12
1521-0103/358/3/504515$25.00 http://dx.doi.org/10.1124/jpet.116.234476 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 358:504515, September 2016 Copyright ª 2016 by The American Society for Pharmacology and Experimental Therapeutics Minireviews Involvement of Neuroinflammation during Brain Development in Social Cognitive Deficits in Autism Spectrum Disorder and Schizophrenia Yutaka Nakagawa and Kenji Chiba Innovative Research Division, Mitsubishi Tanabe Pharma, Yokohama, Japan Received April 18, 2016; accepted July 5, 2016 ABSTRACT Development of social cognition, a unique and high-order func- tion, depends on brain maturation from childhood to adulthood in humans. Autism spectrum disorder (ASD) and schizophrenia have similar social cognitive deficits, although age of onset in each disorder is different. Pathogenesis of these disorders is complex and contains several features, including genetic risk factors, environmental risk factors, and sites of abnormalities in the brain. Although several hypotheses have been postulated, they seem to be insufficient to explain how brain alterations associated with symptoms in these disorders develop at distinct developmental stages. Development of ASD appears to be related to cerebellar dysfunction and subsequent thalamic hyperactiva- tion in early childhood. By contrast, schizophrenia seems to be triggered by thalamic hyperactivation in late adolescence, whereas hippocampal aberration has been possibly initiated in childhood. One of the possible culprits is metal homeostasis disturbances that can induce dysfunction of blood-cerebrospinal fluid barrier. Thalamic hyperactivation is thought to be induced by microglia-mediated neuroinflammation and abnormalities of intra- cerebral environment. Consequently, it is likely that the thalamic hyperactivation triggers dysregulation of the dorsolateral pre- frontal cortex for lower brain regions related to social cognition. In this review, we summarize the brain aberration in ASD and schizophrenia and provide a possible mechanism underlying social cognitive deficits in these disorders based on their distinct ages of onset. Introduction Microglial cells contribute to immune surveillance in the central nervous systems (CNS), and polarization of microglial subsets (M1 and M2) plays an important role in controlling the balance between promotion and suppression in neuroinflam- mation (Ransohoff and Perry, 2009; Kettenmann et al., 2011; Mikita et al., 2011; Saijo and Glass, 2011). M1 microglial cells can promote neuroinflammation and subsequent neural net- work dysfunction in the CNS, whereas M2 microglia are implicated in inhibiting inflammation and restoring homeo- stasis (Mosser and Edwards, 2008; Gordon and Martinez, 2010). As we reported previously, imbalance of M1 and M2 microglia appears to be closely related to mood aberration of major depressive disorder and bipolar disorder (Nakagawa and Chiba, 2014, 2015). Autism spectrum disorder (ASD) is defined by deficits in social communication interaction and behavioral flexibility, manifesting in early childhood (Volkmar et al., 2014). Eighteen surveys conducted according to the Diagnostic and Statistical Manual of Mental Disorders, fourth edition, criteria demonstrated that estimates of the rate of ASD range from 10 in 10,000 to 16 in 10,000, whereas the most recent study estimated the prevalence in the United States as 11.3 in 1,000, suggesting that the prevalence of ASD is increasing (Volkmar et al., 2014). Parents of children with ASD have often reported that the first sign of a problem with their child was the delay of language in the second year of life (Tager-Flusberg, 2000). Social cognitive deficits are a hallmark characteristic of ASD and schizophrenia (Couture et al., 2006; Meyer et al., 2011a). Schizophrenia is characterized by positive, negative, and cognitive symptoms and afflicts approximately 1% worldwide; it develops in late adolescence most frequently (Stahl, 2013). It The authors declare no conflict of interest. Both authors are employees of Mitsubishi Tanabe Pharma and have not received financial support from any other institution. dx.doi.org/10.1124/jpet.116.234476. ABBREVIATIONS: ASD, autism spectrum disorder; BBB, blood-brain barrier; BCB, blood-CSF barrier; [ 11 C](R)-PK11195, [ 11 C] (R)-(1-[2- chlorophenyl]-N-methyl-N-[1-methylpropyl]-3-isoquinoline carboxamide); CCL, CC-chemokine ligand; CNS, central nervous system; CNTNAP2, contactin-associated protein-like 2; CSF, cerebrospinal fluid; CVO, circumventricular organ; CXCL, CXC-chemokine ligand; DLPFC, dorsolateral prefrontal cortex; FFA, fusiform face area; FOXP2, forkhead box P2; IFN-g, interferon-g; IL, interleukin; PrPC, cellular prion protein; PrPSc, a disease-associated isoform of the prion protein; PSD-95, postsynaptic density protein 95; SOD-1, superoxide dismutase-1; STG, superior temporal gyrus; STS, superior temporal sulcus; VLPFC, ventrolateral prefrontal cortex; VMPFC, ventromedial prefrontal cortex. 504 at ASPET Journals on April 1, 2020 jpet.aspetjournals.org Downloaded from

Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

1521-0103/358/3/504–515$25.00 http://dx.doi.org/10.1124/jpet.116.234476THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 358:504–515, September 2016Copyright ª 2016 by The American Society for Pharmacology and Experimental Therapeutics

Minireviews

Involvement of Neuroinflammation during Brain Development inSocial Cognitive Deficits in Autism Spectrum Disorderand Schizophrenia

Yutaka Nakagawa and Kenji ChibaInnovative Research Division, Mitsubishi Tanabe Pharma, Yokohama, Japan

Received April 18, 2016; accepted July 5, 2016

ABSTRACTDevelopment of social cognition, a unique and high-order func-tion, depends on brain maturation from childhood to adulthoodin humans. Autism spectrum disorder (ASD) and schizophreniahave similar social cognitive deficits, although age of onset ineach disorder is different. Pathogenesis of these disorders iscomplex and contains several features, including genetic riskfactors, environmental risk factors, and sites of abnormalitiesin the brain. Although several hypotheses have been postulated,they seem to be insufficient to explain how brain alterationsassociated with symptoms in these disorders develop at distinctdevelopmental stages. Development of ASD appears to be relatedto cerebellar dysfunction and subsequent thalamic hyperactiva-tion in early childhood. By contrast, schizophrenia seems to be

triggered by thalamic hyperactivation in late adolescence,whereas hippocampal aberration has been possibly initiated inchildhood. One of the possible culprits is metal homeostasisdisturbances that can induce dysfunction of blood-cerebrospinalfluid barrier. Thalamic hyperactivation is thought to be induced bymicroglia-mediated neuroinflammation and abnormalities of intra-cerebral environment. Consequently, it is likely that the thalamichyperactivation triggers dysregulation of the dorsolateral pre-frontal cortex for lower brain regions related to social cognition.In this review, we summarize the brain aberration in ASD andschizophrenia and provide a possible mechanism underlyingsocial cognitive deficits in these disorders based on their distinctages of onset.

IntroductionMicroglial cells contribute to immune surveillance in the

central nervous systems (CNS), and polarization of microglialsubsets (M1 andM2) plays an important role in controlling thebalance between promotion and suppression in neuroinflam-mation (Ransohoff and Perry, 2009; Kettenmann et al., 2011;Mikita et al., 2011; Saijo and Glass, 2011). M1 microglial cellscan promote neuroinflammation and subsequent neural net-work dysfunction in the CNS, whereas M2 microglia areimplicated in inhibiting inflammation and restoring homeo-stasis (Mosser and Edwards, 2008; Gordon and Martinez,2010). As we reported previously, imbalance of M1 and M2microglia appears to be closely related to mood aberration of

major depressive disorder and bipolar disorder (Nakagawaand Chiba, 2014, 2015). Autism spectrum disorder (ASD) isdefined by deficits in social communication interaction andbehavioral flexibility, manifesting in early childhood (Volkmaret al., 2014). Eighteen surveys conducted according to theDiagnostic and Statistical Manual of Mental Disorders, fourthedition, criteria demonstrated that estimates of the rate ofASD range from 10 in 10,000 to 16 in 10,000, whereas themostrecent study estimated the prevalence in the United Statesas 11.3 in 1,000, suggesting that the prevalence of ASD isincreasing (Volkmar et al., 2014). Parents of children withASD have often reported that the first sign of a problem withtheir child was the delay of language in the second year of life(Tager-Flusberg, 2000). Social cognitive deficits are a hallmarkcharacteristic of ASD and schizophrenia (Couture et al., 2006;Meyer et al., 2011a).Schizophrenia is characterized by positive, negative, and

cognitive symptoms and afflicts approximately 1% worldwide;it develops in late adolescencemost frequently (Stahl, 2013). It

The authors declare no conflict of interest. Both authors are employees ofMitsubishi Tanabe Pharma and have not received financial support from anyother institution.

dx.doi.org/10.1124/jpet.116.234476.

ABBREVIATIONS: ASD, autism spectrum disorder; BBB, blood-brain barrier; BCB, blood-CSF barrier; [11C](R)-PK11195, [11C] (R)-(1-[2-chlorophenyl]-N-methyl-N-[1-methylpropyl]-3-isoquinoline carboxamide); CCL, CC-chemokine ligand; CNS, central nervous system; CNTNAP2,contactin-associated protein-like 2; CSF, cerebrospinal fluid; CVO, circumventricular organ; CXCL, CXC-chemokine ligand; DLPFC, dorsolateralprefrontal cortex; FFA, fusiform face area; FOXP2, forkhead box P2; IFN-g, interferon-g; IL, interleukin; PrPC, cellular prion protein; PrPSc, adisease-associated isoform of the prion protein; PSD-95, postsynaptic density protein 95; SOD-1, superoxide dismutase-1; STG, superior temporalgyrus; STS, superior temporal sulcus; VLPFC, ventrolateral prefrontal cortex; VMPFC, ventromedial prefrontal cortex.

504

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 2: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

is postulated that dopamine hyperactivity in the nucleusaccumbens induced by hypofunction of glutamate receptorsin the prefrontal cortex and hippocampus is related to positivesymptoms, whereas hypoactivation of dopamine and gluta-mate neurons in the prefrontal cortex contributes to negativesymptoms (Stahl, 2013). In contrast, according to the neuro-developmental hypothesis, the etiology of schizophrenia ap-pears to involve pathologic processes that are caused bygenetic and environmental risk factors and begin beforethe brain fully matures (Weinberger, 1986; Marenco andWeinberger, 2000).Pathogenesis of these disorders is complex and contains

several features, including genetic risk factors, environmentalrisk factors, and sites of abnormalities in the brain (Hultmanet al., 1999; Gardener et al., 2011; Miles, 2011; Onore et al.,2012). Recently, Meyer et al. (2011a), focusing on similaritiesand differences between schizophrenia and ASD, implied thatdisturbed neurodevelopment by maternal infection and suc-ceeding prenatal neuroinflammation is shared by ASD andschizophrenia, whereas persistent and latent neuroinflamma-tion contributes to onsets of ASD and schizophrenia, respec-tively. However, these hypotheses seem not to clarify howbrain alterations associated with symptoms in these dis-orders develop at distinct developmental stages. Therefore,in this article, we summarize the brain aberration and relatedneuroinflammation in early and chronic phases of ASD,discuss similarities and differences between social cognitivedeficits in ASD and those in schizophrenia, and provide apossible mechanism underlying the social cognitive deficits inthese disorders.

ASD and NeuroinflammationGenetic Risk Factors. Although there has been con-

siderable debate in etiology of ASD, the genetic basis of thisdisorder is supported by several twin studies (Rosenberg et al.,2009; Frazier et al., 2014). Much attention has been focusedon a relationship between ASD and a transcriptional gene,forkhead box P2 (FOXP2), which is closely related to languageability (Bowers and Konopka, 2012). FOXP2 protein regulatesgene expression ofMET (Mukamel, et al., 2011) and contactin-associated protein-like 2 (CNTNAP2) (Arking et al., 2008;Bowers and Konopka, 2012). During gestation, MET gene isselectively expressed in the temporal and occipital lobes, whichcontribute to verbal communication, implying that METplays an important role in development of language ability(Mukamel et al., 2011). The CNTNAP2 encodes a cell adhesionmolecule of neurexin subfamily and is highly expressed infrontal lobe circuits in the developing human brain (Arkinget al., 2008). It has been reported that common variants inCNTNAP2 are associated with several allied neurodevelop-mental disorders, including ASD (Scott-Van Zeeland et al.,2010). FOXP2 mutant mice show ASD-like phenotypes, suchas reduced or atypical vocalizations and impairments oflearning performances (Shu et al., 2005; Fujita et al., 2008;Kurt et al., 2012).Other studies demonstrated that ASD symptoms are associ-

ated with gene variations of neuroligin, neurexin, Src homologyand multiple ankyrin repeat domain 3, and cell adhesionmolecule 1 (Zhiling et al., 2008; Bourgeron, 2009; Fujita-Jimboet al., 2015). The variations of these synaptic cell adhesionmolecules seem to induce an imbalance of excitatory and

inhibitory receptors and synaptic pruning deficits (Lisé andEl-Husseini, 2006; LeBlanc and Fagiolini, 2011). In the pro-cess of synaptic pruning, upregulation of myocyte enhancerfactor 2 can lead to ubiquitination of postsynaptic densityprotein 95 (PSD-95) in the postsynaptic density, and, sub-sequently, the degradation of ubiquitinated PSD-95 resultsin a synapse elimination (Caroni et al., 2014). Interestingly,mutated neuroligin 3 or cell adhesion molecule 1 protein upre-gulates C/EBP-homologous protein, a marker of endoplasmicreticulum stress, suggesting that the synaptic pruning deficitsinduce endoplasmic reticulum stress and neuroinflammationin ASD (Fujita et al., 2010; Fujita-Jimbo et al., 2012, 2015).Consequently, these genetic factors presumably influencefunction of neural circuitry of social cognition in combinationwith environmental risk factors.Environmental Risk Factors. Air pollutants, pesticides,

polychlorinated biphenyls, solvents, and glyphosate appearto be associated with development of ASD (Sealey et al., 2016).A possible relationship is shown between ASD severity andblood or hair levels of heavy metals, including mercury andlead (Rossignol et al., 2014). A recent study revealed thatsome of ASD may be caused by maternal antibodies to fetalbrain antigens such as cypin, which, along with PSD-95,can regulate dendritic branching of hippocampal neurons(Braunschweig et al., 2013). Sodium valproate, an anticonvul-sant and a mood stabilizer, induces fetal neurodevelopmentaberration in animals, including humans, higher incidence ofASD in humans, and ASD-like phenotypes such as reducedsocial interaction and increased repetitive behaviors in ro-dents (Roullet et al., 2013). There are many prenatal andpostnatal risk factors, as follows: maternal medication use,C-reactive protein levels, hypertension, hemorrhage, infec-tion, depressive symptomatology, food, gestational diabetes,parental age at birth, prolonged labor, fetal distress, birthinjury or trauma,multiple birth, low birth weight, etc. (Glassonet al., 2004; Gardener et al., 2009, 2011; Lupien et al., 2011;Patterson, 2011; Guinchat et al., 2012; Neggers, 2014; Xianget al., 2015).Metal Homeostasis Disturbances. It is highly probable

that a well-balanced diet plays an essential role in develop-ment and maintenance of brain functions, because goodnutrition is a cornerstone of goodhealth,whereas poor nutritionis associated with reduced immunity, impaired physical devel-opment, and reduced productivity (World Health Organization,2016). Children with ASD often have restricted diets that canlead to nutrient deficiencies with brain metal homeostasisdisturbances (Bilgiç et al., 2010; Sidrak et al., 2014). Irondeficiency is one of the most prevalent types of malnutrition,affecting probable two billion people in the world, and pregnantwomen and young children are affected most severely, becausepregnancy and infant growth demand iron (World HealthOrganization, 2016). Iron is required for basic cellular functionsin all of the tissues, especially in the brain and muscle, andis critically important for the oxygen-carrier function of hemo-globin in red blood cells (Breymann, 2015). In the brain, iron istransported by transferrin or divalentmetal transporter-1 fromthe peripheral blood and is essential for the activity of severalenzymes involved in myelination process and monoamineneurotransmitter synthesis (Dusek et al., 2015). Iron deficiencyis suggested to be related to development of ASD (Bilgiç et al.,2010; Sidrak et al., 2014). Lower iron concentrations in theblood appear to induce transport of neurotoxic manganese

Neuroinflammation in Social Cognitive Deficits 505

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 3: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

instead of iron into the brain (Gunter et al., 2013; Duseket al., 2015).When zinc levels are decreased and conversely copper levels

are increased in the blood during pregnancy, prevention offetal development would be introduced (Faber et al., 2009).The imbalance of zinc and copper levels may deteriorateFOXP2 functions and induce language disability, becauseFOXP2 contains zinc finger domain (Bowers and Konopka,2012). Indeed, it has been reported that ASD children showreduced zinc levels in their hairs and low blood zinc/copperratio (Faber et al., 2009; Yasuda et al., 2011, 2013). Takentogether, it is possible that metal homeostasis disturbancesare one of the primary culprits of ASD.Involvement of Neuroinflammation. A ligand for the

peripheral benzodiazepine receptor, [11C] (R)-(1-[2-chlorophenyl]-N-methyl-N-[1-methylpropyl]-3-isoquinoline carboxamide) ([11C](R)-PK11195), can be used for the imaging of activated micro-glia cells with positron emission tomography (Banati, 2002). Ithas been reported that [11C](R)-PK11195–binding potentialvalues are significantly higher in the brain regions, includingthe cerebellum,midbrain, fusiform gyrus, and prefrontal cortexin ASD adults as compared with controls (Suzuki et al., 2013).In the visual cortex in autopsy brains, microglial densities aresignificantly greater in individuals with ASD versus controls(Tetreault et al., 2012). An immunohistochemical study revealedthat a marked activation of microglia and astrocytes in themiddle frontal and anterior cingulate gyri and cerebellum isobtained at autopsy from ASD subjects (Vargas et al., 2005).Similarly, microglial cells are markedly activated in the dorso-lateral prefrontal cortex (DLPFC) of ASD individuals (Morganet al., 2010). Risperidone in combinationwith a cyclooxygenase-2inhibitor, celecoxib, showed a superior efficacy as comparedwith monotherapy of risperidone in a randomized double-blind placebo-controlled clinical study in ASD children(Asadabadi et al., 2013). From these results, neuroinflam-mation mediated by M1 microglia appears to be associatedwith ASD.Postmortem studies revealed prominent microglia activa-

tion and increased proinflammatory cytokines and chemokines,including interferon-g (IFN-g), interleukin (IL)-1b, IL-6,IL-12p40, tumor necrosis factor-a, and CC-chemokine ligand2 (CCL2) in the brain, particularly in the frontal cortex,cerebellum, and cerebrospinal fluid (CSF) of ASD individuals(Vargas et al., 2005; Li et al., 2009; Morgan et al., 2010; Onoreet al., 2012). Similarly, the levels of IL-1b, IL-6, IL-8, IL-12p40,CCL2, CCL5, and CCL11 are significantly increased in theplasma of ASD individuals (Ashwood et al., 2011a, b). Theincreased cytokine levels were predominantly in children whohad a regressive form of ASD and were associated with moreimpaired communication and aberrant behaviors (Ashwoodet al., 2011a, b). Furthermore, BTBR T1tf/J mice with genemutation in kynurenine 3-hydroxylase (Kmo) related to neuro-protective actions show elevated levels of proinflammatorycytokines, including IL-1b and IL-6 in the cerebellum andASD-like phenotypes such as reduced social interactions andimpaired juvenile play (McFarlane et al., 2008; Heo et al., 2011).In contrast, the levels of IL-4 and IL-10 in the plasma showed nodifference between ASD and control subjects (Ashwood et al.,2011b). The levels of transforming growth factor-b in ASDbloodare significantly lower as compared with typically developingcontrols and are negatively correlated with psychologic symp-tom severity (Okada et al., 2007; Ashwood et al., 2008).

Therefore, it is highly possible that neuroinflammationmediated by M1 microglia is associated with symptoms ofASD based on imbalance of proinflammatory M1 and anti-inflammatory M2 polarization states of microglia. Our view isalong the lines of previous hypotheses that the imbalance ofM1/M2 polarization of microglia is implicated in neuroinflam-mation and disruption of excitatory versus inhibitory balancein the brain of ASD (Gottfried et al., 2015; Koyama andIkegaya, 2015).Disturbances in the Blood-Brain Barrier. The blood-

brain barrier (BBB) has a unique barrier structure betweenthe lumen of cerebral blood vessels and brain parenchyma.The endothelial cells have luminal tight junction and aresurrounded by basement membrane and the pericytes. Aroundall of these structures are the astrocytic end feet processes fromnearby astrocytes. The interaction among vascular endothelialcells, pericytes, and astrocytes plays an important role inmaintenance of BBB functions (Abbott et al., 2006; Bonkowskiet al., 2011). It is possible that BBB dysfunction contributesto major depressive disorder and schizophrenia (Shalev et al.,2009).Maternal hyperglycemia and hypertension are often asso-

ciated with activation of inflammatory cells, inducing abnor-malities in capillary endothelial cells of the brain (Jackson,2011). In the inflamed capillaries, proinflammatory cytokinesand mediators contribute to the polarization and recruitmentof M1 macrophages (Jackson, 2011). At early phase, M1macrophages appear to remove injured vascular endothelialcells by phagocytosis (Jackson, 2011; Phillipson and Kubes,2011); however, under insufficient M2 condition, M1 macro-phages can probably induce prolonged inflammation and de-struction of BBB. The BBB possesses manganese sensitivity(Bornhorst et al., 2012), suggesting that disequilibrium of ironand manganese levels is presumably associated with dysfunc-tion of BBB directly. Imbalance of zinc and copper levelsmay contribute to dysfunction of astrocytes because astrocytesexpress copper, zinc (CuZn) superoxide dismutase-1 (SOD-1)activity (Chen et al., 2006). Taken together, it is likely thatdysfunction of BBB is caused by multiple prenatal and post-natal risk factors. Because BBB function is immature at earlydevelopmental stages (Saunders et al., 2014), cerebral neuralcells in fetuses, infants, and young children are presumablyvulnerable to these factors. Therefore, it is possible that BBBdisturbances are related to ASD.Disturbances in the Blood-CSF Barrier. The blood-

CSF barrier (BCB) lies between the choroid plexus vessels incircumventricular organs (CVOs) and the CSF (Choi and Kim,2008; Benarroch, 2011; Lun et al., 2015). The CVOs are foundto be located around the cerebral ventricles, and the CVOswith BCB are characterized by lack of BBB properties (Choiand Kim, 2008; Benarroch, 2011). In the BCB, tanycytes,highly specialized ependymal cells, have tight junction andplay regulatory roles in interaction among the blood, brainparenchyma, and CSF (Benarroch, 2011; Langlet et al., 2013).Tanycytes contribute to monitoring the CSF composition (Sisóet al., 2010; Benarroch, 2011), and these cells seem to bevulnerable to imbalance of zinc and copper levels becausetanycytes express CuZn SOD-1 (Peluffo et al., 2005). Further-more, the BCB is thought to be a major route for manganeseinto the CSF because the increased blood manganese levelscan induce more severe dysfunction of the BCB rather thanBBB (Bornhorst et al., 2012). Therefore, it is highly probable

506 Nakagawa and Chiba

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 4: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

that maternal disturbances of cellular metal homeostasispredominantly induce dysfunction of tanycytes in the BCBand subsequent neuroinflammation that triggers aberration ofneural network in fetuses, infants, and young children (Fig. 1).

Social Cognitive Deficits in ASDCognitive function such as self-regulation and social cogni-

tion enables humans to make plans and regulate actions insocial situations appropriately (Heatherton and Wagner,2011). This unique cognitive function depends on brainmaturation from childhood to adulthood in humans (Lebelet al., 2008; Catts et al., 2013). Therefore, it is likely thatimmaturity negatively impacts high-order cognition acquiredat each developmental stage.Dysfunction of the Cerebellum. Young children with

ASD (2–4 years of age) show a significant increase in cerebellarvolume as compared with typically developing children,whereas cerebellar volume of ASD schoolchildren and adoles-cents is similar to or rather smaller than controls (Sparks et al.,2002; Allen, 2005; Anagnostou and Taylor, 2011). A diffusion

tensor imaging study to investigate brain anatomic connectivityindicated a significant increase in fractional anisotropy, thedeviation from pure isotropic diffusion of water mobility, in thebrain regions including cerebellum of young children with ASD(1–6 years of age) (Weinstein et al., 2011). In contrast, fractionalanisotropy in the cerebellum is decreased inASDschoolchildren(6–12 years old) (Brito et al., 2009). Serotonin transporterbinding is reduced in several brain regions, including cerebel-lum of ASD adults (Nakamura et al., 2010). Furthermore, [11C](R)-PK11195–binding values are significantly higher in thebrain regions including cerebellum of ASD adults as comparedwith those of controls (Suzuki et al., 2013). From these results,it is possible that neuronal hyperactivation and neuroinflam-mation occur in the cerebellum of young children with ASD,subsequently leading to reduction of neuronal activity andatrophy with persistent neuroinflammation in the cerebellumof ASD schoolchildren, adolescents, and adults.The cerebellum is involved in body and limb movement,

executive and cognitive functions, and language ability (Koziolet al., 2014; Wang et al., 2014). ASD individuals often showmotor abnormalities consistent with cerebellar dysfunctionthat occurs in young children and is persistent over time (LeeandBo, 2015). Although cerebellar lesions are unlikely to resultin profound cognitive deficits in adults, cerebellar abnormal-ities at earlier ages lead to more conspicuous cognitive andaffective changes, diagnosed as cerebellar cognitive-affectivesyndrome (Wang et al., 2014). Cerebellar cognitive-affectivesyndrome is characterized by disturbances of executive func-tion, impaired spatial cognition, personality change, andlinguistic difficulties (Schmahmann and Sherman, 1998). Ithas been hypothesized that the cerebellum plays a pivotal rolein cognition in young children within the critical period (2–4years of age) and contributes to body and limb movement atevery developmental stage (Wang et al., 2014). Thus, one couldspeculate that cerebellar aberration in young children isassociated with initiation of social cognitive deficits in ASD.As mentioned in the above section, impaired cellular metal

homeostasis can induce disturbances of cerebral ventriclesand contributes to functional or structural alterations in thebrain. Because the cerebellum is located adjacent to the fourthventricle and the cerebral ventricles are partly surrounded bythe choroid plexus in young children (Su and Young, 2011;Liddelow, 2015), impaired cellular metal homeostasis proba-bly induces hyperactivation of the cerebellum via dysfunctionof choroid plexus and fourth ventricle in young children withASD. Neuronal hyperactivation can trigger M1 polarization ofmicroglia in the cerebellum, and thenM1microglial-mediatedneuroinflammation and neural network dysfunction mayextend to the related tissues under inhibition in the M2 state.Consequently, it is possible that imbalance between M1 andM2 polarization of microglia plays an important role in theprogression of ASD (Nakagawa and Chiba, 2014, 2015).It is unlikely that symptoms of ASD can be caused by

abnormalities in a single brain region because a significantreduction in total gray matter volume and a marked increasein cerebral ventricular volume are observed in ASD school-children and adolescents (McAlonan et al., 2005). There areneuronal connections between the cerebellum and other brainregions (Mazzola et al., 2013; Rogers et al., 2013; Koziol et al.,2014). To understand symptoms, initiation, and progression ofASD more precisely, it is important to analyze the neuropatho-logical relation between the cerebellum and other brain regions.

Fig. 1. The cerebral ventricles, BBB, and BCB. (A) The lateral, third, andfourth ventricles neighbor brain regions such as hippocampus, amygdala,thalamus, and cerebellum. (B) The BBB has a pivotal role in sufficientsupply of oxygen and nutrients to neurons and consists of the pericytes,end foot of astrocytes, and vascular endothelial cells that have luminaltight junction. (C) In the BCB, tanycytes, highly specialized ependymalcells, have tight junction and play regulatory roles in interaction amongthe blood, brain parenchyma, and CSF. Brain tissues with BCB arecharacterized by lack of BBB properties. Astrocytes and tanycytes expressCuZn SOD-1.

Neuroinflammation in Social Cognitive Deficits 507

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 5: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

Dysfunction of the Thalamus. Like the cerebellum, theamygdala and hippocampus were enlarged in children withASD as compared with control subjects, whereas no differ-ences were found in adolescent amygdala volumes (Sparkset al., 2002; Schumann et al., 2004). In contrast, atrophy andreduction of serotonin transporter binding in the thalamuswere reported in ASD from late childhood to adulthood(Tsatsanis et al., 2003; Hardan et al., 2006; Nakamura et al.,2010; Tamura et al., 2010). Higher microglial activity is foundin the thalamus of ASD adults (Suzuki et al., 2013). Fromthese results, it is presumed that in young children with ASD,neuroinflammation is initially induced in the thalamus, and,subsequently, persistent neuroinflammation induces thalamicatrophy in schoolchildren, adolescent, and adults. Because thethalamus neighbors the third ventricle, dysfunction of thethalamus in ASD may be related to impaired cellular metalhomeostasis and BCB disruption (Sisó et al., 2010).The thalamus plays a key role in sensory gate (Geyer and

Vollenweider, 2008). Both auditory and visual stimuli fromperipheral organs are sent to the thalamus, and then to theprimary auditory or visual area in the cerebral cortex (Tamiettoand de Gelder, 2010; Hackett, 2015). Functions of the thalamusare impaired in ASD schoolchildren, adolescents, and adults(Hardan et al., 2008; Lai et al., 2010). Significant decreases incerebellar activity and cerebellum-thalamus connectivity areinduced in a light or mild motor performance such as 20 fingertaps for each hand in ASD schoolchildren (Mostofsky et al.,2009). In contrast, ASD individuals show significantly increasedactivity in the cerebellum relative to control subjects when theyperform a rather complex motor task, in which they hold ajoystick and press a button with the thumb according toinstruction (Allen et al., 2004). From these results, it is likelythat, in ASD, neural network activities between the cerebellumand thalamus are decreased at resting state, whereas thispathway is activated when a task is provided. In daily life ofASD individuals, activities of the cerebellum and cerebellum-thalamus pathway appear to be increased because socialcommunication is always necessary.Aberration of Verbal Communication. Underconnec-

tivity in the thalamo-frontal pathway is induced in ASD school-children and adolescents (Cheon et al., 2011). In contrast,functional overconnectivity between the thalamus and tem-poral lobe occurs in ASD schoolchildren and adolescents,although underconnectivity is found for other cortical regions(Nair et al., 2013). Therefore, it is possible that hyperactiva-tion of the thalamus-temporal lobe connections is due tothalamo-frontal underconnectivity, and that dysconnectivityof these pathways is associated with ASD symptoms.ASD adults fail to activate the superior temporal sulcus

(STS)/superior temporal gyrus (STG) in response to humanvocal sounds, whereas they show a normal activation patternin response to nonvocal sounds (Gervais et al., 2004). Under-connectivity is induced between the STS/STG and dopami-nergic reward pathway, including the ventral tegmental areaand nucleus accumbens in ASD schoolchildren (Abrams et al.,2013). These results suggest that chronic hyperactivationof the thalamus-temporal lobe connections induces desen-sitization of the STS/STG, and that underconnectivity ofSTS/STG-reward pathway impairs the ability of ASD childrento experience speech as a pleasurable stimulus, subsequentlyleading to deterioration of language and social skills. It ispossible that the definite reduction of STS/STG reactivity to

human vocal sounds in ASD is caused by disturbances of theprefrontal cortex, which regulates functions of the STS/STGand is implicated in social cognition.There are neuronal connections between the STS/STG

and ventrolateral prefrontal cortex (VLPFC) that integratesocial cognitive information (Takahashi et al., 2007; Levy andWagner, 2011; Lai et al., 2012). Underconnectivity betweenthe temporal cortex and VLPFC occurs in adult ASD (Satoet al., 2012). In addition, VLPFC activity is significantlyreduced in schoolchildren and adolescents with ASD relativeto controls during speech stimulation (Lai et al., 2012).Accordingly, it is presumable that in daily life of ASD children,cerebellar and thalamic hyperactivation induces dysfunctionof the STS/STG and VLPFC, leading to a failure to integrateverbal information, which probably persists in adulthood.Aberration of Nonverbal Communication. The thala-

mus, fusiform face area (FFA), STS/STG, andVLPFCalso playa pivotal role in nonverbal communication, including facialexpressions, gestures, eye contact, and body language (Redcay,2008; Sato et al., 2012). These visual signals from the thalamusare sent to the visual cortex and FFA (Tamietto and de Gelder,2010; Blank et al., 2011). Microglial activity is increased in thevisual cortex/FFA of ASD individuals (Tetreault et al., 2012;Suzuki et al., 2013). Therefore, it is probable that thalamichyperactivation induces microglia-mediated neuroinflamma-tion and aberration of the visual cortex and FAA.There are neuronal connections between the visual cortex/FFA,

STS/STG, and VLPFC (Kreifelts et al., 2007; Blank et al.,2011; Sato et al., 2012). The STS/STG plays a key role indirect structural connections between verbal- and nonverbal-recognition areas (Kreifelts et al., 2007; Blank et al., 2011).Functional underconnectivity between the visual cortex andSTS/STG or VLPFC occurs in ASD adults in response toemotional stimuli (Sato et al., 2012; Khan et al., 2013). Takentogether, we hypothesize that abnormalities of verbal andnonverbal communication in ASD are due to cerebellar andthalamic hyperactivation and subsequent dysfunction of thevisual cortex, FAA, STS/STG, and VLPFC.Hippocampus-Amygdala Interaction and Emotional

Responses. The hippocampus is thought to be vulnerable todevelopmental aberration during prenatal stage (Abernethyet al., 2002; Beauchamp et al., 2008). BCB abnormalitiesaccompanying with impaired cellular metal homeostasis maybe related to hippocampal dysfunction because of neighboringthe hippocampus and lateral ventricle (Dziegielewska et al.,2001; Kiernan, 2012).The reciprocal interaction between amygdala and hippo-

campus contributes to emotion, emotion-related memory, andneurocognition, etc. (Potschka et al., 2011; Orsini and Maren,2012). Hyperactivation of the amygdala occurs in ASD adultsduring identification of previously viewed faces (Kleinhanset al., 2008; Monk et al., 2010; Dichter, 2012). Social impair-ment is positively correlated to amygdala activation but isnegatively related to amygdala-FFA connectivity in ASD(Kleinhans et al., 2008; Dichter, 2012). Development of faceperception and social cognitive skills is supported by theamygdala-FFA system (Schultz, 2005; Vuilleumier andPourtois,2007). Therefore, underconnectivity of amygdala-FFA path-way in ASD appears to induce overconnectivity between theamygdala and brain regions other than FFA.The ventromedial prefrontal cortex (VMPFC) plays a crit-

ical role in regulation of amygdala activity (Blair, 2013;

508 Nakagawa and Chiba

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 6: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

Motzkin et al., 2015). ASD subjects show increased neuronalactivities in theVMPFCand amygdala in response to emotionalstimuli and higher microglial activity in the VMPFC (Monket al., 2010; Suzuki et al., 2013). Poor performance on the Iowagambling task is found inASD schoolchildren and adolescents,suggesting lower VMPFC functions (Sawa et al., 2013).Accordingly, it is likely that long-lasting hyperactivation ofthe amygdala induces VMPFC dysfunction, which is mediatedby neuroinflammation. The dysfunction of VMPFC probablycauses dysregulation between the VMPFC and amygdala andmay induce aberration of emotional responses in ASD.The DLPFC and Top-Down Control. Top-down control

of lower brain regions by the DLPFC enables us to makeappropriate planning and action in social situations basedon integration of information, including verbal and non-verbal cognition, workingmemory, and evaluation of immediateas well as future outcomes (Tanaka et al., 2004;Weissman et al.,2008; Heatherton and Wagner, 2011). The number and size ofneuron are significantly increased in theDLPFC of ASD subjects(Courchesne et al., 2011). Compared with typically developingsubjects, ASD schoolchildren and adolescents perform signifi-cantly worse on DLPFC task (Dawson et al., 1998; Sawa et al.,2013). The ratio of N-acetyl-aspartate/creatine/phosphocreatinein the DLPFC is positively correlated with social ability scores inASD children and adolescents (Fujii et al., 2010). From theseresults, hypoactivation of the DLPFC appears to occur in ASDlike other psychiatric disorders (Hassel et al., 2008; Kang et al.,2012; Palaniyappan et al., 2013), and disturbances in top-downcontrol of lower brain regions by DLPFC may result in socialcognitive deficits.Taken together, we hypothesize that impaired cellular

metal homeostasis, one of the primary culprits, can initiallyinduce disturbances of the BCB rather than BBB and

subsequent cerebellar hyperactivation in young children withASD. The cerebellar hyperactivation appears to increasethalamic activity and connectivity between the thalamusand STS/STG because of underconnectivity of the thalamo-frontal pathway, thereby resulting in dysfunction of STS/STGand VLPFC. In contrast, hyperactivation of the hippocampusand amygdala probably causes dysregulation of VMPFC re-sponsible for emotional responses. These events in childhoodmay induce hypoactivation of the DLPFC, which controls lowerbrain regions related to social ability, and then lead to socialcognitive deficits in ASD.

Social Cognitive Deficits in SchizophreniaThe major clinical, biochemical, and genetic features in

ASD and schizophrenia are summarized in Table 1. Althoughcontribution of pathogenic genes is different in these disorders,similar characteristics are seen in social cognitive deficits;disturbances in glutamate, g-aminobutyric acid, and dopamineneurotransmission; and increasing proinflammatory responses.Dysfunction of the Hippocampus and Amygdala. Findings

concerning cerebellar aberration have been controversial inschizophrenia, unlike ASD (Bottmer et al., 2005). In schizo-phrenia, cerebellar disturbances contribute to neurologicalsoft signs, such as observable defects in sensory integration,motor coordination, and behavioral inhibition, which arelargely distinct from the core symptoms (Bottmer et al., 2005;Chan et al., 2010; Shinn, et al., 2015). Accordingly, it is unlikelythat cerebellar aberration is associated with cognitive deficitsin schizophrenia.Individuals with at-risk mental state and patients with the

first-episode or chronic schizophrenia have lower volumes ofseveral brain regions including the hippocampus persistently

TABLE 1Comparison of clinical, biochemical, and genetic characteristics between ASD and schizophrenia

ASD

Clinical Behavioral flexibility, delay of language, social cognitive deficits.a

Worse performance than schizophrenia children in the theory ofmind task.b Manifests in early childhood.a

Biochemical Disturbances in glutamate, GABA, and dopamineneurotransmission.c Increases in proinflammatory cytokine (IL-1b,IL-6, TNF-a, IFN-g) levels in the brain, CSF, and blood.d Noalterations in anti-inflammatory cytokine (IL-4, IL-10) levels in theblood.e

Genetic CNTNAP2, NLGN, NRXN, SHANK3, CADM1, etc.f

SchizophreniaClinical Positive, negative, cognitive symptoms (social cognitive deficits).g

Develops in late adolescence most frequently.g

Biochemical Disturbances in glutamate, GABA, and dopamineneurotransmission.g Increases in proinflammatory cytokine (IL-1b,IL-6, TNF-a) levels in the brain, CSF, and blood.h Reduction of anti-inflammatory cytokine (IL-10, IL-13) levels in the CSF and blood.h

Genetic DISC1, DTNBP1, NRG1, DRD2, HTR2A, COMT, etc.i

CADM1, cell adhesion molecule 1; COMT, catechol-O-methyltransferase; DISC1, disrupted in schizophrenia 1; DRD2,dopamine receptor D2; DTNBP1, dystrobrevin-binding protein 1; GABA, g-aminobutyric acid; HTR2A,5-hydroxytryptamine (serotonin) receptor 2A; NLGN, neuroligin; NRG1, neuregulin 1; NRXN, neurexin; SHANK3, Srchomology 3 and multiple ankyrin repeat domain 3; TNF-a, tumor necrosis factor-a.

aVolkmar et al., 2014.bPilowsky et al., 2000.cMoney and Stanwood, 2013.dOnore et al., 2012.eAshwood et al., 2011b.fBourgeron, 2009.gStahl, 2013.hMeyer et al., 2011b.iGejman et al., 2010.

Neuroinflammation in Social Cognitive Deficits 509

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 7: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

(Borgwardt et al., 2007; Chan et al., 2011). In addition, signifi-cantly reduced hippocampal volume with relatively higherplasmaIL-6 levels is induced inantipsychotic-naive schizophreniapatients with polymorphism IL-6 gene, implying that hippo-campal aberration caused by elevated IL-6 is the neurodevelop-mental origin of schizophrenia (Kalmady et al., 2014). Similarly,

higher IL-6 response bymaternal immune activation in prenatalstage is suggested to be associated with brain alterations toinitiate schizophrenia (Brown, 2011). Therefore, the hippocampusappears to be a possible brain region to initiate schizophrenia.The onset of psychosis is frequently preceded by weeks,

months, or years of prodromal symptoms, including deficits in

Fig. 2. A possible mechanism underlying social cognitive deficits in ASD and schizophrenia on their distinct ages of onset. (A) Social cognitive functionsdevelop accompanying with human brain maturation from childhood to adulthood and are related to increases in myelination of white matter tractsbetween the prefrontal cortex and other brain regions. (B) In ASD, impaired cellular metal homeostasis, one of the primary culprits, can inducedysfunction of the BCB rather than BBB, thereby leading to cerebellar aberration and subsequent thalamic hyperactivation in childhood. In the nextstep, the overconnectivity is induced between the thalamus and temporal lobe (STS/STG) because of underconnectivity of the thalamo-frontal pathway.The thalamic hyperactivation is thought to be induced by microglia-mediated neuroinflammation and presumably leads to dysfunction of the prefrontalcortex, including DLPFC, which regulates functions of lower brain regions contributing to social cognition. In schizophrenia, in contrast, hippocampalaberration can occur in childhood and result in succeeding hyperactivation of the thalamus in late adolescence. The thalamic hyperactivation appears tobe promoted due to immaturity of the fronto-temporal pathway and finally leads to DLPFC dysfunction with a similarity to ASD. (C) The cerebellum andprefrontal cortex are involved differently in social cognitive functions based on the maturation of the brain. The cerebellum appears to play apredominant role in cognitive functions at young childhood because the cerebellum has fully maturated at that time. In contrast, the prefrontal cortex isthought to contribute to cognitive functions along with its maturation progressively.

510 Nakagawa and Chiba

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 8: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

perception, emotion, neurocognition, communication, and sleep(Larson et al., 2010). Individuals who later fulfilled diagnosticcriteria for schizophrenia had shown significant premorbiddeficits in intellectual, language, and behavioral abilities atthe ages of 16–17 (Reichenberg et al., 2002). Symptoms such asproblems of language and cognitive development in children(3–11 years old) can predict schizophreniform disorder in adults(Cannon et al., 2002). From these results, it is presumed thatdisturbances of hippocampus and related brain regions play acentral role in prodromal symptoms of schizophrenia.Schizophrenia patients show hyperactivation of the amyg-

dala and underconnectivity of the VMPFC-amygdala pathway(Fan et al., 2013; Pankow et al., 2013). Therefore, it is probablethat in schizophrenia, like ASD, increased activity in theamygdala caused by hippocampal hyperactivation inducesdesensitization and dysregulation of the VMPFC. Theseimpairments of the VMPFC presumably lead to disturbancesof emotional responses in prodromal period of schizophrenia.Dysfunction of the Thalamus, Temporal Cortex, and

Prefrontal Cortex. There is neuronal connectivity betweenthe hippocampus and thalamus (Aggleton et al., 2010). Thethalamus is one of the damaged brain regions in at-risk mentalstate individuals and schizophrenia patients (Borgwardt et al.,2007; Chan et al., 2011). Schizophrenia patients with thalamicdysfunction show spindle range deficits that may probablycontribute to cognitive impairments (Ferrarelli et al., 2010;Wilson and Argyropoulos, 2012). In contrast, in healthysubjects, the fronto-temporal (VLPFC-STS/STG) connectionsreach 90% development in adulthood (Lebel et al., 2008),implying that immaturity of this pathway can prevent braindevelopment in adolescence. Therefore, it is presumed thatabnormalities of the thalamus caused by hippocampal hyper-activity can be exacerbated under immaturity of the fronto-temporal connections during late adolescence and early adulthoodin schizophrenia. These disturbances can induce STS/STG de-sensitization and VLPFC dysfunction, thereby leading to distur-bances of DLPFC top-down control (Fig. 2; Table 2).

Cuprizone-Treated Mice as an Animal Model ofPsychiatric Disorders

The treatmentwith cuprizone [bis(cyclohexylidenehydrazide)],the copper chelator, severely alters copper and zinc homeo-stasis in various tissues, including the CNS, and induces

demyelination in white and gray matters, such as the corpuscallosum, cerebral cortex, hippocampus, and cerebellum inmice (Zatta et al., 2005; Groebe et al., 2009; Gudi et al.,2009; Koutsoudaki et al., 2009). The treatment of cuprizone inmice is thought to be useful as a model of prion infectionbecause of the involvement of metal homeostasis in functionsof a cellular prion protein (PrPC) (Martins et al., 2001). PrPCis predominately expressed in neuronal cells (Ramasamyet al., 2003) and can bind copper, zinc, iron, and manganese(Brazier et al., 2008; Davies et al., 2009). Interestingly,manganese can induce prion protein transformation fromPrPC to a disease-associated isoform (PrPSc) (Brazier et al.,2008). Furthermore, PrPC but not PrPSc has SOD-1 activity,

TABLE 2Brain regions involved in social cognitive functions

Brain Regions Social Cognitive Functions

Prefrontal cortexDLPFCa Top-down control of social behaviorVLPFCb Integration of verbal and nonverbal informationVMPFCc Regulation for emotional responses

STS/STGd Integration of auditory and visual perceptionThalamuse Sensory gate for auditory and visual stimuliHippocampusf Emotion-associated memory, neurocognitionAmygdalaf Emotional responsesCerebellumg Cognition (early childhood), body and limb movement

aHeatherton and Wagner, 2011.bLevy and Wagner, 2011.cMotzkin et al., 2015.dKreifelts et al., 2007.eGeyer and Vollenweider, 2008.fOrsini and Maren, 2012.gWang et al., 2014.

Fig. 3. Neuroinflammation in cuprizone-treated mice. The treatmentwith cuprizone, the copper chelator, appears to disrupt cellular metalhomeostasis and presumably cause dysfunction of CuZn SOD-1 expressedin tanycytes in the BCB and/or in astrocytes in the BBB. Finally, thedysfunction of CuZn SOD-1 can induce neuroinflammation via reactiveoxygen species release, M1 polarization of macrophages/microglia, andproduction of proinflammatory cytokines, including tumor necrosis factor-a, IL-1b, CXCL10, CCL2, CCL3, CCL4, and CCL5.

Neuroinflammation in Social Cognitive Deficits 511

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 9: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

indicating a high oxidation potential of PrPSc (Brown et al.,1999; Milhavet et al., 2000; Yamamoto and Kuwata, 2009).Therefore, the lacking of SOD-1 activity in PrPSc seems to beassociated with reactive oxygen species production and neuro-inflammation under impaired cellular metal homeostasis.InC57BL/6mice treatedwith cuprizone, themRNA levels of

CCL2, CCL5, and CXC-chemokine ligand 10 (CXCL10), alter-natively termed IFN-g–inducible protein 10, showed thegreatest expression in the brain at 1–2 weeks of treatment,whereas CCL3, CCL4, tumor necrosis factor-a, and IL-1bincreased later (4–5 weeks) (McMahon et al., 2001). Further-more, activation of microglia and infiltration of macrophagesinto the brain are markedly increased during cuprizoneintoxication (McMahon et al., 2002). Recently, it has beenreported that microglial activation is significantly reduced inCXCL10-deficient mice treated with cuprizone, suggesting apivotal role of CXCL10 in cuprizone-induced neuroinflamma-tion and demyelination (Clarner et al., 2015). Similarly,IFN-g, IL-6, IL-8, CCL4, and CXCL10 were shown to besignificantly increased in the CSF of ASD individuals (Vargaset al., 2005). Consequently, impaired cellular metal homeo-stasis appears to induce neuroinflammation and demyelin-ation mediated by M1 phenotype of macrophage/microglia incuprizone-treated mice (Fig. 3).

Pharmacological Approach toNeuroinflammation in ASD and SchizophreniaBecause microglia-mediated neuroinflammation plays an

important role in schizophrenia and ASD, a drug that sup-presses microglial activation may be preferentially useful forthe therapy of these disorders. One of such candidates isminocycline, which inhibits the polarization of proinflamma-tory M1 microglia (Kobayashi et al., 2013; Pusic et al., 2014).Minocycline is a second-generation tetracycline and has beenin therapeutic use for over 30 years because of its antibioticproperties; however, it has been recently shown to exert avariety of biologic actions that are independent of theirantimicrobial activity, including anti-inflammatory and anti-apoptotic activities (Garrido-Mesa et al., 2013).Administration of minocycline attenuated the induction

of M1 microglia markers and inhibited the upregulation ofnuclear factor-kB, whereas it did not affect the expression ofM2 microglia markers in cultured microglia stimulated withlipopolysaccharide and in the spinal cord of SOD-1(G93A)mice (Kobayashi et al., 2013). In cuprizone-fed C57BL/6 mice,minocycline reduced microglial activation and demyelinationin the brain and prevented disturbances inmotor coordination(Pasquini et al., 2007; Skripuletz et al., 2010; Tanaka et al.,2013). Furthermore, minocycline in combination with anantipsychotic such as risperidone, olanzapine, quetiapine, orclozapine significantly improved positive, negative, and cog-nitive symptoms in recent-onset or chronic schizophreniapatients (Levkovitz et al., 2010; Ghanizadeh et al., 2014;Chaves et al., 2015; Kelly et al., 2015). From these results, it islikely that application of minocycline can dampen M1 signal-ing, subsequently resulting in skewing of M2a microglia thatreduce proinflammatory signaling and increase productionof anti-inflammatory cytokines. Consequently, minocyclineadd-on treatment may ameliorate clinical deterioration andbrain alterations in schizophrenia patients.

Although there is no clinical use of minocycline in ASD,recent studies have revealed improvement of ASD symptomsby intranasal administration of oxytocin and demonstratedthe association of polymorphisms in the oxytocin receptor(OXTR) gene in ASD (Andari et al., 2010; Guastella et al.,2010; Aoki et al.; 2014; LoParo and Waldman, 2015). Further-more, abnormal activation of microglia and a reduction ofPSD-95 expression in theOxtr-deficient brain (Miyazaki et al.,2016) have been found. Prenatal minocycline treatment canalter the expression of PSD-95 and ameliorate abnormalmother-infant communication in Oxtr-deficient mice (Miyazakiet al., 2016). These findings suggest that minocycline hasa therapeutic potential for the development of oxytocin/Oxtr-mediated ASD-like phenotypes.

ConclusionIn this review, we compare brain alterations in ASD and

those in schizophrenia and propose a possible mechanismunderlying social cognitive deficits. Development of ASDappearsto be triggered by cerebellar dysfunction and subsequentthalamic hyperactivation in early childhood. In contrast, schizo-phrenia seems to be induced by thalamic hyperactivation in lateadolescence, whereas hippocampal aberration possibly has beeninitiated in childhood. The thalamic hyperactivation can lead todysfunction of the DLPFC, which regulates neural circuitry ofsocial cognition. Consequently, we conclude that in ASD andschizophrenia, the initiating brain regions with abnormalitiesare distinct; however, the aberration of the same brain regioncontributes to social cognitive deficits, a common symptom.Furthermore, we strongly suggest that one of the primaryculprits is a disturbance inmetal homeostasis, which can inducedysfunction of the BCB rather than BBB. It is probable thatdysfunction of CuZn SOD-1 expressed in the BCB leads toneuroinflammation via reactive oxygen species release, M1polarization of macrophages/microglia, and production of proin-flammatory cytokines. M1 microglia-mediated neuroinflamma-tion appears to be associatedwith abnormalities of the initiatingbrain regions located adjacent to the cerebral ventricles andsubsequent dysregulation of the neural circuitry of socialcognition in the DLPFC. Consequently, a drug that selectivelysuppresses polarization ofM1microgliamayprovide a beneficialtherapy for ASD and schizophrenia. From this point of view,cuprizone-treated mice are thought to be a useful model formetal homeostasis disturbance and neuroinflammation inthese disorders. This view may pave the way for treatmentof ASD and schizophrenia.

Acknowledgments

The authors acknowledge Professor Sam J. Enna (University ofKansasMedical Center, Kansas City, KS) for invaluable comments onan early draft of this paper.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Nakagawa,Chiba.

References

Abbott NJ, Rönnbäck L, and Hansson E (2006) Astrocyte-endothelial interactions atthe blood-brain barrier. Nat Rev Neurosci 7:41–53.

Abernethy LJ, Palaniappan M, and Cooke RWI (2002) Quantitative magnetic reso-nance imaging of the brain in survivors of very low birth weight. Arch Dis Child 87:279–283.

512 Nakagawa and Chiba

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 10: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

Abrams DA, Lynch CJ, Cheng KM, Phillips J, Supekar K, Ryali S, Uddin LQ,and Menon V (2013) Underconnectivity between voice-selective cortex and rewardcircuitry in children with autism. Proc Natl Acad Sci USA 110:12060–12065.

Aggleton JP, O’Mara SM, Vann SD, Wright NF, Tsanov M, and Erichsen JT (2010)Hippocampal-anterior thalamic pathways for memory: uncovering a network ofdirect and indirect actions. Eur J Neurosci 31:2292–2307.

Allen G (2005) The cerebellum in autism. Clin Neuropsychiatry 2:321–337.Allen G, Müller R-A, and Courchesne E (2004) Cerebellar function in autism: func-tional magnetic resonance image activation during a simple motor task. BiolPsychiatry 56:269–278.

Anagnostou E and Taylor MJ (2011) Review of neuroimaging in autism spectrumdisorders: what have we learned and where we go from here. Mol Autism 2:4.

Andari E, Duhamel JR, Zalla T, Herbrecht E, Leboyer M, and Sirigu A (2010) Pro-moting social behavior with oxytocin in high-functioning autism spectrum disor-ders. Proc Natl Acad Sci USA 107:4389–4394.

Aoki Y, Yahata N, Watanabe T, Takano Y, Kawakubo Y, Kuwabara H, Iwashiro N,Natsubori T, Inoue H, and Suga M, et al. (2014) Oxytocin improves behaviouraland neural deficits in inferring others’ social emotions in autism. Brain 137:3073–3086.

Arking DE, Cutler DJ, Brune CW, Teslovich TM, West K, Ikeda M, Rea A, Guy M,Lin S, and Cook EH, Jr, et al. (2008) A common genetic variant in the neurexinsuperfamily member CNTNAP2 increases familial risk of autism. Am J Hum Genet82:160–164.

Asadabadi M, Mohammadi M-R, Ghanizadeh A, Modabbernia A, Ashrafi M,Hassanzadeh E, Forghani S, and Akhondzadeh S (2013) Celecoxib as adjunctivetreatment to risperidone in children with autistic disorder: a randomized, double-blind, placebo-controlled trial. Psychopharmacology 225:51–59.

Ashwood P, Enstrom A, Krakowiak P, Hertz-Picciotto I, Hansen RL, Croen LA,Ozonoff S, Pessah IN, and Van de Water J (2008) Decreased transforming growthfactor beta1 in autism: a potential link between immune dysregulation and im-pairment in clinical behavioral outcomes. J Neuroimmunol 204:149–153.

Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah IN, and Van de WaterJ (2011a) Associations of impaired behaviors with elevated plasma chemokines inautism spectrum disorders. J Neuroimmunol 232:196–199.

Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah I, and Van de Water J(2011b) Elevated plasma cytokines in autism spectrum disorders provide evidenceof immune dysfunction and are associated with impaired behavioral outcome.Brain Behav Immun 25:40–45.

Banati RB (2002) Visualising microglial activation in vivo. Glia 40:206–217.Beauchamp MH, Thompson DK, Howard K, Doyle LW, Egan GF, Inder TE,and Anderson PJ (2008) Preterm infant hippocampal volumes correlate with laterworking memory deficits. Brain 131:2986–2994.

Benarroch EE (2011) Circumventricular organs: receptive and homeostatic functionsand clinical implications. Neurology 77:1198–1204.

Bilgiç A, Gürkan K, Türko�glu S, Akça OS, Kılıç GJ, and Uslu R (2010) Iron deficiencyin preschool children with autistic spectrum disorders. Res Autism Spectr Disord4:639–644.

Blair RJR (2013) The neurobiology of psychopathic traits in youths.Nat Rev Neurosci14:786–799.

Blank H, Anwander A, and von Kriegstein K (2011) Direct structural connectionsbetween voice- and face-recognition areas. J Neurosci 31:12906–12915.

Bonkowski D, Katyshev V, Balabanov RD, Borisov A, and Dore-Duffy P (2011) TheCNS microvascular pericyte: pericyte-astrocyte crosstalk in the regulation of tissuesurvival. Fluids Barriers CNS 8:8.

Borgwardt SJ, Riecher-Rössler A, Dazzan P, Chitnis X, Aston J, Drewe M,Gschwandtner U, Haller S, Pflüger M, and Rechsteiner E, et al. (2007) Regionalgray matter volume abnormalities in the at risk mental state. Biol Psychiatry 61:1148–1156.

Bornhorst J, Wehe CA, Hüwel S, Karst U, Galla H-J, and Schwerdtle T (2012) Impactof manganese on and transfer across blood-brain and blood-cerebrospinal fluidbarrier in vitro. J Biol Chem 287:17140–17151.

Bottmer C, Bachmann S, Pantel J, Essig M, Amann M, Schad LR, Magnotta V,and Schröder J (2005) Reduced cerebellar volume and neurological soft signs infirst-episode schizophrenia. Psychiatry Res 140:239–250.

Bourgeron T (2009) A synaptic trek to autism. Curr Opin Neurobiol 19:231–234.Bowers JM and Konopka G (2012) The role of the FOXP family of transcriptionfactors in ASD. Dis Markers 33:251–260.

Braunschweig D, Krakowiak P, Duncanson P, Boyce R, Hansen RL, Ashwood P,Hertz-Picciotto I, Pessah IN, and Van de Water J (2013) Autism-specific maternalautoantibodies recognize critical proteins in developing brain. Transl Psychiatry 3:e277.

Brazier MW, Davies P, Player E, Marken F, Viles JH, and Brown DR (2008) Man-ganese binding to the prion protein. J Biol Chem 283:12831–12839.

Breymann C (2015) Iron deficiency anemia in pregnancy. Semin Hematol 52:339–347.Brito AR, Vasconcelos MM, Domingues RC, Hygino da Cruz LC, Jr, Rodrigues L de S,Gasparetto EL, and Calçada CABP (2009) Diffusion tensor imaging findings inschool-aged autistic children. J Neuroimaging 19:337–343.

Brown AS (2011) The environment and susceptibility to schizophrenia. Prog Neu-robiol 93:23–58.

Brown DR, Wong B-S, Hafiz F, Clive C, Haswell SJ, and Jones IM (1999) Normalprion protein has an activity like that of superoxide dismutase. Biochem J 344:1–5.

Cannon M, Caspi A, Moffitt TE, Harrington H, Taylor A, Murray RM, and Poulton R(2002) Evidence for early-childhood, pan-developmental impairment specific toschizophreniform disorder: results from a longitudinal birth cohort. Arch GenPsychiatry 59:449–456.

Caroni P, Chowdhury A, and Lahr M (2014) Synapse rearrangements upon learning:from divergent-sparse connectivity to dedicated sub-circuits. Trends Neurosci 37:604–614.

Catts VS, Fung SJ, Long LE, Joshi D, Vercammen A, Allen KM, Fillman SG,Rothmond DA, Sinclair D, and Tiwari Y, et al. (2013) Rethinking schizophrenia inthe context of normal neurodevelopment. Front Cell Neurosci 7:60.

Chan RCK, Di X, McAlonan GM, and Gong QY (2011) Brain anatomical abnormal-ities in high-risk individuals, first-episode, and chronic schizophrenia: an activa-tion likelihood estimation meta-analysis of illness progression. Schizophr Bull 37:177–188.

Chan RCK, Xu T, Heinrichs RW, Yu Y, and Wang Y (2010) Neurological soft signs inschizophrenia: a meta-analysis. Schizophr Bull 36:1089–1104.

Chaves C, Marque CR, Maia-de-Oliveira JP, Wichert-Ana L, Ferrari TB, Santos AC,Araújo D, Machado-de-Sousa JP, Bressan RA, and Elkis H, et al. (2015) Effects ofminocycline add-on treatment on brain morphometry and cerebral perfusion inrecent-onset schizophrenia. Schizophr Res 161:439–445.

Chen HB, Chan Y-T, Hung AC, Tsai Y-C, and Sun SH (2006) Elucidation of ATP-stimulated stress protein expression of RBA-2 type-2 astrocytes: ATP potentiateHSP60 and Cu/Zn SOD expression and stimulates pI shift of peroxiredoxin II. JCell Biochem 97:314–326.

Cheon K-A, Kim Y-S, Oh S-H, Park S-Y, Yoon H-W, Herrington J, Nair A, Koh Y-J,Jang D-P, and Kim Y-B, et al. (2011) Involvement of the anterior thalamic radia-tion in boys with high functioning autism spectrum disorders: a Diffusion TensorImaging study. Brain Res 1417:77–86.

Choi YK and Kim K-W (2008) Blood-neural barrier: its diversity and coordinated cell-to-cell communication. BMB Rep 41:345–352.

Clarner T, Janssen K, Nellessen L, Stangel M, Skripuletz T, Krauspe B, Hess F-M,Denecke B, Beutner C, and Linnartz-Gerlach B, et al. (2015) CXCL10 triggers earlymicroglial activation in the cuprizone model. J Immunol 194:3400–3413.

Courchesne E, Mouton PR, Calhoun ME, Semendeferi K, Ahrens-Barbeau C, HalletMJ, Barnes CC, and Pierce K (2011) Neuron number and size in prefrontal cortexof children with autism. JAMA 306:2001–2010.

Couture SM, Penn DL, and Roberts DL (2006) The functional significance of socialcognition in schizophrenia: a review. Schizophr Bull 32 (Suppl 1):S44–S63.

Davies P, Marken F, Salter S, and Brown DR (2009) Thermodynamic and voltam-metric characterization of the metal binding to the prion protein: insights into pHdependence and redox chemistry. Biochemistry 48:2610–2619.

Dawson G, Meltzoff AN, Osterling J, and Rinaldi J (1998) Neuropsychological cor-relates of early symptoms of autism. Child Dev 69:1276–1285.

Dichter GS (2012) Functional magnetic resonance imaging of autism spectrum dis-orders. Dialogues Clin Neurosci 14:319–351.

Dusek P, Roos PM, Litwin T, Schneider SA, Flaten TP, and Aaseth J (2015) Theneurotoxicity of iron, copper and manganese in Parkinson’s and Wilson’s diseases.J Trace Elem Med Biol 31:193–203.

Dziegielewska KM, Ek J, Habgood MD, and Saunders NR (2001) Development of thechoroid plexus. Microsc Res Tech 52:5–20.

Faber S, Zinn GM, Kern JC, 2nd, and Kingston HMS (2009) The plasma zinc/serumcopper ratio as a biomarker in children with autism spectrum disorders. Bio-markers 14:171–180.

Fan F-M, Tan S-P, Yang F-D, Tan Y-L, Zhao Y-L, Chen N, Li B-B, Song C-S, WangY-H, and Jin Z, et al. (2013) Ventral medial prefrontal functional connectivity andemotion regulation in chronic schizophrenia: a pilot study. Neurosci Bull 29:59–74.

Ferrarelli F, Peterson MJ, Sarasso S, Riedner BA, Murphy MJ, Benca RM, Bria P,Kalin NH, and Tononi G (2010) Thalamic dysfunction in schizophrenia suggestedby whole-night deficits in slow and fast spindles. Am J Psychiatry 167:1339–1348.

Frazier TW, Thompson L, Youngstrom EA, Law P, Hardan AY, Eng C, and Morris N(2014) A twin study of heritable and shared environmental contributions to autism.J Autism Dev Disord 44:2013–2025.

Fujii E, Mori K, Miyazaki M, Hashimoto T, Harada M, and Kagami S (2010) Functionof the frontal lobe in autistic individuals: a proton magnetic resonance spectro-scopic study. J Med Invest 57:35–44.

Fujita E, Dai H, Tanabe Y, Zhiling Y, Yamagata T, Miyakawa T, Tanokura M, MomoiMY, and Momoi T (2010) Autism spectrum disorder is related to endoplasmicreticulum stress induced by mutations in the synaptic cell adhesion molecule,CADM1. Cell Death Dis 1:e47.

Fujita E, Tanabe Y, Shiota A, Ueda M, Suwa K, Momoi MY, and Momoi T (2008)Ultrasonic vocalization impairment of Foxp2 (R552H) knockin mice related tospeech-language disorder and abnormality of Purkinje cells. Proc Natl Acad SciUSA 105:3117–3122.

Fujita-Jimbo E, Tanabe Y, Yu Z, Kojima K, Mori M, Li H, Iwamoto S, Yamagata T,Momoi MY, and Momoi T (2015) The association of GPR85 with PSD-95-neuroligincomplex and autism spectrum disorder: a molecular analysis. Mol Autism 6:17.

Fujita-Jimbo E, Yu ZL, Li H, Yamagata T, Mori M, Momoi T, and Momoi MY (2012)Mutation in Parkinson disease-associated, G-protein-coupled receptor 37 (GPR37/PaelR) is related to autism spectrum disorder. PLoS One 7:e51155.

Gardener H, Spiegelman D, and Buka SL (2009) Prenatal risk factors for autism:comprehensive meta-analysis. Br J Psychiatry 195:7–14.

Gardener H, Spiegelman D, and Buka SL (2011) Perinatal and neonatal risk factorsfor autism: a comprehensive meta-analysis. Pediatrics 128:344–355.

Garrido-Mesa N, Zarzuelo A, and Gálvez J (2013) Minocycline: far beyond an anti-biotic. Br J Pharmacol 169:337–352.

Gejman PV, Sanders AR, and Duan J (2010) The role of genetics in the etiology ofschizophrenia. Psychiatr Clin North Am 33:35–66.

Gervais H, Belin P, Boddaert N, Leboyer M, Coez A, Sfaello I, Barthélémy C,Brunelle F, Samson Y, and Zilbovicius M (2004) Abnormal cortical voice processingin autism. Nat Neurosci 7:801–802.

Geyer MA and Vollenweider FX (2008) Serotonin research: contributions to un-derstanding psychoses. Trends Pharmacol Sci 29:445–453.

Ghanizadeh A, Dehbozorgi S, OmraniSigaroodi M, and Rezaei Z (2014) Minocycline asadd-on treatment decreases the negative symptoms of schizophrenia; a randomizedplacebo-controlled clinical trial. Recent Pat Inflamm Allergy Drug Discov 8:211–215.

Glasson EJ, Bower C, Petterson B, de Klerk N, Chaney G, and Hallmayer JF (2004)Perinatal factors and the development of autism: a population study. Arch GenPsychiatry 61:618–627.

Gordon S and Martinez FO (2010) Alternative activation of macrophages: mechanismand functions. Immunity 32:593–604.

Neuroinflammation in Social Cognitive Deficits 513

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 11: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

Gottfried C, Bambini-Junior V, Francis F, Riesgo R, and Savino W (2015) The impactof neuroimmune alterations in autism spectrum disorder. Front Psychiatry 6:121.

Groebe A, Clarner T, Baumgartner W, Dang J, Beyer C, and Kipp M (2009) Cupri-zone treatment induces distinct demyelination, astrocytosis, and microglia cellinvasion or proliferation in the mouse cerebellum. Cerebellum 8:163–174.

Guastella AJ, Einfeld SL, Gray KM, Rinehart NJ, Tonge BJ, Lambert TJ, and HickieIB (2010) Intranasal oxytocin improves emotion recognition for youth with autismspectrum disorders. Biol Psychiatry 67:692–694.

Gudi V, Moharregh-Khiabani D, Skripuletz T, Koutsoudaki PN, Kotsiari A, Skuljec J,Trebst C, and Stangel M (2009) Regional differences between grey and whitematter in cuprizone induced demyelination. Brain Res 1283:127–138.

Guinchat V, Thorsen P, Laurent C, Cans C, Bodeau N, and Cohen D (2012) Pre-, peri-and neonatal risk factors for autism. Acta Obstet Gynecol Scand 91:287–300.

Gunter TE, Gerstner B, Gunter KK, Malecki J, Gelein R, Valentine WM, Aschner M,and Yule DI (2013) Manganese transport via the transferrin mechanism. Neuro-toxicology 34:118–127.

Hackett TA (2015) Anatomic organization of the auditory cortex. Handb Clin Neurol129:27–53.

Hardan AY, Girgis RR, Adams J, Gilbert AR, Keshavan MS, and Minshew NJ (2006)Abnormal brain size effect on the thalamus in autism. Psychiatry Res 147:145–151.

Hardan AY, Minshew NJ, Melhem NM, Srihari S, Jo B, Bansal R, Keshavan MS,and Stanley JA (2008) An MRI and proton spectroscopy study of the thalamus inchildren with autism. Psychiatry Res 163:97–105.

Hassel S, Almeida JRC, Kerr N, Nau S, Ladouceur CD, Fissell K, Kupfer DJ,and Phillips ML (2008) Elevated striatal and decreased dorsolateral prefrontalcortical activity in response to emotional stimuli in euthymic bipolar disorder: noassociations with psychotropic medication load. Bipolar Disord 10:916–927.

Heatherton TF and Wagner DD (2011) Cognitive neuroscience of self-regulationfailure. Trends Cogn Sci 15:132–139.

Heo Y, Zhang Y, Gao D, Miller VM, and Lawrence DA (2011) Aberrant immuneresponses in a mouse with behavioral disorders. PLoS One 6:e20912.

Hultman CM, Sparén P, Takei N, Murray RM, and Cnattingius S (1999) Prenataland perinatal risk factors for schizophrenia, affective psychosis, and reactive psy-chosis of early onset: case-control study. BMJ 318:421–426.

Jackson SP (2011) Arterial thrombosis: insidious, unpredictable and deadly.Nat Med17:1423–1436.

Kalmady SV, Venkatasubramanian G, Shivakumar V, Gautham S, Subramaniam A,Jose DA, Maitra A, Ravi V, and Gangadhar BN (2014) Relationship betweeninterleukin-6 gene polymorphism and hippocampal volume in antipsychotic-naiveschizophrenia: evidence for differential susceptibility? PLoS One 9:e96021.

Kang HJ, Voleti B, Hajszan T, Rajkowska G, Stockmeier CA, Licznerski P, Lepack A,Majik MS, Jeong LS, and Banasr M, et al. (2012) Decreased expression of synapse-related genes and loss of synapses in major depressive disorder. Nat Med 18:1413–1417.

Kelly DL, Sullivan KM, McEvoy JP, McMahon RP, Wehring HJ, Gold JM, Liu F,Warfel D, Vyas G, and Richardson CM, et al. (2015) Adjunctive minocycline inclozapine-treated schizophrenia patients with persistent symptoms. J Clin Psy-chopharmacol 35:374–381.

Kettenmann H, Hanisch U-K, Noda M, and Verkhratsky A (2011) Physiology ofmicroglia. Physiol Rev 91:461–553.

Khan S, Gramfort A, Shetty NR, Kitzbichler MG, Ganesan S, Moran JM, Lee SM,Gabrieli JDE, Tager-Flusberg HB, and Joseph RM, et al. (2013) Local and long-range functional connectivity is reduced in concert in autism spectrum disorders.Proc Natl Acad Sci USA 110:3107–3112.

Kiernan JA (2012) Anatomy of the temporal lobe. Epilepsy Res Treat 2012:176157.Kleinhans NM, Richards T, Sterling L, Stegbauer KC, Mahurin R, Johnson LC,Greenson J, Dawson G, and Aylward E (2008) Abnormal functional connectivity inautism spectrum disorders during face processing. Brain 131:1000–1012.

Kobayashi K, Imagama S, Ohgomori T, Hirano K, Uchimura K, Sakamoto K,Hirakawa A, Takeuchi H, Suzumura A, and Ishiguro N, et al. (2013) Minocyclineselectively inhibits M1 polarization of microglia. Cell Death Dis 4:e525.

Koutsoudaki PN, Skripuletz T, Gudi V, Moharregh-Khiabani D, Hildebrandt H,Trebst C, and Stangel M (2009) Demyelination of the hippocampus is prominent inthe cuprizone model. Neurosci Lett 451:83–88.

Koyama R and Ikegaya Y (2015) Microglia in the pathogenesis of autism spectrumdisorders. Neurosci Res 100:1–5.

Koziol LF, Budding D, Andreasen N, D’Arrigo S, Bulgheroni S, Imamizu H, Ito M,Manto M, Marvel C, and Parker K, et al. (2014) Consensus paper: the cerebellum’srole in movement and cognition. Cerebellum 13:151–177.

Kreifelts B, Ethofer T, Grodd W, Erb M, and Wildgruber D (2007) Audiovisual in-tegration of emotional signals in voice and face: an event-related fMRI study.Neuroimage 37:1445–1456.

Kurt S, Fisher SE, and Ehret G (2012) Foxp2 mutations impair auditory-motor as-sociation learning. PLoS One 7:e33130.

Lai G, Pantazatos SP, Schneider H, and Hirsch J (2012) Neural systems for speechand song in autism. Brain 135:961–975.

Lai M-C, Lombardo MV, Chakrabarti B, Sadek SA, Pasco G, Wheelwright SJ, BullmoreET, Baron-Cohen S, and Suckling J; MRC AIMS Consortium (2010) A shift to ran-domness of brain oscillations in people with autism. Biol Psychiatry 68:1092–1099.

Langlet F, Mullier A, Bouret SG, Prevot V, and Dehouck B (2013) Tanycyte-like cellsform a blood-cerebrospinal fluid barrier in the circumventricular organs of themouse brain. J Comp Neurol 521:3389–3405.

Larson MK, Walker EF, and Compton MT (2010) Early signs, diagnosis and thera-peutics of the prodromal phase of schizophrenia and related psychotic disorders.Expert Rev Neurother 10:1347–1359.

Lebel C, Walker L, Leemans A, Phillips L, and Beaulieu C (2008) Microstructuralmaturation of the human brain from childhood to adulthood. Neuroimage 40:1044–1055.

LeBlanc JJ and Fagiolini M (2011) Autism: a “critical period” disorder? Neural Plast2011:921680.

Lee C-M and Bo J (2015) Motor functioning in children with autism spectrum dis-order. Acta Psychopathologica 1:13.

Levkovitz Y, Mendlovich S, Riwkes S, Braw Y, Levkovitch-Verbin H, Gal G, Fennig S,Treves I, and Kron S (2010) A double-blind, randomized study of minocycline forthe treatment of negative and cognitive symptoms in early-phase schizophrenia. JClin Psychiatry 71:138–149.

Levy BJ and Wagner AD (2011) Cognitive control and right ventrolateral prefrontalcortex: reflexive reorienting, motor inhibition, and action updating. Ann N Y AcadSci 1224:40–62.

Li X, Chauhan A, Sheikh AM, Patil S, Chauhan V, Li X-M, Ji L, Brown T, and MalikM (2009) Elevated immune response in the brain of autistic patients. J Neuro-immunol 207:111–116.

Liddelow SA (2015) Development of the choroid plexus and blood-CSF barrier. FrontNeurosci 9:32.

Lisé MF and El-Husseini A (2006) The neuroligin and neurexin families: fromstructure to function at the synapse. Cell Mol Life Sci 63:1833–1849.

LoParo D and Waldman ID (2015) The oxytocin receptor gene (OXTR) is associatedwith autism spectrum disorder: a meta-analysis. Mol Psychiatry 20:640–646.

Lun MP, Monuki ES, and Lehtinen MK (2015) Development and functions of thechoroid plexus-cerebrospinal fluid system. Nat Rev Neurosci 16:445–457.

Lupien SJ, Parent S, Evans AC, Tremblay RE, Zelazo PD, Corbo V, Pruessner JC,and Séguin JR (2011) Larger amygdala but no change in hippocampal volume in10-year-old children exposed to maternal depressive symptomatology since birth.Proc Natl Acad Sci USA 108:14324–14329.

Marenco S and Weinberger DR (2000) The neurodevelopmental hypothesis ofschizophrenia: following a trail of evidence from cradle to grave. Dev Psychopathol12:501–527.

Martins VR, Mercadante AF, Cabral ALB, Freitas ARO, and Castro RMRPS (2001)Insights into the physiological function of cellular prion protein. Braz J Med BiolRes 34:585–595.

Mazzola V, Vuilleumier P, Latorre V, Petito A, Gallese V, Popolizio T, Arciero G,and Bondolfi G (2013) Effects of emotional contexts on cerebello-thalamo-corticalactivity during action observation. PLoS One 8:e75912.

McAlonan GM, Cheung V, Cheung C, Suckling J, Lam GY, Tai KS, Yip L, MurphyDGM, and Chua SE (2005) Mapping the brain in autism: a voxel-based MRI studyof volumetric differences and intercorrelations in autism. Brain 128:268–276.

McFarlane HG, Kusek GK, Yang M, Phoenix JL, Bolivar VJ, and Crawley JN (2008)Autism-like behavioral phenotypes in BTBR T1tf/J mice. Genes Brain Behav7:152–163.

McMahon EJ, Cook DN, Suzuki K, and Matsushima GK (2001) Absence ofmacrophage-inflammatory protein-1a delays central nervous system demyelinationin the presence of an intact blood-brain barrier. J Immunol 167:2964–2971.

McMahon EJ, Suzuki K, and Matsushima GK (2002) Peripheral macrophage re-cruitment in cuprizone-induced CNS demyelination despite an intact blood-brainbarrier. J Neuroimmunol 130:32–45.

Meyer U, Feldon J, and Dammann O (2011a) Schizophrenia and autism: both sharedand disorder-specific pathogenesis via perinatal inflammation? Pediatr Res 69:26R–33R.

Meyer U, Schwarz MJ, and Müller N (2011b) Inflammatory processes in schizo-phrenia: a promising neuroimmunological target for the treatment of negative/cognitive symptoms and beyond. Pharmacol Ther 132:96–110.

Mikita J, Dubourdieu-Cassagno N, Deloire MS, Vekris A, Biran M, Raffard G,Brochet B, Canron MH, Franconi JM, and Boiziau C, et al. (2011) Altered M1/M2activation patterns of monocytes in severe relapsing experimental rat model ofmultiple sclerosis: amelioration of clinical status by M2 activated monocyte ad-ministration. Mult Scler 17:2–15.

Miles JH (2011) Autism spectrum disorders: a genetics review. Genet Med 13:278–294.

Milhavet O, McMahon HEM, Rachidi W, Nishida N, Katamine S, Mangé A, ArlottoM, Casanova D, Riondel J, and Favier A, et al. (2000) Prion infection impairs thecellular response to oxidative stress. Proc Natl Acad Sci USA 97:13937–13942.

Miyazaki S, Hiraoka Y, Hidema S, and Nishimori K (2016) Prenatal minocyclinetreatment alters synaptic protein expression, and rescues reduced mother callrate in oxytocin receptor-knockout mice. Biochem Biophys Res Commun 472:319–323.

Money KM and Stanwood GD (2013) Developmental origins of brain disorders: rolesfor dopamine. Front Cell Neurosci 7:260.

Monk CS, Weng S-J, Wiggins JL, Kurapati N, Louro HMC, Carrasco M, Maslowsky J,Risi S, and Lord C (2010) Neural circuitry of emotional face processing in autismspectrum disorders. J Psychiatry Neurosci 35:105–114.

Morgan JT, Chana G, Pardo CA, Achim C, Semendeferi K, Buckwalter J, CourchesneE, and Everall IP (2010) Microglial activation and increased microglial densityobserved in the dorsolateral prefrontal cortex in autism. Biol Psychiatry 68:368–376.

Mosser DM and Edwards JP (2008) Exploring the full spectrum of macrophage ac-tivation. Nat Rev Immunol 8:958–969.

Mostofsky SH, Powell SK, Simmonds DJ, Goldberg MC, Caffo B, and Pekar JJ (2009)Decreased connectivity and cerebellar activity in autism during motor task per-formance. Brain 132:2413–2425.

Motzkin JC, Philippi CL, Wolf RC, Baskaya MK, and Koenigs M (2015) Ventromedialprefrontal cortex is critical for the regulation of amygdala activity in humans. BiolPsychiatry 77:276–284.

Mukamel Z, Konopka G, Wexler E, Osborn GE, Dong H, Bergman MY, Levitt P,and Geschwind DH (2011) Regulation of MET by FOXP2, genes implicated inhigher cognitive dysfunction and autism risk. J Neurosci 31:11437–11442.

Nair A, Treiber JM, Shukla DK, Shih P, and Müller R-A (2013) Impaired thalamo-cortical connectivity in autism spectrum disorder: a study of functional and ana-tomical connectivity. Brain 136:1942–1955.

Nakagawa Y and Chiba K (2014) Role of microglial m1/m2 polarization in relapse andremission of psychiatric disorders and diseases. Pharmaceuticals 7:1028–1048.

514 Nakagawa and Chiba

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from

Page 12: Involvement of Neuroinflammation during Brain Development ...jpet.aspetjournals.org/content/jpet/358/3/504.full.pdfthecerebellum,midbrain,fusiformgyrus,andprefrontalcortex in ASD adults

Nakagawa Y and Chiba K (2015) Diversity and plasticity of microglial cells in psy-chiatric and neurological disorders. Pharmacol Ther 154:21–35.

Nakamura T, Furuhashi M, Li P, Cao H, Tuncman G, Sonenberg N, Gorgun CZ,and Hotamisligil GS (2010) Double-stranded RNA-dependent protein kinase linkspathogen sensing with stress and metabolic homeostasis. Cell 140:338–348.

Neggers YH (2014) Increasing prevalence, changes in diagnostic criteria, and nu-tritional risk factors for autism spectrum disorders. ISRN Nutr 2014:514026.

Okada K, Hashimoto K, Iwata Y, Nakamura K, Tsujii M, Tsuchiya KJ, Sekine Y,Suda S, Suzuki K, and Sugihara G, et al. (2007) Decreased serum levels of trans-forming growth factor-b1 in patients with autism. Prog Neuropsychopharmacol BiolPsychiatry 31:187–190.

Onore C, Careaga M, and Ashwood P (2012) The role of immune dysfunction in thepathophysiology of autism. Brain Behav Immun 26:383–392.

Orsini CA and Maren S (2012) Neural and cellular mechanisms of fear and extinctionmemory formation. Neurosci Biobehav Rev 36:1773–1802.

Palaniyappan L, Simmonite M, White TP, Liddle EB, and Liddle PF (2013) Neuralprimacy of the salience processing system in schizophrenia. Neuron 79:814–828.

Pankow A, Friedel E, Sterzer P, Seiferth N, Walter H, Heinz A, and Schlagenhauf F(2013) Altered amygdala activation in schizophrenia patients during emotionprocessing. Schizophr Res 150:101–106.

Pasquini LA, Calatayud CA, Bertone Uña AL, Millet V, Pasquini JM, and Soto EF(2007) The neurotoxic effect of cuprizone on oligodendrocytes depends on thepresence of pro-inflammatory cytokines secreted by microglia. Neurochem Res 32:279–292.

Patterson PH (2011) Maternal infection and immune involvement in autism. TrendsMol Med 17:389–394.

Peluffo H, Acarin L, Faiz M, Castellano B, and Gonzalez B (2005) Cu/Zn superoxidedismutase expression in the postnatal rat brain following an excitotoxic injury. JNeuroinflammation 2:12.

Phillipson M and Kubes P (2011) The neutrophil in vascular inflammation. Nat Med17:1381–1390.

Pilowsky T, Yirmiya N, Arbelle S, and Mozes T (2000) Theory of mind abilities ofchildren with schizophrenia, children with autism, and normally developing chil-dren. Schizophr Res 42:145–155.

Potschka H, Baltes S, Fedrowitz M, and Löscher W (2011) Impact of seizure activityon free extracellular phenytoin concentrations in amygdala-kindled rats. Neuro-pharmacology 61:909–917.

Pusic KM, Pusic AD, Kemme J, and Kraig RP (2014) Spreading depression requiresmicroglia and is decreased by their M2a polarization from environmental enrich-ment. Glia 62:1176–1194.

Ramasamy I, Law M, Collins S, and Brooke F (2003) Organ distribution of prionproteins in variant Creutzfeldt-Jakob disease. Lancet Infect Dis 3:214–222.

Ransohoff RM and Perry VH (2009) Microglial physiology: unique stimuli, specializedresponses. Annu Rev Immunol 27:119–145.

Redcay E (2008) The superior temporal sulcus performs a common function for socialand speech perception: implications for the emergence of autism. Neurosci Bio-behav Rev 32:123–142.

Reichenberg A, Weiser M, Rabinowitz J, Caspi A, Schmeidler J, Mark M, Kaplan Z,and Davidson M (2002) A population-based cohort study of premorbid intellectual,language, and behavioral functioning in patients with schizophrenia, schizoaffectivedisorder, and nonpsychotic bipolar disorder. Am J Psychiatry 159:2027–2035.

Rogers TD, McKimm E, Dickson PE, Goldowitz D, Blaha CD, and Mittleman G (2013)Is autism a disease of the cerebellum? An integration of clinical and pre-clinicalresearch. Front Syst Neurosci 7:15.

Rosenberg RE, Law JK, Yenokyan G, McGready J, Kaufmann WE, and Law PA(2009) Characteristics and concordance of autism spectrum disorders among277 twin pairs. Arch Pediatr Adolesc Med 163:907–914.

Rossignol DA, Genuis SJ, and Frye RE (2014) Environmental toxicants and autismspectrum disorders: a systematic review. Transl Psychiatry 4:e360.

Roullet FI, Lai JKY, and Foster JA (2013) In utero exposure to valproic acid and autism:a current review of clinical and animal studies. Neurotoxicol Teratol 36:47–56.

Saijo K and Glass CK (2011) Microglial cell origin and phenotypes in health anddisease. Nat Rev Immunol 11:775–787.

Sato W, Toichi M, Uono S, and Kochiyama T (2012) Impaired social brain network forprocessing dynamic facial expressions in autism spectrum disorders. BMC Neu-rosci 13:99.

Saunders NR, Dreifuss J-J, Dziegielewska KM, Johansson PA, Habgood MD,Møllgård K, and Bauer H-C (2014) The rights and wrongs of blood-brain barrierpermeability studies: a walk through 100 years of history. Front Neurosci 8:404.

Sawa T, Kodaira M, Oiji A, Sasayama D, Iwadare Y, Ushijima H, Usami M,Watanabe K, and Saito K (2013) Dysfunction of orbitofrontal and dorsolateralprefrontal cortices in children and adolescents with high-functioning pervasivedevelopmental disorders. Ann Gen Psychiatry 12:31.

Schmahmann JD and Sherman JC (1998) The cerebellar cognitive affective syn-drome. Brain 121:561–579.

Schultz RT (2005) Developmental deficits in social perception in autism: the role ofthe amygdala and fusiform face area. Int J Dev Neurosci 23:125–141.

Schumann CM, Hamstra J, Goodlin-Jones BL, Lotspeich LJ, Kwon H, BuonocoreMH, Lammers CR, Reiss AL, and Amaral DG (2004) The amygdala is enlarged inchildren but not adolescents with autism; the hippocampus is enlarged at all ages.J Neurosci 24:6392–6401.

Scott-Van Zeeland AA, Abrahams BS, Alvarez-Retuerto AI, Sonnenblick LI, RudieJD, Ghahremani D, Mumford JA, Poldrack RA, Dapretto M, and Geschwind DH,et al. (2010) Altered functional connectivity in frontal lobe circuits is associatedwith variation in the autism risk gene CNTNAP2. Sci Transl Med 2:56ra80.

Sealey LA, Hughes BW, Sriskanda AN, Guest JR, Gibson AD, Johnson-Williams L,Pace DG, and Bagasra O (2016) Environmental factors in the development of au-tism spectrum disorders. Environ Int 88:288–298.

Shalev H, Serlin Y, and Friedman A (2009) Breaching the blood-brain barrier as agate to psychiatric disorder. Cardiovasc Psychiatry Neurol 2009:278531.

Shinn AK, Baker JT, Lewandowski KE, Öngür D, and Cohen BM (2015) Aberrantcerebellar connectivity in motor and association networks in schizophrenia. FrontHum Neurosci 9:134.

Shu W, Cho JY, Jiang Y, Zhang M, Weisz D, Elder GA, Schmeidler J, De Gasperi R,Sosa MAG, and Rabidou D, et al. (2005) Altered ultrasonic vocalization in micewith a disruption in the Foxp2 gene. Proc Natl Acad Sci USA 102:9643–9648.

Sidrak S, Yoong T, and Woolfenden S (2014) Iron deficiency in children with globaldevelopmental delay and autism spectrum disorder. J Paediatr Child Health 50:356–361.

Sisó S, Jeffrey M, and González L (2010) Sensory circumventricular organs in healthand disease. Acta Neuropathol 120:689–705.

Skripuletz T, Miller E, Moharregh-Khiabani D, Blank A, Pul R, Gudi V, Trebst C,and Stangel M (2010) Beneficial effects of minocycline on cuprizone induced cor-tical demyelination. Neurochem Res 35:1422–1433.

Sparks BF, Friedman SD, Shaw DW, Aylward EH, Echelard D, Artru AA, MaravillaKR, Giedd JN, Munson J, and Dawson G, et al. (2002) Brain structural abnor-malities in young children with autism spectrum disorder. Neurology 59:184–192.

Stahl SM (2013) Stahl’s Essential Psychopharmacology: Neuroscientific Basis andPractical Applications, 4th ed, Cambridge University Press, Cambridge, UK.

Su CH and Young YH (2011) Disorders affecting the fourth ventricle: etiology andclinical correlates. Otol Neurotol 32:1329–1335.

Suzuki K, Sugihara G, Ouchi Y, Nakamura K, Futatsubashi M, Takebayashi K,Yoshihara Y, Omata K, Matsumoto K, and Tsuchiya KJ, et al. (2013) Microglial acti-vation in young adults with autism spectrum disorder. JAMA Psychiatry 70:49–58.

Tager-Flusberg H (2000) Understanding the language and communicative impair-ments in autism, in International Review of Research on Mental Retardation(Glidden LM ed) pp 185–205, Academic Press, San Diego, CA.

Takahashi E, Ohki K, and Kim D-S (2007) Diffusion tensor studies dissociated twofronto-temporal pathways in the human memory system. Neuroimage 34:827–838.

Tamietto M and de Gelder B (2010) Neural bases of the non-conscious perception ofemotional signals. Nat Rev Neurosci 11:697–709.

Tamura R, Kitamura H, Endo T, Hasegawa N, and Someya T (2010) Reduced tha-lamic volume observed across different subgroups of autism spectrum disorders.Psychiatry Res 184:186–188.

Tanaka SC, Doya K, Okada G, Ueda K, Okamoto Y, and Yamawaki S (2004) Pre-diction of immediate and future rewards differentially recruits cortico-basal gan-glia loops. Nat Neurosci 7:887–893.

Tanaka T, Murakami K, Bando Y, and Yoshida S (2013) Minocycline reducesremyelination by suppressing ciliary neurotrophic factor expression after cuprizone-induced demyelination. J Neurochem 127:259–270.

Tetreault NA, Hakeem AY, Jiang S, Williams BA, Allman E, Wold BJ, and Allman JM(2012) Microglia in the cerebral cortex in autism. J Autism Dev Disord 42:2569–2584.

Tsatsanis KD, Rourke BP, Klin A, Volkmar FR, Cicchetti D, and Schultz RT (2003)Reduced thalamic volume in high-functioning individuals with autism. Biol Psy-chiatry 53:121–129.

Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, and Pardo CA (2005)Neuroglial activation and neuroinflammation in the brain of patients with autism.Ann Neurol 57:67–81.

Volkmar F, Siegel M, Woodbury-Smith M, King B, McCracken J, and State M;American Academy of Child and Adolescent Psychiatry (AACAP) Committee onQuality Issues (CQI) (2014) Practice parameter for the assessment and treatmentof children and adolescents with autism spectrum disorder. J Am Acad ChildAdolesc Psychiatry 53:237–257.

Vuilleumier P and Pourtois G (2007) Distributed and interactive brain mechanismsduring emotion face perception: evidence from functional neuroimaging. Neuro-psychologia 45:174–194.

Wang SS-H, Kloth AD, and Badura A (2014) The cerebellum, sensitive periods, andautism. Neuron 83:518–532.

Weinberger DR (1986) The pathogenesis of schizophrenia: a neurodevelopmentaltheory, in The Neurology of Schizophrenia (Nasrallah HAW and Weinberger DReds) pp 397–406, Elsevier, Amsterdam.

Weinstein M, Ben-Sira L, Levy Y, Zachor DA, Ben Itzhak E, Artzi M, Tarrasch R,Eksteine PM, Hendler T, and Ben Bashat D (2011) Abnormal white matter in-tegrity in young children with autism. Hum Brain Mapp 32:534–543.

Weissman DH, Perkins AS, and Woldorff MG (2008) Cognitive control in social sit-uations: a role for the dorsolateral prefrontal cortex. Neuroimage 40:955–962.

Wilson S and Argyropoulos S (2012) Sleep in schizophrenia: time for closer attention.Br J Psychiatry 200:273–274.

World Health Organization (2016) http://www.who.int/whr/2002/chapter4/en/index3.html.Xiang AH, Wang X, Martinez MP, Walthall JC, Curry ES, Page K, Buchanan TA,Coleman KJ, and Getahun D (2015) Association of maternal diabetes with autismin offspring. JAMA 313:1425–1434.

Yamamoto N and Kuwata K (2009) Difference in redox behaviors between copper-bindingoctarepeat and nonoctarepeat sites in prion protein. J Biol Inorg Chem 14:1209–1218.

Yasuda H, Yasuda Y, and Tsutsui T (2013) Estimation of autistic children by met-allomics analysis. Sci Rep 3:1199.

Yasuda H, Yoshida K, Yasuda Y, and Tsutsui T (2011) Infantile zinc deficiency:association with autism spectrum disorders. Sci Rep 1:129.

Zatta P, Raso M, Zambenedetti P, Wittkowski W, Messori L, Piccioli F, Mauri PL,and Beltramini M (2005) Copper and zinc dismetabolism in the mouse brain uponchronic cuprizone treatment. Cell Mol Life Sci 62:1502–1513.

Zhiling Y, Fujita E, Tanabe Y, Yamagata T, Momoi T, and Momoi MY (2008) Mu-tations in the gene encoding CADM1 are associated with autism spectrum disor-der. Biochem Biophys Res Commun 377:926–929.

Address correspondence to: Dr. Yutaka Nakagawa, Innovative ResearchDivision, Mitsubishi Tanabe Pharma, Yokohama 227-0033, Japan. E-mail:[email protected]

Neuroinflammation in Social Cognitive Deficits 515

at ASPE

T Journals on A

pril 1, 2020jpet.aspetjournals.org

Dow

nloaded from