricular white matter, all neuropathological hallmarks of CP (Krägeloh-Mann et al., 2002, Bax et al., 2006, Krägeloh-Mann and Horber, 2007).

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that regulates cellular growth and proliferation in response to various environmental stimuli, including nutrients, oxygen, energy, and growth factors (Hall, 2008). mTOR signaling, altered independently by hypoxia–ischemia and inflammation, plays a critical role in regulating cell death following environmental stress. For example, lipopolysaccharide (LPS) induces an mTOR-dependent release of cytokines and pro-inflammatory mediators, such as IL-1β, 6, 8, and TNFα (Kusaba et al., 2005, Weichhart et al., 2008), which have been implicated in neuronal and white matter damage (Allan and Rothwell, 2001, Kadhim et al., 2001). Interestingly, enhanced mTOR activity, by suppression of the upstream mTOR inhibitors Tsc1 or Tsc2, increases vulnerability of neurons to hypoxic–ischemic injury (Ng et al., 2011, Papadakis et al., 2013), whereas, decreasing mTOR activity via Tsc1 overexpression fosters resistance to ischemia-induced damage (Papadakis et al., 2013). mTOR inhibition with rapamycin treatment prior to injury reduces neuronal death and increases autophagy in an animal model of neonatal stroke (Carloni et al., 2008, Chen et al., 2012). Autophagy, a regulated intracellular degradation process, may play a role in cell survival during bioenergetic stress (Levine and Klionsky, 2004, Kiffin et al., 2006, Wu et al., 2009) and thus, rapamycin-induced autophagy is a potential mechanism of mTOR-dependent neuroprotection (Carloni et al., 2008, Carloni et al., 2010).

While the mTOR signaling cascade has been linked to several pediatric neurological disorders (Curatolo et al., 2001, Goorden et al., 2007, Sharma et al., 2010, Talos et al., 2012, Zeng et al., 2008), manipulation of the mTOR signaling cascade as a pre-clinical therapeutic strategy in CP has not been investigated. While previous studies in neonatal stroke models have used mTOR inhibitors prior to hypoxic–ischemic injury conditions, in a clinically relevant paradigm, we hypothesize that mTOR pathway inhibition with rapamycin following HIL may reduce neuronal death and neuroinflammation in a mouse model of CP. If successful, our approach could provide a completely new cell signaling cascade to investigate for therapeutic development in a subset of infants at high risk for CP.

Section snippets

HIL animal model

HIL surgical procedures were performed as described previously (Shen et al., 2010). On post-natal day 6 (P6), C57BL/6 pups were anesthetized using indirect cooling on ice to the point of unconsciousness. Indirect cooling on ice and is the form of anesthesia recommended by the Temple University IACUC on-line training website for very young mice (Anesthesia and Analgesia of Rodents, http://www.research.temple.edu/iacuc/iaonmodules.asp). The pup was considered fully anesthetized when it was not

HIL model

A consistent finding in MRI analyses and post-mortem human brain tissue in CP is injury to the periventricular white matter (periventricular leukomalacia, PVL) (Bax et al., 2006, Delaporte et al., 1985, Krägeloh-Mann and Horber, 2007). Similar to rat (Hu et al., 2013) and rabbit (Tan et al., 2005) models of CP, the HIL mouse model of CP is characterized by PVL, accompanied by cell death in the hippocampus ipsilateral to the carotid ligation, as well as selective areas of the cortex and thalamus

Discussion

We demonstrate that neuronal death in the HIL mouse model of CP can be prevented with the mTOR inhibitor rapamycin, even when administered after the inciting HIL procedure. Rapamycin inhibited expression of HIF1α, a marker of cellular hypoxic stress, and led to autophagy induction in association with diminished cellular injury. Rapamycin was also associated with a substantial decrease in microglial activation within the injured brain. To further support the effects rapamycin on survival, we

Conflicts-of-interest/disclosures

None.

Acknowledgments and sources of funding

This work was supported by funds from the Shriners Hospital Pediatric Research Center.

References (59)

  • H. Kusaba et al.

    Interleukin-12-induced interferon-gamma production by human peripheral blood T cells is regulated by mammalian target of rapamycin (mTOR)

    J. Biol. Chem.

    (2005)
  • B. Levine et al.

    Development by self-digestion: molecular mechanisms and biological functions of autophagy

    Dev. Cell

    (2004)
  • K.B. Nelson et al.

    Potentially asphyxiating conditions and spastic cerebral palsy in infants of normal birth weight

    Am. J. Obstet. Gynecol.

    (1998)
  • A.T. Pakula et al.

    Cerebral palsy: classification and epidemiology

    Phys. Med. Rehabil. Clin. N. Am.

    (2009)
  • A. Rami et al.

    Spatial resolution of phospholipid scramblase 1 (PLSCR1), caspase-3 activation and DNA-fragmentation in the human hippocampus after cerebral ischemia

    Neurochem. Int.

    (2003)
  • S. Rees et al.

    Fetal and neonatal origins of altered brain development

    Early Hum. Dev.

    (2005)
  • R. Schmidt-Kastner et al.

    Selective vulnerability of the hippocampus in brain ischemia

    Neuroscience

    (1991)
  • T. Weichhart et al.

    The TSC-mTOR signaling pathway regulates the innate inflammatory response

    Immunity

    (2008)
  • S.M. Allan et al.

    Cytokines and acute neurodegeneration

    Nat. Rev. Neurosci.

    (2001)
  • R.N. Auer et al.

    Neurobehavioral deficit due to ischemic brain damage limited to half of the CA1 sector of the hippocampus

    J. Neurosci.

    (1989)
  • M. Bax et al.

    Proposed definition and classification of cerebral palsy, April 2005

    Dev. Med. Child Neurol.

    (2005)
  • M. Bax et al.

    Clinical and MRI correlates of cerebral palsy: the European Cerebral Palsy Study

    JAMA

    (2006)
  • K.M. Buckley et al.

    Rapamycin up-regulation of autophagy reduces infarct size and improves outcomes in both permanent MCAL, and embolic MCAO, murine models of stroke

    Exp. Transl. Stroke Med.

    (2014)
  • S. Carloni et al.

    Activation of autophagy and Akt/CREB signaling play an equivalent role in the neuroprotective effect of rapamycin in neonatal hypoxia–ischemia

    Autophagy

    (2010)
  • W. Chen et al.

    HIF-1 Alpha Inhibition Ameliorates Neonatal Brain Damage After Hypoxic–Ischemic Injury

  • M. Czeh et al.

    The yin and yang of microglia

    Dev. Neurosci.

    (2011)
  • B. Delaporte et al.

    Early echographic findings in non-hemorrhagic periventricular leukomalacia of the premature infant

    Pediatr. Radiol.

    (1985)
  • R. Di et al.

    S6K inhibition renders cardiac protection against myocardial infarction through PDK1 phosphorylation of Akt

    Biochem. J.

    (2012)
  • S. Eklind et al.

    The role of glucose in brain injury following the combination of lipopolysaccharide or lipoteichoic acid and hypoxia–ischemia in neonatal rats

    Dev. Neurosci.

    (2004)
  • Cited by (68)

    • PLXNA2 knockdown promotes M2 microglia polarization through mTOR/STAT3 signaling to improve functional recovery in rats after cerebral ischemia/reperfusion injury

      2021, Experimental Neurology
      Citation Excerpt :

      Mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that is involved in various cellular processes, including energy metabolism, transcription and translation of mRNA, ribosomal biogenesis, cytoskeletal organization, lipid biosynthesis, and autophagy (Liu and Sabatini, 2020; Ryskalin et al., 2017; Wullschleger et al., 2006). Previous studies have shown that activation of mTOR signaling may contribute to the developmental process of neuroinflammation in many neurologic diseases (Huang et al., 2016; Srivastava et al., 2016). In addition, there could be a possible association between mTOR and semaphorin signaling in mammalian immune systems (Ito et al., 2015).

    • HIF1A polymorphisms do not modify the risk of epilepsy nor cerebral palsy after neonatal hypoxic-ischemic encephalopathy

      2021, Brain Research
      Citation Excerpt :

      However, it was recently demonstrated that HIF-1α expression corresponds with the pattern of neuronal cell death in a mouse model of CP. Furthermore, reducing HIF-1α expression prevented neuronal death and diminished neuroinflammation, suggesting HIF-1α could hold neurotoxic properties in the setting of mouse model of CP (Srivastava et al., 2016). It is well known that gender represents a risk factor for ischaemic brain injury in neonates with low birth weight: male infants are at higher risk of cerebral palsy than female infants (Smith et al., 2014).

    View all citing articles on Scopus
    View full text