15
Cytokine dysregulation in autism spectrum disorders (ASD): Possible role of the environment Paula E. Goines a , Paul Ashwood b, a University of California, Davis, School of Veterinary Medicine, Department of Molecular Biosciences, Sacramento, CA, USA b University of California, Davis, School of Medicine, Department of Medical Microbiology and Immunology, Sacramento, CA, USA abstract article info Article history: Received 4 April 2012 Received in revised form 3 July 2012 Accepted 31 July 2012 Available online xxxx Keywords: Autism spectrum disorder Cytokine Toxicant Immunology Environment Neurodevelopment Autism spectrum disorders (ASD) are neurodevelopmental diseases that affect an alarming number of indi- viduals. The etiological basis of ASD is unclear, and evidence suggests it involves both genetic and environ- mental factors. There are many reports of cytokine imbalances in ASD. These imbalances could have a pathogenic role, or they may be markers of underlying genetic and environmental inuences. Cytokines act primarily as mediators of immunological activity but they also have signicant interactions with the nervous system. They participate in normal neural development and function, and inappropriate activity can have a variety of neurological implications. It is therefore possible that cytokine dysregulation contributes directly to neural dysfunction in ASD. Further, cytokine proles change dramatically in the face of infection, disease, and toxic exposures. Imbalances in cytokines may represent an immune response to environmental contrib- utors to ASD. The following review is presented in two main parts. First, we discuss select cytokines implicat- ed in ASD, including IL-1Β, IL-6, IL-4, IFN-γ, and TGF-Β, and focus on their role in the nervous system. Second, we explore several neurotoxic environmental factors that may be involved in the disorders, and focus on their immunological impacts. This review represents an emerging model that recognizes the importance of both genetic and environmental factors in ASD etiology. We propose that the immune system provides crit- ical clues regarding the nature of the gene by environment interactions that underlie ASD pathophysiology. © 2012 Elsevier Inc. All rights reserved. 1. Introduction Autism spectrum disorders (ASD) are clinically variable neurodevel- opmental disorders that arise in early childhood. ASD is diagnosed in 1 out of every 88 children in the United States, and is characterized by stereotypic behaviors and impaired language and communication (American Psychiatric Association, 1994; Lord et al., 1994; Prevention, 2009; CDC, 2012). The biological basis of ASD remains largely elusive. Postmortem investigations have demonstrated abnormal brain growth, including neuronal overgrowth in some regions (Courchesne et al., 2011), and undergrowth in others (Schumann and Amaral, 2006). Al- tered ratios of excitatory to inhibitory signaling in the brain may also contribute to ASD (Fatemi et al., 2002b; Rubenstein and Merzenich, 2003; Fatemi et al., 2010; Yizhar et al., 2011). Inheritance analyses have demonstrated a large genetic component, although a recent twin study suggests that non-genetic environmental factors also make a sub- stantial contribution (Hallmayer et al., 2011; Ronald and Hoekstra, 2011). A multitude of genes have been implicated in autism, but only a few cases can be traced to specic rare genetic variants, and many of the implicated genes are also found in typically developing populations (State and Levitt, 2011). Collectively, these ndings suggest that autism results from complex interactions between multiple susceptibility genes and environmental factors. A growing body of evidence implicates immunological disturbances in ASD. Several of the genes linked to ASD have critical roles in immune signaling, activation, and regulation (Torres et al., 2002; Campbell et al., 2006; Lee et al., 2006; Torres et al., 2006; Varga et al., 2009; Orlova and Crino, 2010). Individuals with autism and their family members (pri- marily mothers) demonstrate increased autoimmunity, altered cellular immunity, and skewed expression of soluble mediators such as cyto- kines (Vargas et al., 2005; Braunschweig et al., 2008; Li et al., 2009; Morgan et al., 2010; Goines et al., 2011a; Onore et al., 2011). Cytokine abnormalities in ASD are an important clue for researchers as they might result from genetic and environmental factors, and may contrib- ute directly to neurological dysfunction. The following considers the neurological signicance of cytokine dysregulation in ASD, and how en- vironmental factors can modulate the cytokine response. 1.1. Cytokines: the common language between the immune and nervous system The immune system and the nervous system interact extensively. It is therefore not surprising that immune dysfunction is often noted in neurological disorders. Immune mediators known as cytokines are Neurotoxicology and Teratology xxx (2012) xxxxxx Corresponding author at: University of California, Davis, UC Davis M.I.N.D. Institute, 2805 Wet Lab Building, Sacramento, CA 95817, USA. Tel.: +1 916 703 0405. E-mail address: [email protected] (P. Ashwood). NTT-06335; No of Pages 15 0892-0362/$ see front matter © 2012 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ntt.2012.07.006 Contents lists available at SciVerse ScienceDirect Neurotoxicology and Teratology journal homepage: www.elsevier.com/locate/neutera Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulation in autism spectrum disorders (ASD): Possible role of the environ- ment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012.07.006

Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

Embed Size (px)

Citation preview

Page 1: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

Neurotoxicology and Teratology xxx (2012) xxx–xxx

NTT-06335; No of Pages 15

Contents lists available at SciVerse ScienceDirect

Neurotoxicology and Teratology

j ourna l homepage: www.e lsev ie r .com/ locate /neutera

Cytokine dysregulation in autism spectrum disorders (ASD): Possible role ofthe environment

Paula E. Goines a, Paul Ashwood b,⁎a University of California, Davis, School of Veterinary Medicine, Department of Molecular Biosciences, Sacramento, CA, USAb University of California, Davis, School of Medicine, Department of Medical Microbiology and Immunology, Sacramento, CA, USA

⁎ Corresponding author at: University of California, Da2805 Wet Lab Building, Sacramento, CA 95817, USA. Tel.:

E-mail address: [email protected] (P. Ashwoo

0892-0362/$ – see front matter © 2012 Elsevier Inc. Allhttp://dx.doi.org/10.1016/j.ntt.2012.07.006

Please cite this article as: Goines PE, Ashwoment, Neurotoxicol Teratol (2012), http://d

a b s t r a c t

a r t i c l e i n f o

Article history:Received 4 April 2012Received in revised form 3 July 2012Accepted 31 July 2012Available online xxxx

Keywords:Autism spectrum disorderCytokineToxicantImmunologyEnvironmentNeurodevelopment

Autism spectrum disorders (ASD) are neurodevelopmental diseases that affect an alarming number of indi-viduals. The etiological basis of ASD is unclear, and evidence suggests it involves both genetic and environ-mental factors. There are many reports of cytokine imbalances in ASD. These imbalances could have apathogenic role, or they may be markers of underlying genetic and environmental influences. Cytokines actprimarily as mediators of immunological activity but they also have significant interactions with the nervoussystem. They participate in normal neural development and function, and inappropriate activity can have avariety of neurological implications. It is therefore possible that cytokine dysregulation contributes directlyto neural dysfunction in ASD. Further, cytokine profiles change dramatically in the face of infection, disease,and toxic exposures. Imbalances in cytokines may represent an immune response to environmental contrib-utors to ASD. The following review is presented in two main parts. First, we discuss select cytokines implicat-ed in ASD, including IL-1Β, IL-6, IL-4, IFN-γ, and TGF-Β, and focus on their role in the nervous system. Second,we explore several neurotoxic environmental factors that may be involved in the disorders, and focus ontheir immunological impacts. This review represents an emerging model that recognizes the importance ofboth genetic and environmental factors in ASD etiology. We propose that the immune system provides crit-ical clues regarding the nature of the gene by environment interactions that underlie ASD pathophysiology.

© 2012 Elsevier Inc. All rights reserved.

1. Introduction

Autism spectrum disorders (ASD) are clinically variable neurodevel-opmental disorders that arise in early childhood. ASD is diagnosed in 1out of every 88 children in the United States, and is characterized bystereotypic behaviors and impaired language and communication(American Psychiatric Association, 1994; Lord et al., 1994; Prevention,2009; CDC, 2012). The biological basis of ASD remains largely elusive.Postmortem investigations have demonstrated abnormal brain growth,including neuronal overgrowth in some regions (Courchesne et al.,2011), and undergrowth in others (Schumann and Amaral, 2006). Al-tered ratios of excitatory to inhibitory signaling in the brain may alsocontribute to ASD (Fatemi et al., 2002b; Rubenstein and Merzenich,2003; Fatemi et al., 2010; Yizhar et al., 2011). Inheritance analyseshave demonstrated a large genetic component, although a recent twinstudy suggests that non-genetic environmental factors alsomake a sub-stantial contribution (Hallmayer et al., 2011; Ronald and Hoekstra,2011). A multitude of genes have been implicated in autism, but onlya few cases can be traced to specific rare genetic variants, and many ofthe implicated genes are also found in typically developing populations

vis, UC Davis M.I.N.D. Institute,+1 916 703 0405.d).

rights reserved.

od P, Cytokine dysregulationx.doi.org/10.1016/j.ntt.2012

(State and Levitt, 2011). Collectively, these findings suggest that autismresults from complex interactions between multiple susceptibilitygenes and environmental factors.

A growing body of evidence implicates immunological disturbancesin ASD. Several of the genes linked to ASD have critical roles in immunesignaling, activation, and regulation (Torres et al., 2002; Campbell et al.,2006; Lee et al., 2006; Torres et al., 2006; Varga et al., 2009; Orlova andCrino, 2010). Individuals with autism and their family members (pri-marily mothers) demonstrate increased autoimmunity, altered cellularimmunity, and skewed expression of soluble mediators such as cyto-kines (Vargas et al., 2005; Braunschweig et al., 2008; Li et al., 2009;Morgan et al., 2010; Goines et al., 2011a; Onore et al., 2011). Cytokineabnormalities in ASD are an important clue for researchers as theymight result from genetic and environmental factors, and may contrib-ute directly to neurological dysfunction. The following considers theneurological significance of cytokine dysregulation in ASD, and how en-vironmental factors can modulate the cytokine response.

1.1. Cytokines: the common language between the immune and nervoussystem

The immune system and the nervous system interact extensively. Itis therefore not surprising that immune dysfunction is often noted inneurological disorders. Immune mediators known as cytokines are

in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 2: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

2 PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

key facilitators of cross-systemic communication. Cytokines are pro-teins that control the nature, duration, and intensity of an immune re-sponse. They are highly pleiotropic, and can act in an autocrine,paracrine, and/or endocrine fashions. Immune cells, including dendriticcells, macrophages, neutrophils, T cells, and B cells, are the primarysource of cytokines; though many additional cell types, including neu-rons, produce and respond to them. Cytokines share structural similar-ities and signaling pathways with neurotrophins and neurologicallyrelevant growth factors. In many ways, cytokines represent a commonlanguage between the immune system and the nervous system.

Cytokines influence both the development and function of the ner-vous system. Their significance varies based on the timing, duration,and intensity of the neuro-immune interaction. For example, cytokinesimpact the developing brain differently than the adult brain; and maybe beneficial at one concentration while harmful at another. Cytokinesare involved in normal aspects of neurodevelopment, including pro-genitor cell differentiation, cellular localization/migration within thenervous system, and synaptic network formation (Deverman andPatterson, 2009). During infection and illness, cytokinesmediate neuro-logical changes associatedwith fever and sickness behavior by signalingdirectly to the hypothalamus (Dantzer, 2001; Skurlova et al., 2006).Emerging evidence also implicates cytokines in higher order neurolog-ical functions, including cognition and memory (McAfoose and Baune,2009; Derecki et al., 2010). Imbalanced cytokine production, signaling,and/or regulation can therefore have a wide range of neurologicalconsequences.

2. Cytokines in ASD

Aberrant expression of cytokines and their signaling intermediariesis often noted in ASD (Table 1). This is observed in the brain (Vargaset al., 2005; Grigorenko et al., 2008; Voineagu et al., 2011; Ziats and

Table 1Cytokines in autism spectrum disorders. A variety of independent clinical studies have linkOften multiple cytokines were associated with ASD in a single study, which is noted in par

Cytokine Findings in autism

IL-1Β Elevated plasma levels in children with ASD, correlated with regressive onset.

Elevated plasma levels in high functioning children with ASD. (IL-1RA, IL-5, ILElevated plasma levels in adults with severe ASD. (IL-6 and endotoxin levels aPeripheral blood cells from ASD subjects produce higher baseline levels. (SimiPeripheral blood cells from ASD subjects produce higher levels with TLR2 or T(Similar trends for IL-6 and TNFα)

IL-6 Elevated plasma levels in children with ASD, correlated with regressive onset.

Elevated plasma levels in adults with severe autism. (IL-Β and endotoxin levelPeripheral blood cells from ASD subjects produce higher baseline levels. (SimiPeripheral blood cells from children with ASD produce higher levels with TLR2lation. (Similar trends for IL-6 and TNFαLymphoblasts from ASD subjects produce more IL-6. (Also TNF-α)Increased IL-6 staining in postmortem cerebellar sections from ASD subjectsIncreased IL-6 in postmortem brain specimens (various regions) from ASD subchemokines).Increased IL-6 in postmortem brain tissue from ASD subjects. (Also increased

IL-4 Increased IL-4 in mid-gestational serum samples from mothers giving birth toIncreased IL-4 in amniotic fluid samples from mothers giving birth to a child w(Also IL-10, TNF-α and TNF-Β)Peripheral blood cells from ASD subjects stimulated with PMA-ionomycin wer

IFN-γ Increased IFN-γ in mid-gestational serum samples from mothers giving birthIncreased plasma levels in individuals with ASD. (Also IL-12)Peripheral blood cells stimulated with PMA-ionomycin are less likely to be IFN(And more likely to be IL-4+)Unstimulated whole blood from ASD subjects produced significantly more IFNand TNF-α)NK cells from children with ASD produced higher IFN-γ under resting conditioperforin and granzyme B)Increased IFN-γ in post mortem brain specimens from ASD subjects. (Also inc

TGF-Β Decreased plasma TGF-Β in children with ASD. Lower levels correlated with mDecreased serum TGF-Β in adults with ASD.Increased TGF-Β levels in postmortem brain specimens (various regions) from

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulatioment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

Rennert, 2011) peripheral blood (Molloy et al., 2006; Ashwood et al.,2011a, 2011b) and the gastrointestinal tract (DeFelice et al., 2003;Ashwood et al., 2004). Cytokine imbalances during development and/or throughout life could impact neural activity and mediate behavioralaspects of the disorder. The following considers the significance of sev-eral cytokines linked to ASD.

2.1. Interleukin (IL)-1B

IL-1Β is an inflammatory cytokine expressed very early in immuneresponses (Jiang et al., 1997). In tissue, IL-1Β propagates inflamma-tion by activating local immune cells and the vascular endothelium.Systemically, IL-1Β stimulates IL-6 production and eventually anacute phase response in the liver. Systemic IL-1Β can cross the bloodbrain barrier (Banks et al., 1991) and stimulate its own expressionin the hypothalamus, which leads to neuroendocrine changes associ-ated with fever and sickness behavior (Dantzer, 2001; Skurlova et al.,2006). IL-1Β receptors are structurally related to toll-like receptors(TLRs), and signaling is achieved through NF-κB and MAP kinase(MAPK) signaling cascades (O'Neill, 2000). IL-1Β belongs to an evolu-tionarily conserved family of proteins that function beyond immunity(Barksby et al., 2007). It shares structural homology with fibroblastgrowth factors (Zhang et al., 1991), which are critical in embryonicneurodevelopment, and are implicated in autism and schizophrenia(Tabares-Seisdedos and Rubenstein, 2009; Stevens et al., 2010).

Genes for IL-1Β, its receptor, and its receptor-associated proteins areassociatedwith intellectual disability, schizophrenia, and autism (Katilaet al., 1999; Piton et al., 2008; Handley et al., 2010). Children and adultswith autism have increased plasma IL-1Β and skewed cellular IL-1Β re-sponses following stimulation (Ashwood et al., 2011a, 2011b; Suzuki etal., 2011). Compared to controls, monocytes from children with ASDproduce excessive IL-1Β following LPS exposure (Jyonouchi et al.,

ed cytokines to ASD. This table presents detailed findings for each individual cytokine.entheses.

Reference

(IL-6, IL-8 and IL-12p40 also elevated) (Ashwood et al., 2011a,2011b)

-8, IL-12p70, IL-13, IL-17 and GRO-α also elevated) (Suzuki et al., 2011)lso elevated) (Emanuele et al., 2010)lar trends for IL-6 and TNF-α) (Jyonouchi et al., 2001)LR4 stimulation, and lower levels with TLR-9 stimulation. (Enstrom et al., 2010)

(IL-1Β, IL-8, and IL-12p40 also elevated) (Ashwood et al., 2011a,2011b)

s also elevated) (Emanuele et al., 2010)lar trends for IL-1Β and TNF-α) (Jyonouchi et al., 2001)or TLR4 stimulation, and lower levels with TLR-9 stimu- (Enstrom et al., 2010)

(Malik et al., 2011)(Wei et al., 2011)

jects. (Also increased TGF-Β and inflammatory (Vargas et al., 2005)

TNF-α, IFN-γ, GM-CSF, and IL-8) (Li et al., 2009)a child with ASD. (Also IL-5 and IFN-γ) (Goines et al., 2011b)ith ASD (Abdallah et al., 2011)

e more likely to be IL-4+ (And less likely to be IFN-γ+) (Gupta et al., 1998)to a child with ASD. (Also IL-4 and IL-5) (Goines et al., 2011b)

(Singh, 1996)-γ+ (Gupta et al., 1998)

-γ compared to controls. (Also increased IL-1RA, IL-6, (Croonenberghs et al.,2002)

ns, and lower levels after stimulation. (Also observed with (Enstrom et al., 2009a)

reased TNF-α, IL-6, GM-CSF, and IL-8) (Li et al., 2009)ore severe behavioral scores. (Ashwood et al., 2008)

(Okada et al., 2007)ASD subjects. (Also IL-6 and inflammatory chemokines) (Vargas et al., 2005)

n in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 3: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

3PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

2001; Enstrom et al., 2010), and lower levels following exposure to TLR9 agonists (Enstrom et al., 2010). The IL-1 antagonist, IL-1ra, is also in-creased amongASD subjects (Suzuki et al., 2011). IL-1ra reduces inflam-mation by competing for the IL-1Β receptor, and increased levels mayrepresent an attempt to counteract inflammation in ASD. Postmortembrains from ASD subjects had normal IL-1Β levels (Li et al., 2009), butgiven that peripheral IL-1Β can enter the brain (Banks et al., 1991), in-creased systemic levels could directly impact neurological processes.

IL-1Β disruption can have a variety of neurological consequences rel-evant to ASD. The cytokine and its receptors are found throughout thenervous system during critical developmental periods (Giulian et al.,1988). IL-1Β induces neural progenitor cell proliferation in someCNS re-gions, while inhibiting it in others (de la Mano et al., 2007). This couldcontribute to the region-specific overgrowth and undergrowth ob-served in the ASD brain. Excitatory synapse formation is partially medi-ated by the IL-1 receptor and receptor-associated proteins (Yoshidaet al., 2011). Altering these proteins can tip the balance between excit-atory and inhibitory signaling, which might underlie neurological fea-tures of ASD (Rubenstein and Merzenich, 2003). Increased IL-1ra inASD suggests an attempt to counterbalance IL-1Β and may or may notbe beneficial. Following brain injury, IL-1ra upregulation serves aneuroprotective role by dampening excessive inflammation (Loddickand Rothwell, 1996). However, if administered during critical windowsof neurodevelopment, IL-1ra can negatively impact neurogenesis, brainmorphology, memory consolidation, and behavior (Spulber et al., 2008,2010, 2011). This shows that some level of IL-1B signaling is essentialduring development. In adulthood, IL-1Β is implicated in CNS disorderslike Alzheimer's disease and the advancement of amyloid-containingplaques (Griffin et al., 1995). While excessive IL-1B contributes to pa-thology in some cases, it may have a protective role in others. For exam-ple, IL-1Β limits neuronal damage following excitotoxic exposures(Strijbos and Rothwell, 1995), and mice lacking IL-1Β fail to undergoremyelination following experimental autoimmune encephalitis (EAE)induction (Mason et al., 2001). IL-1Β is involved in higher order brainprocesses and is induced in the hippocampus during learning processes,and is critical for maintenance of long-term potentiation (LTP) (Rosset al., 2003). Both over expression (Barrientos et al., 2009) and under ex-pression of IL-1 beta (Goshen et al., 2007; Labrousse et al., 2009) are as-sociated with impairments in memory and learning.

In summary, IL-1Β participates in neurological processes, and ap-pears to have a role in both CNS pathology and healing. Normal, ho-meostatic levels of IL-1Β and its antagonist IL-1ra are necessary forproper brain development and function. This “Goldilocks” state is typ-ical of many cytokines, where too much or too little is not desirable.Alterations in IL-1Β systems due to genetic mechanisms or environ-mental exposures may contribute to autism.

2.2. Interleukin (IL)-6

IL-6 is an inflammatory cytokine that shares functional propertieswith IL-1Β. Like IL-1Β, IL-6 is produced early in immune reactions, al-though it appears later and persists longer (Jiang et al., 1997). IL-6 isbest known for stimulating the acute phase response in the liver, gener-ating fever, and activating lymphocytes. Despite the functional similar-ities with IL-1Β, IL-6 differs drastically in terms of structure andsignaling properties. It is a member of the neuropoietic cytokine family,which includes leukemia inhibitory factor (LIF), ciliary neurotrophicfactor (CNTF), and IL-11. These cytokines signal through a gp130 recep-tor complex (Ward et al., 1994), and activate JAK-STAT (specificallySTAT 3) and MAPK signaling pathways (Heinrich et al., 2003). In addi-tion to their inflammatory properties, neuropoietic cytokines have anumber ofwell-described roles in the nervous system, and are intricate-ly involved in neurodevelopment and function (Bauer et al., 2007;McAfoose and Baune, 2009). IL-6 and its receptors are expressed atlow levels in the healthy brain (Gadient and Otten, 1994a, 1994b) andat higher levels in a variety of disease states (Huell et al., 1995; Hang

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulationment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

et al., 2004). Peripheral IL-6 can cross the blood brain barrier and influ-ence a variety of processes in the adult brain (Banks et al., 1994).

Prenatal cytokine imbalancesmay contribute to neurodevelopmentaldisorders like autism and schizophrenia through “fetal programming”.Fetal programming is the concept that maternal factors like inflamma-tion and chronic stress can alter the gestational environment, skewdevelopment, and lead to long termphysiological and behavioral conse-quences (Patterson, 2009; Bilbo, 2010). IL-6 readily crosses the placentaand enters fetal tissues, which is unique among cytokines, and can in-duce changes in placental physiology and gene expression (Zaretskyet al., 2004; Aaltonen et al., 2005; Dahlgren et al., 2006; Hsiao andPatterson, 2011). Animal models show that IL-6 is necessary and suffi-cient to alter neurodevelopmental outcomes, leading to changes in be-havior, cognition, neuropathology, GABA dysregulation, and skewedimmune function among offspring (Samuelsson et al., 2006; Smith etal., 2007). Similar effects are seen with prenatal exposure to infectionor the infectious mimic poly I:C (Smith et al., 2007; Malkova et al.,2012). IL-6 can impact a variety of processes in the developing brain.IL-6 and its familymembers regulate self-renewal among neuronal pre-cursors (Escary et al., 1993; Yoshimatsu et al., 2006), direct neuronalmigration (Wei et al., 2011), promote cell survival (Kushima et al.,1992), and regulate neurite outgrowth (Ihara et al., 1997). IL-6 expo-sure during critical windows can also alter synaptic networks. ChronicIL-6 overexpression reduces expression of glutamate receptors andL-type calcium channels in culture and in vivo (Vereyken et al., 2007),and increases the ratio of excitatory to inhibitory synapses in culturesof cerebellar granular cell cultures (Wei et al., 2011). This is of particularinterest in autism, given that skewed excitatory and inhibitory ratiosmay be an underlying factor in its pathogenesis.

Despite intriguing evidence from animal models, two recent humanstudies question whether gestational IL-6 alone contributes to autism. Aretrospective examination of IL-6 in archived mid-pregnancy maternalserum and amniotic fluid showed that increased levels associated withdevelopmental disorders, but not autism (Abdallah et al., 2011; Goineset al., 2011b). This suggests that gestational IL-6 might be a marker forneurodevelopmental diseases, but is insufficient on its own to causeASD. IL-6 can affect many sequential phases of neurodevelopment, sothe timing of the exposurewill largely dictate the neurological outcome.Therefore, excessive IL-6 during one phase of neurodevelopment couldhave one set of consequences, while similar expression during anotherphase has an entirely different effect. A longitudinal examination ofIL-6 throughout gestation is therefore needed to obtain amore completepicture of its relevance in neurodevelopmental disorders.

IL-6 can also impact processes in the adult brain, and physiologicallevels are critical for homeostasis, cognition, learning, and memory.Physiological levels of IL-6 are critical for normal CNS function, andboth over and under expression leads to neurological problems. Miceoverexpressing IL-6 in the CNS have overt symptoms including tremor,ataxia, and seizure (Campbell et al., 1993), and more subtle alterationsin cognition and avoidance behaviors (Heyser et al., 1997). IL-6 is tran-scribed in the hippocampus during LTP (Balschun et al., 2004).Overexpression of IL-6 reduces LTP (Bellinger et al., 1995; Li et al.,1997), while under expression increases it and improves learning andmemory (Balschun et al., 2004; Braida et al., 2004).With regard to socialbehaviors, mice overexpressing IL-6 are more social than mice that lackthe cytokine, while mice lacking IL-6 demonstrate higher aggressionand emotionality (Alleva et al., 1998; Armario et al., 1998).

Many independent studies show IL-6 dysregulation in individualswith autism. Children and adults with the disorder have higher circulat-ing IL-6 levels compared to typically developing controls (Emanuele etal., 2010; Ashwood et al., 2011a, 2011b). Further, cellular IL-6 produc-tion is increased with and without stimulation (Jyonouchi et al., 2001;Enstrom et al., 2010; Malik et al., 2011). Increased IL-6 is also found inpostmortem brain specimens from ASD subjects. Specifically, immuno-histochemical analysis of cerebellar sections showed significantly moreIL-6 staining in autism postmortem brain specimens (Wei et al., 2011).

in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 4: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

4 PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

Two additional analyses of homogenates of the frontal cortex and ante-rior cingulate gyrus also showed higher IL-6 levels (Vargas et al., 2005;Li et al., 2009). Given the ability of IL-6 to impact processes in the adultbrain, it is conceivable that increased IL-6 in autism could contribute toongoing aspects of the disorder. Alternatively, it might be an epiphe-nomenon, and represent a biomarker of infectious or toxic environmen-tal exposures and altered biological homeostasis.

In summary, there is extensive evidence that IL-6 can alterneurodevelopment and function. While it is unclear whether gesta-tional IL-6 in humans is related to autism, a dysregulation of IL-6 isobserved later in life in individuals with autism. The significance ofthese findings is unclear, and may be the result of other genetic andenvironmental factors in autism. These possibilities warrant furtherinvestigation.

2.3. Interleukin (IL)-4

IL-4 is a class I cytokine that activates Jak/Stat (STAT 6), MAPK, andPI3 kinase signaling cascades (Nelms et al., 1999). Immunologically,IL-4 has a variety of interesting roles, and can 1) induce “alternativelyactivated” macrophages that promote tissue repair over inflammation,2) activate basophils andmast cells, 3) promote B-cell isotype switchingtowards IgG1 and IgE, 4) participate in immune responses againsthelminthes by inducing epithelial cell turnover in the gut, and 5) partic-ipate in allergy and asthma-related immune responses (Kuperman andSchleimer, 2008; Byers and Holtzman, 2011; Oliphant et al., 2011).

The receptors for IL-4 are expressed in the brain under normal con-ditions throughout life (Nolan et al., 2005). During development, IL-4promotes oligodendrogenesis among neuronal progenitor cells,(Butovsky et al., 2006), and improves survival in embryonic hippocam-pal cultures (Araujo and Cotman, 1993). IL-4 influences retinal circuitryby regulating progenitor cell proliferation and differentiation (da Silvaet al., 2008). During later phases of neurodevelopment, IL-4 can altersynapse formation; increasing the proportion of GABAergic synapsesin cell culture models (Sholl-Franco et al., 2002).

Two recent studies have linked developmental IL-4 exposures to au-tism, though its role in pathogenicity versus protection is unclear.Mothers giving birth to a child with autism show higher levels of IL-4in mid-pregnancy serum samples (Goines et al., 2011b) and amnioticfluid (Abdallah et al., 2011) compared to controls. IL-4 is not thoughtto cross the placenta, and maternal serum and amniotic fluid IL-4 mayor may not relate to IL-4 in fetal tissues. Other cytokines were alsoupregulated in these archived samples, including IFN-γ, TNF-α, andthe anti-inflammatory cytokine IL-10. This raises thequestion ofwheth-er IL-4 acts alone, or in concert with other cytokines. Increased IL-4mayrepresent a regulatory reflex to inflammation along with IL-10. IL-4'srole in pregnancy and fetal health is unclear. Increased levels duringpregnancy have been associated with poor outcomes such as pretermlabor (Dudley et al., 1996) but also healthy outcomes such as protectionfrom preeclampsia (Kronborg et al., 2011; Rajakumar et al., 2011).More subtle neurodevelopmental outcomes have not thoroughly beenexplored with respect to gestational IL-4.

In the adult brain, IL-4 largely serves a neuroprotective role, and isassociated with higher order cognitive processes. It is upregulatedduring CNS inflammation, inducing alternative activation of glialcells and protecting from apoptosis (Garg et al., 2009; Sholl-Francoet al., 2009; You et al., 2011). In a mouse model for Alzheimer's dis-ease, IL-4 can attenuate disease progression (Kiyota et al., 2010). Fol-lowing LPS exposure, IL-4 reduces inflammation and improvesmemory and LTP in the aged hippocampus (Nolan et al., 2005). An el-egant study by Derecki et al. showed that IL-4-producing T cells accu-mulate in the meningeal spaces during cognitive tasks. Depletion ofIL-4 led to an inflammatory phenotype among meningeal myeloidcells, and a dramatic decline in cognitive capacity. Remarkably, cogni-tive deficits in IL-4 deficient mice could be reversed by reintroducingthe cytokine in adulthood (Derecki et al., 2010).

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulatioment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

Among individuals diagnosed with autism, plasma and CNS IL-4levels appear to be normal (Vargas et al., 2005; Li et al., 2009;Ashwood et al., 2011a, 2011b). However, IL-4 producing T cells areproportionately higher in children with autism compared to controls(Gupta et al., 1998). Given the evidence that meningeal IL-4 produc-ing T cells are critical for normal cognitive function in adulthood, itis possible that dysregulation in this cell population could contributeto altered behavior throughout life (Derecki et al., 2010).

Collectively, IL-4 serves a variety of neurological roles, and is in-creased in autism. Its role during gestation is unclear due to a dearthof in-vivo studies of pregnancy and neurobehavioral outcomes fol-lowing developmental IL-4 exposures. The significance of increasedIL-4 producing T cells in subjects with autism is also unclear. Exten-sive evidence suggests that IL-4 is neurologically beneficial, so itmay be that increased IL-4 in autism represents an immunological at-tempt to regulate other detrimental processes, and does not contrib-ute to the disease itself. Future studies should explore this possibility.

2.4. Interferon-gamma (IFN-γ)

Interferon gamma (IFN-γ) is the sole type II interferon. It sharessome functional similarities with type I interferons like IFN-α andIFN-Β but has unique structural features, receptors, and signaling path-ways. IFN-γ is producedprimarily by T cells andNatural Killer (NK) cellsduring cell-mediated immune responses, and functions largely to acti-vate macrophages and combat viral infections (Boehm et al., 1997;Schroder et al., 2004). It signals mainly through the JAK/STAT (STAT1),and MAPK cascades (Hu et al., 2001; Platanias, 2005). IFN-γ and IL-4counterbalance one another's activity via TH1/TH2 interactions, sodysregulation in one cytokine often impacts the other. It is thereforenot surprising that both cytokines are implicated in ASD.

Developmental exposure to IFN-γ has been linked to autism.Mothers of children with autism have higher serum IFN-γ duringthe second trimester compared to controls (Goines et al., 2011b).Like IL-4, IFN-γ does not cross the placenta, and the relationship be-tween maternal serum levels and fetal exposure to the cytokine isunclear. If the cytokine is present in fetal tissues, it could interferewith normal neural development and synapse formation. IFN-γ pro-motes neuronal differentiation among neural progenitor cells(Barish et al., 1991; Jonakait et al., 1994; Wong et al., 2004;Butovsky et al., 2006; Zahir et al., 2009; Leipzig et al., 2010; Li etal., 2010), however, these cells appear to be abnormal and exhibitcompromised function and strange patterns of neuronal marker ex-pression (Walter et al., 2011). IFN-γ also impacts dendritic morphol-ogy and synapse formation, leading to long-term changes in cellularconnectivity and communication. Depending on cell culture condi-tions, IFN-γ either promotes or inhibits dendrite outgrowth throughSTAT 1 and MAPK signaling pathways (Barish et al., 1991; Kim et al.,2002a; Wong et al., 2004; Song et al., 2005; Andres et al., 2008). Inculture, excessive IFN-γ alters patterns of excitatory signaling andreceptor expression (Vikman et al., 2001), and animals lacking thecytokine have fewer pre-synaptic terminals (Victorio et al., 2010).Interestingly, mice overexpressing IFN-γ show increased MHC I inthe brain (Corbin et al., 1996). MHC I is critical for T cell and NK cellrecognition of self and foreign entities, and was historically thought tobe absent in the CNS. However, recent studies have demonstrated thatit is expressed in the CNS, and has an essential role in synapse formationand plasticity (Shatz, 2009). IFN-γ may therefore induce abnormalitiesin synaptic organization by altering MHC I expression. Collectively,these studies show that direct exposure to IFN-γ can cause abnormalneurodevelopment, which may explain features of autism.

If excess IFN-γ is not present in fetal tissues, excessive maternallevels could have an indirect impact on fetal development. Interest-ingly, IFN-γ has a variety of critical roles in pregnancy, and directs as-pects of placental development, health, and maintenance (Murphyet al., 2009). Increased gestational IFN-γ is associated with adverse

n in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 5: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

5PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

pregnancy outcomes including recurrent miscarriage (Jenkins et al.,2000). Therefore, IFN-γ might be an indicator of compromised healthin pregnancy, which could lead to neurodevelopmental abnormalities.

Peripheral IFN-γ is up-regulated in a number of neurological disor-ders including multiple sclerosis (Martins et al., 2011) and Down's syn-drome (Torre et al., 1995). Individuals with autism have increasedplasma levels of IFN-γ (Singh, 1996), which correlates with other pe-ripheral inflammatory mediators such as nitric oxide (Sweeten et al.,2004a, 2004b). Peripheral immune cells from ASD subjects producehigher basal levels of IFN-γ but fail to respond further following immu-nological stimulation (Gupta et al., 1998; Croonenberghs et al., 2002;Enstrom et al., 2009a). In addition to peripheral IFN-γ dysregulation,postmortem brain specimens showed increased levels of IFN-γ (Liet al., 2009), suggesting IFN-γmay directly impact CNS processes in au-tism. In the developed nervous system, IFN-γ is historically associatedwith neurodegeneration, although some evidence suggests it mayhave a beneficial role. IFN-γ can cross the blood brain barrier at lowlevels (Pan et al., 1997), but is barely detectable in the healthy nervoussystem (Traugott and Lebon, 1988; De Simone et al., 1998). In the CNS,IFN-γ is up-regulated following infectious exposures (De Simone et al.,1998), and in diseases including cerebral palsy (Folkerth et al., 2004),multiple sclerosis (Traugott and Lebon, 1988), HIV dementia (Noltinget al., 2009), and Parkinson's (Barcia et al., 2011; Mangano et al.,2011). High levels are harmful in cell culture, and cause enhanced glu-tamate induced neurotoxicity (Mizuno et al., 2008). However, in cellculture and in vivo, low IFN-γ levels reduce oxidative-stress induced ap-optosis through activation of astrocytes (Garg et al., 2009; Victorio et al.,2010) and may be neuroprotective in some cases. In a mouse model ofAlzheimer's disease, overexpressing IFN-γ actually attenuated plaqueformation (Chakrabarty et al., 2010), while protective roles for IFN-γhave been suggested during some phases of demyelinating autoim-mune disorders (Kumar and Sercarz, 1998). It is therefore difficult todetermine whether IFN-γ has a pathogenic role in ASD, or if it repre-sents a potentially beneficial immune response to damage associatedwith genetic and/or environmental influences.

2.5. Transforming growth factor-beta (TGF-Β)

TGF-Β is a highly pleiotropic cytokine thatmaintains immune homeo-stasis, directs lymphocyte differentiation, and orchestrates aspects of em-bryonic development. TGF-Β is largely immunosuppressive; limitingexcessive T cell activity and inflammation (Mantel and Schmidt-Weber,2011). It exists in three isoforms, each with distinct and overlappingroles. TGF-Β1 is best characterized, and is the founding member of theTGF-Β superfamily of proteins, which includes growth differentiation fac-tors, bonemorphogenic proteins (BMPs), activins, and inhibins (Kingsley,1994). TGF-Β superfamily signaling occurs largely via SMAD pathways,though MAPK cascades are also triggered (Yu et al., 2002; Shi andMassague, 2003).

TGF-Β superfamily proteins are critical for proper neurodevelopment.For example, BMPs have an important role in early neural induction anddifferentiation (Reissmann et al., 1996; Bachiller et al., 2000; Tropepeet al., 2001). TGF-Β1 is involved in neuronal migration, survival, andsynapse formation. Mice lacking the cytokine demonstrate improperCNS development, including a disorganized extracellular matrix, wide-spread neuronal degeneration, microgliosis, reduced expression ofsynaptophysin, and deficits in both glutamatergic and GABAergic synap-ses (Brionne et al., 2003; Heupel et al., 2008; Vashlishan et al., 2008).Overexpressing TGF-Β in vivo also disrupts the extracellular matrix, andleads to seizures, motor incoordination, hydrocephalus, and behavioralabnormalities (Wyss-Coray et al., 1995; Depino et al., 2011). Changes inCNS expression levels of IL-6, Neuroligin 3 and reelin are also observedwith TGF-Β overexpression (Depino et al., 2011), which is intriguingbecause many of these proteins are altered in autism (Persico et al.,2001; Laumonnier et al., 2004; Fatemi et al., 2005). Interestingly, CNSoverexpression of TGF-Β during development vs. adulthood leads to

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulationment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

opposite behavioral consequences (Depino et al., 2011). Early in life,overexpression led to decreased social behavior and heightened anxi-ety/depression behaviors, while overexpression later in life had theexact opposite effect. This highlights the importance of timingwhen con-sidering the neurological consequences of cytokine imbalances.

There is no evidence for TGF-Β dysregulation during gestational de-velopment in autism. This does not negate the possibility that it is in-volved, as these endpoints are extremely difficult to measure in vivo.However, there is evidence for TGF-Β dysregulation in individuals diag-nosed with the disorder themselves. Plasma TGF-Β is decreased in chil-dren and adults with ASD (Okada et al., 2007; Ashwood et al., 2008),and lower levels of the cytokine correlate with more severe autism be-haviors (Ashwood et al., 2008). The connection between low TGF-Βand behavioral phenotype is unclear, although thisfinding lends promiseto the goal of developing a simple ASD testing regime based on biologicalmarkers in addition to behavioral symptoms. In contrast to peripheralTGF-Β in ASD, postmortem brain specimens show increased levels com-pared to controls (Vargas et al., 2005). The reason for this periphery/brain disparity is unclear. Although, TGF-Β does not cross the bloodbrain barrier (Kastin et al., 2003), the cytokine and its receptors areexpressed normally throughout the nervous system (Gomes et al.,2005) and it is conceivable that peripheral and brain levels are indepen-dent. In the adult brain, TGF-Β is generally thought to be neuroprotective.CNS levels spike following injury and infection, and increase with age,HIV dementia, andAlzheimer's disease,which leads to protection againstdisease-related neural degeneration and apoptosis (Henrich-Noack et al.,1994; Krupinski et al., 1996; Ruocco et al., 1999; Buckwalter andWyss-Coray, 2004; Doyle et al., 2010). However, TGF-Β can also contrib-ute to pathogenicity. For example, high levels of the cytokine cause glialscarring and fibrosis (Moon and Fawcett, 2001), and increase diseasesusceptibility and severity in a murine model of multiple sclerosis(Wyss-Coray et al., 1997).

Overall, the role of TGF-Β in the nervous system is complex, andvaries based on the timing and context of the interaction. IncreasedTGF-Β in the brain of ASD subjects might represent a protective reflexto a disease state, or perhaps contribute to the pathology of the diseaseitself. In the periphery, ASD subjects have decreased TGF-Β aswell as in-creased inflammatory markers (Jyonouchi et al., 2001; Croonenberghset al., 2002; Emanuele et al., 2010; Ashwood et al., 2011a, 2011b). Thissuggests a global immune dysregulation in ASD, with an improper bal-ance between regulation and activation, which could have widereaching consequences for many systems in the body.

3. Environmental factors in autism and immune dysfunction

There is now general consensus that autism has an environmentalcomponent (Pessah, 2008; Hallmayer et al., 2011). For our purposes,the “environment” is a broad term used to define non-genetic, toxic,infectious, and/or immune factors that may contribute to the disor-der. The hypothesis follows that an ill-timed exposure could cause au-tism in genetically susceptible individuals (Fig. 1). For example, somegenes associated with autism may cause inappropriate immune re-sponses (Heuer et al., 2011; Onore et al., 2011), while others reducethe capacity to deal appropriately with toxins (D'Amelio et al.,2005). These individuals are more susceptible to environmental influ-ences, which could lead to ASD.

Environmental toxicants can cause both neural and immune dys-function. This is likely mediated through disrupted cell signaling andhomeostasis. Many toxicants alter calcium homeostasis, which canhave a variety of consequences for immune development and function(Limke et al., 2003; Toscano and Guilarte, 2005; Savignac et al., 2007;Vig and Kinet, 2009; Pessah et al., 2010; Bhatti et al., 2011). Toxicantscan also disrupt endocrine function, which can have a variety of immu-nological consequences (Rivest, 2010; De Vito et al., 2011; Schug et al.,2011). The following section considers environmental factors that

in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 6: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

6 PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

may be related to autism, and focuses on their role in the immunesystem.

3.1. Heavy metals

Heavy metals like lead and mercury are widespread environmentaltoxins. Developmental exposure to these compounds is associated withlower IQ, endocrine disruptions, and behavioral disturbances (Bellingerand Dietrich, 1994; Steuerwald et al., 2000; Cordier et al., 2002;Selevan et al., 2003; Winneke, 2011). Heavy metals also haveimmunotoxic properties, leading in some instances to autoantibody pro-duction (Waterman et al., 1994; Bagenstose et al., 1999; Rowley andMonestier, 2005) and skewed cytokine profiles (discussed below). Bothof these immunological phenomena are observed in ASD (Enstromet al., 2009b; Onore et al., 2011). These features have made them possi-ble, though controversial, candidates in autism.

3.2. Heavy metals: lead

Lead has a variety of toxicmechanisms, and shares structural featureswith calcium which allows it to compete for binding sites (Toscano andGuilarte, 2005). In addition to its neurotoxic activity, lead is highlyimmunotoxic (Toscano andGuilarte, 2005;Mishra, 2009). At high levels,it is immunosuppressive, leading to increased production of regulatorycytokines and enhanced susceptibility to infection (Valentino et al.,2007). At lower levels, lead appears to be immunostimulatory (Floheet al., 2002). One study showed that lead can tip the balance between in-flammation and regulation by increasing expression of IFN-γ and reduc-ing TGF-Β (Goebel et al., 2000): a cytokine profile which has beenobserved in autism (Singh, 1996; Gupta et al., 1998; Croonenberghset al., 2002; Ashwood et al., 2008; Enstrom et al., 2009a; Li et al., 2009).Lead and pro-inflammatory cytokines can function in concert to alterthe nervous system. When co-administered to glial cells, lead and cyto-kines such as IL-1Β changed matrix metalloproteinase expression in amanner that was not observed when either was administered alone(Lahat et al., 2002) and suggests that immune and environmental factorscould act synergistically on tissue remodeling.

There are conflicting reports regarding lead in autism. Higher serumlead levels have been documented in a few studies (Cohen et al., 1976;Shannon and Graef, 1996; Filipek et al., 1999), although more recentstudies show no difference between autism and control populations(Tian et al., 2011; Albizzati et al., 2012). Polymorphisms in ALAD, agene associated with heavy metal toxicity, have been described inASD (Rose et al., 2008) leading, some authors to speculate that childrenwith ASD have higher lead levels due to a decreased capacity to elimi-nate lead from the body (Kern et al., 2007). Others suggest increasedfrequency of pica (hand-to-mouth behaviors) in ASDmay also alter ex-posures to lead (Cohen et al., 1976; Shannon and Graef, 1996). Despite

Fig. 1. Multifactorial model for ASD etiology involving genes, the environment, and immunspond inappropriately to environmental influences, like an infection or toxic exposure. An evous and immune system. This disruption could lead to ASD, as well as the neurological, be

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulatioment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

lead's well-established neurotoxic and immunotoxic mechanisms, it iscurrently debatable whether it contributes to autism.

3.3. Heavy metals: mercury

Mercury exerts its toxicity broadly throughout the body. It binds awide range of molecular groups including thiols, hydroxyls, and car-boxyls (Bridges and Zalups, 2010), and can dramatically increase intra-cellular calcium levels (Sakamoto et al., 1996; Limke et al., 2003).In the immune system, mercury's effects depend heavily on geneticbackground. For example, certain strains of mice reliably developanti-nuclear antibodies after mercury exposure, while others do not(Bagenstose et al., 1999; Rowley andMonestier, 2005). Mercury also im-pacts cytokine profiles; and some strains of mice strongly up-regulatethe TH2 cytokine IL-4 in response to mercury, while other strainsup-regulate TH1 cytokines like IFN-γ (Wu et al., 2001; Hemdan et al.,2007). In two humanmast cell lines,mercury exposurewas shown to in-crease IL-6 production (Kempuraj et al., 2010). Further, mercury inter-feres with cytokine-related signaling cascades including NF-κB and p38MAPK (Dieguez-Acuna et al., 2001; Kim et al., 2002b). Altered cytokinerepertoires in response to mercury exposure may contribute to the de-velopment of autoimmunity. Interestingly, IL-4 deficient mice produceautoantibodies in response to mercury exposure (Bagenstose et al.,1998; Kono et al., 1998), while IFN-γ deficient mice do not (Kono et al.,1998), suggesting that autoantibody production is IFN-γ dependent.

The evidence implicatingmercury in autism is somewhat contradic-tory. Several recent studies have shown no link between mercury bodyburdens and autism (Ip et al., 2004; Hertz-Picciotto et al., 2010;Albizzati et al., 2012; Wright et al., 2012). However, a reanalysis ofdata generated by Ip et al. showed there was in fact an association be-tweenmercury levels and autism that had been overlooked due to a sta-tistical error (Desoto andHitlan, 2007). Ethylmercury is a component inthe vaccine preservative thimerosal, which has received attention in re-cent years. It has neurotoxic capacities, and can alter calcium signalingand cytokine production (specifically IL-6) (Goth et al., 2006). Whilein vitro studies suggest toxic potential for thimerosal, a large numberof independent epidemiological studies show no link to autism(DeStefano, 2007; Miller and Reynolds, 2009; Price et al., 2010).

Two recent studies examined the correlation between gene tran-scription profiles (35,000+ genes) and heavy metal body burdens inchildren with autism and controls (Stamova et al., 2011; Tian et al.,2011). Mercury loads correlated with the expression of several immu-nologically relevant genes across all study participants. Further, therewere some unique correlations in the autism group for genes involvedin antigen presentation and recognition of self. This suggests that indi-viduals with ASD may have a unique immunological susceptibility toheavymetals, but the significance of these findings is not clear. Althoughgenomic expression profiles may suggest correlative changes in autism,

ity. This model begins with a genetic background that predisposes an individual to re-nvironmental exposure during a critical period could disrupt development of the ner-havioral, and immune dysfunctions observed in the disorder.

n in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 7: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

7PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

analysis of common polymorphisms associated with mercury transportand excretion, namelyMT1a, DMT1, LAT1, andMTF1,were unable to de-tect any differences in autism (Owens et al., 2011). However, in contrast,a different study showed that polymorphisms in MTF1 and the heavymetal transport gene (SLC11A3) are associated with the ASD (Serajeeet al., 2004). Additional genetic analyses are needed to rectify these dis-parate findings.

3.4. Pesticides

Pesticides are fairly non-persistent toxic compounds that are delib-erately spread throughout the environment in mass quantities. Whileminimizing off-target toxicity is a primary goal in pesticide develop-ment, several products have been banned once their toxic potential inhumans was recognized. Developmental exposure to several types ofpesticides, including organophosphates (OPs), organochlorines (OCs),and pyrethroids, is associated with neurological dysfunction and an in-creased risk for ASD (Garry et al., 2002; Kamel and Hoppin, 2004;Eskenazi et al., 2007; Roberts et al., 2007; Eskenazi et al., 2010;Bouchard et al., 2011). Genetic analyses also suggest that individualswith ASDmay be less capable of excreting pesticides, due to expressionof a less-active variant of the OP-metabolizing enzyme paroxonase(D'Amelio et al., 2005; Pasca et al., 2006). In addition to their neurotox-icity, many pesticides impact the immune system and cytokine produc-tion, which may be relevant for ASD (Banerjee et al., 1996; Li, 2007).

3.5. Pesticides: organochlorines

Organochlorine (OC) pesticides are structurally and functionally var-iable toxic compounds that include members like hexachlorobenzene,dicofol, and DDT; many of which have been banned (Crinnion, 2009).OCs interfereswith calcium signaling, voltage sensitive sodium channels,and GABA receptors, leading to neuro- and immunotoxicity (Casida,2009; Crinnion, 2009; Heusinkveld and Westerink, 2012). Immunologi-cally, these compounds impact both humoral and cell-mediated process-es, and reduce the host response to infectious challenges (Banerjee et al.,1996; Reed et al., 2004; Nagayama et al., 2007). A handful of studies haveexplored the impact of OCs on cytokine profiles. DDT reduced IL-2 pro-duction in cell culture by interfering with the transcription factorNF-κB (Ndebele et al., 2004). Given the central role of NF-κB in cytokineproduction and function, this is likely to have a wide-ranging immuno-logical impact. In contrast, cases of DDT or lindane poisoning in humansis associated with increased serum levels of IL-2, IL-4, and TNF-alpha, aswell as decreased levels of IFN-γ (Daniel et al., 2002; Seth et al., 2005). Itis not clear why IL-2 production in response to OCs differs in cell cultureversus in vivo. However, the findings of increased IL-4 and decreasedIFN-γ in humans suggest a TH2 immune bias following OC exposure,which could have downstream consequences for allergic and asthmaticdisorders. Similar immune profiles have been reported in some studiesof autism (Gupta et al., 1998).

3.6. Pesticides: organophosphates

Organophosphates (OP) are esters of phosphoric acid that were in-troduced as replacements for various banned OC pesticides. They actprimarily through acetylcholinesterase (AChE) inhibition, leading to al-tered cholinergic signaling, parasympathetic and sympathetic perturba-tions, seizures, and/or respiratory arrest. OPs can also be toxic in theabsence of AChE inhibition, and may induce higher order neural andcognitive dysfunction (Duysen et al., 2001; Costa, 2006; Pancetti et al.,2007). Some OP developmental effects are more severe in males thanfemales (Levin et al., 2001, 2010), which is intriguing given that autismalso has a heavy male bias (Baron-Cohen et al., 2005).

OPs induce a variety of immunological phenomena relevant to au-tism (Galloway andHandy, 2003; Li, 2007). In general, OPs appear to in-duce a prolonged inflammatory state that may evolve into an adaptive

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulationment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

response characterized by up-regulation of both TH1 and TH2 cytokines.Acute OP intoxication is related to system-wide production of inflam-matory mediators (Hamaguchi et al., 2006; Roeyen et al., 2008; Anandet al., 2009). Within the CNS, acute and chronic exposure to OPs resultsin increased inflammatory cytokines including IL-1Β and IL-6 in multi-ple brain regions (Svensson et al., 2001; Henderson et al., 2002;Williams et al., 2003; Dhote et al., 2007; Dillman et al., 2009; Johnsonand Kan, 2010); similar to findings in the autism brain (Vargas et al.,2005; Li et al., 2009). OP induced inflammation can be long term,re-emerging and persisting long after the initial exposure (Chapmanet al., 2006). In primary cultures of human peripheral blood cells or as-trocytes, the OP pesticide chlorpyrifos up-regulates IL-6 and IFN-γ pro-duction and the expression of related genes (Mense et al., 2006).Children born tomothers working in agriculture had higher productionof TH2 cells at 12 and 24months of age. (Duramad et al., 2006a, 2006b).An immunosuppressive response can also be induced following expo-sure to various OPs, perhaps as a reflex to the toxin's initial inflammato-ry properties (Williams et al., 2003;Damodaran et al., 2006; Dhote et al.,2007). Collectively, these data suggest that OPs impact cytokine profilesin the short and long term, increasing inflammatory cytokines, TH1 andTH2 profiles and compensatory regulatory activity.

3.7. Pesticides: pyrethroids

Pyrethroids are a group of insecticides and repellants derived fromnatural compounds in the Chrysanthemum genus of plants. Theymedi-ate their toxicity by disrupting calcium signaling, interfering with volt-age sensitive sodium channels, and inducing oxidative stress (Shaferet al., 2005; Soderlund, 2012). Exposure to these compounds is associat-ed with a wide range of neurodevelopmental problems in mammalianmodels (Shafer et al., 2005; Wolansky and Harrill, 2008). Immunologi-cal abnormalities are also linked to pyrethroids. In human peripheralblood mononuclear cells, exposure to several different pyrethroid com-pounds suppressed both IFN-γ and IL-4 expression in a time and con-centration dependent manner (Diel et al., 2003). In a monocytic cellline, various synthetic pyrethroids and their metabolites reduced ex-pression of immunoregulatory IL-10, and increased production ofmore inflammatory cytokines IL-12 and TNF-α (Zhang et al., 2010). InaXenopus laevismodel, application of environmentally relevant concen-trations of various pyrethroids increased IL-1Β expression (Martiniet al., 2010). In primary human fetal astrocytes, the pyrethroid pesticidecyfluthrin was found to have an activating effect, and increased the ex-pression of genes involved in IFN-γ and IL-6 production and signaling(Mense et al., 2006).

3.8. Halogenated aromatic hydrocarbons

Halogenated aromatic hydrocarbons are toxic compounds that arehighly resistant to degradation. Two examples are polychlorinated bi-phenyls (PCBs) and polybrominated diphenyl ethers (PBDEs). PCBsand PBDEs consist of two aromatic rings with various chlorine(PCBs) or bromine (PBDEs) substitutions. There are over two hundreddifferent congeners each of PCBs and PBDEs, which differ based onthe number and orientation of halogen substitutions. Toxicity in thisclass of molecules is congener-specific, and involves varying degreesof 1) interaction with the Aryl hydrocarbon receptor (Ahr) (Mitchelland Elferink, 2009; Gu et al., 2012), 2) disruption of endocrine sys-tems (Morse et al., 1993; Van Birgelen et al., 1995; Stoker et al.,2005; Kuriyama et al., 2007; Lema et al., 2008), and/or 3) interferencewith calcium homeostasis through interactions with the ryanodinereceptor (Coburn et al., 2008; Pessah et al., 2009, 2010; Kim et al.,2011; Langeveld et al., 2012). These toxic mechanisms have both neu-rological and immunological significance.

Children with ASDmay be uniquely susceptible to halogenated aro-matic hydrocarbons. Postmortem analysis showed altered ryanodinereceptor expression in the brain of autism subjects compared to

in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 8: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

8 PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

controls, which could alter their sensitivity to Ryr-reactive compounds(Voineagu et al., 2011). Further, in a mouse model of Rett syndrome, agenetic disorder that shares features with autism, developmental expo-sure to PBDE-47 caused epigenetic, cognitive, and social differences thatwere not observed in wild type mice (Amir et al., 1999; Woods et al.,2012). Finally, children with ASD have unique immune responses toPBDEs, which is discussed in detail below (Ashwood et al., 2009).

3.9. Halogenated aromatic hydrocarbons: polychlorinated biphenyls(PCBs)

Polychlorinated biphenyls (PCBs) are ubiquitous in the environmentand in animal and human tissues. Theywerewidely used as additives toindustrial oils and lubricants until the late 1970s when their adversehealth effects became apparent. PCB exposure is linked to adverse preg-nancy outcomes and neurobehavioral deficits (Kuratsune et al., 1971;Rogan et al., 1988; Chen and Hsu, 1994; Eriksson and Fredriksson,1998; Howard et al., 2003; Kenet et al., 2007; Lein et al., 2007;Tsukimori et al., 2008; Kim et al., 2009; Boix et al., 2010; Kim andPessah, 2011), as well as immune dysfunction. In studies involving ma-rine animals, murinemodels, and humans, PCBs lead to generalized im-mune suppression, characterized by diminished cellular immunity andatrophied lymphoid organs (Davis and Safe, 1990; Narayanan et al.,1998; Fournier et al., 2000; Shin et al., 2000; Tan et al., 2003; Beinekeet al., 2005; Leijs et al., 2009). Cytokine profiles are also impacted byPCBs. In a murine model, perinatal exposure to a PCB mixture inducedinflammatory cytokine expression (primarily IL-6) in the brain ofadult offspring (Hayley et al., 2011). In human blood cells, PCB 52 andPCB 133 induced transcriptional changes in several cytokine signalingand regulation pathways (Wens et al., 2011). This effect was only ob-served in cells from male donors, suggesting a male-bias for some PCBeffects. Another study showed that PCB 118 enhanced IL-4 producingT-cell development (Gaspar-Ramirez et al., 2012), which correlateswith findings in children with ASD and their mothers (Gupta et al.,1998; Abdallah et al., 2011; Goines et al., 2011b).

PCBs likely exert their immunotoxicity by interfering withimmunologically-relevant signaling pathways. For example, dioxin-likePCBs interfere with Ahr signaling, which is critical for maintaining ahealthy immune balance in the skin and gut (Li et al., 2011;Monteleone et al., 2011). PCBs also disrupt the key cytokine signalingpathways JAK/STAT and MAPK. Immune cells exposed to PCB 47 and153 have compromised function and altered STAT 5 and ERK phosphor-ylation (Canesi et al., 2003). STAT 5 activation is central for regulatory Tcell development (Burchill et al., 2003; Adamson et al., 2009), and acti-vation of this molecule by PCBs might mediate some of their immuno-suppressive effects. Finally, some PCB congeners interfere withryanodine receptors,which are expressedwidely in the immune system.These receptors are regulated by cytokines including TGF-Β (Hosoi et al.,2001; Pessah et al., 2010), and can induce IL-6 production after engage-ment (Treves et al., 2011). In summary, PCBs alter immune functionthrough a variety of mechanisms that may be relevant to immune pro-files observed in autism.

3.10. Halogenated aromatic hydrocarbons: polybrominated diphenylethers (PBDEs)

PBDEs are a group of flame retardants that are the subject of grow-ing concern due to their structural and functional similarities to PCBs.They are widely dispersed in the environment and bioaccumulate upthe food chain (Johnson-Restrepo and Kannan, 2009). EnvironmentalPBDE levels have jumped dramatically in the last several decades, andare increasingly found in human tissues and breast milk (Noren andMeironyte, 2000; Darnerud et al., 2001). Of note, this heightenedprevalence is concurrent with the apparent rise in ASD diagnoses(Hertz-Picciotto and Delwiche, 2009; Messer, 2010). While not linkedspecifically to ASD, PBDE exposure is associated with improper

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulatioment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

neurodevelopment, hormonal disruptions, and a variety of behavior-al, motor, and cognitive issues (Alm et al., 2008; He, He et al., 2008;Roze et al., 2009; Herbstman et al., 2010; Kodavanti et al., 2010;Schreiber et al., 2010; Dingemans et al., 2011).

Some evidence shows that PBDEs can impact immune activity.Studies involving marine and murine models link PBDEs to changesin the immune system, including thymic and splenic atrophy, in-creased production of IL-10, lymphocyte depletion, reduced antibodyrecall responses, and decreased responses to pathogens (Fowles et al.,1994; Thuvander, 1999; Beineke et al., 2005; Zhou et al., 2006;Beineke et al., 2007a, 2007b; Lundgren et al., 2009; Watanabe et al.,2010; Bondy et al., 2011; Fair et al., 2012). There is little data regard-ing the impact of PBDEs on cytokine production and signaling, and fu-ture studies should examine these endpoints in more detail. Onerecent study considered the effect of BDE-47 on cytokine responsesin children with autism and controls. Peripheral blood mononuclearcells obtained from ASD and typically developing controls werepretreated with PBDE-47 and stimulated with LPS. Analysis of super-natant cytokines showed that BDE-47 had a divergent impact on cellsfrom ASD versus typical controls. Among controls, BDE-47 caused asignificant decrease in inflammatory cytokines production (IL-6,IL-12, GM-CSF, TNF-α), indicating broad immune suppression. In con-trast, children with ASD only down-regulated IL-6 in the presence ofBDE-47, and had significantly higher IL-1Β responses (Ashwoodet al., 2009). This suggests that BDE-47 can enhance some aspects ofinflammation in ASD. This diagnosis-specific immunotoxic effect sug-gests that children with ASD respond differently to PBDEs than typicalchildren. Robust inflammation in response to such exposures duringcritical developmental windows could have long term neurologicaleffects, and may be a possible mechanism in ASD. Future studiesshould continue to examine a potential role for PBDEs in immunedysfunction and neurodevelopmental disorders like autism.

3.11. Pathogenic exposures in autism

Early-life infections can skew fetal development, leading to aber-rant neural and immune activity. This is a widely suggested etiologi-cal mechanism in schizophrenia, and is increasingly implicated inautism as well (Patterson, 2009). Several infections, including mea-sles, cytomegalovirus, and rubella during pre- and perinatal periodshave been associated with autism (Chess, 1977; Markowitz, 1983;Ivarsson et al., 1990; Sweeten et al., 2004a, 2004b; Libbey et al.,2005; Meyer et al., 2011). A recent large-scale epidemiologicalstudy showed that infection-related hospitalizations during pregnan-cy significantly increased the risk of ASD (Atladottir et al., 2010). In-terestingly, the risk was not associated with any specific type ofinfection, suggesting that the general immunemechanism controllingthe response to the pathogen rather than the pathogen itself were in-volved. This response is likely guaranteed to include cytokines. In-deed, there is clinical evidence for altered gestational cytokinemilieus in autism (Abdallah et al., 2011; Goines et al., 2011b), whichcould be related to infectious exposures, and may mediate aspectsof the disorder.

As discussed in previous sections, several studies have linked earlyimmune challenges to long-term changes in behavioral and immuneparameters. For example, mice prenatally exposed to influenza(Fatemi et al., 2002a; Shi et al., 2003), the viral mimic Poly I:C(Vuillermot et al., 2010), or the bacterial component LPS (Romeroet al., 2010) demonstrate long term neurological and behavioral ab-normalities. This effect is largely mediated by cytokines like IL-6 andIL-1Β (Samuelsson et al., 2006; Smith et al., 2007; Bilbo et al., 2008).Prenatal infectious exposures can also impact the developing immunesystem, and lead to long term immune dysregulation. This was dem-onstrated by increased levels of IL-6, IL-2, and TNF-α in adult animalsthat had been prenatally exposed to LPS (Romero et al., 2010). Anoth-er interesting set of studies showed that an early life infection led to

n in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 9: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

9PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

neurological deficits in adulthood that only became apparent after animmunological stimulation (Bilbo et al., 2005a, 2005b; Bilbo andSchwarz, 2009). This suggests that early life infection can change im-mune responses later in life, and that this has neurological conse-quences. The concept that prenatal infections can impact both brainand immune development is intriguing, and should be explored fur-ther in autism.

Overall, when considering environmental exposures, it is impor-tant to take time to consider that the prevalence rates for ASD haveincreased dramatically over the last 10–20 years. These rates continueto increase year-on-year. Arguably concentrations for some com-pounds such as lead and chlorinated pesticides have fallen in the pop-ulation since their removal or reduction from the environment in the1970s. In contrast, levels of PBDEs and OPs have increased. Althoughit may be tempting to link compounds as risk factors for ASD basedsolely on similar time trends there is a need for more extensive re-search to begin to understand whether such temporal relationshipsare associated with risk for ASD. Future research should focus onthe relationships between environmental exposures and risk forASD diagnosis and whether environmental exposures to such com-pounds as PBDE induce cytokine responses that could modulate neu-ronal function in the pediatric population. Moreover, future researchshould discern whether children who develop a ASD are more sensi-tive to specific environmental exposures using cytokine production asreadouts. More research focused on environmental exposures andASD is warranted.

4. Conclusions

Cytokine imbalances are well documented in autism and havemany interesting implications. Cytokines are intricately involved inneurodevelopment and neuronal function, and an ill-timed cytokinedisruption can have long term neurological consequences. Further,cytokine expression is largely dependent on genetic and environmentalinfluences. Therefore, they may represent a biomarker for genetic or en-vironmental factors at play in autism. To illustrate the connection be-tween immunity, genes, the environment, and neurodevelopmentaloutcome, consider two scenarios: First, an individual may be geneticallypoised to mount an inappropriate immune response to an infectious ortoxic exposure. This individual might respond either too robustly or tooweakly to resolve the threat without collateral damage to the brain andother body systems (including the immune system). Second, an individ-ual may lack appropriate genetic machinery to excrete toxins; leading totheir accumulation in tissue. This could lead to an amplification of thetoxin's effects on a variety of body systems, including the brain and im-mune system. For each child, an environmental challenge during a criti-cal window of development could have especially severe consequences,causing abnormal CNS function, altered immune phenotypes, and per-haps autism. These scenarios represent an emerging global view of au-tism that considers a broad contribution of several factors, includinggenes, the environment, and the immune system. Cross-disciplinary in-vestigations that consider diverse biological contributions will be essen-tial to untangle ASD.

Conflict of interest statement

The authors report no conflict of interest.

Acknowledgements and financial disclosures

This study was supported by the National Institute of NeurologicalDisorders and Stroke (R21HD065269), Autism Speaks Foundation(7567), the Jane Botsford Johnson Foundation, National Alliance forResearch on Schizophrenia and Depression, the Peter Emch Founda-tion, and the Hartwell Foundation. The authors report no conflicts ofinterest for this study.

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulationment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

References

Aaltonen R, Heikkinen T, Hakala K, Laine K, Alanen A. Transfer of proinflammatory cy-tokines across term placenta. Obstet Gynecol 2005;106(4):802–7.

Abdallah MW, Larsen N, Grove J, Norgaard-Pedersen B, Thorsen P, Mortensen EL, et al.Amniotic fluid inflammatory cytokines: Potential markers of immunologic dys-function in autism spectrum disorders. World J Biol Psychiatry 2011.

Adamson AS, Collins K, Laurence A, O'Shea JJ. The Current STATus of lymphocyte signal-ing: new roles for old players. Curr Opin Immunol 2009;21(2):161–6.

Albizzati A, More L, Di Candia D, Saccani M, Lenti C. Normal concentrations of heavymetals in autistic spectrum disorders. Minerva Pediatr 2012;64(1):27–31.

Alleva E, Cirulli F, Bianchi M, Bondiolotti GP, Chiarotti F, De Acetis L, et al. Behaviouralcharacterization of interleukin-6 overexpressing or deficient mice during agonisticencounters. Eur J Neurosci 1998;10(12):3664–72.

Alm H, Kultima K, Scholz B, Nilsson A, Andren PE, Fex-Svenningsen A, et al. Exposure tobrominated flame retardant PBDE-99 affects cytoskeletal protein expression in theneonatal mouse cerebral cortex. Neurotoxicology 2008;29(4):628–37.

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disor-ders. Washington DC: American Psychiatric Association; 1994.

Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome iscaused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2.Nat Genet 1999;23(2):185–8.

Anand S, Singh S, Nahar Saikia U, Bhalla A, Paul Sharma Y, Singh D. Cardiac abnormal-ities in acute organophosphate poisoning. Clin Toxicol (Phila) 2009;47(3):230–5.

Andres DA, Shi GX, Bruun D, Barnhart C, Lein PJ. Rit signaling contributes tointerferon-gamma-induced dendritic retraction via p38 mitogen-activated proteinkinase activation. J Neurochem 2008;107(5):1436–47.

Araujo DM, Cotman CW. Trophic effects of interleukin-4, -7 and -8 on hippocampalneuronal cultures: potential involvement of glial-derived factors. Brain Res1993;600(1):49–55.

Armario A, Hernandez J, Bluethmann H, Hidalgo J. IL-6 deficiency leads to increasedemotionality in mice: evidence in transgenic mice carrying a null mutation forIL-6. J Neuroimmunol 1998;92(1–2):160–9.

Ashwood P, Anthony A, Torrente F, Wakefield AJ. Spontaneous mucosal lymphocyte cy-tokine profiles in children with autism and gastrointestinal symptoms: mucosalimmune activation and reduced counter regulatory interleukin-10. J Clin Immunol2004;24(6):664–73.

Ashwood P, Enstrom A, Krakowiak P, Hertz-Picciotto I, Hansen RL, Croen LA, et al. De-creased transforming growth factor beta1 in autism: a potential link between im-mune dysregulation and impairment in clinical behavioral outcomes. JNeuroimmunol 2008;204(1–2):149–53.

Ashwood P, Schauer J, Pessah IN, Van de Water J. Preliminary evidence of the in vitroeffects of BDE-47 on innate immune responses in children with autism spectrumdisorders. J Neuroimmunol 2009;208(1–2):130–5.

Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah I, Van deWater J. Elevatedplasma cytokines in autism spectrum disorders provide evidence of immune dys-function and are associated with impaired behavioral outcome. Brain BehavImmun 2011a;25(1):40–5.

Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah IN, Van deWater J. AlteredT cell responses in children with autism. Brain Behav Immun 2011b;25(5):840–9.

Atladottir HO, Thorsen P, Ostergaard L, Schendel DE, Lemcke S, Abdallah M, et al. Ma-ternal infection requiring hospitalization during pregnancy and autism spectrumdisorders. J Autism Dev Disord 2010;40(12):1423–30.

Bachiller D, Klingensmith J, Kemp C, Belo JA, Anderson RM, May SR, et al. The organizerfactors Chordin and Noggin are required for mouse forebrain development. Nature2000;403(6770):658–61.

Bagenstose LM, Salgame P, Monestier M. Mercury-induced autoimmunity in the ab-sence of IL-4. Clin Exp Immunol 1998;114(1):9-12.

Bagenstose LM, Salgame P, Monestier M. Murine mercury-induced autoimmunity: amodel of chemically related autoimmunity in humans. Immunol Res 1999;20(1):67–78.

Balschun D, Wetzel W, Del Rey A, Pitossi F, Schneider H, Zuschratter W, et al. Interleu-kin-6: a cytokine to forget. FASEB J 2004;18(14):1788–90.

Banerjee BD, Koner BC, Ray A. Immunotoxicity of pesticides: perspectives and trends.Indian J Exp Biol 1996;34(8):723–33.

Banks WA, Ortiz L, Plotkin SR, Kastin AJ. Human interleukin (IL) 1 alpha, murine IL-1alpha and murine IL-1 beta are transported from blood to brain in the mouse bya shared saturable mechanism. J Pharmacol Exp Ther 1991;259(3):988–96.

Banks WA, Kastin AJ, Gutierrez EG. Penetration of interleukin-6 across the murineblood-brain barrier. Neurosci Lett 1994;179(1–2):53–6.

Barcia C, Ros CM, Annese V, Gomez A, Ros-Bernal F, Aguado-Yera D, et al. IFN-gammasignaling, with the synergistic contribution of TNF-alpha, mediates cell specificmicroglial and astroglial activation in experimental models of Parkinson's disease.Cell Death Dis 2011;2:e142.

Barish ME, Mansdorf NB, Raissdana SS. Gamma-interferon promotes differentiation ofcultured cortical and hippocampal neurons. Dev Biol 1991;144(2):412–23.

Barksby HE, Lea SR, Preshaw PM, Taylor JJ. The expanding family of interleukin-1 cyto-kines and their role in destructive inflammatory disorders. Clin Exp Immunol2007;149(2):217–25.

Baron-Cohen S, Knickmeyer RC, Belmonte MK. Sex differences in the brain: implica-tions for explaining autism. Science 2005;310(5749):819–23.

Barrientos RM, Frank MG, Hein AM, Higgins EA, Watkins LR, Rudy JW, et al. Time courseof hippocampal IL-1 beta and memory consolidation impairments in aging rats fol-lowing peripheral infection. Brain Behav Immun 2009;23(1):46–54.

Bauer S, Kerr BJ, Patterson PH. The neuropoietic cytokine family in development, plas-ticity, disease and injury. Nat Rev Neurosci 2007;8(3):221–32.

in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 10: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

10 PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

Beineke A, Siebert U, McLachlan M, Bruhn R, Thron K, Failing K, et al. Investigations ofthe potential influence of environmental contaminants on the thymus and spleenof harbor porpoises (Phocoena phocoena). Environ Sci Technol 2005;39(11):3933–8.

Beineke A, Siebert U, Muller G, Baumgartner W. Increased blood interleukin-10 mRNAlevels in diseased free-ranging harbor porpoises (Phocoena phocoena). VetImmunol Immunopathol 2007a;115(1–2):100–6.

Beineke A, Siebert U, Stott J, Muller G, Baumgartner W. Phenotypical characterizationof changes in thymus and spleen associated with lymphoid depletion infree-ranging harbor porpoises (Phocoena phocoena). Vet Immunol Immunopathol2007b;117(3–4):254–65.

Bellinger D, Dietrich KN. Low-level lead exposure and cognitive function in children.Pediatr Ann 1994;23(11):600–5.

Bellinger FP, Madamba SG, Campbell IL, Siggins GR. Reduced long-term potentiation inthe dentate gyrus of transgenic mice with cerebral overexpression of interleukin-6.Neurosci Lett 1995;198(2):95–8.

Bhatti GK, Bhatti JS, Kiran R, Sandhir R. Alterations in Ca(2) homeostasis and oxidativedamage induced by ethion in erythrocytes of Wistar rats: ameliorative effect of vi-tamin E. Environ Toxicol Pharmacol 2011;31(3):378–86.

Bilbo SD. Early-life infection is a vulnerability factor for aging-related glial alterationsand cognitive decline. Neurobiol Learn Mem 2010;94(1):57–64.

Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a criticalrole for the immune system. Front Behav Neurosci 2009;3:14.

Bilbo SD, Biedenkapp JC, Der-Avakian A, Watkins LR, Rudy JW, Maier SF. Neonatalinfection-induced memory impairment after lipopolysaccharide in adulthood isprevented via caspase-1 inhibition. J Neurosci 2005a;25(35):8000–9.

Bilbo SD, Levkoff LH, Mahoney JH, Watkins LR, Rudy JW, Maier SF. Neonatal infectioninduces memory impairments following an immune challenge in adulthood.Behav Neurosci 2005b;119(1):293–301.

Bilbo SD, Barrientos RM, Eads AS, Northcutt A, Watkins LR, Rudy JW, et al. Early-life in-fection leads to altered BDNF and IL-1beta mRNA expression in rat hippocampusfollowing learning in adulthood. Brain Behav Immun 2008;22(4):451–5.

Boehm U, Klamp T, Groot M, Howard JC. Cellular responses to interferon-gamma. AnnuRev Immunol 1997;15:749–95.

Boix J, Cauli O, Felipo V. Developmental exposure to polychlorinated biphenyls 52, 138or 180 affects differentially learning or motor coordination in adult rats. Mecha-nisms involved. Neuroscience 2010;167(4):994-1003.

Bondy GS, Lefebvre DE, Aziz S, Cherry W, Coady L, Maclellan E, et al. Toxicologic andimmunologic effects of perinatal exposure to the brominated diphenyl ether(BDE) mixture DE-71 in the Sprague-Dawley rat. Environ Toxicol 2011. May 4epub ahead of print. http://dx.doi.org/10.1002/tox.20713.

Bouchard MF, Chevrier J, Harley KG, Kogut K, Vedar M, Calderon N, et al. Prenatal expo-sure to organophosphate pesticides and IQ in 7-year-old children. Environ HealthPerspect 2011;119(8):1189–95.

Braida D, Sacerdote P, Panerai AE, Bianchi M, Aloisi AM, Iosue S, et al. Cognitive functionin young and adult IL (interleukin)-6 deficient mice. Behav Brain Res 2004;153(2):423–9.

Braunschweig D, Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Croen LA, et al.Autism: maternally derived antibodies specific for fetal brain proteins.Neurotoxicology 2008;29(2):226–31.

Bridges CC, Zalups RK. Transport of inorganic mercury andmethylmercury in target tis-sues and organs. J Toxicol Environ Health B Crit Rev 2010;13(5):385–410.

Brionne TC, Tesseur I, Masliah E, Wyss-Coray T. Loss of TGF-beta 1 leads to increasedneuronal cell death and microgliosis in mouse brain. Neuron 2003;40(6):1133–45.

Buckwalter MS, Wyss-Coray T. Modelling neuroinflammatory phenotypes in vivo. JNeuroinflammation 2004;1(1):10.

Burchill MA, Goetz CA, Prlic M, O'Neil JJ, Harmon IR, Bensinger SJ, et al. Distinct effectsof STAT5 activation on CD4+ and CD8+ T cell homeostasis: development ofCD4+CD25+ regulatory T cells versus CD8+ memory T cells. J Immunol2003;171(11):5853–64.

Butovsky O, Ziv Y, Schwartz A, Landa G, Talpalar AE, Pluchino S, et al. Microglia activatedby IL-4 or IFN-gamma differentially induce neurogenesis and oligodendrogenesisfrom adult stem/progenitor cells. Mol Cell Neurosci 2006;31(1):149–60.

Byers DE, Holtzman MJ. Alternatively activated macrophages and airway disease. Chest2011;140(3):768–74.

Campbell IL, Abraham CR, Masliah E, Kemper P, Inglis JD, Oldstone MB, et al. Neurologicdisease induced in transgenic mice by cerebral overexpression of interleukin 6.Proc Natl Acad Sci U S A 1993;90(21):10061–5.

Campbell DB, Sutcliffe JS, Ebert PJ, Militerni R, Bravaccio C, Trillo S, et al. A genetic var-iant that disrupts MET transcription is associated with autism. Proc Natl Acad Sci US A 2006;103(45):16834–9.

Canesi L, Ciacci C, Betti M, Scarpato A, Citterio B, Pruzzo C, et al. Effects of PCB congenerson the immune function of Mytilus hemocytes: alterations of tyrosinekinase-mediated cell signaling. Aquat Toxicol 2003;63(3):293–306.

Casida JE. Pest toxicology: the primary mechanisms of pesticide action. Chem ResToxicol 2009;22(4):609–19.

CDC. Prevalence of autism spectrum disorders - autism and developmental disabilitiesmonitoring network, 14 sites, United States, 2008. MMWR Surveill Summ2012;61(3):1-19.

Centers for Disease Control and Prevention. Prevalence of autism spectrum disorders -Autism and Developmental Disabilities Monitoring Network, United States, 2006.MMWR Surveill Summ 2009;58(10):1-20.

Chakrabarty P, Ceballos-Diaz C, Beccard A, Janus C, Dickson D, Golde TE, et al.IFN-gamma promotes complement expression and attenuates amyloid plaque de-position in amyloid beta precursor protein transgenic mice. J Immunol2010;184(9):5333–43.

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulatioment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

Chapman S, Kadar T, Gilat E. Seizure duration following sarin exposure affectsneuro-inflammatory markers in the rat brain. Neurotoxicology 2006;27(2):277–83.

Chen YJ, Hsu CC. Effects of prenatal exposure to PCBs on the neurological function ofchildren: a neuropsychological and neurophysiological study. Dev Med ChildNeurol 1994;36(4):312–20.

Chess S. Follow-up report on autism in congenital rubella. J Autism Child Schizophr1977;7(1):69–81.

Coburn CG, Curras-Collazo MC, Kodavanti PR. In vitro effects of environmentally rele-vant polybrominated diphenyl ether (PBDE) congeners on calcium bufferingmechanisms in rat brain. Neurochem Res 2008;33(2):355–64.

Cohen DJ, Johnson WT, Caparulo BK. Pica and elevated blood lead level in autistic andatypical children. Am J Dis Child 1976;130(1):47–8.

Corbin JG, Kelly D, Rath EM, Baerwald KD, Suzuki K, Popko B. Targeted CNS expressionof interferon-gamma in transgenic mice leads to hypomyelination, reactive gliosis,and abnormal cerebellar development. Mol Cell Neurosci 1996;7(5):354–70.

Cordier S, Garel M, Mandereau L, Morcel H, Doineau P, Gosme-Seguret S, et al.Neurodevelopmental investigations among methylmercury-exposed children inFrench Guiana. Environ Res 2002;89(1):1-11.

Costa LG. Current issues in organophosphate toxicology. Clin Chim Acta 2006;366(1–2):1-13.

Courchesne E, Mouton PR, Calhoun ME, Semendeferi K, Ahrens-Barbeau C, Hallet MJ,et al. Neuron number and size in prefrontal cortex of children with autism. JAMA2011;306(18):2001–10.

Crinnion WJ. Chlorinated pesticides: threats to health and importance of detection.Altern Med Rev 2009;14(4):347–59.

Croonenberghs J, Bosmans E, Deboutte D, Kenis G, Maes M. Activation of the inflamma-tory response system in autism. Neuropsychobiology 2002;45(1):1–6.

da Silva AG, Campello-Costa P, Linden R, Sholl-Franco A. Interleukin-4 blocks prolifer-ation of retinal progenitor cells and increases rod photoreceptor differentiationthrough distinct signaling pathways. J Neuroimmunol 2008;196(1–2):82–93.

Dahlgren J, Samuelsson AM, Jansson T, Holmang A. Interleukin-6 in the maternal circu-lation reaches the rat fetus in mid-gestation. Pediatr Res 2006;60(2):147–51.

D'Amelio M, Ricci I, Sacco R, Liu X, D'Agruma L, Muscarella LA, et al. Paraoxonase genevariants are associated with autism in North America, but not in Italy: possible re-gional specificity in gene-environment interactions. Mol Psychiatry 2005;10(11):1006–16.

Damodaran TV, Greenfield ST, Patel AG, Dressman HK, Lin SK, Abou-Donia MB.Toxicogenomic studies of the rat brain at an early time point following acutesarin exposure. Neurochem Res 2006;31(3):367–81.

Daniel V, Huber W, Bauer K, Suesal C, Conradt C, Opelz G. Associations ofdichlorodiphenyltrichloroethane (DDT) 4.4 and dichlorodiphenyldichloroethylene(DDE) 4.4 blood levels with plasma IL-4. Arch Environ Health 2002;57(6):541–7.

Dantzer R. Cytokine-induced sickness behavior: where do we stand? Brain BehavImmun 2001;15(1):7-24.

Darnerud PO, Eriksen GS, Johannesson T, Larsen PB, Viluksela M. Polybrominateddiphenyl ethers: occurrence, dietary exposure, and toxicology. Environ HealthPerspect 2001;109(Suppl. 1):49–68.

Davis D, Safe S. Immunosuppressive activities of polychlorinated biphenyls inC57BL/6N mice: structure-activity relationships as Ah receptor agonists and partialantagonists. Toxicology 1990;63(1):97-111.

de la Mano A, Gato A, Alonso MI, Carnicero E, Martin C, Moro JA. Role ofinterleukin-1beta in the control of neuroepithelial proliferation and differentiationof the spinal cord during development. Cytokine 2007;37(2):128–37.

De Simone R, Levi G, Aloisi F. Interferon gamma gene expression in rat central nervoussystem glial cells. Cytokine 1998;10(6):418–22.

De Vito P, Incerpi S, Pedersen JZ, Luly P, Davis FB, Davis PJ. Thyroid hormones as mod-ulators of immune activities at the cellular level. Thyroid 2011;21(8):879–90.

DeFelice ML, Ruchelli ED, Markowitz JE, Strogatz M, Reddy KP, Kadivar K, et al. Intesti-nal cytokines in children with pervasive developmental disorders. Am JGastroenterol 2003;98(8):1777–82.

Depino AM, Lucchina L, Pitossi F. Early and adult hippocampal TGF-beta1overexpression have opposite effects on behavior. Brain Behav Immun2011;25(8):1582–91.

Derecki NC, Cardani AN, Yang CH, Quinnies KM, Crihfield A, Lynch KR, et al. Regulationof learning and memory by meningeal immunity: a key role for IL-4. J Exp Med2010;207(5):1067–80.

Desoto MC, Hitlan RT. Blood levels of mercury are related to diagnosis of autism: areanalysis of an important data set. J Child Neurol 2007;22(11):1308–11.

DeStefano F. Vaccines and autism: evidence does not support a causal association. ClinPharmacol Ther 2007;82(6):756–9.

Deverman BE, Patterson PH. Cytokines and CNS development. Neuron 2009;64(1):61–78.

Dhote F, Peinnequin A, Carpentier P, Baille V, Delacour C, Foquin A, et al. Prolonged in-flammatory gene response following soman-induced seizures in mice. Toxicology2007;238(2–3):166–76.

Dieguez-Acuna FJ, Ellis ME, Kushleika J, Woods JS. Mercuric ion attenuates nuclearfactor-kappaB activation and DNA binding in normal rat kidney epithelial cells: im-plications for mercury-induced nephrotoxicity. Toxicol Appl Pharmacol2001;173(3):176–87.

Diel F, Horr B, Borck H, Irman-Florjanc T. Pyrethroid insecticides influence the signaltransduction in T helper lymphocytes from atopic and nonatopic subjects. InflammRes 2003;52(4):154–63.

Dillman III JF, Phillips CS, Kniffin DM, Tompkins CP, Hamilton TA, Kan RK. Gene expres-sion profiling of rat hippocampus following exposure to the acetylcholinesteraseinhibitor soman. Chem Res Toxicol 2009;22(4):633–8.

n in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 11: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

11PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

Dingemans MM, van den Berg M, Westerink RH. Neurotoxicity of brominated flame re-tardants: (in)direct effects of parent and hydroxylated polybrominated diphenylethers on the (developing) nervous system. Environ Health Perspect2011;119(7):900–7.

Doyle KP, Cekanaviciute E, Mamer LE, Buckwalter MS. TGFbeta signaling in the brainincreases with aging and signals to astrocytes and innate immune cells in theweeks after stroke. J Neuroinflammation 2010;7:62.

Dudley DJ, Hunter C, Varner MW, Mitchell MD. Elevation of amniotic fluid interleu-kin-4 concentrations in women with preterm labor and chorioamnionitis. Am JPerinatol 1996;13(7):443–7.

Duramad P, Harley K, Lipsett M, Bradman A, Eskenazi B, Holland NT, et al. Early envi-ronmental exposures and intracellular Th1/Th2 cytokine profiles in 24-month-oldchildren living in an agricultural area. Environ Health Perspect 2006a;114(12):1916–22.

Duramad P, Tager IB, Leikauf J, Eskenazi B, Holland NT. Expression of Th1/Th2 cytokinesin human blood after in vitro treatment with chlorpyrifos, and its metabolites, incombination with endotoxin LPS and allergen Der p1. J Appl Toxicol2006b;26(5):458–65.

Duysen EG, Li B, Xie W, Schopfer LM, Anderson RS, Broomfield CA, et al. Evidence fornonacetylcholinesterase targets of organophosphorus nerve agent:supersensitivity of acetylcholinesterase knockout mouse to VX lethality. JPharmacol Exp Ther 2001;299(2):528–35.

Emanuele E, Orsi P, Boso M, Broglia D, Brondino N, Barale F, et al. Low-gradeendotoxemia in patients with severe autism. Neurosci Lett 2010;471(3):162–5.

Enstrom AM, Van deWater JA, Ashwood P. Autoimmunity in autism. Curr Opin InvestigDrugs 2009a;10(5):463–73.

Enstrom AM, Lit L, Onore CE, Gregg JP, Hansen RL, Pessah IN, et al. Altered gene expres-sion and function of peripheral blood natural killer cells in children with autism.Brain Behav Immun 2009b;23(1):124–33.

Enstrom AM, Onore CE, Van de Water JA, Ashwood P. Differential monocyte responsesto TLR ligands in children with autism spectrum disorders. Brain Behav Immun2010;24(1):64–71.

Eriksson P, Fredriksson A. Neurotoxic effects in adult mice neonatally exposed to3,3'4,4'5-pentachlorobiphenyl or 2,3,3'4,4'-pentachlorobiphenyl. Changes in brainnicotinic receptors and behaviour. Environ Toxicol Pharmacol 1998;5(1):17–27.

Escary JL, Perreau J, Dumenil D, Ezine S, Brulet P. Leukaemia inhibitory factor is neces-sary for maintenance of haematopoietic stem cells and thymocyte stimulation. Na-ture 1993;363(6427):361–4.

Eskenazi B, Marks AR, Bradman A, Harley K, Barr DB, Johnson C, et al. Organophosphatepesticide exposure and neurodevelopment in young Mexican-American children.Environ Health Perspect 2007;115(5):792–8.

Eskenazi B, Huen K, Marks A, Harley KG, Bradman A, Barr DB, et al. PON1 andneurodevelopment in children from the CHAMACOS study exposed toorganophosphate pesticides in utero. Environ Health Perspect 2010;118(12):1775–81.

Fair PA, Stavros HC, Mollenhauer MA, Dewitt JC, Henry N, Kannan K, et al. Immunefunction in female B(6)C(3)F(1) mice is modulated by DE-71, a commercialpolybrominated diphenyl ether mixture. J Immunotoxicol 2012;9(1):96-107.

Fatemi SH, Earle J, Kanodia R, Kist D, Emamian ES, Patterson PH, et al. Prenatal viral in-fection leads to pyramidal cell atrophy and macrocephaly in adulthood: implica-tions for genesis of autism and schizophrenia. Cell Mol Neurobiol 2002a;22(1):25–33.

Fatemi SH, Halt AR, Stary JM, Kanodia R, Schulz SC, Realmuto GR. Glutamic acid decar-boxylase 65 and 67 kDa proteins are reduced in autistic parietal and cerebellar cor-tices. Biol Psychiatry 2002b;52(8):805–10.

Fatemi SH, Snow AV, Stary JM, Araghi-Niknam M, Reutiman TJ, Lee S, et al. Reelin sig-naling is impaired in autism. Biol Psychiatry 2005;57(7):777–87.

Fatemi SH, Reutiman TJ, Folsom TD, Rooney RJ, Patel DH, Thuras PD. mRNA and proteinlevels for GABAAalpha4, alpha5, beta1 and GABABR1 receptors are altered inbrains from subjects with autism. J Autism Dev Disord 2010;40(6):743–50.

Filipek PA, Accardo PJ, Baranek GT, Cook Jr EH, Dawson G, Gordon B, et al. The screeningand diagnosis of autistic spectrum disorders. J Autism Dev Disord 1999;29(6):439–84.

Flohe SB, Bruggemann J, Herder C, Goebel C, Kolb H. Enhanced proinflammatory re-sponse to endotoxin after priming of macrophages with lead ions. J Leukoc Biol2002;71(3):417–24.

Folkerth RD, Keefe RJ, Haynes RL, Trachtenberg FL, Volpe JJ, Kinney HC.Interferon-gamma expression in periventricular leukomalacia in the humanbrain. Brain Pathol 2004;14(3):265–74.

Fournier M, Degas V, Colborn T, Omara FO, Denizeau F, Potworowski EF, et al. Immuno-suppression in mice fed on diets containing beluga whale blubber from the St Law-rence estuary and the Arctic populations. Toxicol Lett 2000;112–113:311–7.

Fowles JR, Fairbrother A, Baecher-Steppan L, Kerkvliet NI. Immunologic and endocrineeffects of the flame-retardant pentabromodiphenyl ether (DE-71) in C57BL/6Jmice. Toxicology 1994;86(1–2):49–61.

Gadient RA, Otten U. Expression of interleukin-6 (IL-6) and interleukin-6 receptor(IL-6R) mRNAs in rat brain during postnatal development. Brain Res1994a;637(1–2):10–4.

Gadient RA, Otten U. Identification of interleukin-6 (IL-6)-expressing neurons in thecerebellum and hippocampus of normal adult rats. Neurosci Lett 1994b;182(2):243–6.

Galloway T, Handy R. Immunotoxicity of organophosphorous pesticides. Ecotoxicology2003;12(1–4):345–63.

Garg SK, Kipnis J, Banerjee R. IFN-gamma and IL-4 differentially shape metabolic re-sponses and neuroprotective phenotype of astrocytes. J Neurochem 2009;108(5):1155–66.

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulationment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

Garry VF, Harkins ME, Erickson LL, Long-Simpson LK, Holland SE, Burroughs BL. Birthdefects, season of conception, and sex of children born to pesticide applicators liv-ing in the Red River Valley of Minnesota, USA. Environ Health Perspect2002;110(Suppl. 3):441–9.

Gaspar-Ramirez O, Perez-Vazquez FJ, Pruneda-Alvarez LG, Orta-Garcia ST,Gonzalez-Amaro R, Perez-Maldonado IN. Effect of polychlorinated biphenyls 118and 153 on Th1/Th2 cells differentiation. Immunopharmacol Immunotoxicol2012;34(4):627–32.

Giulian D, Young DG, Woodward J, Brown DC, Lachman LB. Interleukin-1 is an astroglialgrowth factor in the developing brain. J Neurosci 1988;8(2):709–14.

Goebel C, Flohe SB, Kirchhoff K, Herder C, Kolb H. Orally administered lead chloride in-duces bias of mucosal immunity. Cytokine 2000;12(9):1414–8.

Goines P, Haapanen L, Boyce R, Duncanson P, Braunschweig D, Delwiche L, et al. Auto-antibodies to cerebellum in children with autism associate with behavior. BrainBehav Immun 2011a;25(3):514–23.

Goines PE, Croen LA, Braunschweig D, Yoshida CK, Grether J, Hansen R, et al. Increasedmidgestational IFN-gamma, IL-4 and IL-5 in women bearing a child with autism: Acase-control study. Mol Autism 2011b;2:13.

Gomes FC, Sousa Vde O, Romao L. Emerging roles for TGF-beta1 in nervous system de-velopment. Int J Dev Neurosci 2005;23(5):413–24.

Goshen I, Kreisel T, Ounallah-Saad H, Renbaum P, Zalzstein Y, Ben-Hur T, et al. A dualrole for interleukin-1 in hippocampal-dependent memory processes.Psychoneuroendocrinology 2007;32(8–10):1106–15.

Goth SR, Chu RA, Gregg JP, Cherednichenko G, Pessah IN. Uncoupling of ATP-mediatedcalcium signaling and dysregulated interleukin-6 secretion in dendritic cells bynanomolar thimerosal. Environ Health Perspect 2006;114(7):1083–91.

Griffin WS, Sheng JG, Roberts GW, Mrak RE. Interleukin-1 expression in differentplaque types in Alzheimer's disease: significance in plaque evolution. JNeuropathol Exp Neurol 1995;54(2):276–81.

Grigorenko EL, Han SS, Yrigollen CM, Leng L, Mizue Y, Anderson GM, et al. Macrophagemigration inhibitory factor and autism spectrum disorders. Pediatrics2008;122(2):e438–45.

Gu C, Goodarzi M, Yang X, Bian Y, Sun C, Jiang X. Predictive insight into the relationshipbetween AhR binding property and toxicity of polybrominated diphenyl ethers byPLS-derived QSAR. Toxicol Lett 2012;208(3):269–74.

Gupta S, Aggarwal S, Rashanravan B, Lee T. Th1- and Th2-like cytokines in CD4+ andCD8+ T cells in autism. J Neuroimmunol 1998;85(1):106–9.

Hallmayer J, Cleveland S, Torres A, Phillips J, Cohen B, Torigoe T, et al. Genetic heritabil-ity and shared environmental factors among twin pairs with autism. Arch Gen Psy-chiatry 2011;68(11):1095–102.

Hamaguchi M, Namera A, Tsuda N, Uejima T, Maruyama K, Kanai T, et al. Severe acutepancreatitis caused by organophosphate poisoning. Chudoku Kenkyu 2006;19(4):395–9.

Handley MT, Lian LY, Haynes LP, Burgoyne RD. Structural and functional deficits in aneuronal calcium sensor-1 mutant identified in a case of autistic spectrum disor-der. PLoS One 2010;5(5):e10534.

Hang CH, Shi JX, Tian J, Li JS, Wu W, Yin HX. Effect of systemic LPS injection on corticalNF-kappaB activity and inflammatory response following traumatic brain injury inrats. Brain Res 2004;1026(1):23–32.

Hayley S, Mangano E, Crowe G, Li N, Bowers WJ. An in vivo animal study assessinglong-term changes in hypothalamic cytokines following perinatal exposure to achemical mixture based on Arctic maternal body burden. Environ Health2011;10:65.

He P, HeW,Wang A, Xia T, Xu B, Zhang M, et al. PBDE-47-induced oxidative stress, DNAdamage and apoptosis in primary cultured rat hippocampal neurons.Neurotoxicology 2008;29(1):124–9.

Heinrich PC, Behrmann I, Haan S, Hermanns HM, Muller-Newen G, Schaper F. Princi-ples of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J2003;374(Pt 1):1-20.

Hemdan NY, Lehmann I, Wichmann G, Lehmann J, Emmrich F, Sack U.Immunomodulation by mercuric chloride in vitro: application of different cell acti-vation pathways. Clin Exp Immunol 2007;148(2):325–37.

Henderson RF, Barr EB, Blackwell WB, Clark CR, Conn CA, Kalra R, et al. Response of ratsto low levels of sarin. Toxicol Appl Pharmacol 2002;184(2):67–76.

Henrich-Noack P, Prehn JH, Krieglstein J. Neuroprotective effects of TGF-beta 1. J NeuralTransm Suppl 1994;43:33–45.

Herbstman JB, Sjodin A, Kurzon M, Lederman SA, Jones RS, Rauh V, et al. Prenatal expo-sure to PBDEs and neurodevelopment. Environ Health Perspect 2010;118(5):712–9.

Hertz-Picciotto I, Delwiche L. The rise in autism and the role of age at diagnosis. Epide-miology 2009;20(1):84–90.

Hertz-Picciotto I, Green PG, Delwiche L, Hansen R, Walker C, Pessah IN. Blood mercuryconcentrations in CHARGE Study children with and without autism. EnvironHealth Perspect 2010;118(1):161–6.

Heuer L, Braunschweig D, Ashwood P, Van de Water J, Campbell DB. Association of aMET genetic variant with autism-associated maternal autoantibodies to fetalbrain proteins and cytokine expression. Transl Psychiatry 2011;1.

Heupel K, Sargsyan V, Plomp JJ, Rickmann M, Varoqueaux F, Zhang W, et al. Loss oftransforming growth factor-beta 2 leads to impairment of central synapse func-tion. Neural Dev 2008;3:25.

Heusinkveld HJ, Westerink RH. Organochlorine insecticides lindane and dieldrin andtheir binary mixture disturb calcium homeostasis in dopaminergic PC12 cells. En-viron Sci Technol 2012;46(3):1842–8.

Heyser CJ, Masliah E, Samimi A, Campbell IL, Gold LH. Progressive decline in avoidancelearning paralleled by inflammatory neurodegeneration in transgenic mice ex-pressing interleukin 6 in the brain. Proc Natl Acad Sci U S A 1997;94(4):1500–5.

in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 12: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

12 PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

Hosoi E, Nishizaki C, Gallagher KL, Wyre HW, Matsuo Y, Sei Y. Expression of theryanodine receptor isoforms in immune cells. J Immunol 2001;167(9):4887–94.

Howard AS, Fitzpatrick R, Pessah I, Kostyniak P, Lein PJ. Polychlorinated biphenyls in-duce caspase-dependent cell death in cultured embryonic rat hippocampal butnot cortical neurons via activation of the ryanodine receptor. Toxicol ApplPharmacol 2003;190(1):72–86.

Hsiao EY, Patterson PH. Activation of the maternal immune system induces endocrinechanges in the placenta via IL-6. Brain Behav Immun 2011;25(4):604–15.

Hu J, Roy SK, Shapiro PS, Rodig SR, Reddy SP, Platanias LC, et al. ERK1 and ERK2 activateCCAAAT/enhancer-binding protein-beta-dependent gene transcription in responseto interferon-gamma. J Biol Chem 2001;276(1):287–97.

Huell M, Strauss S, Volk B, Berger M, Bauer J. Interleukin-6 is present in early stages ofplaque formation and is restricted to the brains of Alzheimer's disease patients.Acta Neuropathol 1995;89(6):544–51.

Ihara S, Nakajima K, Fukada T, Hibi M, Nagata S, Hirano T, et al. Dual control of neuriteoutgrowth by STAT3 and MAP kinase in PC12 cells stimulated with interleukin-6.EMBO J 1997;16(17):5345–52.

Ip P, Wong V, Ho M, Lee J, Wong W. Mercury exposure in children with autistic spec-trum disorder: case-control study. J Child Neurol 2004;19(6):431–4.

Ivarsson SA, Bjerre I, Vegfors P, Ahlfors K. Autism as one of several disabilities in twochildren with congenital cytomegalovirus infection. Neuropediatrics 1990;21(2):102–3.

Jenkins C, Roberts J, Wilson R, MacLean MA, Shilito J, Walker JJ. Evidence of a T(H) 1type response associated with recurrent miscarriage. Fertil Steril 2000;73(6):1206–8.

Jiang J, Tian K, Diao Y, Chen H, Zhu P, Wang Z. Expression of TNF alpha, IL-1 beta, IL-6mRNA, release of TNF alpha in vital organs and their relationship with endotoxintranslocation following hemorrhagic shock. Chin Med Sci J 1997;12(1):41–6.

Johnson EA, Kan RK. The acute phase response and soman-induced status epilepticus:temporal, regional and cellular changes in rat brain cytokine concentrations. JNeuroinflammation 2010;7:40.

Johnson-Restrepo B, Kannan K. An assessment of sources and pathways of human ex-posure to polybrominated diphenyl ethers in the United States. Chemosphere 2009Jul;76(4):542–8.

Jonakait GM, Wei R, Sheng ZL, Hart RP, Ni L. Interferon-gamma promotes cholinergicdifferentiation of embryonic septal nuclei and adjacent basal forebrain. Neuron1994;12(5):1149–59.

Jyonouchi H, Sun S, Le H. Proinflammatory and regulatory cytokine production associ-ated with innate and adaptive immune responses in children with autism spec-trum disorders and developmental regression. J Neuroimmunol 2001;120(1–2):170–9.

Kamel F, Hoppin JA. Association of pesticide exposure with neurologic dysfunction anddisease. Environ Health Perspect 2004;112(9):950–8.

Kastin AJ, Akerstrom V, Pan W. Circulating TGF-beta1 does not cross the intactblood-brain barrier. J Mol Neurosci 2003;21(1):43–8.

Katila H, Hanninen K, Hurme M. Polymorphisms of the interleukin-1 gene complex inschizophrenia. Mol Psychiatry 1999;4(2):179–81.

Kempuraj D, Asadi S, Zhang B, Manola A, Hogan J, Peterson E, et al. Mercury induces in-flammatory mediator release from humanmast cells. J Neuroinflammation 2010;7:20.

Kenet T, Froemke RC, Schreiner CE, Pessah IN, Merzenich MM. Perinatal exposure to anoncoplanar polychlorinated biphenyl alters tonotopy, receptive fields, and plas-ticity in rat primary auditory cortex. Proc Natl Acad Sci U S A 2007;104(18):7646–51.

Kern JK, Grannemann BD, Trivedi MH, Adams JB. Sulfhydryl-reactive metals in autism. JToxicol Environ Health A 2007;70(8):715–21.

Kim KH, Pessah IN. Perinatal exposure to environmental polychlorinated biphenylssensitizes hippocampus to excitotoxicity ex vivo. Neurotoxicology 2011;32(6):981–5.

Kim IJ, Beck HN, Lein PJ, Higgins D. Interferon gamma induces retrograde dendritic re-traction and inhibits synapse formation. J Neurosci 2002a;22(11):4530–9.

Kim SH, Johnson VJ, Sharma RP. Mercury inhibits nitric oxide production but activatesproinflammatory cytokine expression in murine macrophage: differential modula-tion of NF-kappaB and p38 MAPK signaling pathways. Nitric Oxide 2002b;7(1):67–74.

Kim KH, Inan SY, Berman RF, Pessah IN. Excitatory and inhibitory synaptic transmissionis differentially influenced by two ortho-substituted polychlorinated biphenyls inthe hippocampal slice preparation. Toxicol Appl Pharmacol 2009;237(2):168–77.

Kim KH, Bose DD, Ghogha A, Riehl J, Zhang R, Barnhart CD, et al. Para- andortho-substitutions are key determinants of polybrominated diphenyl ether activ-ity toward ryanodine receptors and neurotoxicity. Environ Health Perspect2011;119(4):519–26.

Kingsley DM. The TGF-beta superfamily: new members, new receptors, and new ge-netic tests of function in different organisms. Genes Dev 1994;8(2):133–46.

Kiyota T, Okuyama S, Swan RJ, Jacobsen MT, Gendelman HE, Ikezu T. CNS expression ofanti-inflammatory cytokine interleukin-4 attenuates Alzheimer's disease-likepathogenesis in APP+PS1 bigenic mice. FASEB J 2010;24(8):3093–102.

Kodavanti PR, Coburn CG, Moser VC, MacPhail RC, Fenton SE, Stoker TE, et al. Develop-mental exposure to a commercial PBDE mixture, DE-71: neurobehavioral, hormon-al, and reproductive effects. Toxicol Sci 2010;116(1):297–312.

Kono DH, Balomenos D, Pearson DL, Park MS, Hildebrandt B, Hultman P, et al. The pro-totypic Th2 autoimmunity induced by mercury is dependent on IFN-gamma andnot Th1/Th2 imbalance. J Immunol 1998;161(1):234–40.

Kronborg CS, Gjedsted J, Vittinghus E, Hansen TK, Allen J, Knudsen UB. Longitudinalmeasurement of cytokines in pre-eclamptic and normotensive pregnancies. ActaObstet Gynecol Scand 2011;90(7):791–6.

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulatioment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

Krupinski J, Kumar P, Kumar S, Kaluza J. Increased expression of TGF-beta 1 in brain tis-sue after ischemic stroke in humans. Stroke 1996;27(5):852–7.

Kumar V, Sercarz E. Induction or protection from experimental autoimmune encepha-lomyelitis depends on the cytokine secretion profile of TCR peptide-specific regu-latory CD4 T cells. J Immunol 1998;161(12):6585–91.

Kuperman DA, Schleimer RP. Interleukin-4, interleukin-13, signal transducer and acti-vator of transcription factor 6, and allergic asthma. Curr Mol Med 2008;8(5):384–92.

Kuratsune M, Yoshimura T, Matsuzaka J, Yamaguchi A. Yusho, a poisoning caused byrice oil contaminated with polychlorinated biphenyls. HSMHA Health Rep1971;86(12):1083–91.

Kuriyama SN, Wanner A, Fidalgo-Neto AA, Talsness CE, Koerner W, Chahoud I. Devel-opmental exposure to low-dose PBDE-99: tissue distribution and thyroid hormonelevels. Toxicology 2007;242(1–3):80–90.

Kushima Y, Hama T, Hatanaka H. Interleukin-6 as a neurotrophic factor for promotingthe survival of cultured catecholaminergic neurons in a chemically defined medi-um from fetal and postnatal rat midbrains. Neurosci Res 1992;13(4):267–80.

Labrousse VF, Costes L, Aubert A, Darnaudery M, Ferreira G, Amedee T, et al. Impairedinterleukin-1beta and c-Fos expression in the hippocampus is associated with aspatial memory deficit in P2X(7) receptor-deficient mice. PLoS One 2009;4(6):e6006.

Lahat N, Shapiro S, Froom P, Kristal-Boneh E, Inspector M, Miller A. Inorganic lead en-hances cytokine-induced elevation of matrix metalloproteinase MMP-9 expressionin glial cells. J Neuroimmunol 2002;132(1–2):123–8.

Langeveld WT, Meijer M, Westerink RH. Differential effects of 20 non-dioxin-like PCBson basal and depolarisation-evoked intracellular calcium levels in PC12 cells.Toxicol Sci 2012;126(2):487–96.

Laumonnier F, Bonnet-Brilhault F, Gomot M, Blanc R, David A, Moizard MP, et al.X-linked mental retardation and autism are associated with a mutation in theNLGN4 gene, a member of the neuroligin family. Am J Hum Genet 2004;74(3):552–7.

Lee LC, Zachary AA, Leffell MS, Newschaffer CJ, Matteson KJ, Tyler JD, et al. HLA-DR4 infamilies with autism. Pediatr Neurol 2006;35(5):303–7.

Leijs MM, Koppe JG, Olie K, van Aalderen WM, de Voogt P, ten Tusscher GW. Effects ofdioxins, PCBs, and PBDEs on immunology and hematology in adolescents. EnvironSci Technol 2009;43(20):7946–51.

Lein PJ, Yang D, Bachstetter AD, Tilson HA, Harry GJ, Mervis RF, et al. Ontogenetic alter-ations in molecular and structural correlates of dendritic growth after develop-mental exposure to polychlorinated biphenyls. Environ Health Perspect2007;115(4):556–63.

Leipzig ND, Xu C, Zahir T, Shoichet MS. Functional immobilization of interferon-gammainduces neuronal differentiation of neural stem cells. J Biomed Mater Res A2010;93(2):625–33.

Lema SC, Dickey JT, Schultz IR, Swanson P. Dietary exposure to2,2',4,4'-tetrabromodiphenyl ether (PBDE-47) alters thyroid status and thyroidhormone-regulated gene transcription in the pituitary and brain. Environ HealthPerspect 2008;116(12):1694–9.

Levin ED, Addy N, Nakajima A, Christopher NC, Seidler FJ, Slotkin TA. Persistent behav-ioral consequences of neonatal chlorpyrifos exposure in rats. Brain Res Dev BrainRes 2001;130(1):83–9.

Levin ED, Timofeeva OA, Yang L, Petro A, Ryde IT, Wrench N, et al. Early postnatal para-thion exposure in rats causes sex-selective cognitive impairment and neurotrans-mitter defects which emerge in aging. Behav Brain Res 2010;208(2):319–27.

Li Q. Newmechanism of organophosphorus pesticide-induced immunotoxicity. J NihonMed Sch 2007;74(2):92-105.

Li AJ, Katafuchi T, Oda S, Hori T, Oomura Y. Interleukin-6 inhibits long-term potentia-tion in rat hippocampal slices. Brain Res 1997;748(1–2):30–8.

Li X, Chauhan A, Sheikh AM, Patil S, Chauhan V, Li XM, et al. Elevated immune responsein the brain of autistic patients. J Neuroimmunol 2009;207(1–2):111–6.

Li L, Walker TL, Zhang Y, Mackay EW, Bartlett PF. Endogenous interferon gamma direct-ly regulates neural precursors in the non-inflammatory brain. J Neurosci2010;30(27):9038–50.

Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR, Grigorieva EF, et al. Exoge-nous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptoractivation. Cell 2011;147(3):629–40.

Libbey JE, Sweeten TL, McMahon WM, Fujinami RS. Autistic disorder and viral infec-tions. J Neurovirol 2005;11(1):1-10.

Limke TL, Otero-Montanez JK, Atchison WD. Evidence for interactions between intra-cellular calcium stores during methylmercury-induced intracellular calciumdysregulation in rat cerebellar granule neurons. J Pharmacol Exp Ther2003;304(3):949–58.

Loddick SA, Rothwell NJ. Neuroprotective effects of human recombinant interleukin-1receptor antagonist in focal cerebral ischaemia in the rat. J Cereb Blood FlowMetab1996;16(5):932–40.

Lord C, Rutter M, Le Couteur A. Autism Diagnostic Interview-Revised: a revised versionof a diagnostic interview for caregivers of individuals with possible pervasive de-velopmental disorders. J Autism Dev Disord 1994;24(5):659–85.

Lundgren M, Darnerud PO, Blomberg J, Friman G, Ilback NG. Polybrominated diphenylether exposure suppresses cytokines important in the defence to coxsackievirus B3infection in mice. Toxicol Lett 2009;184(2):107–13.

Malik M, Sheikh AM, Wen G, Spivack W, Brown WT, Li X. Expression of inflammatorycytokines, Bcl2 and cathepsin D are altered in lymphoblasts of autistic subjects.Immunobiology 2011;216(1–2):80–5.

Malkova NV, Yu CZ, Hsiao EY, Moore MJ, Patterson PH. Maternal immune activationyields offspring displaying mouse versions of the three core symptoms of autism.Brain Behav Immun 2012;26(4):607–16.

n in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 13: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

13PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

Mangano EN, Litteljohn D, So R, Nelson E, Peters S, Bethune C, et al. Interferon-gammaplays a role in paraquat-induced neurodegeneration involving oxidative andproinflammatory pathways. Neurobiol Aging 2011;33(7):1411–26.

Mantel PY, Schmidt-Weber CB. Transforming growth factor-beta: recent advances onits role in immune tolerance. Methods Mol Biol 2011;677:303–38.

Markowitz PI. Autism in a child with congenital cytomegalovirus infection. J AutismDev Disord 1983;13(3):249–53.

Martini F, Fernandez C, Segundo LS, Tarazona JV, Pablos MV. Assessment of potentialimmunotoxic effects caused by cypermethrin, fluoxetine, and thiabendazoleusing heat shock protein 70 and interleukin-1beta mRNA expression in the anuranXenopus laevis. Environ Toxicol Chem 2010;29(11):2536–43.

Martins TB, Rose JW, Jaskowski TD, Wilson AR, Husebye D, Seraj HS, et al. Analysis ofproinflammatory and anti-inflammatory cytokine serum concentrations in pa-tients with multiple sclerosis by using a multiplexed immunoassay. Am J ClinPathol 2011;136(5):696–704.

Mason JL, Suzuki K, Chaplin DD, Matsushima GK. Interleukin-1beta promotes repair ofthe CNS. J Neurosci 2001;21(18):7046–52.

McAfoose J, Baune BT. Evidence for a cytokine model of cognitive function. NeurosciBiobehav Rev 2009;33(3):355–66.

Mense SM, Sengupta A, Lan C, Zhou M, Bentsman G, Volsky DJ, et al. The common in-secticides cyfluthrin and chlorpyrifos alter the expression of a subset of geneswith diverse functions in primary human astrocytes. Toxicol Sci 2006;93(1):125–35.

Messer A. Mini-review: polybrominated diphenyl ether (PBDE) flame retardants as po-tential autism risk factors. Physiol Behav 2010;100(3):245–9.

Meyer U, Feldon J, Dammann O. Schizophrenia and autism: both shared anddisorder-specific pathogenesis via perinatal inflammation? Pediatr Res2011;69(5 Pt 2):26R–33R.

Miller L, Reynolds J. Autism and vaccination-the current evidence. J Spec Pediatr Nurs2009;14(3):166–72.

Mishra KP. Lead exposure and its impact on immune system: a review. Toxicol In Vitro2009;23(6):969–72.

Mitchell KA, Elferink CJ. Timing is everything: consequences of transient and sustainedAhR activity. Biochem Pharmacol 2009;77(6):947–56.

Mizuno T, Zhang G, Takeuchi H, Kawanokuchi J, Wang J, Sonobe Y, et al.Interferon-gamma directly induces neurotoxicity through a neuron specific,calcium-permeable complex of IFN-gamma receptor and AMPA GluR1 receptor.FASEB J 2008;22(6):1797–806.

Molloy CA, Morrow AL, Meinzen-Derr J, Schleifer K, Dienger K, Manning-Courtney P,et al. Elevated cytokine levels in children with autism spectrum disorder. JNeuroimmunol 2006;172(1–2):198–205.

Monteleone I, Rizzo A, Sarra M, Sica G, Sileri P, Biancone L, et al. Aryl hydrocarbonreceptor-induced signals up-regulate IL-22 production and inhibit inflammationin the gastrointestinal tract. Gastroenterology 2011;141(1):237–48.

Moon LD, Fawcett JW. Reduction in CNS scar formation without concomitant increasein axon regeneration following treatment of adult rat brain with a combination ofantibodies to TGFbeta1 and beta2. Eur J Neurosci 2001;14(10):1667–77.

Morgan JT, Chana G, Pardo CA, Achim C, Semendeferi K, Buckwalter J, et al. Microglialactivation and increased microglial density observed in the dorsolateral prefrontalcortex in autism. Biol Psychiatry 2010;68(4):368–76.

Morse DC, Groen D, Veerman M, van Amerongen CJ, Koeter HB, Smits van Prooije AE,et al. Interference of polychlorinated biphenyls in hepatic and brain thyroid hor-mone metabolism in fetal and neonatal rats. Toxicol Appl Pharmacol1993;122(1):27–33.

Murphy SP, Tayade C, Ashkar AA, Hatta K, Zhang J, Croy BA. Interferon gamma in suc-cessful pregnancies. Biol Reprod 2009;80(5):848–59.

Nagayama J, Tsuji H, Iida T, Nakagawa R, Matsueda T, Hirakawa H, et al. Immunologiceffects of perinatal exposure to dioxins, PCBs and organochlorine pesticides in Jap-anese infants. Chemosphere 2007;67(9):S393–8.

Narayanan PK, Carter WO, Ganey PE, Roth RA, Voytik-Harbin SL, Robinson JP. Impair-ment of human neutrophil oxidative burst by polychlorinated biphenyls: inhibi-tion of superoxide dismutase activity. J Leukoc Biol 1998;63(2):216–24.

Ndebele K, Tchounwou PB, McMurray RW. Coumestrol, bisphenol-A, DDT, and TCDDmodulation of interleukin-2 expression in activated CD+4 Jurkat T cells. Int J Envi-ron Res Public Health 2004;1(1):3-11.

Nelms K, Keegan AD, Zamorano J, Ryan JJ, Paul WE. The IL-4 receptor: signaling mech-anisms and biologic functions. Annu Rev Immunol 1999;17:701–38.

Nolan Y, Maher FO, Martin DS, Clarke RM, Brady MT, Bolton AE, et al. Role of interleu-kin-4 in regulation of age-related inflammatory changes in the hippocampus. J BiolChem 2005;280(10):9354–62.

Nolting T, Lindecke A, Koutsilieri E, Maschke M, Husstedt IW, Sopper S, et al. Measure-ment of soluble inflammatory mediators in cerebrospinal fluid of human immuno-deficiency virus-positive patients at distinct stages of infection by solid-phaseprotein array. J Neurovirol 2009;15(5–6):390–400.

Noren K, Meironyte D. Certain organochlorine and organobromine contaminants inSwedish human milk in perspective of past 20-30 years. Chemosphere2000;40(9–11):1111–23.

Okada K, Hashimoto K, Iwata Y, Nakamura K, Tsujii M, Tsuchiya KJ, et al. Decreasedserum levels of transforming growth factor-beta1 in patients with autism. ProgNeuropsychopharmacol Biol Psychiatry 2007;31(1):187–90.

Oliphant CJ, Barlow JL, McKenzie AN. Insights into the initiation of type 2 immune re-sponses. Immunology 2011;134(4):378–85.

O'Neill LA. The interleukin-1 receptor/Toll-like receptor superfamily: signal transduc-tion during inflammation and host defense. Sci STKE 2000;2000(44):re1.

Onore C, Careaga M, Ashwood P. The role of immune dysfunction in the pathophysiol-ogy of autism. Brain Behav Immun 2011;26(3):383–92.

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulationment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

Orlova KA, Crino PB. The tuberous sclerosis complex. Ann N Y Acad Sci 2010;1184:87-105.

Owens SE, Summar ML, Ryckman KK, Haines JL, Reiss S, Summar SR, et al. Lack of asso-ciation between autism and four heavy metal regulatory genes. Neurotoxicology2011;32(6):769–75.

Pan W, Banks WA, Kastin AJ. Permeability of the blood-brain and blood-spinal cordbarriers to interferons. J Neuroimmunol 1997;76(1–2):105–11.

Pancetti F, Olmos C, Dagnino-Subiabre A, Rozas C, Morales B. Noncholinesterase effectsinduced by organophosphate pesticides and their relationship to cognitive pro-cesses: implication for the action of acylpeptide hydrolase. J Toxicol Environ HealthB Crit Rev 2007;10(8):623–30.

Pasca SP, Nemes B, Vlase L, Gagyi CE, Dronca E, Miu AC, et al. High levels of homocys-teine and low serum paraoxonase 1 arylesterase activity in children with autism.Life Sci 2006;78(19):2244–8.

Patterson PH. Immune involvement in schizophrenia and autism: etiology, pathologyand animal models. Behav Brain Res 2009;204(2):313–21.

Persico AM, D'Agruma L, Maiorano N, Totaro A, Militerni R, Bravaccio C, et al. Reelingene alleles and haplotypes as a factor predisposing to autistic disorder. Mol Psy-chiatry 2001;6(2):150–9.

Pessah I, Lein PJ. Chapter 19: Evidence for Environmental Susceptivility in Autism.What we know what we need to know about gene x environment interactions.In: Zimmerman AW, editor. Autism: Current Theories and Evidence. HumanaPress; 2008.

Pessah IN, Lehmler HJ, Robertson LW, Perez CF, Cabrales E, Bose DD, et al. Enantiomericspecificity of (-)-2,2',3,3',6,6'-hexachlorobiphenyl toward ryanodine receptortypes 1 and 2. Chem Res Toxicol 2009;22(1):201–7.

Pessah IN, Cherednichenko G, Lein PJ. Minding the calcium store: Ryanodine receptoractivation as a convergent mechanism of PCB toxicity. Pharmacol Ther2010;125(2):260–85.

Piton A, Michaud JL, Peng H, Aradhya S, Gauthier J, Mottron L, et al. Mutations in thecalcium-related gene IL1RAPL1 are associated with autism. Hum Mol Genet2008;17(24):3965–74.

Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. NatRev Immunol 2005;5(5):375–86.

Price CS, Thompson WW, Goodson B, Weintraub ES, Croen LA, Hinrichsen VL, et al. Pre-natal and infant exposure to thimerosal from vaccines and immunoglobulins andrisk of autism. Pediatrics 2010;126(4):656–64.

Rajakumar A, Chu T, Handley DE, Bunce KD, Burke B, Hubel CA, et al. Maternal gene ex-pression profiling during pregnancy and preeclampsia in human peripheral bloodmononuclear cells. Placenta 2011;32(1):70–8.

Reed A, Dzon L, Loganathan BG, Whalen MM. Immunomodulation of human naturalkiller cell cytotoxic function by organochlorine pesticides. Hum Exp Toxicol2004;23(10):463–71.

Reissmann E, Ernsberger U, Francis-West PH, Rueger D, Brickell PM, Rohrer H. Involve-ment of bone morphogenetic protein-4 and bone morphogenetic protein-7 in thedifferentiation of the adrenergic phenotype in developing sympathetic neurons.Development 1996;122(7):2079–88.

Rivest S. Interactions between the immune and neuroendocrine systems. Prog BrainRes 2010;181:43–53.

Roberts EM, English PB, Grether JK, Windham GC, Somberg L, Wolff C. Maternal resi-dence near agricultural pesticide applications and autism spectrum disordersamong children in the California Central Valley. Environ Health Perspect2007;115(10):1482–9.

Roeyen G, Chapelle T, Jorens P, de Beeck BO, Ysebaert D. Necrotizing pancreatitis due topoisoning with organophosphate pesticides. Acta Gastroenterol Belg 2008;71(1):27–9.

Rogan WJ, Gladen BC, Hung KL, Koong SL, Shih LY, Taylor JS, et al. Congenital poisoningby polychlorinated biphenyls and their contaminants in Taiwan. Science1988;241(4863):334–6.

Romero E, Guaza C, Castellano B, Borrell J. Ontogeny of sensorimotor gating and im-mune impairment induced by prenatal immune challenge in rats: implicationsfor the etiopathology of schizophrenia. Mol Psychiatry 2010;15(4):372–83.

Ronald A, Hoekstra RA. Autism spectrum disorders and autistic traits: A decade ofnew twin studies. Am J Med Genet B Neuropsychiatr Genet 2011;156B(3):255–74.

Rose S, Melnyk S, Savenka A, Hubanks A, Jernigan S, Cleves M, et al. The Frequency ofPolymorphisms affecting Lead and Mercury Toxicity among Children with Autism.Am J Biochem Biotechnol 2008;4(2):84–95.

Ross FM, Allan SM, Rothwell NJ, Verkhratsky A. A dual role for interleukin-1 in LTP inmouse hippocampal slices. J Neuroimmunol 2003;144(1–2):61–7.

Rowley B, Monestier M. Mechanisms of heavy metal-induced autoimmunity. MolImmunol 2005;42(7):833–8.

Roze E, Meijer L, Bakker A, Van Braeckel KN, Sauer PJ, Bos AF. Prenatal exposure toorganohalogens, including brominated flame retardants, influences motor, cogni-tive, and behavioral performance at school age. Environ Health Perspect2009;117(12):1953–8.

Rubenstein JL, Merzenich MM. Model of autism: increased ratio of excitation/inhibitionin key neural systems. Genes Brain Behav 2003;2(5):255–67.

Ruocco A, Nicole O, Docagne F, Ali C, Chazalviel L, Komesli S, et al. A transforminggrowth factor-beta antagonist unmasks the neuroprotective role of this endoge-nous cytokine in excitotoxic and ischemic brain injury. J Cereb Blood Flow Metab1999;19(12):1345–53.

Sakamoto M, Ikegami N, Nakano A. Protective effects of Ca2+ channel blockers againstmethyl mercury toxicity. Pharmacol Toxicol 1996;78(3):193–9.

Samuelsson AM, Jennische E, Hansson HA, Holmang A. Prenatal exposure to interleu-kin-6 results in inflammatory neurodegeneration in hippocampus with

in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 14: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

14 PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

NMDA/GABA(A) dysregulation and impaired spatial learning. Am J Physiol RegulIntegr Comp Physiol 2006;290(5):R1345–56.

Savignac M, Mellstrom B, Naranjo JR. Calcium-dependent transcription of cytokinegenes in T lymphocytes. Pflugers Arch 2007;454(4):523–33.

Schreiber T, Gassmann K, Gotz C, Hubenthal U, Moors M, Krause G, et al.Polybrominated diphenyl ethers induce developmental neurotoxicity in a humanin vitro model: evidence for endocrine disruption. Environ Health Perspect2010;118(4):572–8.

Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals,mechanisms and functions. J Leukoc Biol 2004;75(2):163–89.

Schug TT, Janesick A, Blumberg B, Heindel JJ. Endocrine disrupting chemicals and dis-ease susceptibility. J Steroid Biochem Mol Biol 2011;127(3–5):204–15.

Schumann CM, Amaral DG. Stereological analysis of amygdala neuron number in au-tism. J Neurosci 2006;26(29):7674–9.

Selevan SG, Rice DC, Hogan KA, Euling SY, Pfahles-Hutchens A, Bethel J. Blood lead con-centration and delayed puberty in girls. N Engl J Med 2003;348(16):1527–36.

Serajee FJ, Nabi R, Zhong H, Huq M. Polymorphisms in xenobiotic metabolism genesand autism. J Child Neurol 2004;19(6):413–7.

Seth V, Ahmad RS, Suke SG, Pasha ST, Bhattacharya A, Banerjee BD. Lindane-inducedimmunological alterations in human poisoning cases. Clin Biochem 2005;38(7):678–80.

Shafer TJ, Meyer DA, Crofton KM. Developmental neurotoxicity of pyrethroid insecti-cides: critical review and future research needs. Environ Health Perspect2005;113(2):123–36.

Shannon M, Graef JW. Lead intoxication in children with pervasive developmental dis-orders. J Toxicol Clin Toxicol 1996;34(2):177–81.

Shatz CJ. MHC class I: an unexpected role in neuronal plasticity. Neuron 2009;64(1):40–5.

Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucle-us. Cell 2003;113(6):685–700.

Shi L, Fatemi SH, Sidwell RW, Patterson PH. Maternal influenza infection causes markedbehavioral and pharmacological changes in the offspring. J Neurosci 2003;23(1):297–302.

Shin KJ, Bae SS, Hwang YA, Seo JK, Ryu SH, Suh PG. 2,2',4,6,6'-pentachlorobiphenyl in-duces apoptosis in human monocytic cells. Toxicol Appl Pharmacol 2000;169(1):1–7.

Sholl-Franco A, Marques PM, Ferreira CM, de Araujo EG. IL-4 increases GABAergic phe-notype in rat retinal cell cultures: involvement of muscarinic receptors and proteinkinase C. J Neuroimmunol 2002;133(1–2):20–9.

Sholl-Franco A, da Silva AG, Adao-Novaes J. Interleukin-4 as a neuromodulatory cyto-kine: roles and signaling in the nervous system. Ann N Y Acad Sci 2009;1153:65–75.

Singh VK. Plasma increase of interleukin-12 and interferon-gamma. Pathological signif-icance in autism. J Neuroimmunol 1996;66(1–2):143–5.

Skurlova M, Stofkova A, Jurcovicova J. Exogenous IL-1beta induces its own expression,but not that of IL-6 in the hypothalamus and activates HPA axis and prolactin re-lease. Endocr Regul 2006;40(4):125–8.

Smith SE, Li J, Garbett K, Mirnics K, Patterson PH. Maternal immune activation altersfetal brain development through interleukin-6. J Neurosci 2007;27(40):10695–702.

Soderlund DM. Molecular mechanisms of pyrethroid insecticide neurotoxicity: recentadvances. Arch Toxicol 2012;86(2):165–81.

Song JH, Wang CX, Song DK, Wang P, Shuaib A, Hao C. Interferon gamma inducesneurite outgrowth by up-regulation of p35 neuron-specific cyclin-dependent ki-nase 5 activator via activation of ERK1/2 pathway. J Biol Chem 2005;280(13):12896–901.

Spulber S, Oprica M, Bartfai T, Winblad B, Schultzberg M. Blunted neurogenesis andgliosis due to transgenic overexpression of human soluble IL-1ra in the mouse.Eur J Neurosci 2008;27(3):549–58.

Spulber S, Bartfai T, Winblad B, Schultzberg M. Morphological and behavioral changesinduced by transgenic overexpression of interleukin-1ra in the brain. J NeurosciRes 2010;89(2):142–52.

Spulber S, Bartfai T, Winblad B, Schultzberg M. Morphological and behavioral changesinduced by transgenic overexpression of interleukin-1ra in the brain. J NeurosciRes 2011;89(2):142–52.

Stamova B, Green PG, Tian Y, Hertz-Picciotto I, Pessah IN, Hansen R, et al. Correlationsbetween gene expression and mercury levels in blood of boys with and withoutautism. Neurotox Res 2011;19(1):31–48.

State MW, Levitt P. The conundrums of understanding genetic risks for autism spec-trum disorders. Nat Neurosci 2011;14(12):1499–506.

Steuerwald U, Weihe P, Jorgensen PJ, Bjerve K, Brock J, Heinzow B, et al. Maternal sea-food diet, methylmercury exposure, and neonatal neurologic function. J Pediatr2000;136(5):599–605.

Stevens HE, Smith KM, Rash BG, Vaccarino FM. Neural stem cell regulation, fibroblastgrowth factors, and the developmental origins of neuropsychiatric disorders.Front Neurosci 2010;4.

Stoker TE, Cooper RL, Lambright CS, Wilson VS, Furr J, Gray LE. In vivo and in vitroanti-androgenic effects of DE-71, a commercial polybrominated diphenyl ether(PBDE) mixture. Toxicol Appl Pharmacol 2005;207(1):78–88.

Strijbos PJ, Rothwell NJ. Interleukin-1 beta attenuates excitatory amino acid-inducedneurodegeneration in vitro: involvement of nerve growth factor. J Neurosci1995;15(5 Pt 1):3468–74.

Suzuki K, Matsuzaki H, Iwata K, Kameno Y, Shimmura C, Kawai S, et al. Plasma cytokineprofiles in subjects with high-functioning autism spectrum disorders. PLoS One2011;6(5):e20470.

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulatioment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

Svensson I, Waara L, Johansson L, Bucht A, Cassel G. Soman-induced interleukin-1 betamRNA and protein in rat brain. Neurotoxicology 2001;22(3):355–62.

Sweeten TL, Posey DJ, McDougle CJ. Brief report: autistic disorder in three children withcytomegalovirus infection. J Autism Dev Disord 2004a;34(5):583–6.

Sweeten TL, Posey DJ, Shankar S, McDougle CJ. High nitric oxide production in autisticdisorder: a possible role for interferon-gamma. Biol Psychiatry 2004b;55(4):434–7.

Tabares-Seisdedos R, Rubenstein JL. Chromosome 8p as a potential hub for develop-mental neuropsychiatric disorders: implications for schizophrenia, autism andcancer. Mol Psychiatry 2009;14(6):563–89.

Tan Y, Li D, Song R, Lawrence D, Carpenter DO. Ortho-substituted PCBs kill thymocytes.Toxicol Sci 2003;76(2):328–37.

Thuvander A, Darnerud PO. Effects of polybrominated diphenyl ether (PBDE) andpolychlorinated biphenyl (PCB) on some immunological parameters after oral ex-posure in rats and mice. Toxicol Environ Chem 1999;70:229–42.

Tian Y, Green PG, Stamova B, Hertz-Picciotto I, Pessah IN, Hansen R, et al. Correlationsof gene expression with blood lead levels in children with autism compared to typ-ically developing controls. Neurotox Res 2011;19(1):1-13.

Torre D, Broggini M, Zeroli C, Agrifoglio L, Botta V, Casalone R, et al. Serum levels ofgamma interferon in patients with Down's syndrome. Infection 1995;23(1):66–7.

Torres AR, Maciulis A, Stubbs EG, Cutler A, Odell D. The transmission disequilibriumtest suggests that HLA-DR4 and DR13 are linked to autism spectrum disorder.Hum Immunol 2002;63(4):311–6.

Torres AR, Sweeten TL, Cutler A, Bedke BJ, Fillmore M, Stubbs EG, et al. The associationand linkage of the HLA-A2 class I allele with autism. Hum Immunol 2006;67(4–5):346–51.

Toscano CD, Guilarte TR. Lead neurotoxicity: from exposure to molecular effects. BrainRes Brain Res Rev 2005;49(3):529–54.

Traugott U, Lebon P. Interferon-gamma and Ia antigen are present on astrocytes in ac-tive chronic multiple sclerosis lesions. J Neurol Sci 1988;84(2–3):257–64.

Treves S, Vukcevic M, Jeannet PY, Levano S, Girard T, Urwyler A, et al. Enhancedexcitation-coupled Ca(2+) entry induces nuclear translocation of NFAT and con-tributes to IL-6 release from myotubes from patients with central core disease.Hum Mol Genet 2011;20(3):589–600.

Tropepe V, Hitoshi S, Sirard C, Mak TW, Rossant J, van der Kooy D. Direct neural fatespecification from embryonic stem cells: a primitive mammalian neural stem cellstage acquired through a default mechanism. Neuron 2001;30(1):65–78.

Tsukimori K, Tokunaga S, Shibata S, Uchi H, Nakayama D, Ishimaru T, et al. Long-termeffects of polychlorinated biphenyls and dioxins on pregnancy outcomes inwomen affected by the Yusho incident. Environ Health Perspect 2008;116(5):626–30.

Valentino M, Rapisarda V, Santarelli L, Bracci M, Scorcelletti M, Di Lorenzo L, et al. Effectof lead on the levels of some immunoregulatory cytokines in occupationally ex-posed workers. Hum Exp Toxicol 2007;26(7):551–6.

Van Birgelen AP, Smit EA, Kampen IM, Groeneveld CN, Fase KM, Van der Kolk J, et al.Subchronic effects of 2,3,7,8-TCDD or PCBs on thyroid hormone metabolism: usein risk assessment. Eur J Pharmacol 1995;293(1):77–85.

Varga EA, Pastore M, Prior T, Herman GE, McBride KL. The prevalence of PTEN muta-tions in a clinical pediatric cohort with autism spectrum disorders, developmentaldelay, and macrocephaly. Genet Med 2009;11(2):111–7.

Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA. Neuroglial activationand neuroinflammation in the brain of patients with autism. Ann Neurol2005;57(1):67–81.

Vashlishan AB, Madison JM, Dybbs M, Bai J, Sieburth D, Ch'ng Q, et al. An RNAi screenidentifies genes that regulate GABA synapses. Neuron 2008;58(3):346–61.

Vereyken EJ, Bajova H, Chow S, de Graan PN, Gruol DL. Chronic interleukin-6 alters thelevel of synaptic proteins in hippocampus in culture and in vivo. Eur J Neurosci2007;25(12):3605–16.

Victorio SC, Havton LA, Oliveira AL. Absence of IFNgamma expression induces neuronaldegeneration in the spinal cord of adult mice. J Neuroinflammation 2010;7:77.

Vig M, Kinet JP. Calcium signaling in immune cells. Nat Immunol 2009;10(1):21–7.Vikman KS, Owe-Larsson B, Brask J, Kristensson KS, Hill RH. Interferon-gamma-induced

changes in synaptic activity and AMPA receptor clustering in hippocampal cul-tures. Brain Res 2001;896(1–2):18–29.

Voineagu I, Wang X, Johnston P, Lowe JK, Tian Y, Horvath S, et al. Transcriptomic anal-ysis of autistic brain reveals convergent molecular pathology. Nature2011;474(7351):380–4.

Vuillermot S, Weber L, Feldon J, Meyer U. A longitudinal examination of theneurodevelopmental impact of prenatal immune activation in mice reveals prima-ry defects in dopaminergic development relevant to schizophrenia. J Neurosci2010;30(4):1270–87.

Walter J, Honsek SD, Illes S, Wellen JM, Hartung HP, Rose CR, et al. A new role for inter-feron gamma in neural stem/precursor cell dysregulation. Mol Neurodegener2011;6:18.

Ward LD, Howlett GJ, Discolo G, Yasukawa K, Hammacher A, Moritz RL, et al. High af-finity interleukin-6 receptor is a hexameric complex consisting of two moleculeseach of interleukin-6, interleukin-6 receptor, and gp-130. J Biol Chem1994;269(37):23286–9.

Watanabe W, Shimizu T, Sawamura R, Hino A, Konno K, Kurokawa M. Functional disor-der of primary immunity responding to respiratory syncytial virus infection in off-spring mice exposed to a flame retardant, decabrominated diphenyl ether,perinatally. J Med Virol 2010;82(6):1075–82.

Waterman SJ, el-Fawal HA, Snyder CA. Lead alters the immunogenicity of two neuralproteins: a potential mechanism for the progression of lead-induced neurotoxicity.Environ Health Perspect 1994;102(12):1052–6.

n in autism spectrum disorders (ASD): Possible role of the environ-.07.006

Page 15: Neurotoxicology and Teratology - VaccinePapers.orgvaccinepapers.org/wp-content/uploads/Cytokine-dysregulation-in... · key facilitators of cross-systemic communication. Cytokines

15PE Goines, P. Ashwood / Neurotoxicology and Teratology xxx (2012) xxx–xxx

Wei H, Zou H, Sheikh AM, Malik M, Dobkin C, Brown WT, et al. IL-6 is increased in thecerebellum of autistic brain and alters neural cell adhesion, migration and synapticformation. J Neuroinflammation 2011;8:52.

Wens B, De Boever P, Maes M, Hollanders K, Schoeters G. Transcriptomics identifies dif-ferences between ultrapure non-dioxin-like polychlorinated biphenyls (PCBs) anddioxin-like PCB126 in cultured peripheral blood mononuclear cells. Toxicology2011;287(1–3):113–23.

Williams AJ, Berti R, Yao C, Price RA, Velarde LC, Koplovitz I, et al. Centralneuro-inflammatory gene response following soman exposure in the rat. NeurosciLett 2003;349(3):147–50.

Winneke G. Developmental aspects of environmental neurotoxicology: lessons fromlead and polychlorinated biphenyls. J Neurol Sci 2011;308(1–2):9-15.

Wolansky MJ, Harrill JA. Neurobehavioral toxicology of pyrethroid insecticides in adultanimals: a critical review. Neurotoxicol Teratol 2008;30(2):55–78.

Wong G, Goldshmit Y, Turnley AM. Interferon-gamma but not TNF alpha promotesneuronal differentiation and neurite outgrowth of murine adult neural stemcells. Exp Neurol 2004;187(1):171–7.

Woods R, Vallero RO, Golub M, Suarez JK, Ta TA, Yasui DH, et al. Long-lived epigeneticinteractions between perinatal PBDE exposure and Mecp2308 mutation. Hum MolGenet 2012;21(11):2399–411.

Wright B, Pearce H, Allgar V, Miles J, Whitton C, Leon I, et al. A Comparison of UrinaryMercury between Children with Autism Spectrum Disorders and Control Children.PLoS One 2012;7(2):e29547.

Wu Z, Turner DR, Oliveira DB. IL-4 gene expression up-regulated by mercury in ratmast cells: a role of oxidant stress in IL-4 transcription. Int Immunol 2001;13(3):297–304.

Wyss-Coray T, Feng L, Masliah E, Ruppe MD, Lee HS, Toggas SM, et al. Increased centralnervous system production of extracellular matrix components and developmentof hydrocephalus in transgenic mice overexpressing transforming growthfactor-beta 1. Am J Pathol 1995;147(1):53–67.

Wyss-Coray T, Borrow P, Brooker MJ, Mucke L. Astroglial overproduction of TGF-beta 1enhances inflammatory central nervous system disease in transgenic mice. JNeuroimmunol 1997;77(1):45–50.

Please cite this article as: Goines PE, Ashwood P, Cytokine dysregulationment, Neurotoxicol Teratol (2012), http://dx.doi.org/10.1016/j.ntt.2012

Yizhar O, Fenno LE, Prigge M, Schneider F, Davidson TJ, O'Shea DJ, et al. Neocorticalexcitation/inhibition balance in information processing and social dysfunction. Na-ture 2011;477(7363):171–8.

Yoshida T, Yasumura M, Uemura T, Lee SJ, Ra M, Taguchi R, et al. IL-1 receptor accessoryprotein-like 1 associated with mental retardation and autism mediates synapseformation by trans-synaptic interaction with protein tyrosine phosphatase delta.J Neurosci 2011;31(38):13485–99.

Yoshimatsu T, Kawaguchi D, Oishi K, Takeda K, Akira S, Masuyama N, et al.Non-cell-autonomous action of STAT3 in maintenance of neural precursor cells inthe mouse neocortex. Development 2006;133(13):2553–63.

You Z, Luo C, ZhangW, Chen Y, He J, Zhao Q, et al. Pro- and anti-inflammatory cytokinesexpression in rat's brain and spleen exposed to chronic mild stress: involvement indepression. Behav Brain Res 2011;225(1):135–41.

Yu L, Hebert MC, Zhang YE. TGF-beta receptor-activated p38 MAP kinase mediatesSmad-independent TGF-beta responses. EMBO J 2002;21(14):3749–59.

Zahir T, Chen YF, MacDonald JF, Leipzig N, Tator CH, Shoichet MS. Neuralstem/progenitor cells differentiate in vitro to neurons by the combined action ofdibutyryl cAMP and interferon-gamma. Stem Cells Dev 2009;18(10):1423–32.

Zaretsky MV, Alexander JM, Byrd W, Bawdon RE. Transfer of inflammatory cytokinesacross the placenta. Obstet Gynecol 2004;103(3):546–50.

Zhang JD, Cousens LS, Barr PJ, Sprang SR. Three-dimensional structure of human basicfibroblast growth factor, a structural homolog of interleukin 1 beta. Proc Natl AcadSci U S A 1991;88(8):3446–50.

Zhang Y, Zhao M, Jin M, Xu C, Wang C, Liu W. Immunotoxicity of pyrethroid metabo-lites in an in vitro model. Environ Toxicol Chem 2010;29(11):2505–10.

Zhou J, Chen DJ, Liao QP, Yu YH. Impact of PBDE-209 exposure during pregnancy andlactation on immune function of offspring rats. Nan Fang Yi Ke Da Xue Xue Bao2006;26(6):738–41.

Ziats MN, Rennert OM. Expression profiling of autism candidate genes during humanbrain development implicates central immune signaling pathways. PLoS One2011;6(9):e24691.

in autism spectrum disorders (ASD): Possible role of the environ-.07.006