Elsevier

Brain Research Bulletin

Volume 137, March 2018, Pages 53-70
Brain Research Bulletin

CaMKIIα expression in a mouse model of NMDAR hypofunction schizophrenia: Putative roles for IGF-1R and TLR4

https://doi.org/10.1016/j.brainresbull.2017.11.007Get rights and content

Highlights

  • Induced NMDAR hypofunction caused hippocampal CaMKIIα loss.

  • Depletion of hippocampal IGF-1R was linked with CaMKIIα loss in NMDAR hypofunction.

  • Cre-lox knockdown of TLR4 rescued IGF-1R and CaMKIIα loss after an induced NMDAR hypofunction.

Abstract

Schizophrenia (SCZ) is a neuropsychiatric disorder that is linked to social behavioral deficits and other negative symptoms associated with hippocampal synaptic dysfunction. Synaptic mechanism of schizophrenia is characterized by loss of hippocampal N-Methyl-d-Aspartate Receptor (NMDAR) activity (NMDAR hypofunction) and dendritic spines. Previous studies show that genetic deletion of hippocampal synaptic regulatory calcium-calmodulin dependent kinase II alpha (CaMKIIα) cause synaptic and behavioral defects associated with schizophrenia in mice. Although CaMKIIα is involved in modulation of NMDAR activity, it is equally linked to inflammatory and neurotropin signaling in neurons. Based on these propositions, we speculate that non-neurotransmitter upstream receptors associated with neurotropic and inflammatory signaling activities of CaMKIIα may alter its synaptic function. Besides, how these receptors (i.e. inflammatory and neurotropic receptors) alter CaMKIIα function (phosphorylation) relative to hippocampal NMDAR activity in schizophrenia is poorly understood. Here, we examined the relationship between toll-like receptor (TLR4; inflammatory), insulin-like growth factor receptor 1 (IGF-1R; neurotropic) and CaMKIIα expression in the hippocampus of behaviorally deficient schizophrenic mice after we induced schizophrenia through NMDAR inhibition.

Schizophrenia was induced in WT (C57BL/6) mice through intraperitoneal administration of 30 mg/Kg ketamine (NMDAR antagonist) for 5 days (WT/SCZ). Five days after the last ketamine treatment, wild type schizophrenic mice show deficiencies in sociability and social novelty behavior. Furthermore, there was a significant decrease in hippocampal CaMKIIα (p < 0.001) and IGF-1R (p < 0.001) expression when assessed through immunoblotting and confocal immunofluorescence microscopy. Additionally, WT schizophrenic mice show an increased percentage of phosphorylated CaMKIIα in addition to upregulated TLR4 signaling (TLR4, NF-κB, and MAPK/ErK) in the hippocampus. To ascertain the functional link between TLR4, IGF-1R and CaMKIIα relative to NMDAR hypofunction in schizophrenia, we created hippocampal-specific TLR4 knockdown mouse using AAV-driven Cre-lox technique (TLR4 KD). Subsequently, we inhibited NMDAR function in TLR4 KD mice in an attempt to induce schizophrenia (TLR4 KD SCZ). Interestingly, IGF-1R and CaMKIIα expressions were preserved in the TLR4 KD hippocampus after attenuation of NMDAR function. Furthermore, TLR4 KD SCZ mice showed no prominent defects in sociability and social novelty behavior when compared with the control (WT).

Our results show that a sustained IGF-1R expression may preserve the synaptic activity of CaMKIIα while TLR4 signaling ablates hippocampal CaMKIIα expression in NMDAR hypofunction schizophrenia. Together, we infer that IGF-1R depletion and increased TLR4 signaling are non-neurotransmitter pro-schizophrenic cues that can reduce synaptic CaMKIIα activity in a pharmacologic mouse model of schizophrenia.

Introduction

Schizophrenia affects approximately 1.1% (∼83 Million people) of the world’s population across all sex, age and background (NIH-NIMH, 2017, SARDAA, 2017). About 3.5 Million people are affected in the United States and it is one of the leading causes of disability (NIH-NIMH, 2017, SARDAA, 2017). While the disorder is partially genetic, stress and drugs are predominant causes of schizophrenia. Importantly, 75% of individuals that become schizophrenic show the symptoms in late adolescence to early adulthood (age 16–25yrs) (NIH-NIMH, 2017, SARDAA, 2017, Arain et al., 2013). This coincides with the peak of brain wiring, then re-wiring (age 22–25yrs) required for transitioning from childhood to adult brain (Arain et al., 2013, Owens et al., 2012, Owen and O'Donovan, 2012, Eckfeld et al., 2017). In schizophrenia, this generally involves changes in expression of neurotransmitters (glutamate and dopamine), receptors (NMDAR; N-Methyl-d-Aspartate receptor and D2R; Dopaminergic D2 receptor), and proteins involved in the regulation of their synaptic activities (Nakazawa et al., 2017, Cohen et al., 2015, Dias, 2012, Barkus et al., 2012, Seillier and Giuffrida, 2009). As such, most available treatment methods involve the use of antipsychotics that target these receptors (i.e. NMDAR and D2R). Side effects of these drugs are a limitation as it often leads to depression, movement disorders and catalepsy (Amodeo et al., 2017, Nakata et al., 2017, Nikvarz et al., 2017, Freyberg et al., 2017, Ibi et al., 2017, Meltzer, 2017, Ostinelli et al., 2017, Su et al., 2017). Therefore, there is a need for assessment of other synaptic targets that can reduce synaptic and behavioral defects of schizophrenia while circumventing neurotransmitter receptor sensitivity (Yang and Tsai, 2017).

Despite the limitations, treatment methods that target dopamine and glutamate receptors are laudable based on vastly studied dopamine and glutamate hypothesis of schizophrenia (Yang and Tsai, 2017, Purves-Tyson et al., 2017, Howes et al., 2017). However, the significance of synaptic regulatory calcium-calmodulin-depended kinase 2 alpha (CaMKIIα) − linked with synaptic function of NMDAR (Hagihara et al., 2014, Hagihara et al., 2013, Purkayastha et al., 2012, Dhavan et al., 2002, Robison et al., 2005) and D2R (Ng et al., 2010, Takeuchi et al., 2002, Vekshina et al., 2017) − cannot be over emphasized in schizophrenia. Substantive evidence now exists to confirm that genetic deletion of CaMKIIα is a cause of synaptic and behavioral changes that are characteristic of schizophrenia (Frankland et al., 2008, Matsuo et al., 2009, Yamasaki et al., 2008). Roles of CaMKIIα in neurons are wide and varied. CaMKIIα is involved in inflammatory signaling that is linked with toll-like receptor 4 (TLR4/NF-κB) (Kaltschmidt et al., 2005, Mémet, 2006). Additionally, it acts downstream of IGF-1R-Ras/Raf pathway to regulate calcium-mediated synaptic activity of NMDAR (Hu et al., 2016a, Hu et al., 2016b, Le Grevès et al., 2005, Dou et al., 2005). Based on these propositions, it is logical to speculate that synaptic and inflammatory activity of CaMKIIα are not mutually exclusive. Although CaMKIIα depletion has been implicated in schizophrenia (Matsuo et al., 2009, Yamasaki et al., 2008), the mechanism through which IGF-1R and TLR4 can putatively affect synaptic CaMKIIα expression in pathophysiology of schizophrenia is poorly understood.

Localization of CaMKIIα at post-synaptic sites enables interactions with neurotrophic factors and receptors involved in synaptic development and maintenance (Hami et al., 2014, Kiray et al., 2014, Sun, 2006, Gunnell et al., 2007, Venkatasubramanian et al., 2010, Demirel et al., 2014). Moreover, loss of these proteins have been implicated in several developmental neuropsychiatric disorders including schizophrenia (Nelson et al., 2006, Ribasés et al., 2008). Among these, IGF-1R may alter CaMKIIα expression through Wnt/GSK3 and Ras/Raf signaling in growth, synaptic function, inflammation, and cell death (Pereira et al., 2008). Furthermore, both IGF-1R and CaMKIIα promotes pre- and post-synaptic calcium movement linked to NMDAR function in the hippocampus during long-term potentiation (LTP) and cognition (Cassilhas et al., 2012, Hu et al., 2016a, Hu et al., 2016b). In addition to the control of growth and aging, a recent study demonstrates the significance of IGF-1R signaling in presynaptic calcium release and glutamatergic neurotransmission. In their study, Gazit and co-workers show that IGF-1R modulates calcium release from the mitochondria and regulate presynaptic activity of synaptophysin (Gazit et al., 2016). Equally, IGF-1R signaling alters protein kinase systems (Akt/mTOR and MAPK/ErK) (Dyer et al., 2016, Netchine et al., 2011, Salani et al., 2015) involved in the regulation of neurodevelopment and LTP (Zheng et al., 2012, Kéri et al., 2011, Romanelli et al., 2007). Owing to its effect on synaptic calcium currents (Gazit et al., 2016) and gene expression (Le Grevès et al., 2005, Le Grevès et al., 2006), IGF-1R may significantly affect the activity of ionotropic glutamate receptors in normal LTP and disease conditions (Jiang et al., 2015, Demirel et al., 2014, Gunnell et al., 2007).

The function of CaMKIIα is not restricted to neurotropic regulation of growth, and synaptic function. Downstream of IGF-1R, CaMKIIα can reduce the activity of proinflammatory proteins like NF-κB and MAPK/ErK, and purigenic receptors such as TLR4 and P2RX7 (Meffert and Baltimore, 2005, Luo et al., 2008, Gómez-Villafuertes et al., 2015, Huang et al., 2009). Therefore, a reduction of CaMKIIα activity can lead to inflammation and progression of synaptic dysfunction associated with schizophrenia. Evidently, the expression of TLR4 and associated pro-inflammatory proteins increases in the blood of clinically-diagnosed schizophrenia patients (Venkatasubramanian and Debnath, 2013, Chang et al., 2011, Hansen et al., 2008, McQuillin et al., 2009). In addition to their involvement in inflammation, proteins like NF-κB and MAPK/ErK − in the TLR4 signaling pathway − are known to reduce synaptic activity of CaMKIIα by increasing CaMKIIα phosphorylation (Meffert and Baltimore, 2005, Han et al., 2009). Furthermore, MAPK/ErK and CaMKIIα occupy similar sites at post-synaptic densities where MAPK/ErK phosphorylates CaMKIIα during early and late LTP phases (Du et al., 2004, Giovannini et al., 2001, Tsui et al., 2005, Derkach et al., 2007). Although primarily inflammatory, TLR4 is involved in synaptogenesis as it increases glutamatergic function in the developing brain (Shen et al., 2016, Henneberger and Steinhäuser, 2016). This is required for the development and formation of synapses by migrating neurons (Shen et al., 2016, Venkatasubramanian and Debnath, 2013). However, in excess, TLR4-linked glutamate release may induce epileptogenic effects and other forms of excitotoxicity through an upregulated MAPK/ErK signaling (Jiang et al., 2015, Shen et al., 2016, Henneberger and Steinhäuser, 2016). Since MAPK/ErK also act downstream of IGF-1R (Xing et al., 2016, Dyer et al., 2016, Vahdatpour and Dyer, 2016), neural activity of TLR4 may overlap with neurotropic cues of IGF-1R and other kinase receptors involved in nervous system development and neural circuit formation (Fig. 1). Thus, an increased TLR4 signaling may upregulate MAPK/ErK which can abolish IGF-1R signaling and CaMKIIα activities in the brain.

Based on these propositions, we hypothesize that NMDAR-linked CaMKIIα activity in synaptic function and neurotropic signaling are not exclusive of its fate in inflammation. In brief, CaMKIIα modulates synaptic NMDAR function (Mao et al., 2014, Ma et al., 2015, Park et al., 2008, Johnston and Morris, 1995), and itself (i.e CaMKIIα) can be altered through TLR4 and IGF-1R signaling. Through upregulated MAPK/ErK, TLR4 may alter synaptic function by reducing IGF-1R signaling. This may lead to a reduced CaMKIIα activation downstream of IGF-1R (Ras/Raf), and increased CaMKIIα phosphorylation (inactivation) by MAPK/ErK. Therefore, decreased TLR4 and upregulated IGF-1R signaling are possible mechanisms through which CaMKIIα activity can be preserved in NMDAR hypofunction. Here, we show that induced loss of NMDAR function can cause hippocampal CaMKIIα loss and decrease IGF-1R signaling. Furthermore, we tested the effectiveness of hippocampal-specific TLR4 knockdown as an intervention method to preserve IGF-1R and CaMKIIα in mice after a persistent NMDAR inhibition.

Section snippets

Animal strains

Adult C57BL/6 (WT) and transgenic C57BL/TLR4loxp/loxp (TLR4 floxed mice) weighing between 22-25 g were used for this study. All Animals were procured from Jackson Lab and housed in the LSU School of Veterinary Medicine vivarium. Mice were handled in accordance with the Institutional Animal Care and Use Committee at the Louisiana State University.

Adeno-associated viral gene expression

AAV-CMV-eGFP and AAV-CMV-Cre-eGFP were procured from the University of Iowa Vector Core (Iowa City, IA) and stored at −80 °C. Transgenic TLR4loxp/loxp

Persistent NMDAR blockade caused social behavioral defects in WT mice

After an induced NMDAR hypofunction, we assessed behavioral change in pharmacologic mouse model of schizophrenia. Mice were tested in a social interaction chamber in order to evaluate sociability and social novelty behavior as illustrated in Fig. 2a. Using the scoring methods previously described by Kaidanovich-Beilin et al. (2011), schizophrenic animals show no preference for a strange mouse (S1a) over an empty compartment (E) in the sociability chamber (sociability test) when compared with

Discussion

Pathophysiology of schizophrenia has long been perceived from “neurotransmitter imbalance” and “neurotransmitter receptor sensitivity” perspectives. Based on these school of thoughts, the dopamine hypothesis, and later a glutamate hypothesis of schizophrenia was proposed. These principles were also reflected in the treatment approach to schizophrenia. To date, most drugs available to treat schizophrenia target either dopamine and or glutamate receptors in the brain (Amodeo et al., 2017, Su et

Conclusions

We have shown the significance of TLR4 in upregulation of MAPK/ErK in NMDAR hypofunction schizophrenic-mouse model. This is characterized by a decrease in hippocampal IGF-1R/CaMKIIα, and social interaction behavioral deficits. Ultimately, hippocampal TLR4 knockdown rescued IGF-1R/CaMKIIα decline and prevented behavioral deficits by reducing MAPK/ErK expression.

Competing interests

The author states that the present manuscript presents no conflict of interest.

Acknowledgements

This study was supported by the IBRO-ISN Fellowship 2015 awarded to OOM and Louisiana Board of Regents RCS Grant RD-A-09 and NIH/NIMH Grant R03 MH 104851 awarded to CCL.

References (106)

  • I.V. Grishagin

    Automatic cell counting with ImageJ

    Anal. Biochem.

    (2015)
  • S.J. Han et al.

    Multi-modulation of nuclear receptor coactivators through posttranslational modifications

    Trends Endocrinol. Metab.

    (2009)
  • T. Hansen et al.

    Variation in the purinergic P2RX7 receptor gene and schizophrenia

    Schizophr. Res.

    (2008)
  • O.D. Howes et al.

    The role of genes, stress, and dopamine in the development of schizophrenia

    Biol. Psychiatry

    (2017)
  • A. Hu et al.

    The effect of constitutive over-expression of insulin-like growth factor 1 on the cognitive function in aged mice

    Brain Res.

    (2016)
  • A. Hu et al.

    The effect of constitutive over-expression of insulin-like growth factor 1 on the cognitive function in aged mice

    Brain Res.

    (2016)
  • W. Huang et al.

    Transcriptional integration of TLR2 and TLR4 signaling at the NCoR derepression checkpoint

    Mol. Cell

    (2009)
  • H.M. Johnston et al.

    N-methyl-D-aspartate and nitric oxide regulate the expression of calcium/calmodulin-dependent kinase II in the hippocampal dentate gyrus

    Brain Res. Mol. Brain Res.

    (1995)
  • B. Kaltschmidt et al.

    Signaling via NF-kappaB in the nervous system

    Biochim. Biophys. Acta

    (2005)
  • M. Le Grevès et al.

    Age-related effects of IGF-1 on the NMDA-, GH- and IGF-1-receptor mRNA transcripts in the rat hippocampus

    Brain Res. Bull.

    (2005)
  • J.E. Lisman et al.

    A model of synaptic memory: a CaMKII/PP1 switch that potentiates transmission by organizing an AMPA receptor anchoring assembly

    Neuron

    (2001)
  • S. Mémet

    NF-kappaB functions in the nervous system: from development to disease

    Biochem. Pharmacol.

    (2006)
  • J. Ma et al.

    Overexpression of αCaMKII impairs behavioral flexibility and NMDAR-dependent long-term depression in the medial prefrontal cortex

    Neuroscience

    (2015)
  • M.K. Meffert et al.

    Physiological functions for brain NF-κB

    Trends Neurosci.

    (2005)
  • P.G. Nelson et al.

    Selected neurotrophins, neuropeptides, and cytokines: developmental trajectory and concentrations in neonatal blood of children with autism or Down syndrome

    Int. J. Dev. Neurosci.

    (2006)
  • I. Netchine et al.

    IGF1 molecular anomalies demonstrate its critical role in fetal, postnatal growth and brain development

    Best Pract Res. Clin. Endocrinol. Metab.

    (2011)
  • J. Ng et al.

    Activation of calcium/calmodulin-dependent protein kinase IIalpha in the striatum by the heteromeric D1-D2 dopamine receptor complex

    Neuroscience

    (2010)
  • C.S. Park et al.

    alpha-Isoform of calcium-calmodulin-dependent protein kinase II and postsynaptic density protein 95 differentially regulate synaptic expression of NR2A- and NR2B-containing N-methyl-d-aspartate receptors in hippocampus

    Neuroscience

    (2008)
  • M. Ribasés et al.

    Association study of 10 genes encoding neurotrophic factors and their receptors in adult and child attention-deficit/hyperactivity disorder

    Biol. Psychiatry

    (2008)
  • A.J. Robison et al.

    Multivalent interactions of calcium/calmodulin-dependent protein kinase II with the postsynaptic density proteins NR2B, densin-180, and alpha-actinin-2

    J. Biol. Chem.

    (2005)
  • R.J. Romanelli et al.

    Insulin-like growth factor type-I receptor internalization and recycling mediate the sustained phosphorylation of Akt

    J. Biol. Chem.

    (2007)
  • A. Seillier et al.

    Evaluation of NMDA receptor models of schizophrenia: divergences in the behavioral effects of sub-chronic PCP and MK-801

    Behav. Brain Res.

    (2009)
  • Y.A. Su et al.

    Anticholinergic use trends in 14,013 patients with schizophrenia from three national surveys on the use of psychotropic medications in China (2002–2012)

    Psychiatry Res.

    (2017)
  • J. Tsui et al.

    Calcium/calmodulin-dependent protein kinase II (CaMKII) localization acts in concert with substrate targeting to create spatial restriction for phosphorylation

    J. Biol. Chem.

    (2005)
  • G. Venkatasubramanian et al.

    The TRIPS (Toll-like receptors in immuno-inflammatory pathogenesis) Hypothesis: a novel postulate to understand schizophrenia

    Prog. Neuro-Psychopharmacol. Biol. Psychiatry

    (2013)
  • D. Allen et al.

    Organization and regulation of small conductance Ca2+-activated K + Channel multiprotein complexes

    J. Neurosci.

    (2007)
  • G. Amodeo et al.

    Older and newer strategies for the pharmacological treatment of agitation in schizophrenia and bipolar disorder

    CNS Neurol. Disord. Drug Targets

    (2017)
  • Mariam Arain et al.

    Maturation of the adolescent brain

    Neuropsychiat. Dis. Rreat.

    (2013)
  • D.T. Balu et al.

    Multiple risk pathways for schizophrenia converge in serine racemase knockout mice, a mouse model of NMDA receptor hypofunction

    Proc. Natl. Acad. Sci. U. S. A.

    (2013)
  • C. Barkus et al.

    GluN1 hypomorph mice exhibit wide-ranging behavioral alterations

    Genes Brain Behav.

    (2012)
  • F. Barros et al.

    Cytoplasmic domains and voltage-dependent potassium channel gating

    Front. Pharmacol.

    (2012)
  • A. Burgess et al.

    Loss of human Greatwall results in G2 arrest and multiple mitotic defects due to deregulation of the cyclin B-Cdc2/PP2A balance

    Proc. Natl. Acad. Sci. U. S. A.

    (2010)
  • S. Chiu et al.

    Insulin receptor signaling in the development of neuronal structure and function

    Chiu and Cline Neural Dev.

    (2010)
  • J.T. Coyle

    NMDA receptor and schizophrenia: a brief history

    Schizophr. Bull.

    (2012)
  • S. Dai et al.

    Supramolecular assemblies and localized regulation of voltage-gated ion channels

    Physiol. Rev.

    (2009)
  • V.A. Derkach et al.

    Regulatory mechanisms of AMPA receptors in synaptic plasticity

    Nat. Rev. Neurosci.

    (2007)
  • R. Dhavan et al.

    The cyclin-dependent kinase 5 activators p35 and p39 interact with the alpha-subunit of Ca2+/calmodulin-dependent protein kinase II and alpha-actinin-1 in a calcium-dependent manner

    Neurosci

    (2002)
  • A.M. Dias

    The integration of the glutamatergic and the white matter hypotheses of schizophrenia's etiology

    Curr. Neuropharmacol.

    (2012)
  • J.T. Dou et al.

    Insulin receptor signaling in long-term memory consolidation following spatial learning

    Learn. Mem.

    (2005)
  • J. Du et al.

    Focus on CaMKII: a molecular switch in the pathophysiology and treatment of mood and anxiety disorders

    Int. J. Neuropsychopharmacol.

    (2004)
  • Cited by (23)

    • Long-term effects of stress early in life on microRNA-30a and its network: Preventive effects of lurasidone and potential implications for depression vulnerability

      2020, Neurobiology of Stress
      Citation Excerpt :

      Previous studies have shown that genetic deletion of hippocampal CAMK2A in mice caused synaptic and behavioral defects associated with schizophrenia (Matsuo et al., 2009; Yamasaki et al., 2008). Moreover, since CAMK2A facilitates N-methyl-D-aspartate receptors (NMDARs), its reduced activity may contribute to the hypofunction of NMDAR that has been associated to the pathophysiology of this disorder (Ogundele and Lee, 2018). Interestingly, a reduced expression of CAMK2A was also found in the hippocampus of depressed suicides, which provides further support to the role of this kinase in different psychiatric conditions (Fuchsova et al., 2015).

    View all citing articles on Scopus
    View full text