Autism Research and Treatment

Autism: Where Genetics Meets the Immune System

Guest Editors: Antonio M. Persico, Judy Van de Water, and Carlos A. Pardo Autism: Where Genetics Meets the Immune System Autism Research and Treatment Autism: Where Genetics Meets the Immune System

Guest Editors: Antonio M. Persico, Judy Van de Water, and Carlos A. Pardo Copyright © 2012 Hindawi Publishing Corporation. All rights reserved.

This is a special issue published in “Autism Research and Treatment.” All articles are open access articles distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Editorial Board

Jean-Louis Adrien, France Mohammad Ghaziuddin, USA Klaus-Peter Ossenkopp, Canada M. Aman, USA Barry Gordon, USA Alan K. Percy, USA Elizabeth Aylward, USA H. Jyonouchi, USA Mikhail V. Pletnikov, USA R. F. Berman, USA Connie Kasari, USA Herbert Roeyers, Belgium Roberto Canitano, Italy Walter E. Kaufmann, USA John A. Sweeney, USA Manuel F. Casanova, USA Bryan King, USA Akio Wakabayashi, UK Jamel Chelly, France N. Kraus, USA Geraldine Dawson, USA Bennett L. Leventhal, USA Valsamma Eapen, Australia M.-Reza Mohammadi, Iran Contents

Autism: Where Genetics Meets the Immune System, Antonio M. Persico, Judy Van de Water, and Carlos A. Pardo Volume 2012, Article ID 486359, 2 pages

Is Autism a Member of a Family of Diseases Resulting from Genetic/Cultural Mismatches? Implications for Treatment and Prevention, Staci D. Bilbo, John P. Jones, and William Parker Volume 2012, Article ID 910946, 11 pages

Prenatal and Postnatal Epigenetic Programming: Implications for GI, Immune, and Neuronal Function in Autism, Mostafa I. Waly, Mady Hornig, Malav Trivedi, Nathaniel Hodgson, Radhika Kini, Akio Ohta, and Richard Deth Volume 2012, Article ID 190930, 13 pages

Decreased Levels of EGF in Plasma of Children with Autism Spectrum Disorder,CharityOnore, Judy Van de Water, and Paul Ashwood Volume 2012, Article ID 205362, 4 pages

HLA Immune Function Genes in Autism, Anthony R. Torres, Jonna B. Westover, and Allen J. Rosenspire Volume 2012, Article ID 959073, 13 pages

Intracellular and Extracellular Redox Status and Free Radical Generation in Primary Immune Cells from Children with Autism, Shannon Rose, Stepan Melnyk, Timothy A. Trusty, Oleksandra Pavliv, Lisa Seidel, Jingyun Li, Todd Nick, and S. Jill James Volume 2012, Article ID 986519, 10 pages

High Complement Factor I Activity in the Plasma of Children with Autism Spectrum Disorders, Naghi Momeni, Lars Brudin, Fatemeh Behnia, Berit Nordstrom,¨ Ali Yosefi-Oudarji, Bengt Sivberg, Mohammad T. Joghataei, and Bengt L. Persson Volume 2012, Article ID 868576, 6 pages Hindawi Publishing Corporation Autism Research and Treatment Volume 2012, Article ID 486359, 2 pages doi:10.1155/2012/486359

Editorial Autism: Where Genetics Meets the Immune System

Antonio M. Persico,1, 2 Judy Van de Water,3 and Carlos A. Pardo4

1 Child and Adolescent Neuropsychiatry Unit, University Campus Bio-Medico, Via Alvaro del Portillo 21, 00128 Rome, Italy 2 Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Via Alvaro del Portillo 21, 00128 Rome, Italy 3 Division of Rheumatology/Allergy and Clinical Immunology, UC Davis, 451 Health Science Drive, Suite 6510, GBSF, Davis, CA 95616, USA 4 Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA

Correspondence should be addressed to Antonio M. Persico, [email protected]

Received 24 July 2012; Accepted 24 July 2012 Copyright © 2012 Antonio M. Persico et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Individuals with autism often display immune abnormalities toward microbial infections) or as a secondary event part in the form of altered cytokine profiles, autoantibodies, and of a broader immunomodulatory response aiming to blunt changesinimmunecellfunction.Aretheymere“smoke” an inflammatory process. F1 plasma levels suggest that this or real “fire”? Given what we now know about the cross- process could play a more relevant role in males and in small talk between the immune and nervous systems, one must ASD children. ask: are these immune abnormalities simply bystander The contribution by S. Rose et al. describes reduced effects of genetic/genomic variants directly responsible for glutathione-mediated redox/antioxidant capacity both inside abnormal neurodevelopment, or is there a sizable subgroup primary leukocytes and in the plasma of ASD children. Con- of patients with autism spectrum disorder (ASD), in which a sistently with this intracellular and extracellular imbalance of dysfunctional immune system plays a critical mediator role the glutathione redox status, intracellular production of free in the pathogenic chain of events leading to the disorder? radicals is also enhanced, especially in 30% of ASD cases. Is autism one of many genetic/genomic conditions, Down This and several previous studies provide converging evi- syndrome representing the best-known example, accompa- dence of excessive oxidative stress likely leading to abnormal nied by immune abnormalities not primarily responsible mitochondrial function in a similar percentage of autistic for changing the trajectory of neurodevelopment? Or could individuals. conceivably an unfortunate combination of genetic/genomic Along similar lines, M. I. Waly et al. direct their attention susceptibility and environmental factors converging onto the on the complex array of pathophysiological consequences same individual affect neurodevelopment through immune- produced by enhanced oxidative stress. In addition to mediated mechanisms, such as fetomaternal incompatibility, imbalanced glutathione redox status, reduced methionine autoimmunity, abnormal neuroinflammation, and so on? synthase activity is particularly interesting, as it results This special issue contains six contributions aimed at in lower availability of S-adenosyl-methionine and con- addressing different aspects of this question. sequently blunted DNA methylation. Curiously, a similar The paper by N. Momeni et al. documents significantly abnormality underlies autistic features also in two entirely elevated plasma levels of factor I among ASD children different contexts, namely, children exposed prenatally to compared to controls. Factor I is a plasma enzyme respon- valproic acid and in Rett syndrome girls, carrying MECP2 sible for degrading complement factor 3b, which in turn mutations. is the major opsonin in the complement system enabling The review by S. D. Bilbo et al. presents a stimulating phagocytosisofmicrobialagents.HigherfactorIlevelscan hypothesis, linking ASD to inflammatory, allergic, and be interpreted as either primary (i.e., conferring vulnerability autoimmune diseases. These “hyperimmune” disorders are 2 Autism Research and Treatment known to share steeply increasing incidence rates over the last decades, as well as significant loading in many families with children with autism. In the authors’ view, they also share an inappropriate activation of the immune system due to biome depletion in postindustrial societies, possibly suggesting preventive strategies based on biome reconstitution. The paper by C. Onore et al. reports an impressive threefold decrease in EGF plasma levels among small ASD children compared to controls. EGF is involved in wound healing in the skin, in the gastrointestinal, and respiratory systems, as well as in the central nervous system where this neurotrophic factor supports both adult subventricular zone and midbrain dopaminergic neurons, in addition to stem cell proliferation. A. R. Torres et al. review immune abnormalities in autism, focusing on HLA gene roles as potential contributors to autism pathogenesis through several distinct mechanisms. HLA genes or haplotypes have been found associated with several autoimmune diseases, but strong associations with autism have also been reported. These contributions, while not providing the ultimate answers, do add some new pieces to the autism puzzle, providing further support to the idea that immune abnor- malities may play a pathophysiologically relevant role in a subgroup of families with autistic children. Future studies of the interaction of the immune and central nervous system are needed to clarify in more detail how neuroimmune mecha- nisms influence the enhancement of the neurodevelopmental disarray and disturbances of neurobehavioral trajectories determined by genetic and environmental factors. The heuristic potential of this line of investigation, both in terms of prevention and clinical therapeutics, ensures continued efforts until a more definitive answer can be given to our initial question, so crucial in today’s autism field. Antonio M. Persico Judy Van de Water Carlos A. Pardo Hindawi Publishing Corporation Autism Research and Treatment Volume 2012, Article ID 910946, 11 pages doi:10.1155/2012/910946

Review Article Is Autism a Member of a Family of Diseases Resulting from Genetic/Cultural Mismatches? Implications for Treatment and Prevention

StaciD.Bilbo,1 John P. Jones,2 and William Parker2

1 Systems and Integrative Neuroscience Group, Department of Psychology and Neuroscience, Duke University, Durham, NC 27710, USA 2 Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA

Correspondence should be addressed to William Parker, [email protected]

Received 12 September 2011; Revised 18 January 2012; Accepted 10 April 2012

Academic Editor: Antonio M. Persico

Copyright © 2012 Staci D. Bilbo et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Several lines of evidence support the view that autism is a typical member of a large family of immune-related, noninfectious, chronic diseases associated with postindustrial society. This family of diseases includes a wide range of inflammatory, allergic, and autoimmune diseases and results from consequences of genetic/culture mismatches which profoundly destabilize the immune system. Principle among these consequences is depletion of important components, particularly helminths, from the ecosystem of the human body, the human biome. Autism shares a wide range of features in common with this family of diseases, including the contribution of genetics/epigenetics, the identification of disease-inducing triggers, the apparent role of immunity in pathogenesis, high prevalence, complex etiologies and manifestations, and potentially some aspects of epidemiology. Fortunately, using available resources and technology, modern medicine has the potential to effectively reconstitute the human biome, thus treating or even avoiding altogether the consequences of genetic/cultural mismatches which underpin this entire family of disease. Thus, if indeed autism is an epidemic of postindustrial society associated with immune hypersensitivity, we can expect that the disease is readily preventable.

1. Introduction: Autism as a Member of a Large lupus, multiple sclerosis, hay fever, appendicitis, chronic Family of Postindustrial Epidemics fatigue syndrome, inflammatory bowel disease, asthma, Involving a Hyperimmune Response celiac disease, type 1 diabetes, Graves’ disease, some types of eczema, and a wide range of food allergies. Here, based In this paper, we outline a paradigm that points toward on a variety of evidence, we argue that autism is yet another autism as one disease among many other well-known member of this family of diseases, despite the slowness diseases which all share a common origin and, most of the medical community to recognize it as such. Since likely, a common prevention strategy [1]. These emerging, the pathogenesis of at least one form of autism has been noninfectious diseases are all epidemics of postindustrial directly linked to autoantibody production [2], and since culture, which are absent in antiquity in any culture, seven out of every eight cases of severe autism are associated and absent in present day developing cultures. Members with antineuronal antibodies in the serum [3], this idea of this family of disease are invariably associated with a has already been demonstrated, at least in part. Further, hyperimmune response and can have a very high preva- as described below, autism is associated with a wide range lence in postindustrial populations, with prevalence often of immune abnormalities. Even though some children with greater than 0.1% and sometimes greater than 1.0%. These autism appear to have “normal” immune systems, it is argued hyperimmune-associated diseases include a wide range of that adverse immune events early in development might lead allergic, autoimmune, and inflammatory diseases such as to an autistic phenotype. 2 Autism Research and Treatment

A primary consideration in this view of autism as a 4. Glial Cells Direct Normal hyperimmune-associated disease is the connection between Brain Development immunity and brain development in general, and between hyperimmune responses and autism in particular. Although Microglia are the resident macrophages of the central these connections have little bearing on the overall model nervous system, are associated with the pathogenesis of describing induction of disease by immune hypersensitivity many inflammatory diseases of the brain, and derive from in postindustrial society, the connections provide reason to primitive yolk sac macrophage precursors, which are of expect that the developing brain is sensitive to the same mesodermal origin and enter the neuroectoderm during postindustrial changes in the immune system which are embryogenesis [21]. Early in development, microglia are known to affect virtually all other organs of the human body highly mobile and primarily amoeboid, consistent with (e.g., kidneys, pancreas, epidermis, adult nervous tissues, their role in the phagocytosis of apoptotic cells [22]. The large and small bowel, cardiovascular system, thyroid, lungs, expression of many cytokines within the developing brain, andothers).Thenextfoursectionswillsummarizemuch including IL-1β and TGF-β, depends on the presence of of what is currently known regarding those very extensive amoeboid microglia [23]. Microglia transform into a highly connections. branched, ramified morphology by adulthood in most brain regions. This morphological transition occurs in parallel with neural cell genesis and migration, synaptogenesis, and 2. Pervasive Immune System synaptic pruning, suggesting functions for microglia in Abnormalities in Autism each of these processes, though these are just beginning to be explored [24–26]. Oligodendrocytes myelinate axons Immune system abnormalities exist throughout the body primarily during the postnatal period, and disruption of and brain of autistic children. These include evidence their function can be profoundly debilitating as in the of brain specific auto-antibodies, altered T, B, and NK case of periventricular leukomalacia leading to cerebral cell responses to antigen, altered cytokine production, an palsy [27]. Astrocytes mediate synapse formation within increased incidence of allergies and other autoimmune dis- the developing brain [28], in part via the secretion of orders, and functional changes in brain glial cells (microglia extracellular matrix proteins called thrombospondins (TSPs) and astrocytes) [4–11]. Microglia are the primary immuno- [29, 30]. Alterations in spine density via a putative TSP competent cells of the brain and rapidly respond to any mechanism are implicated in neurodevelopmental disorders infection, injury or other perturbation of homeostasis via such as Down’s and Rett Syndrome [31]. Notably, astrocyte a dynamic process of activation [12]. Notably, increased maturation marks the end of the perinatal synaptogenic microglial activation has been observed in several brain period when the brain is most plastic [32]. regions of autistic patients [13]. Once activated, glia produce a wide number of immune signaling molecules, including cytokines (e.g., interleukin [IL]-1β, tumor necrosis factor [TNF]-α), chemokines (e.g., monocyte chemoattractant 5. Long-Term Consequences of Immune protein [MCP]-1), and other inducible factors (e.g., nitric Activation during Early Development oxide), which may profoundly influence neural function Developmental or “fetal programming” is a growing field of [14]. science based on evidence that experiences during critical or sensitive periods of perinatal life may modulate or “program” 3. Immune System-Central Nervous the normal course of development, with the result that adult System Communication outcomes, including behavior, are significantly and often permanently altered [33]. Given the many ways in which the Beyond its traditional role in host defense and tissue repair, immune system is important for normal brain development, the immune system is now considered a diffuse sensory the capacity for immune-inducing events to influence the organ that works in concert with the endocrine, metabolic, long-term trajectory and function of these processes is likely and nervous systems to achieve and maintain homeostasis profound, perhaps more so than at any other stage of life throughout the body [15, 16]. In essence, the immune [34]. Notably, there is strong evidence that early-life infection system serves as an interface between the human body and can permanently alter stress reactivity, disease susceptibility, the environment, coordinating the response of the body and notably, vulnerability to cognitive and neuropsychiatric to the environment. Immunocompetent cells are located disorders, including Alzheimer’s disease, Parkinson’s disease, throughout every organ of the body, including the brain, , and autism [35–38]. One of the most dra- and regular communication occurs between the central matic and direct illustrations of this effect is the induction of nervous system and immune tissues during both health autism-like symptoms in mice by stimulation of the maternal and disease processes. Many excellent reviews have been immune system during pregnancy [39]. written on these topics [17–20]. Importantly, bidirectional Neuroinflammatory-induced alterations in neurogene- communication between the brain and immune system has sis, migration, axon growth, myelination, synaptogenesis, significant consequences for plasticity mechanisms within and dendritic spine density/function are all candidate mech- the brain, including cognition and emotion, which are anisms by which long-term changes in behavior might markedly altered in autism. result. Further, the long lifespan of microglia and their Autism Research and Treatment 3 pattern of central nervous system colonization during immune system. This effect is closely intertwined with an development suggest that these immune cells may have impact on metabolism and transport, as will be described particular significance in terms of neuroinflammatory effects later. These mismatches of postmodern culture involve some on developmental programming. Diverse early-life events of the most cherished attributes of postmodern culture, may have enduring consequences for the brain and behavior including the widespread use of toilets, water treatment facil- of organisms via an influence on these resident immune ities, indoor working environments, soaps and shampoos, cells, both by impacting their own intrinsic function as well modern medicine, and the loss of factors which demand as their interactions with ongoing developmental processes exercise for survival (e.g., human-eating predators, hardship [40, 41]. in finding food and procuring shelter for most of the population). These mismatches are not a target for therapy, because reversal of any of these mismatches is untenable. 6. Approaching Autism as Another Member of Thus, these mismatches can be described as belonging to the Family of Hyperimmune-Associated the “pretherapeutic zone” in the course of pathogenesis Diseases: Addressing the Underlying Causes (Figure 1). However, these mismatches lead directly to readily measurable consequences that, in turn, lead to disease The study of individual postindustrial associated diseases has if left untended. Fortunately, these direct consequences of often failed to enlighten researchers as to the underlying mismatches are readily treated or even avoided altogether. cause of diseases. While genetic factors associated with the Given that these consequences can be treated or avoided, epidemics of postindustrial society abound, these genetic this part of the pathogenesis of hyperimmune disease can factors have not changed over many generations, and thus be described as the “optimal therapeutic zone” (Figure 1). are not responsible for newly emerging epidemics of disease. In this paper we describe how mismatches between culture Almost paradoxically, even when specific triggers are iden- and genetics/biology have led to specific consequences which tified, these often cannot be said to cause newly emerging cause immune system hypersensitivity and/or destabiliza- epidemics. For example, peanuts and ragweed pollen are tion. All of these consequences are associated with a wide two well-known triggers for allergy, but these substances range of diseases that include allergic, autoimmune, and have been present in the environment for millennia without inflammatory diseases. triggering disease in humans. Similarly, viral infections and The single consequence of genetic/culture mismatches potentially some milk proteins [42] have been identified as that most profoundly impacts hyperimmune-associated dis- triggers for particular autoimmune diseases, but these factors ease has been described as a depletion of components have been present for much longer than the diseases they from the natural ecosystem of the human body, or simply trigger. Thus, as will be described in detail, it is not the “biome depletion” (Figure 1). The use of modern medicine, genetics of the disease which point toward a cure, and it is toilets, water treatment facilities, and cleaning agents in our generally not identification of the triggers for disease that culture has led to a substantial alteration or even loss of is most helpful. Rather, we suggest that the most effective living organisms that have coevolved with humans. Three and insightful approach toward defeating autism for future primary components of the biome which are potentially generations will be the neutralization of underlying factors in important have been affected [43]: (a) loss of interactions postindustrial culture which render postindustrial humans with soil bacteria, (b) alterations of the microbiome, the uniquely susceptible to disease, despite the presence of the normal bacteria that are associated with the human body, same genetics and many of the same triggers as found in and (c) loss of helminths, worms that typically inhabit preindustrial cultures. These factors have been identified the gut of mammalian species. Substantial evidence points as mismatches between normal human genetics and the toward all three of these factors as being important in environment of postindustrial culture. With that in mind, stabilization of the human immune system. Although not an examination of the factors in postindustrial culture which widely appreciated, several lines of evidence point very trigger the plagues of hyperimmune-associated diseases will strongly if not conclusively to the idea that helminths in be presented. particular are necessary for a stable immune system. This evidence has been reviewed extensively [1, 43]andcanbe very briefly summarized as follows. 7. Mismatches between Normal Human Genetics and Postindustrial Culture (i) Helminths are found almost ubiquitously in prein- dustrial humans [44] and in nondomesticated Leading to Hyperimmune Epidemics: mammals, including chimpanzees [45]. Even when Biome Depletion helminths cannot be found, the immune system shows signs of stimulation by helminths [46]. If indeed autism fits into the family of hyperimmune- associated diseases plaguing postindustrial culture, then the (ii) Helminths produce and secrete a number of pathogenesis or origins of these diseases merit detailed molecules which regulate the host immune system analysis in any consideration of autism. As shown in Figure 1, [47]. postindustrial culture has several mismatches with normal (iii) Addition of helminths to laboratory animals blocks human genetics (known as “evolutionary mismatches”) allergic, inflammatory, and autoimmune diseases which have a profoundly destabilizing effect on the human [47]. 4 Autism Research and Treatment

Postindustrial culture Indoor work, soap, Exercise not Pretherapeutic toilets, water treatment, shampoo, bathing essential for survival modern medicine zone

Optimal Biome Vitamin D Uncontrolled therapeutic depletion deficiency stress zone

Genetics, Immune hypersensitivity epigenetics, Alteration of and/or destabilization “triggers” transport and Allergy, Phase I/Phase II autoimmunity, metabolism chronic A wide range of Postoptimal inflammation Metabolic demands on therapeutic dysfunction, transport and zone toxicity issues, Phase I/Phase II oxidative stress metabolism Risk of cognitive dysfunction

Figure 1: The pathogenesis of epidemics of allergic, inflammatory, and autoimmune disease. Cultural mismatches with human biology (the pretherapeutic zone) have consequences that can be readily avoided (the optimal therapeutic zone). If biome depletion in particular is not avoided, the individual is left susceptible to a wide range of often complex immune-related diseases (the postoptimal therapeutic zone), sometimes associated with imbalances in metabolite transport and/or Phase I/Phase II metabolism. Other consequences such as vitamin D deficiency and uncontrolled stress may, in some but not all cases, add to the problem. Factors contributing to disease which are often naturally occurring and generally difficult or impossible to avoid are underlined and fit within the postoptimal therapeutic zone.

(iv) Addition of helminths to humans cures inflamma- infection, genetics or epigenetics) in the induction of tory bowel disease [48] and stops the progression of autism. multiple sclerosis [49]. 8. Mismatches between Normal Human (v) The effects of biome depletion on the immune system are not necessarily instantaneous because Genetics and Post-Industrial Culture long-term effects of immune interactions with the Leading to Hyperimmune Epidemics: biome can span decades or, through epigenetics, even Vitamin D Deficiency generations [1]. Nevertheless, the epidemiology of hyperimmune disease reveals an inverse correlation Based on the available information outlined above, it between helminths and disease. seems very likely that biome depletion is by far the most significant and most profoundly influential consequence (vi) Humans with helminths and rodents colonized with of genetic/cultural mismatches in postindustrial society. helminths have profoundly less reactive immune sys- However, other consequences which apparently exacerbate tems than do either without helminths [44, 50, 51]. the effects of biome depletion are evident. For example, (vii) A long coevolutionary history of helminths and ver- vitamin D deficiency has reached epidemic proportions in tebrates, in conjunction with the potent immunoreg- postindustrial society, is known to impact inflammation ulatory effects of the former, is consistent with the during early development [52], and, like biome depletion, idea that the human immune system is literally, has been linked to a spectrum of allergic, autoimmune, and physically dependent on helminths [1]. inflammatory diseases [53–55]. Further, epidemiology and other circumstantial evidence link vitamin D deficiency to We propose that biome depletion is changing the autism [56, 57]. Interestingly, the widely publicized asso- immune system at a population-wide level, which in ciation between rainfall and autism [58]canbeaccounted turn will undoubtedly impact the brain, particularly dur- for, perhaps more than in part, by decreased exposure to ing sensitive periods of development. Importantly, this sunshine in areas with high rainfall, and the subsequent modeldoesnotcompetewithanypriorhypothesesor impact of lower vitamin D levels. known risk factors for neuroinflammatory conditions, but Vitamin D deficiency is another consequence of rather complements them. For instance, biome deple- postindustrial culture’s mismatches with human biology tion may set the stage for exaggerated levels of neu- (Figure 1). Vitamin D production requires exposure of oils roinflammation, which interacts with other known risk on the body’s surface to sunlight. The resulting photochem- factors or triggers (psychological or metabolic stress, ical reaction and production of vitamin D is greatly reduced Autism Research and Treatment 5 by components of postindustrial culture that reduce expo- consequences of postindustrial culture can also be found sure to sunlight (e.g., indoor work environments, sunscreen). which are associated with a wide range of disease, but limits The association of vitamin D deficiency with a spectrum in space prevent a complete discussion. For example, chronic of hyperimmune-associated diseases places this consequence uncontrollable psychosocial stress is yet another factor that is of biology/culture mismatch alongside biome depletion as an generally associated with a range of hyperimmune-associated underlying agent destabilizing immune system function in diseases that include allergy, autoimmunity, and inflamma- postindustrial populations. However, vitamin D deficiency is tion [63–66]. Such stress derives from a complex range of an ancient problem, as indicated by the ancient occurrence biology/culture mismatches, including, for example, the loss of rickets, whereas epidemics of hyperimmune-associated of physical exercise [67–69] as a requirement for survival in diseases are more recent in nature. Thus, it seems likely postindustrial culture (Figure 1). As another example of con- that vitamin D deficiency is a contributor to hyperimmune- sequences of biology/culture mismatches which destabilizes associated disease, but not sufficient by itself to lead to immune function, deprivation from mother’s milk during disease. Further, the prevalence of vitamin D deficiency in infancy (not included in Figure 1) is associated with a wide postindustrial society is somewhat less than the prevalence range of allergic and autoimmune conditions [70–74]. This of biome depletion: although the prevalence of vitamin D genetic/culture mismatch, like other mismatches, involves an deficiency is substantial, with one study finding 24% of aspect of postindustrial society which should not be reversed individuals deficient (≤20 ng/mL) with an additional 34% (the survival of infants despite the unavailability of mother’s having levels that were insufficient (≤29 ng/mL) [59], the milk), and thus fits into the “pretherapeutic zone” of prevalence of biome depletion in postindustrial populations Figure 1. The extent to which factors such as uncontrollable is essentially 100%. Thus, it is expected that the impact of stress and lack of mother’s milk can contribute to disease vitamin D deficiency on immunity in postmodern culture in the presence of an otherwise normal biome remains to may be a matter of exacerbating the problems associated with be elucidated, although, as outlined above, biome depletion biome depletion more so than a significant problem when appears to be the single most important factor contributing considered in isolation. to the increased incidence of hyperimmune-associated dis- Some mechanisms by which vitamin D might stabilize eases. the immune system have been elucidated. Vitamin D acts as a signaling molecule to enhance immune cell function in the presence of infection and is important for maintaining the 10. Metabolism and Transport of Toxins and normal interface between the microbiome and the immune Hyperimmune-Associated Disease system [60]. However, the mechanisms by which vitamin D interacts within the body are complex, with the molecule The connection between metabolism and immunity is being intertwined either directly or indirectly in virtually important in a consideration of hyperimmune-associated all aspects of human biology. The active form of vitamin diseases. A variety of enzymes have been identified which are D binds and activates the Vitamin D receptor, altering the useful in the biotransformation of endogenous (produced expression of a variety of genes involved in such diverse by the body or the associated biome) as well as exogenous processes as cell growth and proliferation, bone remodeling, (pharmaceutical) substrates. This system of enzymes pro- and calcium homeostasis [61]. It has been estimated that vides the body with a means of processing a wide range vitamin D binds human DNA in more than 2,500 places of substances, some of which are toxins, and is profoundly and changes the expression of more than 200 genes [62]. affected by the immune system [75]. The metabolic portion Thus, while it is known that vitamin D is required for normal of this system is readily divided into two components: immune function, it is likely that vitamin D deficiency Phase I biotransformation includes the oxidative reactions could cause a general destabilization of the human biome of the microsomal cytochrome P450 monooxygenase system independent of the immune system, particularly during early that is expressed in the liver and gastrointestinal tract, development. Fortunately, regardless of the extent to which and to some extent in a variety of extrahepatic tissues. vitamin D deficiency destabilizes the human biome, dietary Phase II biotransformation includes conjugation reactions of supplements are readily available, and thus this consequence substrates with endogenous cofactors such as glucuronate, of culture/biology mismatch is easily avoided. sulphate, acetate, or glycine. In addition to Phase I and Phase II biotransformation, the transport of their substrates and byproducts is also an 9. Mismatches between Normal Human important component of this system. A variety of membrane Genetics and Postindustrial Culture spanning polypeptides have been identified in the liver, Leading to Hyperimmune Epidemics: gastrointestinal tract, kidney as well as other tissues in the Other Factors body, each responsible for the active trafficking of substrates across cell membranes. These transport proteins are in fact The above discussion points out two factors, primarily regulated by the same pathways that regulate the Phase I and biome depletion and secondarily vitamin D deficiency, II enzymes, and they can be thought of as being regulated in which generally destabilize the human immune system. tandem [76–80]. These factors can be identified by their association with Immune activation results in a downregulation of Phase a wide range of hyperimmune-associated diseases. Other I components [81, 82], transporters [83, 84] and potentially 6 Autism Research and Treatment

Phase II components in what might be viewed as a tradeoff Thus, if porcine whipworms do not work, it does not between immunity and metabolism. In time of duress, when suggest that the hypothesis is wrong. the immune system is geared to inflammation and defense, (v) This model does not suggest that helminths are the energy devoted to metabolism/transport may need to the only organisms involved in biome depletion. be temporarily diverted to host defense. This is apparently Normalization of the microbiome may also be a an effective strategy, but as a consequence, chronic immune ffi critical factor. In the face of modern practices in activation could result in metabolic di culties as outlined obstetrics and of widespread antibiotic use, this is an in Figure 1. Unfortunately, vitamin D deficiency is expected issue that must be considered. to make this problem worse, since vitamin D is required toupregulatebothPhaseIandPhaseIIcomponents[85] (vi) Pharmaceuticals may never prove adequate. The of metabolism. Further, accumulation of toxins as a result complexity of the immune system and the effects of of reduced Phase I, Phase II, or transport activity may biome depletion suggest that efforts to readjust the also stimulate the immune system, adding further to the system using drugs may be naive and overly opti- propensity for hyperimmune activity. mistic. Biome reconstitution may be the easiest and most straight forward way to bring the system into homeostasis. Similarly, metabolic factors may not 11. Implications of Autism as a Member of easily be compensated for by pharmaceuticals. Nor- the Family of Hyperimmune-Associated, malization and regulation rather than compensation Postindustrial Diseases and suppression are probably the better approaches. The implications of this model are several-fold and deserve (vii) Genetics may be relatively unimportant from a careful consideration. The consideration of some possible clinical perspective because it may not provide a assertions that are not supported by this model is probably means to prevent or treat disease. Triggers may also equally as important. be unimportant from a clinical perspective because (a) they may simply be unavoidable, and (b) even (i) This model indicates that prophylactic normalization if triggers for a particular disease can be identified ff of the biome will result in a profound decrease and avoided, such e orts may simply decrease the in the incidence of autism. Results with other prevalence of one disease at the expense of an hyperimmune-associated diseases have been promis- increased prevalence of another. ing, although a true normalization of the biome as a means of prevention has never been attempted for 12. The Required Approach any disease. Clinical trials are urgently needed to conclusively test a (ii) This model is consistent with the idea that a variety variety of questions. The approach needs to be extensive of factors affecting immunity and/or metabolism/ and systematic rather than timid and piecemeal. The tests transport may strongly influence the incidence of required will be on a large scale, although this scale pales autism. in comparison to the current amount of effort and energy (iii) There are several potential therapeutic approaches being poured into research on allergic, inflammatory, and that this model does not suggest which will necessar- autoimmune issues. This approach to medicine will not ily be successful. For example, this model does not likely be supported by any committee of experts containing predict that helminths will necessarily work as a cure members with vested interests in traditional immunological for autism. Whether helminths can be used to treat approaches (either in humans or in rodents) or in the autism will depend on the extent to which autism is pharmaceutical industry. Rather, this approach is much more reversible and the extent to which ongoing problems likely to be appreciated by those individuals with a broad with biome stability can be reversed. understanding of biology and whose primary interest is in improving health care and, ultimately, the health of the (iv) Although porcine whipworms are currently being ff population as a whole. With that in mind, the following used to treat the e ects of biome depletion in some questions need to be answered. studies, the model described in this paper does not predict that the porcine whipworm will prevent (i) Can autism be cured or effectively treated with autism, even prophylactically. (Although the use of biome reconstitution, or only prevented by biome porcine whipworms is currently underway for a few reconstitution? clinical studies, the use of porcine whipworms as a means of prophylaxis is probably not economically (ii) How important for children is the biome of the feasible because the costs of giving the necessary mother prior to and during pregnancy, and while biweekly doses would be prohibitive. This problem breastfeeding? (Does prevention require reconstitu- is not expected when using helminths adapted to tion of the mother’s biome?) humans, since one dose lasts for years.) It might, but (iii) Which helminths or combination of helminths is this potentially depends on the extent to which this both safe and effective? Although some concerns species recapitulates the presence of a normal biome. dealing with the use of helminths for therapy have Autism Research and Treatment 7

been expressed, these objections are easily dealt with P = 0.05) that autism is a consequence of evolutionary [1]. Several candidate species are well adapted to mismatches in postindustrial society and is consistent with humans, asymptomatic in low numbers, and thus are the idea that the parental immune status is important in obvious candidates for initial and immediate test- the development of autism. Further assessment of autism ing. Further, new technologies might be developed in preindustrial cultures may, however, prove unnecessary; which could improve on species that are naturally the easiest way to evaluate the idea that autism is related occurring. For example, irradiation of organisms to to biome depletion is probably to conduct the experiment. achieve sterility and eliminate the possibility of trans- Whether prophylactic biome reconstitution does or does mission, or cultivation of human-specific helminths not affect the incidence of autism will answer all questions in ultraclean and immunodeficient rodents or even conclusively, just as the effects of biome reconstitution in vitro might improve the utility of helminths for [49, 87] and vitamin D supplementation [88] on multiple widespread medical use. sclerosis, if confirmed by future studies, will effectively end (iv) What medical conditions such as a suppressed long-standing debates about the etiology of that disease. immune system, anemia, or coagulopathy might be The idea that autism is a result of evolutionary mis- contraindications for biome reconstitution? matches is predicated on the idea that autism often occurs in genetically “normal” individuals. Thus, if autism can (v) Does optimal biome reconstitution vary with human be attributed to genetic factors which dictate the presence genotype or other factors such as age, gender, and of disease even in individuals living in a preindustrial body size/composition? society, then biome depletion is probably not a factor in (vi) In addition to the restoration of helminths to the the pathogenesis of autism, and biome reconstitution will biome, what steps should be taken for restoration and probably not affect the incidence of autism. Certainly there maintenance of the microbiome? are cases where autism is associated with genetic mutations ff which are deleterious, such as , but at (vii) How will a reconstituted biome a ect other areas present most individuals with autism appear genetically of medicine, including aging, immunosuppression, normal [89]. However, it remains unknown what percentage infectious disease, cancer biology, and vaccine tech- of autism can be attributed directly to genetics, and the idea nology? that biome depletion and other evolutionary mismatches affect mutation rates is speculative and unexplored. Again, 13. Is Autism a Postindustrial, the most effective approach to resolving the debate regarding Hyperimmune-Associated Epidemic? the role of genetics may be to prophylactically reconstitute the biome of a sample population and await the results. The idea that autism is a result of evolutionary mismatches The epidemiology and the genetics of autism are is predicated on the idea that autism is epidemic in postin- inextricably woven together. It is anticipated that one of dustrial society. Thus, if the incidence of autism is the same two pictures will emerge. Either a postindustrial epidemic in preindustrial societies as it is in postindustrial societies, of autism with normal genetics will be identified, or then biome depletion is not a factor in the pathogenesis autism will be eventually defined as a “normal” part of of autism, and biome reconstitution will not affect the the human condition, potentially associated with certain incidence of autism. The epidemiology of autism has been genes or combinations of genes that predispose to disease hotly debated. Some changes in diagnostic criteria and regardless of the environment. In the former view, autism awareness of autism have certainly affected changes in the is due to an evolutionary mismatch, whereas in the latter reported incidence of autism over time. However, debates view, autism is due to evolution itself. It is of critical regarding the changing diagnosis of autism over the past few importance not to accept the latter view prematurely since decades have little bearing on the idea that biome depletion such acceptance may disable efforts at compensating for profoundly influences autism. Rather, the pertinent question evolutionary mismatches or discourage work on identifying is whether the prevalence of autism has changed over the postindustrial triggers for disease, with potentially tragic past 150 years as various cultures have transitioned from consequences. Fortunately, several lines of evidence argue pre to postindustrial over the course of several generations. against the idea that autism is a “normal” part of human Although it may be very difficult if not impossible to estimate biology. First, a general consideration of the nature of human the prevalence of autism prior to the industrial revolution evolution argues that evolution is not responsible for autism. in countries that are currently postindustrial, it might still The social interactions disrupted by autism are fundamental be possible to examine the prevalence of autism in some to survival of not only humans, but also a wide range of preindustrial societies. One very recent study by Schieve et mammalian and nonmammalian species, pointing to the al. is particularly informative in this regard: using a phone ancient evolutionary origins of those interactions. Given the survey technique to obtain data on 4,690 Hispanic children, intense selection pressures on early human populations, it a “striking heterogeneity” in the prevalence of autism seems unlikely that an error rate of 1% in social interactions spectrum disorder between US-born Hispanic children with would have developed and then persisted for many tens 2 US-born parents (autism spectrum disorder prevalence of thousands of years. However, the idea that autism was 2.39%) and otherwise similar children with 2 foreign- tolerated during human evolution because it is essentially a born parents (autism spectrum disorder prevalence 0.31%; tradeoff, an inherent “side-effect” of human brain function 8 Autism Research and Treatment that is required for survival, cannot be ruled out. A millions are affected by a wide range of allergic, inflam- second line of evidence pointing in favor of autism as a matory, and autoimmune disorders, and inflammation as a result of evolutionary mismatch rather than evolution is result of biome destabilization may play a key role in such the study, described above, showing that parental birth in common maladies as heart disease and dementia. With that a postindustrial culture seems to predispose offspring to in mind, the need for normalization of the biome cannot be autism [86]. Third, the observations (a) that the immune overemphasized, regardless of particular diseases that might system is profoundly destabilized by postindustrial culture be prevented by identifying and avoiding the environmental and (b) that brain development is closely tied with immune trigger. Thus, while it is hoped that a trigger that causes function, when considered together, argue strongly for the autism might be confidently identified in the near future, and idea that it is an evolutionary mismatch, not evolution, new cases of autism might be greatly reduced as a result, it is which is responsible for autism. Indeed, to the extent hypothesized that normalization of the human biome will be that human brain development is strongly tied to the necessary to avoid the full milieu of chronic immune-related immune system, it would be difficult to explain the idea diseases that plague postindustrial society. that postindustrial culture and the resulting consequences of evolutionary mismatches such as biome depletion do not profoundly affect brain development. Finally, the strong Acknowledgments association of autism with autoantibody production [3] The authors gratefully acknowledge the contribution of the and other immune abnormalities points toward the etiology Research Coordination Network in Ecological Immunology of autism as straight forward: another epidemic in the for facilitating discussion, and the Coalition for SafeMinds long list of hyperimmune associated epidemics caused by for providing the publication fee. The authors also thank biome depletion in postindustrial society. The fact that this Sharon L. Brenner, Zoie E. Holzknecht, and Susanne Meza- particular disease affects primarily the brain rather than Keuthen for helpful discussion. other organs or tissues undoubtedly leads to the tremendous complexity in the pathology associated with autism, but this complexity should not blind the medical community to the References simple roots that apparently underlie epidemics of all allergic and autoimmune diseases, or to the apparent solutions that [1] S. D. Bilbo, G. A. Wray, S. E. Perkins, and W. Parker, can currently be implemented to prevent those diseases [1]. “Reconstitution of the human biome as the most reasonable solution for epidemics of allergic and autoimmune diseases,” Medical Hypotheses, vol. 77, no. 4, pp. 494–504, 2011. 14. Summary/Conclusions [2]V.T.Ramaekers,N.Blau,J.M.Sequeira,M.C.Nassogne,and E. V. Quadros, “Folate receptor autoimmunity and cerebral Autism has many of the characteristic features of folate deficiency in low-functioning autism with neurological hyperimmune-associated diseases which result from deficits,” Neuropediatrics, vol. 38, no. 6, pp. 276–281, 2007. biome depletion and other consequences of biology/cultural [3] G. A. Mostafa and L. Y. Al-Ayadhi, “The relationship between mismatches in postindustrial culture, potentially sharing the increased frequency of serum antineuronal antibodies a common cause and potentially a common solution with and the severity of autism in children,” European Journal of those diseases. The identification of triggers for autism Pediatric Neurology. In press. (e.g., association with viral infection during pregnancy [4] P. Ashwood and A. J. Wakefield, “Immune activation of [90]) parallels that of other diseases in this family. However, peripheral blood and mucosal CD3+ lymphocyte cytokine the trigger that causes disease is often unrelated to the profiles in children with autism and gastrointestinal symp- cultural mismatches with human biology which destabilize toms,” Journal of Neuroimmunology, vol. 173, no. 1-2, pp. 126– 134, 2006. the human biome and make the trigger dangerous. The [5]P.Ashwood,S.Wills,andJ.VanDeWater,“Theimmune association of autism with genetic and probably epigenetic response in autism: a new frontier for autism research,” Journal factors also parallels that of other hyperimmune-associated of Leukocyte Biology, vol. 80, no. 1, pp. 1–15, 2006. diseases, but offers little in the hope of prevention, treatment, [6] R. L. Blaylock, “A possible central mechanism in autism or a cure. spectrum disorders, part 2: immunoexcitotoxicity,” Alternative The ecosystem of the human body, the human biome, Therapies in Health and Medicine, vol. 15, no. 1, pp. 60–67, is a tightly interwoven collection of factors which includes 2009. immunity, brain function, sunlight, hormones, metabolism, [7] R. L. Blaylock and A. Strunecka, “Immune-glutamatergic transport of metabolites, the microbiome, and helminths. It dysfunction as a central mechanism of the autism spectrum stands firmly to reason that if one or more of these com- disorders,” Current Medicinal Chemistry,vol.16,no.2,pp. ponents are profoundly altered or even deleted, the entire 157–170, 2009. system may become destabilized. This view of ecosystems [8] N. C. Derecki, E. Privman, and J. Kipnis, “Rett syndrome and other autism spectrum disordersbrain diseases of immune has long been appreciated by ecologists [91]. Although some malfunction,” Molecular Psychiatry, vol. 15, no. 4, pp. 355–363, hyperimmune diseases might be eliminated if the respective 2010. triggers for those diseases are identified and avoided, such [9]A.M.Enstrom,C.E.Onore,J.A.VandeWater,andP. as approach cannot be successful in eliminating the overall Ashwood, “Differential monocyte responses to TLR ligands problem; roughly 50% of children in postindustrial society in children with autism spectrum disorders,” Brain, Behavior, have some form of chronic health condition [92], many and Immunity, vol. 24, no. 1, pp. 64–71, 2010. Autism Research and Treatment 9

[10] I. Maezawa and L. W. Jin, “Rett syndrome microglia damage promote CNS synaptogenesis,” Cell, vol. 120, no. 3, pp. 421– dendrites and synapses by the elevated release of glutamate,” 433, 2005. Journal of Neuroscience, vol. 30, no. 15, pp. 5346–5356, 2010. [30] C¸.Eroglu,N.J.Allen,M.W.Susmanetal.,“Gabapentin [11] C. A. Pardo, D. L. Vargas, and A. W. Zimmerman, “Immunity, receptor alpha2delta-1 is a neuronal thrombospondin receptor neuroglia and neuroinflammation in autism,” International responsible for excitatory CNS synaptogenesis,” Cell, vol. 139, Review of Psychiatry, vol. 17, no. 6, pp. 485–495, 2005. no. 2, pp. 380–392, 2009. [12] W. J. Streit, “Microglia as neuroprotective, immunocompetent [31] O. Garcia, M. Torres, P. Helguera, P. Coskun, and J. Busciglio, cells of the CNS,” GLIA, vol. 40, no. 2, pp. 133–139, 2002. “A role for thrombospondin-1 deficits in astrocyte-mediated [13] D. L. Vargas, C. Nascimbene, C. Krishnan, A. W. Zim- spine and synaptic pathology in down’s syndrome,” PLoS merman, and C. A. Pardo, “Neuroglial activation and ONE, vol. 5, no. 12, Article ID e14200, 2010. neuroinflammation in the brain of patients with autism,” [32] C. M. Muller and J. Best, “Ocular dominance plasticity in adult Annals of Neurology, vol. 57, no. 1, pp. 67–81, 2005. cat visual cortex after transplantation of cultured astrocytes,” [14] W. J. Streit, “Microglia and neuroprotection: implications for Nature, vol. 342, no. 6248, pp. 427–430, 1989. Alzheimer’s disease,” Brain Research Reviews, vol. 48, no. 2, pp. [33] L. Bennet and A. Gunn, “The fetal origins of adult mental 234–239, 2005. illness,” in Early Life Origins of Health and Disease (Advances [15] A. J. Husband, “The immune system and integrated home- in Experimental Medicine and Biology), M. Wintour-Coghlan ostasis,” Immunology and Cell Biology, vol. 73, no. 4, pp. 377– and J. Owens, Eds., pp. 204–211, Springer, New York, NY, 382, 1995. USA, 2006. [16] L. Vitkovic, J. Bockaert, and C. Jacque, “’Inflammatory’ [34] D. Rice and S. Barone Jr, “Critical periods of vulnerability for cytokines’ neuromodulators in normal brain?” Journal of the developing nervous system: evidence from humans and Neurochemistry, vol. 74, no. 2, pp. 457–471, 2000. animal models,” Environmental Health Perspectives, vol. 108, [17] R. Dantzer and K. W. Kelley, “Twenty years of research supplement 3, pp. 511–533, 2000. on cytokine-induced sickness behavior,” Brain, Behavior, and [35] M. Hornig, H. Weissenbock,¨ N. Horscroft, and W. I. Lipkin, Immunity, vol. 21, no. 2, pp. 153–160, 2007. “An infection-based model of neurodevelopmental damage,” [18] S. F. Maier and L. R. Watkins, “Cytokines for psychologists: Proceedings of the National Academy of Sciences of the United implications of bidirectional immune-to-brain communi- States of America, vol. 96, no. 21, pp. 12102–12107, 1999. cation for understanding behavior, mood, and cognition,” [36] K. B. Nelson and R. E. Willoughby, “Infection, inflammation Psychological Review, vol. 105, no. 1, pp. 83–107, 1998. and the risk of cerebral palsy,” Current Opinion in Neurology, vol. 13, no. 2, pp. 133–139, 2000. [19] F. Mignini, V. Streccioni, and F. Amenta, “Autonomic inner- [37] P. Rantakallio, P. Jones, J. Moring, and L. Von Wendt, vation of immune organs and neuroimmune modulation,” “Association between central nervous system infections during Autonomic and Autacoid Pharmacology, vol. 23, no. 1, pp. 1– childhood and adult onset schizophrenia and other psychoses: 25, 2003. a 28-year follow-up,” International Journal of Epidemiology, [20] D. Wrona, “Neural-immune interactions: an integrative view vol. 26, no. 4, pp. 837–843, 1997. of the bidirectional relationship between the brain and [38] L. Shi, S. H. Fatemi, R. W. Sidwell, and P. H. Patterson, immune systems,” Journal of Neuroimmunology, vol. 172, no. “Maternal influenza infection causes marked behavioral and 1-2, pp. 38–58, 2006. pharmacological changes in the offspring,” Journal of Neuro- [21] F. Ginhoux, M. Greter, M. Leboeuf et al., “Fate mapping science, vol. 23, no. 1, pp. 297–302, 2003. analysis reveals that adult microglia derive from primitive [39]N.V.Malkova,C.Z.Yu,E.Y.Hsiao,M.J.Moore,andP. macrophages,” Science, vol. 330, no. 6005, pp. 841–845, 2010. H. Patterson, “Maternal immune activation yields offspring [22] P. Rezaie and D. Male, “Mesoglia and microglia—a historical displaying mouse versions of the three core symptoms of review of the concept of mononuclear phagocytes within autism,” Brain, Behavior, and Immunity,vol.26,no.4,pp. the central nervous system,” Journal of the History of the 607–616, 2012. Neurosciences, vol. 11, no. 4, pp. 325–374, 2002. [40] S. D. Bilbo and J. M. Schwarz, “Early-life programming of [23] D. Giulian, D. G. Young, J. Woodward, D. C. Brown, and L. B. later-life brain and behavior: a critical role for the immune Lachman, “Interleukin-1 is an astroglial growth factor in the system,” Frontiers in Behavioral Neuroscience, vol. 3, p. 14, developing brain,” Journal of Neuroscience, vol. 8, no. 2, pp. 2009. 709–714, 1988. [41] S. D. Bilbo, S. H. Smith, and J. M. Schwarz, “A lifespan [24] S. Rakic and N. Zecevic, “Programmed cell death in the approach to neuroinflammatory and cognitive disorders: a developing human telencephalon,” EuropeanJournalofNeu- critical role for glia,” Journal of Neuroimmune Pharmacology, roscience, vol. 12, no. 8, pp. 2721–2734, 2000. pp. 1–18, 2011. [25] W. J. Streit, “Microglia and macrophages in the developing [42] R. B. Elliott, H. E. Wasmuth, N. J. Bibby, A. K. H. Macgibbon, CNS,” NeuroToxicology, vol. 22, no. 5, pp. 619–624, 2001. and J. P. Hill, “The effect of beta-casein polymorphism on [26] G. A. Garden and T. Moller,¨ “Microglia biology in health and the stimulation of diabetes in the non-obese diabetic mouse disease,” Journal of Neuroimmune Pharmacology, vol. 1, no. 2, and effect of beta-casomorphin-7 on immune cell activity,” pp. 127–137, 2006. International Dairy Journal, vol. 8, p. 580, 1998. [27] M. J. Bell and J. M. Hallenbeck, “Effects of intrauterine [43] G. A. W. Rook, “Review series on helminths, immune inflammation on developing rat brain,” Journal of Neuroscience modulation and the hygiene hypothesis: the broader impli- Research, vol. 70, no. 4, pp. 570–579, 2002. cations of the hygiene hypothesis,” Immunology, vol. 126, no. [28] E. M. Ullian, S. K. Sapperstein, K. S. Christopherson, and B. A. 1, pp.3–11, 2009. Barres,“Controlofsynapsenumberbyglia,”Science, vol. 291, [44] G. Borkow, Q. Leng, Z. Weisman et al., “Chronic immune acti- no. 5504, pp. 657–661, 2001. vation associated with intestinal helminth infections results in [29] K. S. Christopherson, E. M. Ullian, C. C. A. Stokes impaired signal transduction and anergy,” Journal of Clinical et al., “Thrombospondins are astrocyte-secreted proteins that Investigation, vol. 106, no. 8, pp. 1053–1060, 2000. 10 Autism Research and Treatment

[45] M. P.Muehlenbein, “Parasitological analyses of the male chim- [62] S. V. Ramagopalan, A. Heger, A. J. Berlanga et al., “A ChIP- panzees (Pan troglodytes schweinfurthii) at Ngogo, Kibale seq defined genome-wide map of vitamin D receptor binding: National Park, Uganda,” American Journal of Primatology, vol. associations with disease and evolution,” Genome Research, 65, no. 2, pp. 167–179, 2005. vol. 20, no. 10, pp. 1352–1360, 2010. [46] M. Scaglia, M. Tinelli, R. Revoltella et al., “Relationship [63] J. P. Gouin, L. V. Hantsoo, and J. K. Kiecolt-Glaser, “Stress, between serum IgE levels and intestinal parasite load in negative emotions, and inflammation,” in Handbook of Social African populations,” International Archives of Allergy and Neurosciences, J. T. C. J. Decety, Ed., pp. 814–829, John Wiley Applied Immunology, vol. 59, no. 4, pp. 465–468, 1979. and Sons, New York, NY, USA, 2011. [47] J. P. Hewitson, J. R. Grainger, and R. M. Maizels, “Helminth [64] J. K. Kiecolt-Glaser, K. L. Heffner, R. Glaser et al., “How immunoregulation: the role of parasite secreted proteins stress and anxiety can alter immediate and late phase skin test in modulating host immunity,” Molecular and Biochemical responses in allergic rhinitis,” Psychoneuroendocrinology, vol. Parasitology, vol. 167, no. 1, pp. 1–11, 2009. 34, no. 5, pp. 670–680, 2009. [48]R.W.Summers,D.E.Elliott,K.Qadir,J.F.UrbanJr,R. [65] R. Glaser and J. K. Kiecolt-Glaser, “Stress damages immune Thompson, and J. V. Weinstock, “Trichuris suis seems to be ssytem and health,” Discovery Medicine, vol. 5, pp. 165–169, safe and possibly effective in the treatment of inflammatory 2005. bowel disease,” American Journal of Gastroenterology, vol. 98, [66]P.Møller,H.Wallin,andL.E.Knudsen,“Oxidativestress no. 9, pp. 2034–2041, 2003. associated with exercise, psychological stress and life-style [49] J. Correale and M. Farez, “Association between parasite factors,” Chemico-Biological Interactions, vol. 102, no. 1, pp. infection and immune responses in multiple sclerosis,” Annals 17–36, 1996. of Neurology, vol. 61, no. 2, pp. 97–108, 2007. [67] E. Puterman, J. Lin, E. Blackburn, A. O’Donovan, N. Adler, [50] A. P. Devalapalli, A. Lesher, K. Shieh et al., “Increased levels and E. Epel, “The power of exercise: buffering the effect of of IgE and autoreactive, polyreactive IgG in wild rodents: chronic stress on telomere length,” PLoS ONE, vol. 5, no. 5, implications for the hygiene hypothesis,” Scandinavian Journal Article ID e10837, 2010. of Immunology, vol. 64, no. 2, pp. 125–136, 2006. [68] P. Salmon, “Effects of physical exercise on anxiety, depression, [51] A. Lesher, B. Li, P. Whitt et al., “Increased IL-4 production and sensitivity to stress: a unifying theory,” Clinical Psychology and attenuated proliferative and pro-inflammatory responses Review, vol. 21, no. 1, pp. 33–61, 2001. of splenocytes from wild-caught rats (Rattus norvegicus),” [69] U. Rimmele, R. Seiler, B. Marti, P. H. Wirtz, U. Ehlert, Immunology and Cell Biology, vol. 84, no. 4, pp. 374–382, 2006. and M. Heinrichs, “The level of physical activity affects [52] N. Q. Liu, A. T. Kaplan, V.Lagishetty et al., “Vitamin D and the adrenal and cardiovascular reactivity to psychosocial stress,” regulation of placental inflammation,” Journal of Immunology, Psychoneuroendocrinology, vol. 34, no. 2, pp. 190–198, 2009. vol. 186, no. 10, pp. 5968–5974, 2011. [70] T. G. Merrett, M. L. Burr, B. K. Butland, J. Merrett, F. [53] R. M. Lucas, A. L. Ponsonby, K. Dear et al., “Sun exposure and G. Miskelly, and E. Vaughan-Williams, “Infant feeding and vitamin D are independent risk factors for CNS demyelina- allergy: 12-month prospective study of 500 babies born into tion,” Neurology, vol. 76, no. 6, pp. 540–548, 2011. allergic families,” Annals of Allergy, vol. 61, no. 6, pp. 13–20, [54] S. Sharief, S. Jariwala, J. Kumar, P. Muntner, and M. L. 1988. Melamed, “Vitamin D levels and food and environmental [71] R. K. Chandra, S. Puri, and A. Hamed, “Influence of maternal allergies in the United States: results from the National Health diet during lactation and use of formula feeds on development and Nutrition Examination Survey 2005-2006,” Journal of of atopic eczema in high risk infants,” British Medical Journal, Allergy and Clinical Immunology, vol. 127, no. 5, pp. 1195– vol. 298, no. 6693, pp. 228–230, 1989. 1202, 2011. [72]S.Koletzko,P.Sherman,M.Corey,A.Griffiths, and C. Smith, [55] M. F. Holick, “Vitamin D deficiency,” The New England Journal “Role of infant feeding practices in development of Crohn’s of Medicine, vol. 357, pp. 266–281, 2007. disease in childhood,” British Medical Journal, vol. 298, no. [56] M. J. Dealberto, “Prevalence of autism according to maternal 6688, pp. 1617–1618, 1989. immigrant status and ethnic origin,” Acta Psychiatrica Scandi- [73] G. Dick, “President’s address. The etiology of multiple sclero- navica, vol. 123, no. 5, pp. 339–348, 2011. sis,” Proceedings of the Royal Society of Medicine, vol. 69, no. 8, [57] J. J. Cannell, “Autism and vitamin D,” Medical Hypotheses, vol. pp. 611–615, 1976. 70, no. 4, pp. 750–759, 2008. [74] D. Fava, R. D. G. Leslie, and P. Pozzilli, “Relationship between [58] M. Waldman, S. Nicholson, N. Adilov, and J. Williams, dairy product consumption and incidence of IDDM in “Autism prevalence and precipitation rates in California, childhood in Italy,” Diabetes Care, vol. 17, no. 12, pp. 1488– Oregon, and Washington counties,” Archives of Pediatrics and 1490, 1994. Adolescent Medicine, vol. 162, no. 11, pp. 1026–1034, 2008. [75] L. B. Ray, “Linking inflammation and metabolism,” Science’s [59] V. Gilsanz, A. Kremer, A. O. Mo, T. A. L. Wren, and R. Kremer, STKE, vol. 386, Article ID tw165, 2007. “Vitamin D status and its relation to muscle mass and muscle [76] M. Assem, E. G. Schuetz, M. Leggas et al., “Interactions fat in young women,” Journal of Clinical Endocrinology and between hepatic Mrp4 and Sult2a as revealed by the constitu- Metabolism, vol. 95, no. 4, pp. 1595–1601, 2010. tive androstane receptor and Mrp4 knockout mice,” Journal of [60] A. L. Kau, P. P. Ahern, N. W. Griffin, A. L. Goodman, Biological Chemistry, vol. 279, no. 21, pp. 22250–22257, 2004. and J. I. Gordon, “Human nutrition, the gut microbiome [77]O.Burk,K.A.Arnold,A.K.Nussleretal.,“Antimalar- and the immune system,” Nature, vol. 474, no. 7351, pp. ial artemisinin drugs induce cytochrome P450 and MDR1 327–336, 2011. expression by activation of xenosensors pregnane X receptor [61]M.H.deBorst,R.A.deBoer,R.P.Stolk,J.P.J.Slaets,B.H.R. and constitutive androstane receptor,” Molecular Pharmacol- Wolffenbuttel, and G. Navis, “Vitamin D deficiency: universal ogy, vol. 67, no. 6, pp. 1954–1965, 2005. risk factor for multifactorial diseases?” Current Drug Targets, [78] A. Geick, M. Eichelbaum, and O. Burk, “Nuclear receptor vol. 12, no. 1, pp. 97–106, 2011. response elements mediate induction of Intestinal MDR1 by Autism Research and Treatment 11

Rifampin,” Journal of Biological Chemistry, vol. 276, no. 18, pp. 14581–14587, 2001. [79] H. R. Kast, B. Goodwin, P. T. Tarr et al., “Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane X receptor, farnesoid X-activated receptor, and constitutive androstane receptor,” Journal of Biological Chemistry, vol. 277, no. 4, pp. 2908–2915, 2002. [80] E. G. Schuetz, A. H. Schinkel, M. V. Relling, and J. D. Schuetz, “P-glycoprotein: a major determinant of rifampicin-inducible expression of cytochrome P4503A in mice and humans,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 9, pp. 4001–4005, 1996. [81] K. W. Renton, “Alteration of drug biotransformation and elimination during infection and inflammation,” Pharmacol- ogy and Therapeutics, vol. 92, no. 2-3, pp. 147–163, 2001. [82] A. P. Beigneux, A. H. Moser, J. K. Shigenaga, C. Grunfeld, and K. R. Feingold, “Reduction in cytochrome P-450 enzyme expression is associated with repression of CAR (constitutive androstane receptor) and PXR (pregnane X receptor) in mouse liver during the acute phase response,” Biochemical and Biophysical Research Communications, vol. 293, no. 1, pp. 145– 149, 2002. [83] G. Hartmann, A. K. Y. Cheung, and M. Piquette-Miller, “Inflammatory cytokines, but not bile acids, regulate expres- sion of murine hepatic anion transporters in endotoxemia,” Journal of Pharmacology and Experimental Therapeutics, vol. 303, no. 1, pp. 273–281, 2002. [84] E. Siewert, C. G. Dietrich, F. Lammert et al., “Interleukin-6 regulates hepatic transporters during acute-phase response,” Biochemical and Biophysical Research Communications, vol. 322, no. 1, pp. 232–238, 2004. [85] C. S. Song, I. Echchgadda, Y. K. Seo et al., “An essential role of the CAAT/enhancer binding protein-α in the vita- min D-induced expression of the human steroid/bile acid- sulfotransferase (SULT2A1),” Molecular Endocrinology, vol. 20, no. 4, pp. 795–808, 2006. [86] L. A. Schieve, S. Boulet, S. J. Blumberg et al., “Association between parental nativity and autism spectrum disorder among US-born non-Hispanic white and Hispanic children, 2007 National Survey of Children’s Health,” Disability and Health Journal, vol. 5, no. 1, pp. 18–25, 2012. [87] J. Correale, M. Farez, and G. Razzitte, “Helminth infections associated with multiple sclerosis induce regulatory B cells,” Annals of Neurology, vol. 64, no. 2, pp. 187–199, 2008. [88] J. M. Burton, S. Kimball, R. Vieth et al., “A phase I/II dose-escalation trial of vitamin D3 and calcium in multiple sclerosis,” Neurology, vol. 74, no. 23, pp. 1852–1859, 2010. [89] Y. Shen, K. A. Dies, I. A. Holm et al., “Clinical genetic testing for patients with autism spectrum disorders,” Pediatrics, vol. 125, no. 4, pp. e727–e735, 2010. [90] H. O.´ Atladottir,´ P. Thorsen, L. Østergaard et al., “Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders,” Journal of Autism and Develop- mental Disorders, vol. 40, no. 12, pp. 1423–1430, 2010. [91] L. S. Mills, M. F. Soule, and D. F. Doak, “The keystone-species concept in ecology and conservation,” Bioscience, vol. 43, no. 4, pp. 219–224, 1993. [92] C. D. Bethell, M. D. Kogan, B. B. Strickland, E. L. Schor, J. Robertson, and P. W. Newacheck, “A national and state profile of leading health problems and health care quality for US children: key insurance disparities and across-state variations,” Academic Pediatrics, vol. 11, no. 3, pp. S22–S33, 2011. Hindawi Publishing Corporation Autism Research and Treatment Volume 2012, Article ID 190930, 13 pages doi:10.1155/2012/190930

Research Article Prenatal and Postnatal Epigenetic Programming: Implications for GI, Immune, and Neuronal Function in Autism

Mostafa I. Waly,1 Mady Hornig,2 Malav Trivedi,3 Nathaniel Hodgson,3 Radhika Kini,3 Akio Ohta,3 and Richard Deth3

1 Department of Food Science and Nutrition, Sultan Qaboos University, Alkoudh 123, Muscat, Oman 2 Department of Epidemiology, Columbia University, New York, NY 10032, USA 3 Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA

Correspondence should be addressed to Richard Deth, [email protected]

Received 23 January 2012; Accepted 3 May 2012

Academic Editor: Antonio M. Persico

Copyright © 2012 Mostafa I. Waly et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Although autism is first and foremost a disorder of the central nervous system, comorbid dysfunction of the gastrointestinal (GI) and immune systems is common, suggesting that all three systems may be affected by common molecular mechanisms. Substantial systemic deficits in the antioxidant glutathione and its precursor, cysteine, have been documented in autism in association with oxidative stress and impaired methylation. DNA and histone methylation provide epigenetic regulation of gene expression during prenatal and postnatal development. Prenatal epigenetic programming (PrEP) can be affected by the maternal metabolic and nutritional environment, whereas postnatal epigenetic programming (PEP) importantly depends upon nutritional support provided through the GI tract. Cysteine absorption from the GI tract is a crucial determinant of antioxidant capacity, and systemic deficits of glutathione and cysteine in autism are likely to reflect impaired cysteine absorption. Excitatory amino acid transporter 3 (EAAT3) provides cysteine uptake for GI epithelial, neuronal, and immune cells, and its activity is decreased during oxidative stress. Based upon these observations, we propose that neurodevelopmental, GI, and immune aspects of autism each reflect manifestations of inadequate antioxidant capacity, secondary to impaired cysteine uptake by the GI tract. Genetic and environmental factors that adversely affect antioxidant capacity can disrupt PrEP and/or PEP, increasing vulnerability to autism.

1. Introduction As all cells possess the same DNA, their differentiation into various cell types reflects stable suppression of some Neurological and behavioral symptoms implicate abnormal genes and activation of others, accomplished in large part brain development as a core pathophysiological feature of by epigenetic regulation. Such regulation involves reversible autism, but increasing evidence also indicates immune [1–8] modifications of both DNA nucleotides and histone proteins and gastrointestinal (GI) abnormalities in a significant subset [13]. These modifications or epigenetic marks favor or [1, 8–12]. This triad of dysfunctional systems provides not disfavor formation of nucleosomes, DNA/histone complexes only a more complete description of autism and autism spec- that maintain genes in a compacted, inactive state (hete- trum disorders (ASDs), but also an important opportunity rochromatin). Methylation of DNA is the most fundamental to consider the mechanisms which could result in the shared involvement of these three particular systems, especially epigenetic mark, enabling binding of proteins containing in the context of development. Recent elucidation of the methylDNA binding domains, such as methyl CpG binding central role of epigenetic regulation of gene expression in protein 2 (MeCP2). Proteins with methylDNA binding development presents a molecular framework within which domains recruit other proteins, including those capable of prenatal and postnatal maturation of neuronal, immune, and modifying histones at their tail regions [14]. Multiple sites GI systems can be viewed. and forms of histone modification (e.g., methylation, acety- 2 Autism Research and Treatment lation, and phosphorylation) make this a highly complex 2. Materials and Methods mode of regulation, affected by diverse signaling pathways + that adjust gene expression to local cellular metabolic 2.1. Purification of Regulatory T Cells. To purify CD4 CD25+ regulatory T cells, spleen cells and lymph node conditions [15]. Whereas certain epigenetic marks are to cells from C57BL/6 mice were combined as a cell source. some degree reversible, others are not readily reversed and, The cells were labeled with FITC-conjugated anti-CD24 once in place, these may be sustained for an entire lifespan and anti-CD8 mAbs and with anti-FITC microbeads. After and/or be transmitted across generations through germline the removal of CD8+ T cells and CD24-expressing B cells modifications [16]. Accordingly, epigenetic marking or pro- using an AutoMACS separator (Miltenyi Biotec, Auburn, gramming early in development has the potential to exert CA), regulatory T cells were purified by positive selection of long-lasting effects [17]. CD25+ cells using PE-conjugated anti-CD25 mAb and anti- There is considerable evidence that epigenetic pro- PE microbeads. All antibodies except for anti-CD25 mAb gramming is highly sensitive to changes in the cellular were from BD Biosciences (San Jose, CA). Other materials environment, as broadly defined [18]. Indeed, it appears that were from Miltenyi Biotec. Purity of CD4+ CD25+ cells was epigenetic regulation is a widely utilized adaptive mechanism higher than 95 %. to allow cells to maintain a favorable metabolic status under different conditions, including differential exposure to physiological substances (e.g., hormones, neurotransmitters, 2.2. RNA Isolation. RNA was isolated using the RNAqueous- or growth-regulation factors), xenobiotics (e.g., pollutants, 4PCR kit (Ambion, Naugatuck, CT), then treated with toxic chemicals), or even infectious agents (e.g., bacteria or DNase to eliminate contaminating genomic DNA, and viruses, fungi or parasites) [19]. Epigenetic regulation may quantified using an ND-1000 NanoDrop spectrophotometer. facilitate adaptation to changes in the cellular environment through stable alterations of cellular phenotype, potentially 2.3. cDNA Synthesis. cDNA synthesis was performed using resulting in progressive differentiation and maturation dur- the First strand cDNA synthesis (Roche, Nutley, NJ). 1 μg ing fetal and possibly postnatal development [20]. RNA was used as template, along with 1 mM dNTP mix, During prenatal development, nutritional support to 60 uM random hexamer primers and dH20 to make a final μ ◦ the fetus is provided via the transplacental circulation volume of 13 L. Samples were denatured at 65 C for 5 min- μ as a function of the available maternal nutriture, with utes and then placed on ice. Transcriptor RT (20 units/ L), μ metabolic consequences for both the mother and developing Protector RNase inhibitor (40 U/ L), 5x Transcriptor Reverse Transcriptase Reaction Buffer, and dH 0inafinalvolume child. During postnatal development, nutritional support is 2 of 7 μL were used to bring the final volume to 20 μL. This provided by oral food intake into the GI tract: breast-milk- ◦ was followed by incubation at 25 C for 10 min followed by or cow-milk-based formula at first, followed by introduction ◦ 30 min incubation at 55 C. The reverse transcriptase enzyme of other foods, usually in a controlled, gradual manner, but was inhibited by incubating at 85◦C for 5 min and then the provision of adequate nutritional resources to support cooling on ice. further development of the newborn is not assured. Thus, the prenatal/postnatal transition is a critical juncture in metabolic adaptation. This transition is particularly linked 2.4. Primers. Primers were designed using the Invitrogen OligoPerfect Designer to have approximately 50–60% GC to aerobic metabolism, as the delivery of oxygen via newborn content, an annealing temperature of 60◦C, and a length lungs and its ultimate rate of utilization must be balanced by of 20 bases. Glyceraldehyde phosphate dehydrogenase the available antioxidant capacity of body tissues. Epigenetic ff (GAPDH) expression was used for normalization. Primers regulation o ers an opportunity for adaptation during this for EAAT3 (designated EAAC1 in mice) and GAPDH were metabolic transition, allowing the level of ongoing aerobic EAAT3-Forward: 5-TTACAGCCACCGCTGCCAGC-3; metabolism in each organ system, tissue, and cell type to EAAT3-Reverse: 5-GGCAGCCCCACAGCACTCAG-3; be maintained in homeostatic equilibrium with antioxidant GAPDH-Forward: 5-TCTCCACACCTATGGTGCAA-3; metabolism. GAPDH-Reverse: 5-CAAGAAACAGGGGAGCTGAG-3. We use the term postnatal epigenetic programming (PEP) to describe the ongoing adaptive changes in gene 2.5. qRT-PCR Analysis. qRT-PCR was performed on dupli- expression occurring in response to the transition from fetal cate samples using the LightCycler 480 from Roche. 5 μLof to postnatal metabolism, as distinct from prenatal epigenetic cDNA was used for qRTPCR, along with 10 μM sense and programming (PrEP), at term that recognizes the dynamic antisense primers, 10 μL SYBR Green I Master from Roche, occurrence of similar changes which occur in utero. While anddH20inafinalvolumeof20μL. The following thermal it is clear that autism can result from a variety of genetic parameters were used: incubation for 5 min at 95◦C, followed and environmental factors, by focusing on factors affecting by 45 cycles of 95◦C for 10 sec, 60◦C for 20 sec and 72◦Cfor antioxidant and methylation status in the developing GI 30 sec, and lastly a single cycle of 95◦Cfor5sec,1minat65◦C tract, immune system, and brain, we hope to illuminate the and 97◦C for the melting curve. No template controls were pathological mechanisms underlying ASDs and other related run on each plate and dissociation curves were generated to disorders. determine any nonspecific products. Data was normalized to Autism Research and Treatment 3

GAPDH, and the ΔCt second derivative method was used for of intracellular redox status. The relatively high concentra- analysis of EAAT3 expression. tion of GSH within the cell imposes a pervasive influence on essentially all aspects of metabolism. Although all cell types and tissues rely upon the same core antioxidant mechanisms, 2.6. MS Assay. Brain cortex samples were obtained from variations on this central theme are recognized. For example, thimerosal-treated or untreated C57BL/6 and SJL/J mice, as the intracellular level of GSH in liver hepatocytes is close to per the previously published protocol [21]. MS activity in 10 mM [25], whereas in neurons it is 0.2 mM, some 50-fold mouse cortex was determined by measuring incorporation lower [26]. of radiolabel from [methyl-14C]methyltetrahydrofolate into As the rate of ROS formation must be quantitatively methionine, as previously described [22]. Enzyme assays balanced by the available antioxidant capacity, a multitude were performed under anaerobic conditions by bubbling of mechanisms have evolved to restrict aerobic metabolism nitrogen gas through stoppered vials for 1 hour at 37◦Cand when antioxidant status is low and vice versa. One prominent terminated by heating at 98◦C for 2 min after which samples example involves glutathionylation of cysteine residues in were cooled on ice. Radiolabeled methionine was separated complex I of the mitochondrial electron transport chain, from unreacted radiolabeled methylfolate by passing the which limits the flow of electrons, thereby restricting down- reaction mixture through an anion exchange column. The stream ROS formation [27]. In glutathionylation, GSSG, column was washed with 2 mL of water and the aqueous the oxidized form of glutathione, reacts with a reduced samples were collected and counted. Reported values are cysteine, resulting in GSH release and modification of the normalized to protein content and corrected for the counts protein in proportion to the prevailing redox state [28]. This observed in control assays in which sample enzyme was regulatory mechanism not only controls ROS formation but omitted. also provides an auxiliary pathway for glutathione reductase activity and production of GSH. Additional intracellular 2.7. GSH Measurement. A 100 μLaliquotofcortexsuper- control over the balance between ROS and antioxidant natant was incubated with 2 μL of monochlorobimane reservoirs is achieved by the capacity for reversal of the glu- (25 mmol/L) and 2 μL of glutathione-S-transferase reagent tathionylation of complex I by the action of glutaredoxin 2, was added, as provided by a commercial kit (Biovision, in conjunction with the selenoprotein thioredoxin reductase, Mountain View, CA). After a 30 min incubation at 37◦C, using NADPH-derived electrons [27]. fluorescence was read at 380 nm excitation and 460 nm More than ten studies have reported significantly lower emission. GSH content was determined by comparison with plasma levels of GSH in autism, accompanied by lower values from a standard curve using freshly prepared GSH. levels of cysteine [29–39]. Among these studies, the average decrease in GSH was 37%, representing a substantial reduc- tion in antioxidant capacity. Several studies also reported a 2.8. Statistical Methods. Statistical analysis was carried out significant decrease in the GSH to GSSG ratio [8–20, 22–36], using Graph Pad Prism version 5.01. Data was expressed indicative of oxidative stress, consistent with other reports as mean ± standard error of the mean (SEM). Student’s ff of increased biomarkers of oxidative stress [29, 32, 40]. t-test was conducted to determine the di erences between One consequence of persistent oxidative stress, an increase individual groups. in glutathionylation of complex I of the mitochondrial electron transport chain, provides a potential explanation 3. Results and Discussion for the mitochondrial dysfunction frequently reported in autism [7]. Genetic factors affecting mitochondrial function 3.1. Antioxidant Capacity and Aerobic Metabolism. Aerobic may interact with oxidative stress. For example, SLC25A12, metabolism is unavoidably accompanied by the risk of the gene encoding the Ca2+-dependent aspartate-glutamate oxidative damage, most notably caused by reactive oxygen carrier isoform 1 (AGC1), has been identified as a putative species (ROS) generated as by-products of mitochondrial autism susceptibility gene [41]. AGC1 facilitates the supply oxidative phosphorylation and ATP production. Increased of NADH for oxidative phosphorylation, and elevated Ca2+ metabolic activity requires higher rates of ATP production levels will, therefore, increase ROS formation via increased and increased ROS formation. Maintenance of redox equilib- AGC1 activity. Higher Ca2+-dependent AGC1 transport rates rium requires inactivation of ROS before these by-products were demonstrated in autistic subjects [41], and it has been can oxidize cellular components, as well as repair of oxidized proposed that dyregulated Ca2+ levels may be a broadly biomolecules (proteins, lipids, DNA, and RNA). Catalase, influential factor in causing autism [42]. superoxide dismutase and the selenoprotein glutathione peroxidase inactivate ROS, and a wide array of redox-active proteins and enzymes participate in the repair of oxidized 3.2. Intestinal Absorption of Cysteine. Starting from birth, molecules and the maintenance of thiols in their reduced the systemic availability of cysteine to support GSH synthesis state, with glucose-derived NADPH serving as the source of is dependent upon absorption of the food-derived sulfur- reducing electrons [23, 24]. containing amino acids cysteine and methionine from the As elegantly reviewed by Schafer and Buettner [25], the GI tract (Figure 1). Adequate GI uptake of selenium is equilibrium between reduced and oxidized forms of glu- also critical for maintaining GSH in its reduced state. tathione (GSH and GSSG, resp.) is the primary determinant Intestinal epithelial cells express different transporters on 4 Autism Research and Treatment

GI tract Blood Brain

Astrocytes GI Blood epithelial cells brain barrier Cystine GSH GSH GSH EAAT3

Cysteine Cysteine Cysteine Cysteine EAAT3 Methionine Cystine Cystine

GSH Cysteine Dietary protein Neurons

Cysteine Cystine Liver

Figure 1: Absorption and systemic distribution of dietary cysteine and methionine. GI epithelial cells take up cysteine and methionine, with EAAT3 being particularly important for cysteine uptake in the distal ileum. A portion of methionine is converted to cysteine via the methionine cycle and transsulfuration of homocysteine. Absorbed cysteine is taken up by the liver, which releases oxidized cystine. Cystine, but not cysteine, is able to traverse the blood brain barrier and is taken up by astroctyes, which convert it to GSH. Cysteine from astrocyte- derived GSH is available for EAAT3-mediated neuronal uptake, supporting neuronal GSH synthesis. their luminal brush border surface that are critical for proportion of active transporters in the plasma membrane. uptake of sulfur and selenium-containing amino acids. These Oxidative stress inhibits EAAT3 transport activity [47], and amino acids are then subject to intracellular metabolism several cysteine residues have been identified as being critical before being ultimately released on the contralateral surface, for this mode of regulation, raising the possibility that either in their free form or incorporated into proteins or they might be targets for glutathionylation. Oxidative stress peptides, including GSH. Availability of transported cysteine affecting gut epithelium would, therefore, decrease EAAT3- ff or selenium can, therefore, a ect the GSH levels and redox dependent absorption of cysteine, with the local and systemic status of the epithelial cells themselves, as well as that of consequences of lower GSH levels. the adjacent cells located within the intestinal wall, including Cells express a multitude of adaptive mechanisms that immune cells. allow them to maintain homeostatic redox equilibrium, Among amino acid transporters in GI epithelial cells, of which the Keap1/Nrf2 system is a prominent example. EAAT3 (excitatory amino acid transporter 3, EAAC1) is Nrf2 (nuclear factor erythroid 2-like 2) is a transcription selective for cysteine and was initially cloned from GI factor protein with a high turnover rate, that is, tightly epithelial cells [43]. Subsequent studies revealed that EAAT3 bound to Keap-1 (Kelch-like ECH-associated protein 1); is most prominently expressed in the small intestine, espe- however, in response to a more oxidative redox state, Nrf2 cially in the terminal ileum [43, 44], a prominent site of is released from Keap-1 and moves to the nucleus where it inflammation in subjects with autism [45]. The highest levels promotes expression of a large number of genes via binding were found in crypt cells and lower villus regions, the locus to antioxidant response element (ARE) sites [48]. It has of multipotent stem cells that sustain the epithelial lining of been recently demonstrated that Keap1/Nrf2 mediates up- + + the gut [44]. EAAT3 is a member of a family of Na -andH - regulation of EAAT3 in response to oxidative stress [49]. dependent amino acid transporters named for their ability to Similar to autism, GI disorders such as celiac disease transport glutamate and aspartate, but EAAT3 is unique in its and inflammatory bowel disease have an autoimmune preference for cysteine. EAAT3 exists in equilibrium between component [50], and diets excluding gluten and/or casein the endoplasmic reticulum and the plasma membrane, (i.e., GF/CF diet) are frequently beneficial in both conditions. and its transport activity is regulated by the PI3 kinase Prompted by the recognition that digestion of both gluten signaling pathway, similar to insulin regulation of glucose and casein yields peptides with opiate activity [51, 52], we transporters [46]. In resting cells, about 75% of EAAT3 recently examined the activity of these peptides on EAAT3- resides inside the cell, where it is inactive, but activation mediated cysteine uptake and redox status in human GI of either PI3 kinase or protein kinase C increases the epithelial and neuronal cells and found that they significantly Autism Research and Treatment 5

Astrocyte Oxidative stress or Cystine Cysteine gluten/casein Growth Cysteine Cysteinylglycine GSH GSH factors (−) EAAT3 Neuron (+) GSSG GSH PI3-kinase γ-glutamylcysteine

Cysteine Partially blocked in neuronal cells Cystathionine Adenosine Adenosine D4SAH D4HCY HCY SAH (−) MethylTHF MethylTHF > Phospholipid Methionine 200 methylation synthase Methylation reactions THF THF e.g. D4SAM D4MET MET SAM DNA methylation PP+Pi ATP ATP PP+Pi

Dopamine

Figure 2: Redox and methylation pathways in neurons. The amino acid cysteine is rate-limiting for glutathione (GSH) synthesis and it is provided either by uptake from astrocyte-derived cysteine or by transsulfuration of homocysteine (HCY). The methionine cycle of methylation (lower right) depends upon both dietary methionine (MET) and remethylation of HCY by methionine synthase. Since formation of HCY from S-adenosylhomocysteine (SAH) is reversible and SAH inhibits methylation, decreased methionine synthase activity (e.g., caused by oxidative stress) both augments GSH synthesis and inhibits methylation reactions. Thus, redox status and methylation activity are closely linked. The D4 dopamine receptor carries out a cycle of dopamine-stimulated phospholipid methylation which is completely dependent upon methionine synthase and is therefore also sensitive to oxidative stress. Growth factors promote EAAT3 activity by increasing its location on the cell surface. EAAT3 activity is inhibited by oxidative stress and by food-derived opiate peptides. inhibited uptake [53]. Their action was associated with oxidizing conditions) are able to exert epigenetic effects a decrease in GSH levels and the SAM/SAH ratio, along via their influence on MS and the SAM/SAH ratio. Recog- with alteration in the expression of redox-related genes. nizing this metabolic relationship, we previously proposed Important differences, however, were observed between pep- a “redox/methylation hypothesis” of autism, linking this tides derived from bovine versus human casein, which may neurodevelopmental disorder to oxidative stress induced by contribute to the well-recognized benefits of breastfeeding. xenobiotic exposures [55], and now we apply this perspective to epigenetic programming of GI and immune function. 3.3. Postnatal Epigenetic Regulation (PEP). Methylation of During oxidative stress associated with GI inflammation, DNA and histones is fundamental to epigenetic regulation impaired EAAT3 activity will decrease availability of cys- of gene expression, providing factors affecting methylation teine for GSH synthesis, leading to increased ROS levels, capacity with an opportunity to affect development. S- diminished MS activity, a decrease in the SAM/SAH ratio adenosylmethionine (SAM) is the donor of methyl groups and a global decrease in methylation capacity, altering the to more than 200 methyltransferase reactions, including pattern of DNA and histone methylation in epithelial cells. DNA and histone methyltransferases [54], whereas its de- The distribution of EAAT3 suggests that this inflammation- methylated form, S-adenosylhomocysteine (SAH), inhibits induced epigenetic response will be most prominent in crypt methylation by virtue of its competition with SAM. The stem cells of the terminal ileum, where it can influence both SAM to SAH ratio determines methylation capacity for the proliferation and the functionality of the GI epithelium. all methylation reactions, providing an exceptionally broad Notably, the terminal ileum is also a critical location for folate influence over cellular metabolism. and vitamin B12 absorption [56, 57]. The SAM/SAH ratio is responsive to the activity of Postnatal infant nutrition is most commonly provided by methionine synthase (MS), the folate, and vitamin B12- either human breast milk or bovine milk. Human breast milk dependent enzyme that converts homocysteine (HCY) to is comparatively rich in sulfur-containing amino acids, and methionine (MET) as part of the methionine cycle of colostrum contains 2.5-fold higher levels versus mature milk methylation (Figure 2). Its vitamin B12 (cobalamin) cofactor [58]. A comparison of plasma cysteine levels in 12-week- renders MS activity highly sensitive to oxidative stress; old infants found significantly higher levels for breast-fed changes in cellular redox status (shifts toward reducing or versus formula-fed infants [59]. Thus, breast feeding appears 6 Autism Research and Treatment to provide a superior supply of the essential raw materials for GI tract can be matched to the ongoing level of metabolic synthesis of the antioxidant GSH. As this source of cysteine activity. In other words, the supply of antioxidant must be is made available, EAAT3 and the factors that modulate its matched to the demand for antioxidant, and the rate of activityserveascriticalregulatorsoftheextenttowhich postnatal growth and development must be restricted so as cysteine may be absorbed. to not exceed the supply provided by the GI tract. Breast milk contains a number of different growth fac- As a developmental disorder in which the availability tors, including insulin-like growth factor-1 (IGF-1), epider- of GSH is reported to be reduced by almost 40%, autism mal growth factor (EGF), vascular endothelial growth factor can be viewed as a syndrome resulting from an imbalance (VEGF), hepatocyte growth factor (HGF), and transform- between antioxidant supply and demand—essentially, a state ing growth factor-β1andβ2(TGF-β1/β2), among others of oxidative stress that interrupts the normal epigenetically [60, 61]. These factors activate PI3 kinase and mitogen- based program of development. In the absence of a large- activated protein (MAP) kinase signaling pathways to achieve scale prospective study, it is not possible to determine their growth-promoting effects, which are associated with whether the autism-associated GSH deficit is present from increased metabolic activity and its attendant increase in birth or begin during postnatal development. The former ROS production. We recently demonstrated the ability case would likely reflect a significant maternal deficit in of IGF-1 and other PI3 kinase-activating growth factors antioxidant resources, and the finding that plasma levels to stimulate EAAT3-mediated cysteine uptake in cultured of GSH and the SAM/SAH ratio are significantly decreased human neuronal cells, accompanied by an increase in GSH in mothers of autistic children [36] is consistent with levels, an increased SAM/SAH ratio, and increased DNA such a possibility. Under the latter scenario, the imbalance methylation [62]. HGF, acting via the MAP kinase pathway, might develop in a progressive manner during postnatal increases expression of the two enzymes which convert development if escalating metabolic activity and ROS pro- cysteine to GSH [63]. Thus, growth factors promote GSH duction outstripped the ability of the GI tract to provide synthesis via both increased cysteine uptake and transcrip- sufficient antioxidant, and inhibition of EAAT3-mediated tional effects, corresponding to short-term and longer-term cysteine uptake in response to developing oxidative stress effects, respectively. These observations imply that breast- would also exacerbate this self-reinforcing cycle. Postnatal milk-derived growth factors can stimulate cysteine uptake exposure to agents which interfere with antioxidant capacity, and increase GSH in the GI epithelium, coincident with including mercury, lead, pesticides, and other xenobiotics, the initiation of postnatal digestion. The extent to which could shift the redox balance sufficiently to initiate develop- antioxidant resources (i.e., cysteine and GSH) are made mental regression secondary to impaired methylation and its available to the rest of the body depends upon the efficiency epigenetic consequences. of this process. Microbes within the GI tract compete for nutrient Human breast milk is also rich in selenium, and breast- resources in a normally symbiotic manner, but dysbiosis fed infants have higher selenium levels than formula-fed can intervene when the normal array of organisms is infants [64]. A significant proportion (up to 30%) of breast displaced or skewed. GI dysfunction, which is common in milk selenium is in the form of glutathione peroxidase (GPx), autism [8–12], may be associated with a failure of the GI which is hydrolyzed to selenocysteine during digestion. epithelium to absorb antioxidant nutrients, such as cysteine The importance of GPx activity in breast milk may be to or selenocysteine. Increased availability of these critical combat oxidation, especially of vulnerable omega-3 fatty antioxidant nutrients for intestinal microbes may encourage acids. However, GPx appears to also represent an important the growth of previously restricted microorganisms, while reservoir for delivery of selenium to the developing infant. As diminishing the growth of others. Once established, the a structural analog of cysteine, breast-milk-derived seleno- dysbiotic microbial population will decrease availability of cysteine can also be transported by EAAT3 and contributes antioxidant nutrients for absorption by further competing to the antioxidant capacity and redox status [65]. with the host, perpetuating the antioxidant deficit. As MS activity and the SAM/SAH ratio are responsive The importance of PEP varies by cell and tissue types, to redox status, it is reasonable to propose that the post- depending upon the extent of the developmental matu- natal transition to independent nutrition is associated with ration of specific cells and organs at birth. Delaying the epigenetic responses and that growth factor stimulation of development of selected neural networks (e.g., cortex and cysteine uptake is a key event in this transition, which we hippocampus) until after birth allows their programming call postnatal epigenetic programming or PEP.Although PEP to be guided by experience, in contrast to other networks is an ongoing body-wide process affecting the development (e.g., brainstem respiratory centers) which must function of all tissues, but we specifically propose that the GI immediately upon birth. Indeed, unique features of human tract plays a uniquely important role as the source of brain allow PEP to be dynamically exploited throughout the nutritional support for metabolism, and that availability of lifespan, providing the capacity for memory [66]. In a similar antioxidant resources is the critical determinant of the level manner, maturation of the immune system is restrained of metabolic activity that may be achieved without cellular until birth so as to optimize its response to environmental damage. Epigenetic regulation of gene expression provides a exposures, while at the same time minimizing its poten- molecular mechanism by which the functional state of the tial for autoimmunity. While this perspective encompasses Autism Research and Treatment 7 the triad of neuronal, immune, and GI symptom domains in reliance on and retention of selenoproteins are an insufficient autism, the prominence of each component may vary among means of maintaining antioxidant defenses under certain individuals, reflecting, in part, genetic differences in the conditions, such as exposure to mercury. Selenoproteins vulnerability to oxidative stress and impaired methylation. are exquisitely sensitive to inhibition by mercury, and any mercury that penetrates the brain will interfere with redox 3.4. Redox Signaling in Human Brain. As previously noted, signaling and disrupt normal epigenetic regulation [70, 71]. cysteine availability is limited for GSH synthesis. However, Very tight binding of mercury by selenoproteins, especially the level of cysteine in cerebrospinal fluid (CSF) is only to selenoprotein P, contributes to the long-term retention one tenth the level in blood [67], meaning that the raw of mercury in the brain and its accumulation across the material for making antioxidants is much scarcer in the lifespan, and it has been proposed that selenoprotein P,which brain. Indeed, the GSH level in neurons is the lowest contains ten selenocysteine residues, has evolved to serve a reported for any cell type [25], despite the fact that the specific role to protect against mercury toxicity [72]. brain utilizes oxygen at a ten times higher rate than other In addition to conversion of HCY to MET, MS carries tissues. As illustrated in Figure 2, neurons obtain their out a second reaction, providing methyl groups to the D4 cysteine from neighboring astrocytes, which release GSH dopamine receptor; the D4 receptor, when stimulated by that is subsequently broken down to cysteine, which is then dopamine, subsequently transfers these methyl groups to available for uptake by neurons, depending upon the level membrane phospholipids—a unique activity carried out of EAAT3 activity, which is controlled by growth factors. by the D4 dopamine receptor [73]. Genetic variants of Growth factors utilize redox status to regulate neuronal the D4 receptor (such as the 7-repeat variant) have been function via the intermediate involvement of MS, a form linked to novelty-seeking behavior and are important risk of “redox signaling.” By allowing more cysteine into cells factors for attention-deficit/hyperactivity disorder (ADHD) and increasing GSH synthesis, growth factors increase MS [74]. Although the role of dopamine-stimulated phospho- activity and increase DNA methylation. These increases in lipid methylation remains incompletely understood, we first MS activity and DNA methylation lead to epigenetic changes proposed that it facilitates attention by modulating the in gene expression as well as many other metabolic effects frequency of neural networks, promoting their synchro- through >200 methylation reactions, all affected by the nization [75]; others subsequently confirmed dopamine- SAM/SAH ratio. The increase in antioxidant level also allows stimulated phospholipid methylation to be critically involved cells to safely increase their oxidative metabolism (that is, in neuronal synchronization during attention [76]. As D4 their mitochondrial activity); the resultant increase in ATP receptor-mediated phospholipid methylation is absolutely supports a higher level of neuronal activity. In this way, redox dependent upon MS activity, it is thus also dependent on signaling not only regulates gene expression but also controls EAAT3, raising the possibility that ADHD might involve a the level of neuronal function. decrease in MS activity secondary to oxidative stress. A second brain-specific feature that augments redox signaling is a brain-specific limitation in the level of trans- 3.5. Redox and the Immune Response. In the past several sulfuration activity, which restricts conversion of HCY to years, a previously unappreciated role for redox regulation cysteine [68]. This unique feature contributes to the low of the immune response has been elucidated, involving GSH level in neurons, making them more responsive to extrusion of GSH from activated antigen presenting cells growth factor-induced EAAT3 activation. Together with the (APCs, e.g., dendritic cells, macrophages, or B cells), fol- limited availability of extracellular cysteine, this makes epi- lowed by release of its cysteine, which is then taken up by T genetic regulation exceptionally dynamic. Along with other cells [77] This relationship is analogous to the relationship methylation-dependent consequences of redox signaling, between astrocytes and neurons illustrated in Figure 2. the long-term nature of epigenetic effects endows neurons Uptake of this cysteine by na¨ıve CD4+ CD25− effector T with the ability to create “metabolic memories” that are cells (Teff) leads to their activation and proliferation, in coordinated with experience via the input of sensory systems. association with increased GSH synthesis and the epigenetic This view is consistent with recent studies identifying the consequences thereof. Activated Teff cells release cytokines central role of epigenetic changes in learning and memory that attract other cells and promote inflammation, as well as formation [66, 69]. Compromised function of these redox- stimulate B cell activation and antibody production (Helper dependent systems could contribute to the types of cognitive T cells). However, the extent of Teff cell activation is limited function deficits and neurodevelopmental delays that are by CD4+ CD25+ Foxp3+ regulatory T cells (Treg) that com- primary to autism. pete with Teff cells for available extracellular cysteine, thereby The restricted availability of GSH in the brain is partly suppressing immune and autoimmune responses [77]. compensated for by the presence of alternate mechanisms Recognizing this newly described mode of T cell regu- for sustaining antioxidant capacity, including an increased lation, we hypothesized that EAAT3 might be involved in dependence upon selenoproteins. The brain has developed cysteine uptake. To evaluate this possibility, we used qRT- the capacity to retain selenium under conditions of scarcity, PCR to determine whether EAAT3 was expressed in murine even when other tissues become depleted. Nonetheless, T cells and compared the level of expression in T cell subsets, 8 Autism Research and Treatment

2 40

30 ∗ 1.5 20 ∗∗ ∗∗ g/mg protein) μ

GSH ( 10 1

0 C57BL6/J SJL/J Thimerosal C57BL6/J 0.5 Thimerosal SJL/J

EAAT3 expression normalized to GAPDH normalized to expression EAAT3 Figure 4: GSH levels in frontal cortex of SJL/J and C57BL6/J mice with or without thimerosal treatment. GSH levels were significantly lower (P<0.05) in cortex of SJL/J autoimmune-prone mice compared to C57BL6/J strain mice. GSH levels were not affected 0 by thimerosal treatment. UNSEP CD4+ T cells CD8+ T cells B cells (a) expression by immune cells provides a mechanism for them to take up cysteine in a competitive manner, and higher EAAT3 expression in Treg cells may underlie the ability of ∗ this cell subset to limit the immune response and restrict autoimmunity. 4 The SJL/J strain of mice has been widely employed as a model for autoimmunity, but these mice also exhibit deficits in GSH, similar to those reported in autistic children. In SJL/J mice, the deficit is due to mutations in the enzymes which synthesize GSH; factors underlying GSH deficits in autism are as yet unknown. In a previous study, we showed that the thiol-reactive compound thimerosal caused 2 growth impairment, as well as neurodevelopmental and neurochemical effects in SJL/J mice, but not in C57BL6/J mice, including upregulated brain levels of EAAT3 [21]. To EAAT3 expression normalized to GAPDH normalized to expression EAAT3 further characterize the role of redox and methylation status in autoimmunity, we measured both GSH levels and MS activity in the liver and brain of control and thimerosal- 0 treated SJL/J mice, as compared to C57BL6/J mice. GSH Treg CD4+ levels were significantly lower in both liver and brain of SJL/J (b) as compared with the levels found in C57BL6/J mice, but they did not differ as a function of thimerosal treatment Figure 3: Expression of EAAT3 in murine lymphocyte subsets. in either strain (Figure 4). Methionine synthase activity Spleen and lymph node-derived lymphocytes were separated by was also significantly lower in SJL/J versus C57BL6/J mice, magnetic cell sorting, followed by qRT-PCR analysis for EAAT3 ff (EAAC1) expression. (a) EAAT3 expression was significantly higher but not di erent in thimerosal-treated animals (Figure 5). (P<0.05) in CD4+ T cells as compared to unseparated lymphocytes These results confirm the occurrence of low GSH in the (UNSEP), CD8+ T cells, or B cells. (b) EAAT3 expression was autoimmune-prone SJL/J strain, along with the novel finding significantly higher (P<0.05) in FoxP3+ Treg cells. that MS activity is reduced, although neonatal exposure to thimerosal did not alter these features. Expression of EAAT3 was significantly lower in CD4+ T-cells from SJL/J mice separated on the basis of surface epitopes by magnetic cell versus C57BL6/J mice, suggesting that autoimmunity is asso- sorting. As illustrated in Figure 3(a), EAAT3 expression was ciated with impaired capacity for cysteine uptake (Figure 6). detected in unseparated cells, as well as in CD4+ and CD8+ T Together these findings provide support for the roles of cells and B cell populations, with the highest expression level oxidative stress and impaired methylation in autoimmunity. in CD4+ cells. Further analysis showed that the expression Abnormal immune function, including, but not limited of EAAT3 by Treg cells is almost two-fold higher than to, increased autoimmunity, has been extensively docu- that in unseparated CD4+ cells (Figure 3(b)). Thus, EAAT3 mented in autism. Of particular relevance are autoantibodies Autism Research and Treatment 9

20 2

15

1.5 10 ∗∗ ∗∗

5 ∗ ∗∗ 1 MS activity (pmol/min/mg protein)

0 expression EAAT3 MethylB12 HydroxoB12 0.5 C57BL6/J SJL/J Thimerosal C57BL6/J Thimerosal SJL/J ∗ Figure 5: Methionine synthase activity in cortex of SJl/J and C57BL6/J mice with or without thimerosal treatment. Methionine 0 synthase activity was measured in the presence of either methylco- C57 SJL balamin (MethylB12) or hydroxocobalamin (HydroxoB12). Activity was significantly lower in SJL/J autoimmune-prone mice compared C57BL6/J to the C57BL6/J strain, irrespective of thimerosal treatment. SJL/J Activity was higher in the presence of methylcobalamin. ∗P<0.02 compared to C57BL6/J group, ∗∗P<0.001 compared to C57BL6/J Figure 6: Expression of EAAT3 in CD4+ T-cells from C57BL6/J group. andSJL/Jmice.Spleenandlymphnode-derivedlymphocyteswere separated by FACS analysis cell sorting, followed by qRT-PCR analysis for EAAT3 (EAAC1) expression. EAAT3 expression was significantly higher in CD4+ T-cells from C57BL6/J mice versus targeting brain and neuronal proteins [2, 3, 5]aswellas autoimmune-prone SJL/J mice (P<0.05). autoantibodies targeting the folate receptor [78–80]. The latter were initially linked to the relatively rare cerebral folate deficiency syndrome [78] but recently were found to 4. Conclusions be present in a high proportion (75%) of autistic subjects [80], and a casein-free diet greatly reduced the circulating The prominence of neurological, GI, and immune symptoms level of anti-folate receptor antibodies [79]. The number in autism suggests a shared mechanism of dysregulation that of circulating Treg cells was reported to be significantly may relate to the significant deficits in systemic reservoirs lower in autistic subjects, with 73% showing abnormally low of antioxidant GSH reported in this neurodevelopmental levels [4]. The Treg cell decrease was more prominent in disorder. Impaired GI absorption of cysteine, the essential the more severe cases, as well as among individuals with GSH precursor, is proposed as a crucial factor in the allergic manifestations such as asthma and atopic dermatitis. pathogenesis of this triad of symptoms, wherein reduced Although autistic subjects with low Treg cells did not cysteine availability leads to a condition of local and sys- manifest clinical or laboratory-based signs of autoimmunity temic oxidative stress, and subsequent disruption of normal (arthritis or arthralgia, elevated erythrocyte sedimentation epigenetic regulation of gene expression. In some cases, the rate, or leucopenia), the frequency of autoimmunity was extent of impaired cysteine absorption may be severe enough higher in their families. The same researchers found ele- to result in overt GI inflammation, while in other cases vated markers of oxidative stress (increased plasma F2- the restriction may only alter immune and/or neurological isoprostane and/or decreased GPx activity) in 89% of autistic development and function. Awareness of the redox-based subjects and evidence of antineuronal antibodies in 55%, linkage between GI, brain, and immune systems serves to leading them to suggest a link between oxidative stress illuminate a number of diseases whose origins may reflect and autoimmunity [5]. Notably, expression of Foxp3, the the critically important roles of PrEP and PEP. Indeed, transcription factor which defines Treg cells, is epigenetically adaptive epigenetic responses to changes in redox status are regulated, with increased expression when DNA methylation likely to play a critical role in diseases arising across the is decreased [81], and Treg cell levels are elevated in lifespan, especially those that can be traced to environmental male, but not female, SJL mice [82]. As illustrated in exposures that interfere with antioxidant homeostasis. Figure 7, these observations suggest that autoimmunity in autism may be related to inflammation in the distal ileum, Acknowledgment where impaired activity of EAAT3 in Treg cells can lead to formation of autoantibodies directed against the folate This report was supported in part by a research grant from receptor. SafeMinds to R Deth. 10 Autism Research and Treatment

TerminalTerminalalal partparpaa t of ileocolicileoiil colic c artearteryr CecaCecalal brbbranchesan IleaIleall branchesbrb

Ileum Terminal ileum

Cecum AAppendicularpp ararteryte

Vermiformf process

GI Anti-FR-Ab Body lumen

APC Epigenetic regulation Growth factors (+) + FR CD4 Redox status Opiate peptides (−) Na¨ıve T-cell GSH GSH GSSG GSH CYS CYS Redox CYS status Se-CYS Treg CYS Se-proteins Se-CYS cell EAAT3 Mucosal immune cells GI epithelial cell Figure 7: Cysteine and selenocysteine uptake in the terminal ileum regulates autoimmunity. EAAT3 transports diet-derived cysteine (CYS) and selenocysteine (Sel-CYS) into GI epithelial cells in the terminal ileum. Within the cell, cysteine is directed to GSH synthesis and selenocysteine is incorporated into selenoproteins, which maintain GSH in its reduced state. EAAT3 activity is regulated by multiple factors, including intracellular redox status, Nrf2-dependent transcription, growth-factor-dependent translocation to the cell surface, and the inhibitory effects of casein and gluten-derived opiate peptides. Systemic availability of cysteine and GSH depends upon these terminal ileum events, as does the local mucosal environment. Immune cells in the terminal ileum can be a source of auto-antibodies, including anti-folate receptor (FR) antibodies, which are commonly present in subjects with autism [80]. Under oxidative stress conditions, EAAT3- mediated uptake of cysteine by Treg cells is impaired, limiting their suppression of na¨ıve CD4+ T cells, and increasing autoimmune responses to antigens present locally, such as the folate receptor.

References stress, mitochondrial dysfunction and environmental toxicant exposures,” Molecular Psychiatry, vol. 17, no. 4, pp. 389–401, [1]P.Ashwood,A.Anthony,F.Torrente,andA.J.Wake- 2012. field, “Spontaneous mucosal lymphocyte cytokine profiles [8] H. Jyonouchi, L. Geng, D. L. Streck, and G. A. Toruner, in children with autism and gastrointestinal symptoms: “Children with autism spectrum disorders (ASD) who exhibit mucosal immune activation and reduced counter regulatory chronic gastrointestinal (GI) symptoms and marked fluctua- interleukin-10,” Journal of Clinical Immunology, vol. 24, no. 6, tion of behavioral symptoms exhibit distinct innate immune pp. 664–673, 2004. abnormalities and transcriptional profiles of peripheral blood [2] C. A. Pardo, D. L. Vargas, and A. W. Zimmerman, “Immunity, (PB) monocytes,” Journal of Neuroimmunology, vol. 238, pp. neuroglia and neuroinflammation in autism,” International 73–80, 2011. Review of Psychiatry, vol. 17, no. 6, pp. 485–495, 2005. [9] B. L. Williams, M. Hornig, T. Buie et al., “Impaired car- [3] P. Goines and J. van de Water, “The immune system’s role in bohydrate digestion and transport and mucosal dysbiosis in the biology of autism,” Current Opinion in Neurology, vol. 23, the intestines of children with autism and gastrointestinal no. 2, pp. 111–117, 2010. disturbances,” PLoS ONE, vol. 6, no. 9, Article ID e24585, [4]G.A.Mostafa,A.AlShehab,andN.R.Fouad,“Frequency 2011. of CD4+CD25 high regulatory T cells in the peripheral blood of Egyptian children with autism,” Journal of Child Neurology, [10] T. Buie, G. J. Fuchs III, G. T. Furuta et al., “Recommendations vol. 25, no. 3, pp. 328–335, 2010. for evaluation and treatment of common gastrointestinal [5] G. A. Mostafa, E. S. El-Hadidi, D. H. Hewedi, and M. M. problems in children with ASDs,” Pediatrics, vol. 125, no. 1, Abdou, “Oxidative stress in Egyptian children with autism: pp. S19–S29, 2010. relation to autoimmunity,” Journal of Neuroimmunology, vol. [11] T. Buie, D. B. Campbell, G. J. Fuchs et al., “Evaluation, 219, no. 1-2, pp. 114–118, 2010. diagnosis, and treatment of gastrointestinal disorders in [6] C. Onore, M. Careaga, and P. Ashwood, “The role of individuals with ASDs: a consensus report,” Pediatrics, vol. immune dysfunction in the pathophysiology of autism,” Brain, 125, no. 1, pp. S1–S18, 2010. Behavior, and Immunity, vol. 26, no. 3, pp. 383–392, 2012. [12] R. N. Nikolov, K. E. Bearss, J. Lettinga et al., “Gastrointestinal [7] D. A. Rossignol and R. E. Frye, “A review of research symptoms in a sample of children with pervasive developmen- trends in physiological abnormalities in autism spectrum tal disorders,” Journal of Autism and Developmental Disorders, disorders: immune dysregulation, inflammation, oxidative vol. 39, no. 3, pp. 405–413, 2009. Autism Research and Treatment 11

[13] A. P. Feinberg, “Epigenetics at the epicenter of modern glutathione levels,” Journal of Toxicology, vol. 2009, Article ID medicine,” Journal of the American Medical Association, vol. 532640, 7 pages, 2009. 299, no. 11, pp. 1345–1350, 2008. [31] E.´ Pastural, S. Ritchie, Y. Lu et al., “Novel plasma phospholipid [14] J. M. Lasalle and D. H. Yasui, “Evolving role of MeCP2 in Rett biomarkers of autism: mitochondrial dysfunction as a putative syndrome and autism,” Epigenomics, vol. 1, pp. 119–130, 2009. causative mechanism,” Prostaglandins Leukotrienes and Essen- [15] Y. Natsume-Kitatani, M. Shiga, and H. Mamitsuka, “Genome- tial Fatty Acids, vol. 81, no. 4, pp. 253–264, 2009. wide integration on transcription factors, histone acetylation [32] Y. Al-Gadani, A. El-Ansary, O. Attas, and L. Al-Ayadhi, and gene expression reveals genes Co-regulated by histone “Metabolic biomarkers related to oxidative stress and antiox- modification patterns,” PLoS ONE, vol. 6, no. 7, Article ID idant status in Saudi autistic children,” Clinical Biochemistry, e22281, 2011. vol. 42, no. 10-11, pp. 1032–1040, 2009. [16] M. K. Skinner and C. Guerrero-Bosagna, “Environmental [33] S. P. Pas¸ca, E. Dronca, T. Kaucsar´ et al., “One carbon signals and transgenerational epigenetics,” Epigenomics, vol. 1, metabolism disturbances and the C677T MTHFR gene pp. 111–117, 2009. polymorphism in children with autism spectrum disorders,” [17] F. Perera and J. Herbstman, “Prenatal environmental expo- Journal of Cellular and Molecular Medicine, vol. 13, no. 10, pp. sures, epigenetics, and disease,” Reproductive Toxicology, vol. 4229–4238, 2009. 31, no. 3, pp. 363–373, 2011. [34] D. A. Geier, J. K. Kern, C. R. Garver et al., “Biomarkers of [18] R. Feil and M. F. Fraga, “Epigenetics and the environment: environmental toxicity and susceptibility in autism,” Journal of emerging patterns and implications,” Nature Reviews Genetics, the Neurological Sciences, vol. 280, no. 1-2, pp. 101–108, 2009. vol. 13, pp. 97–109, 2012. [35] D. A. Geier, J. K. Kern, C. R. Garver, J. B. Adams, T. Audhya, [19] Z. Hochberg, R. Feil, M. Constancia et al., “Child health, and M. R. Geier, “A prospective study of transsulfuration developmental plasticity, and epigenetic programming,” biomarkers in autistic disorders,” Neurochemical Research, vol. Endocrine Reviews, vol. 32, no. 2, pp. 159–224, 2011. 34, no. 2, pp. 386–393, 2009. [20] J. M. LaSalle, “A genomic point-of-view on environmental [36]S.JillJames,S.Melnyk,S.Jernigan,A.Hubanks,S.Rose,and factors influencing the human brain methylome,” Epigenetics, D. W. Gaylor, “Abnormal transmethylation/transsulfuration vol. 6, no. 7, pp. 862–869, 2011. metabolism and DNA hypomethylation among parents of ff [21] M. Hornig, D. Chian, and W. I. Lipkin, “Neurotoxic e ects of children with autism,” Journal of Autism and Developmental postnatal thimerosal are mouse strain dependent,” Molecular Disorders, vol. 38, no. 10, pp. 1966–1975, 2008. Psychiatry, vol. 9, no. 9, pp. 833–845, 2004. [37] S. J. James, S. Melnyk, S. Jernigan et al., “Metabolic endophe- [22] M. I. Waly, K. K. Kharbanda, and R. C. Deth, “Ethanol lowers notype and related genotypes are associated with oxidative glutathione in rat liver and brain and inhibits methionine stress in children with autism,” American Journal of Medical synthase in a cobalamin-dependent manner,” Alcoholism, vol. Genetics B, vol. 141, no. 8, pp. 947–956, 2006. 35, no. 2, pp. 277–283, 2011. [38] S. J. James, P.Cutler, S. Melnyk et al., “Metabolic biomarkers of [23] D. P. Jones and Y. M. Go, “Redox compartmentalization and increased oxidative stress and impaired methylation capacity cellular stress,” Diabetes, Obesity and Metabolism, vol. 12, no. in children with autism,” The American Journal of Clinical 2, pp. 116–125, 2010. Nutrition, vol. 80, no. 6, pp. 1611–1617, 2004. [24] E. S. J. Arner´ and A. Holmgren, “Physiological functions of [39] S. Melnyk, G. J. Fuchs, E. Schulz et al., “Metabolic imbal- thioredoxin and thioredoxin reductase,” European Journal of ance associated with methylation dysregulation and oxidative Biochemistry, vol. 267, no. 20, pp. 6102–6109, 2000. damage in children with autism,” Journal of Autism and [25] F. Q. Schafer and G. R. Buettner, “Redox environment of the Developmental Disorders, vol. 42, no. 3, pp. 367–377, 2011. cell as viewed through the redox state of the glutathione disul- fide/glutathione couple,” Free Radical Biology and Medicine, [40] A. Chauhan and V. Chauhan, “Oxidative stress in autism,” vol. 30, no. 11, pp. 1191–1212, 2001. Pathophysiology, vol. 13, no. 3, pp. 171–181, 2006. [26] X. Sun, A. Y. Shih, H. C. Johannssen, H. Erb, P. Li, and T. [41] L. Palmieri, V. Papaleo, V. Porcelli et al., “Altered calcium H. Murphy, “Two-photon imaging of glutathione levels in homeostasis in autism-spectrum disorders: evidence from intact brain indicates enhanced redox buffering in developing biochemical and genetic studies of the mitochondrial aspar- neurons and cells at the cerebrospinal fluid and blood-brain tate/glutamate carrier AGC1,” Molecular Psychiatry, vol. 15, interface,” Journal of Biological Chemistry, vol. 281, no. 25, pp. no. 1, pp. 38–52, 2010. 17420–17431, 2006. [42] V. Napolioni, A. M. Persico, V. Porcelli, and L. Palmieri, “The [27] S. M. Beer, E. R. Taylor, S. E. Brown et al., “Glutaredoxin mitochondrial aspartate/glutamate carrier AGC1 and calcium 2 catalyzes the reversible oxidation and glutathionylation homeostasis: physiological links and abnormalities in autism,” of mitochondrial membrane thiol proteins: implications for Molecular Neurobiology, vol. 44, no. 1, pp. 83–92, 2011. mitochondrial redox regulation and antioxidant defense,” [43] Y. Kanai and M. A. Hediger, “Primary structure and functional Journal of Biological Chemistry, vol. 279, no. 46, pp. 47939– characterization of a high-affinity glutamate transporter,” 47951, 2004. Nature, vol. 360, no. 6403, pp. 467–471, 1992. [28] I. Dalle-Donne, R. Rossi, G. Colombo, D. Giustarini, and [44] T. Iwanaga, M. Goto, and M. Watanabe, “Cellular distribution A. Milzani, “Protein S-glutathionylation: a regulatory device of glutamate transporters in the gastrointestinal tract of from bacteria to humans,” Trends in Biochemical Sciences, vol. mice. An immunohistochemical and in situ hybridization 34, no. 2, pp. 85–96, 2009. approach,” Biomedical Research, vol. 26, no. 6, pp. 271–278, [29] J. B. Adams, T. Audhya, S. McDonough-Means et al., 2006. “Nutritional and metabolic status of children with autism [45] A. J. Wakefield, P. Ashwood, K. Limb, and A. Anthony, “The vs. children, and the association with autism significance of ileo-colonic lymphoid nodular hyperplasia in severity,” Nutrition and Metabolism, vol. 8, article 34, 2011. children with autistic spectrum disorder,” European Journal of [30] J. B. Adams, M. Baral, E. Geis et al., “The severity of autism Gastroenterology and Hepatology, vol. 17, no. 8, pp. 827–836, is associated with toxic metal body burden and red blood cell 2005. 12 Autism Research and Treatment

[46] K. Aoyama, M. Watabe, and T. Nakaki, “Regulation of [62] N. Hodgson, M. Waly, and R. C. Deth, “EAAT3-mediated neuronal glutathione synthesis,” Journal of Pharmacological cysteine uptake in human neuroblastoma cells is PI3 kinase- Sciences, vol. 108, no. 3, pp. 227–238, 2008. dependent,” in Proceedings of the Society for Neuroscience [47] D. Trotti, N. C. Danbolt, and A. Volterra, “Glutamate Meeting, November 2008. transporters are oxidant-vulnerable: a molecular link between [63] H. Yang, N. Magilnick, M. Xia, and S. C. Lu, “Effects of oxidative and excitotoxic neurodegeneration?” Trends in Phar- hepatocyte growth factor on glutathione synthesis, growth, macological Sciences, vol. 19, no. 8, pp. 328–334, 1998. and apoptosis is cell density-dependent,” Experimental Cell [48] S. Singh, S. Vrishni, B. K. Singh, I. Rahman, and P. Kakkar, Research, vol. 314, no. 2, pp. 398–412, 2008. “Nrf2-ARE stress response mechanism: a control point in [64] J. G. Dorea, “Selenium and breast-feeding,” British Journal of oxidative stress-mediated dysfunctions and chronic inflam- Nutrition, vol. 88, no. 5, pp. 443–461, 2002. matory diseases,” Free Radical Research, vol. 44, no. 11, pp. [65] S. D Watts, D. Torres-Salazar, C. B. Divito, and S. G. Amara, 1267–1288, 2010. “EAAT3 cysteine transport: potential mechanism of cysteine [49] C. Escartin, S. J. Won, C. Malgorn et al., “Nuclear factor binding and translocation,” in Proceedings of the Society for erythroid 2-related factor 2 facilitates neuronal glutathione Neuroscience Meeting, November 2009. synthesis by upregulating neuronal excitatory amino acid [66] J. J. Day and J. D. Sweatt, “Epigenetic mechanisms in transporter 3 expression,” Journal of Neuroscience, vol. 31, no. cognition,” Neuron, vol. 70, no. 5, pp. 813–829, 2011. 20, pp. 7392–7401, 2011. [67] A. Castagna, C. Le Grazie, A. Accordini et al., “Cerebrospinal fluid S-adenosylmethionine (SAMe) and glutathione concen- [50] S. Caja, M. Maki,¨ K. Kaukinen, and K. Lindfors, “Antibodies in trations in HIV infection: effect of parenteral treatment with celiac disease: implications beyond diagnostics,” Cellular and SAMe,” Neurology, vol. 45, no. 9, pp. 1678–1683, 1995. Molecular Immunology, vol. 8, no. 2, pp. 103–109, 2011. [68] H. H. Tallan, S. Moore, and W. H. Stein, “L-cystathionine in [51] F. R. Huebner, K. W. Lieberman, R. P. Rubino, and J. S. human brain,” The Journal of Biological Chemistry, vol. 230, Wall, “Demonstration of high opioid-like activity in isolated no. 2, pp. 707–716, 1958. peptides from wheat gluten hydrolysates,” Peptides, vol. 5, no. [69] J. Gra¨ff, D. Kim, M. M. Dobbin, and T. Li-Huei, “Epigenetic 6, pp. 1139–1147, 1984. regulation of gene expression in physiological and patholog- [52] H. Teschemacher, “Opioid receptor ligands derived from food ical brain processes,” Physiological Reviews, vol. 91, no. 2, pp. proteins,” Current Pharmaceutical Design, vol. 9, no. 16, pp. 603–649, 2011. 1331–1344, 2003. [70] C. M. L. Carvalho, E. H. Chew, S. I. Hashemy, J. Lu, and [53] M. S. Trivedi, N. W. Hodgson, and R. C. Deth, “Casein and A. Holmgren, “Inhibition of the human thioredoxin system: gluten-derived peptides modulate cysteine uptake and cellular a molecular mechanism of mercury toxicity,” Journal of thiol levels via activation of opiate receptors,” Society for Biological Chemistry, vol. 283, no. 18, pp. 11913–11923, 2008. Neuroscience Meeting. In press. [71] N. V. C. Ralston and L. J. Raymond, “Dietary selenium’s [54] T. C. Petrossian and S. G. Clarke, “Uncovering the human protective effects against methylmercury toxicity,” Toxicology, methyltransferasome,” Molecular and Cellular Proteomics.In vol. 278, no. 1, pp. 112–123, 2010. press. [72] K. T. Suzuki, C. Sasakura, and S. Yoneda, “Binding sites for the [55] R. Deth, C. Muratore, J. Benzecry, V. A. Power-Charnitsky, (Hg-Se) complex on selenoprotein P,” Biochimica et Biophysica and M. Waly, “How environmental and genetic factors Acta, vol. 1429, no. 1, pp. 102–112, 1998. combine to cause autism: a redox/methylation hypothesis,” [73] A. Sharma, M. L. Kramer, P. F. Wick et al., “D4 dopamine NeuroToxicology, vol. 29, no. 1, pp. 190–201, 2008. receptor-mediated phospholipid methylation and its implica- [56] T. Ganesan, M. H. Khadra, J. Wallis, and D. E. Neal, “Vitamin tions for mental illnesses such as schizophrenia,” Molecular B12 malabsorption following bladder reconstruction or diver- Psychiatry, vol. 4, no. 3, pp. 235–246, 1999. sion with bowel segments,” ANZ Journal of Surgery, vol. 72, no. [74] J. M. Swanson, M. Kinsbourne, J. Nigg et al., “Etiologic 7, pp. 479–482, 2002. subtypes of attention-deficit/hyperactivity disorder: brain imaging, molecular genetic and environmental factors and the [57] S. Vasilopoulos, K. Saiean, J. Emmons et al., “Terminal ileum dopamine hypothesis,” Neuropsychology Review, vol. 17, no. 1, resection is associated with higher plasma homocysteine levels pp. 39–59, 2007. in Crohn’s disease,” Journal of Clinical Gastroenterology, vol. [75] A. Y. Kuznetsova and R. C. Deth, “A model for modulation of 33, no. 2, pp. 132–136, 2001. neuronal synchronization by D4 dopamine receptor-mediated [58] M. E. McNally, S. A. Atkinson, and D. E. C. Cole, “Contribu- phospholipid methylation,” Journal of Computational Neuro- tion of sulfate and sulfoesters to total sulfur intake in infants science, vol. 24, no. 3, pp. 314–329, 2008. fed human milk,” Journal of Nutrition, vol. 121, no. 8, pp. [76] T. Demiralp, C. S. Herrmann, M. E. Erdal et al., “DRD4 1250–1254, 1991. and DAT1 polymorphisms modulate human gamma band [59] J. C. Miner, E. Bisse,´ C. P. Aebischer, A. Bell, H. Wieland, and responses,” Cerebral Cortex, vol. 17, no. 5, pp. 1007–1019, J. Lutschg, “Assessment of vitamin B-12, folate, and vitamin 2007. B-6 status and relation to sulfur amino acid metabolism in [77] Z. Yan, S. K. Garg, and R. Banerjee, “Regulatory T cells neonates,” The American Journal of Clinical Nutrition, vol. 72, interfere with glutathione metabolism in dendritic cells and no. 3, pp. 751–757, 2000. Tcells,”Journal of Biological Chemistry, vol. 285, no. 53, pp. [60] T. Ozgurtas, I. Aydin, O. Turan et al., “Vascular endothelial 41525–41532, 2010. growth factor, basic fibroblast growth factor, insulin-like [78] V. T. Ramaekers, N. Blau, J. M. Sequeira, M. C. Nassogne, and growth factor-I and platelet-derived growth factor levels in E. V. Quadros, “Folate receptor autoimmunity and cerebral human milk of mothers with term and preterm neonates,” folate deficiency in low-functioning autism with neurological Cytokine, vol. 50, no. 2, pp. 192–194, 2010. deficits,” Neuropediatrics, vol. 38, no. 6, pp. 276–281, 2007. [61] M. S. Murphy, “Growth factors and the gastrointestinal tract,” [79] V. T. Ramaekers, J. M. Sequeira, N. Blau, and E. V. Quadros, “A Nutrition, vol. 14, no. 10, pp. 771–774, 1998. milk-free diet downregulates folate receptor autoimmunity in Autism Research and Treatment 13

cerebral folate deficiency syndrome,” Developmental Medicine and Child Neurology, vol. 50, no. 5, pp. 346–352, 2008. [80] R. E. Frye, J. M. Sequeira, E. V. Quadros, S. J. James, and D. A. Rossignol, “Cerebral folate receptor autoantibodies in autism spectrum disorder,” Molecular Psychiatry. In press. [81] J. K. Polansky, L. Schreiber, C. Thelemann et al., “Methylation matters: Binding of Ets-1 to the demethylated Foxp3 gene con- tributes to the stabilization of Foxp3 expression in regulatory T cells,” Journal of Molecular Medicine, vol. 88, no. 10, pp. 1029– 1040, 2010. [82] S. Hussain, S. J. Kirwin, and S. A. Stohlman, “Increased T regulatory cells lead to development of Th2 immune response in male SJL mice,” Autoimmunity, vol. 44, no. 3, pp. 219–228, 2011. Hindawi Publishing Corporation Autism Research and Treatment Volume 2012, Article ID 205362, 4 pages doi:10.1155/2012/205362

Research Article Decreased Levels of EGF in Plasma of Children with Autism Spectrum Disorder

Charity Onore,1, 2 Judy Van de Water,2, 3 and Paul Ashwood1, 2

1 Department of Medical Microbiology and Immunology, University of California, Davis, USA 2 The M.I.N.D. Institute, University of California, Davis, Sacramento, CA 95817, USA 3 Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, USA

Correspondence should be addressed to Paul Ashwood, [email protected]

Received 14 September 2011; Revised 3 November 2011; Accepted 16 December 2011

Academic Editor: Antonio M. Persico

Copyright © 2012 Charity Onore et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder estimated to affect 1 in 110 children in the U.S., yet the pathology of this disorder is not fully understood. Abnormal levels of several growth factors have been demonstrated in adults with ASD, including epidermal growth factor (EGF) and hepatocyte growth factor (HGF). Both of these growth factors serve important roles in neurodevelopment and immune function. In this study, concentrations of EGF and HGF were assessed in the plasma of 49 children with ASD aged 2–4 years old, and 31 typically developing controls of a similar age as part of the Autism Phenome Project (APP). Levels of EGF were significantly reduced in the ASD group compared to typically developing controls (P = 0.003). There were no significant differences in HGF levels in young children with ASD and typically developing controls. EGF plays an important role in regulating neural growth, proliferation, differentiation and migration, and reduced levels of this molecule may negatively impact neurodevelopment in young children with ASD.

1. Introduction [10–12], which may be influenced by atypical growth factor activity. Growth factor dysregulation may contribute to ASD Autism Spectrum Disorder (ASD) is a developmental disor- pathology by directly affecting CNS development, and/or by der characterized by impairments in social interaction and augmenting immune function. communication, and the presence of restricted behaviors or Epidermalgrowthfactor(EGF)andhepatocytegrowth interests [1]. According to the most current CDC estimate, factor (HGF) are both involved in the growth and prolifera- ASD affects 1 in 110 children in the US [2], yet the patho- tion of several cell types, including neurons and glia of the physiology of the disorder is largely unknown. Recently, sev- CNS. EGF is present at high levels in the central nervous eral growth factors have been found to be dysregulated in a system (CNS) and plays a critical role in controlling pro- substantial proportion of adults with ASD [3]. liferation and differentiation of nervous tissue during neu- In the central nervous system, growth factors can regulate rogenesis [13, 14]. In addition to the function of EGF as a the processes of neuronal growth, differentiation, and pro- CNS growth factor, it is a central factor in promoting wound liferation, as well as regulating neuronal survival, neuronal healing. EGF is expressed at sites of injury and inhibits the migration, and the formation or elimination of synapses [3]. activity of nitric oxide synthase, preventing inflammation In addition to their central function in regulating neuro- [4, 15]. EGF deficiency results in several neurological, development, current literature has also illustrated the dual gastrointestinal, dermal, and pulmonary abnormalities in nature of many growth factors as immune modulators and animal models [16]. An increased frequency of EGF single highlighted their involvement in crosstalk between the im- nucleotide polymorphisms has been reported in ASD, as well mune system and the central nervous system (CNS) [4–7]. as lower plasma EGF levels in adults with autism [17, 18]. Many studies suggest the presence of aberrant immune HGF also plays a dual role as a neurological growth factor activity in ASD, in the CNS [8, 9] and in the periphery and an immune modulator. HGF can modulate immune 2 Autism Research and Treatment responses by signaling through the MET receptor on antigen was measured on a Wallac Victor3 multilabel-plate reader presenting cells, which results in a tolerogenic phenotype (PerkinElmer, Boston, MA) at 450 nm. with reduced proinflammatory cytokine production and cellular activity [5]. HGF is also essential for normal neu- 2.3. Statistical Analysis. Statistical analysis to compare levels rodevelopment, and disruption of HGF signaling results in of growth factors between ASD and TD groups was con- complex alterations in GABAergic neuron development in ducted with unpaired Student’s t-test. All analyses were con- the forebrain of animal models [19]. Lower levels of HGF ducted with GraphPad Prism statistical software (GraphPad in sera of autistic adults have been described [20], as well Software Inc., San Diego, CA). as decreased expression of the HGF receptor in postmortem brain samples [21]. To determine if there exists a differential profile for 3. Results peripheral blood growth factor levels in ASD, we analyzed plasma EGF and HGF in well-characterized young children Plasma levels of EGF were approximately 3-fold lower in . ± . aged 2–4 years old with a diagnosis of ASD and unrelated the ASD group (23 1 6 2 pg/mL) compared with the TD . ± . P = . typically developing children who were frequency-matched group (76 9 20 0pg/mL)( 0 003). Plasma levels of HGF ff for age. In addition, levels of plasma EGF and HGF were were not statistically di erent between children with ASD . ± . . ± . investigated for any associations with clinical behavioral and (423 5 20 9 pg/mL) and TD controls (436 6 19 2pg/mL) developmental outcomes. (Figure 1).

2. Methods and Materials 4. Discussion In this study, we investigated the levels of EGF and HGF, 2.1. Subjects and Behavioral Assessments. Eighty study par- two growth factors involved in neurodevelopment. Previous ticipants aged between 2–4 years of age were recruited as studies have found decreased levels of EGF in high function- part of the APP [22]. Participants consisted of 49 children ing adults with autism [18], but no studies have looked at with ASD (median age of 2.88 years, interquartile range children with ASD who are close to the onset of the disorder. 2.66–3.41 years, 42 males) and 31 typically developing (TD) We found that levels of plasma EGF were significantly children (median age of 2.96 years, interquartile range reduced in young children with ASD as compared with 2.85–3.27, 20 males). Diagnostic instruments included the similarly age-matched typically developing control children. Autism Diagnostic Observation Schedule-Generic (ADOS- This finding is consistent with that seen in adults with high G) [23] and the Autism Diagnostic Interview-Revised (ADI- functioning autism and may suggest that a deficiency in EGF R) [24]. All diagnostic assessments were conducted or is persistent throughout the time course of ASD. directly observed by trained, licensed clinical psychologists Current research has demonstrated that EGF is involved who specialize in autism and had been trained according in growth, differentiation, and maintenance of several tissues to research standards for these tools. Inclusion criteria for including the CNS and the gastrointestinal tract (GI) [14]. ASD were taken from the diagnostic definition of ASD Notably, abnormalities of both the CNS and the GI have been in young children formulated and agreed upon by the reported in ASD [26].IntheCNS,EGFservesasapotent Collaborative Programs of Excellence in Autism. Inclusion neurotrophic factor, and in vivo studies have demonstrated criteria for TD controls included developmental scores the effect of EGF in promoting proliferation, differentiation, within two standard deviations of the mean on all subscales survival, and migration of multipotent neural progenitor of the MSEL. Exclusion criteria for TD controls included cells and the differentiation of these cells into astrocytes and a diagnosis of mental retardation, pervasive developmental neurons [27]. In this paper, we were limited to investigating disorder or specific language impairment, or any known EGF in plasma and not in cerebral spinal fluid. However, developmental, neurological, or behavioral problems. TD EGF rapidly transports across the blood-brain barrier [28], children were screened and excluded for autism with the suggesting peripheral EGF levels could be representative of Social Communication Questionnaire (scores > 11) (SCQ— levels in the CNS that may impact neurodevelopment. Lifetime Edition) [25]. In the GI tract, EGF is necessary for normal development of the intestinal mucosa, and mice deficient in EGF receptor 2.2. Measurement of EGF. Peripheral blood was collected in suffer from symptoms similar to those of necrotizing ente- acid-citrate-dextrose Vacutainers (BD Biosciences, San Jose, rocolitis, with gradual destruction of villi, become severely CA). The plasma fraction was immediately harvested by malnourished, and typically die before postnatal day 8 centrifugationandstoredasaliquotsat−80◦C until the date [16]. EGF promotes wound healing in animal models of of assay. Plasma levels of EGF were determined by Human ulcerative colitis [29, 30]. Although GI symptoms affect a EGF enzyme-linked immunosorbant assay (ELISA) kit (R&D large proportion of children with ASD, the exact nature and Systems, Minneapolis, MN). Plasma levels of HGF were extent of GI inflammation in ASD are still controversial [31– determined by Human HGF enzyme-linked immunosorbant 34]. Interestingly, preliminary investigations in this study assay (ELISA) kit (R&D Systems). The assays were performed show that EGF levels are associated with increased bloating according to the protocols provided by the manufacturer, in children with ASD, as well as with raw, standard, and and all samples were assayed in duplicate. Optical density percentile rank scores on the Peabody picture vocabulary Autism Research and Treatment 3

P = 0.0032 500 100

400

75

300

50

HGF (pg/mL) 200 EGF (pg/mL)

25 100

0 0 ASD Control ASD Control

ASD ASD Control Control (a) (b)

Figure 1: Plasma levels of EGF and HGF. Levels of EGF and HGF in peripheral blood plasma from ASD participants and similarly aged typically developing controls as measured by ELISA (data is shown as mean and SEM). EGF levels are significantly lower in ASD subjects in comparison to typically developing controls (P = 0.003). HGF levels did not differ significantly between the two groups. Significance was determined by two-tailed unpaired Student’s t-test.

test-III and with the composite and nonverbal development [3] T. Nickl-Jockschat and T. M. Michel, “The role of neurotroph- quotient of the Mullen’s test (P<0.05, data not shown). ic factors in autism,” Molecular Psychiatry, vol. 16, pp. 478– However, the meaning of this data is not clear and we will 490, 2011. validate these findings in a larger replication cohort. [4] D. E. Heck, D. L. Laskin, C. R. Gardner, and J. D. Laskin, “Epi- It has been previously reported that HGF was decreased dermal growth factor suppresses nitric oxide and hydrogen peroxide production by keratinocytes. Potential role for nitric in the plasma of adults with autism [20]; however, we did not ff oxide in the regulation of wound healing,” Journal of Biological find a significant di erence between children with ASD and Chemistry, vol. 267, no. 30, pp. 21277–21280, 1992. typically developing children for plasma HGF levels. This ff [5] K. Okunishi, M. Dohi, K. Nakagome et al., “A novel role of may be due to age di erences between the participants of our hepatocyte growth factor as an immune regulator through study and those in the previous reports. To our knowledge, suppressing dendritic cell function,” Journal of Immunology, the relationship between age and serum HGF levels has not vol. 175, no. 7, pp. 4745–4753, 2005. been thoroughly established, but there is evidence that levels [6] J. A. Vega, O. Garc´ıa-Suarez,J.Hannestad,M.P´ erez-P´ erez,´ and maydecreasewithage[35]. A. Germana,` “Neurotrophins and the immune system,” Jour- Collectively, our data suggest that reduced levels of EGF nal of Anatomy, vol. 203, no. 1, pp. 1–19, 2003. are present in ASD. The roles of EGF and ASD require further [7] R. Tabakman, S. Lecht, S. Sephanova, H. Arien-Zakay, and P. research to better elucidate the relationship between this Lazarovici, “Interactions between the cells of the immune and potent growth factor and ASD pathophysiology. nervous system: neurotrophins as neuroprotection mediators in CNS injury,” Progress in Brain Research, vol. 146, pp. 387– 401, 2004. References [8] D. L. Vargas, C. Nascimbene, C. Krishnan, A. W. Zimmerman, and C. A. Pardo, “Neuroglial activation and neuroinflamma- [1] APA, Diagnostic and Statistical Manual of Mental Disorders, tion in the brain of patients with autism,” Annals of Neurology, American Psychiatric Publishing, Inc., Arlington, Va, USA, vol. 57, no. 1, pp. 67–81, 2005. 2000. [9] X. Li, A. Chauhan, A. M. Sheikh et al., “Elevated immune re- [2] C. Rice, “Prevalence of autism spectrum disorders—autism sponse in the brain of autistic patients,” Journal of Neuroim- and developmental disabilities monitoring network, United munology, vol. 207, no. 1-2, pp. 111–116, 2009. States, 2006,” Morbidity and Mortality Weekly Report, vol. 58, [10]P.Ashwood,P.Krakowiak,I.Hertz-Picciotto,R.Hansen,I. no. 10, pp. 1–20, 2009. N. Pessah, and J. Van de Water, “Altered T cell responses in 4 Autism Research and Treatment

children with autism,” Brain, Behavior, and Immunity, vol. 25, of endogenous subependymal neural precursor cell popula- no. 5, pp. 840–849, 2011. tions in the adult mouse brain,” Journal of Neuroscience, vol. [11] A. M. Enstrom, C. E. Onore, J. A. Van de Water, and P. 16, no. 8, pp. 2649–2658, 1996. Ashwood, “Differential monocyte responses to TLR ligands [28] W. Pan and A. J. Kastin, “Entry of EGF into brain is rapid and in children with autism spectrum disorders,” Brain, Behavior, saturable,” Peptides, vol. 20, no. 9, pp. 1091–1098, 1999. and Immunity, vol. 24, no. 1, pp. 64–71, 2010. [29] R. J. Farrell, “Epidermal growth factor for ulcerative colitis,” [12]P.Ashwood,P.Krakowiak,I.Hertz-Picciotto,R.Hansen,I. New England Journal of Medicine, vol. 349, no. 4, pp. 395–397, Pessah, and J. Van de Water, “Elevated plasma cytokines in 2003. autism spectrum disorders provide evidence of immune dys- [30] A. Sinha, J. Nightingale, K. P. West, J. Berlanga-Acosta, and function and are associated with impaired behavioral out- R. J. Playford, “Epidermal growth factor enemas with oral come,” Brain, Behavior, and Immunity,vol.25,no.1,pp.40– mesalamine for mild-to-moderate left-sided ulcerative colitis 45, 2011. or proctitis,” New England Journal of Medicine, vol. 349, no. 4, [13] C. J. Xian and X. F. Zhou, “Roles of transforming growth fac- pp. 350–357, 2003. tor-α and related molecules in the nervous system,” Molecular [31] K. Horvath and J. A. Perman, “Autism and gastrointestinal Neurobiology, vol. 20, no. 2-3, pp. 157–183, 1999. symptoms,” Current Gastroenterology Reports,vol.4,no.3,pp. [14] C. J. Xian and X. F. Zhou, “EGF family of growth factors: 251–258, 2002. essential roles and functional redundancy in the nerve system,” [32]P.Ashwood,A.Anthony,A.A.Pellicer,F.Torrente,J.A. Frontiers in Bioscience, vol. 9, pp. 85–92, 2004. Walker-Smith, and A. J. Wakefield, “Intestinal lymphocyte [15] S. Pastore and F. Mascia, “Novel acquisitions on the immuno- populations in children with regressive autism: evidence for protective roles of the EGF receptor in the skin,” Expert Review extensive mucosal immunopathology,” Journal of Clinical of Dermatology, vol. 3, no. 5, pp. 525–527, 2008. Immunology, vol. 23, no. 6, pp. 504–517, 2003. [16] P. J. Miettinen, J. E. Berger, J. Meneses et al., “Epithelial im- [33]P.Ashwood,A.Anthony,F.Torrente,andA.J.Wakefield, maturity and multiorgan failure in mice lacking epidermal “Spontaneous mucosal lymphocyte cytokine profiles in chil- growth factor receptor,” Nature, vol. 376, no. 6538, pp. 337– dren with autism and gastrointestinal symptoms: mucosal im- 341, 1995. mune activation and reduced counter regulatory interleukin- [17] T. Toyoda, K. Nakamura, K. Yamada et al., “SNP analyses of 10,” Journal of Clinical Immunology, vol. 24, no. 6, pp. 664–673, growth factor genes EGF, TGFβ-1, and HGF reveal haplotypic 2004. association of EGF with autism,” Biochemical and Biophysical [34] P. Ashwood and A. J. Wakefield, “Immune activation of pe- Research Communications, vol. 360, no. 4, pp. 715–720, 2007. ripheral blood and mucosal CD3+ lymphocyte cytokine pro- [18] K. Suzuki, K. Hashimoto, Y. Iwata et al., “Decreased serum lev- files in children with autism and gastrointestinal symptoms,” els of epidermal growth factor in adult subjects with high- Journal of Neuroimmunology, vol. 173, no. 1-2, pp. 126–134, functioning autism,” Biological Psychiatry,vol.62,no.3,pp. 2006. 267–269, 2007. [35] R. Ramirez, D. Hsu, A. Patel et al., “Over-expression of hepat- [19] P. Levitt, K. L. Eagleson, and E. M. Powell, “Regulation of ne- ocyte growth factor/scatter factor (HGF/SF) and the HGF/SF ocortical interneuron development and the implications for receptor (cMET) are associated with a high risk of metastasis neurodevelopmental disorders,” Trends in Neurosciences, vol. and recurrence for children and young adults with papillary 27, no. 7, pp. 400–406, 2004. thyroid carcinoma,” Clinical Endocrinology,vol.53,no.5,pp. [20] G. Sugihara, K. Hashimoto, Y. Iwata et al., “Decreased serum 635–644, 2000. levels of hepatocyte growth factor in male adults with high- functioning autism,” Progress in Neuro-Psychopharmacology and Biological Psychiatry, vol. 31, no. 2, pp. 412–415, 2007. [21] D. B. Campbell, R. D’Oronzio, K. Garbett et al., “Disruption of cerebral cortex MET signaling in autism spectrum disorder,” Annals of Neurology, vol. 62, no. 3, pp. 243–250, 2007. [22] E. Singer, ““Phenome” project set to pin down subgroups of autism,” Nature Medicine, vol. 11, no. 6, p. 583, 2005. [23] C. Lord, M. Rutter, S. Goode et al., “Autism diagnostic obser- vation schedule: a standardized observation of communicative and social behavior,” Journal of Autism and Developmental Disorders, vol. 19, no. 2, pp. 185–212, 1989. [24]C.Lord,M.Rutter,andA.L.Couteur,“Autismdiagnostic interview-revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive devel- opmental disorders,” Journal of Autism and Developmental Disorders, vol. 24, no. 5, pp. 659–685, 1994. [25] S. K. Berument, M. Rutter, C. Lord, A. Pickles, and A. Bailey, “Autism screening questionnaire: diagnostic validity,” British Journal of Psychiatry, vol. 175, pp. 444–451, 1999. [26] M. Careaga, J. Van de Water, and P. Ashwood, “Immune dysfunction in autism: a pathway to treatment,” Neurothera- peutics, vol. 7, no. 3, pp. 283–292, 2010. [27] C. G. Craig, V. Tropepe, C. M. Morshead, B. A. Reynolds, S. Weiss, and D. Van Der Kooy, “In vivo growth factor expansion Hindawi Publishing Corporation Autism Research and Treatment Volume 2012, Article ID 959073, 13 pages doi:10.1155/2012/959073

Review Article HLA Immune Function Genes in Autism

Anthony R. Torres,1 Jonna B. Westover,1 and Allen J. Rosenspire2

1 Center for Persons with Disabilities, Utah State University, 6804 Old Main Hill, Logan, UT 84322, USA 2 Department of Immunology and Microbiology, School of Medicine, Wayne State University, 221 Lande Building, Detroit, MI 48201, USA

Correspondence should be addressed to Anthony R. Torres, [email protected]

Received 10 October 2011; Accepted 11 November 2011

Academic Editor: Judy Van de Water

Copyright © 2012 Anthony R. Torres et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

The human leukocyte antigen (HLA) genes on chromosome 6 are instrumental in many innate and adaptive immune responses. The HLA genes/haplotypes can also be involved in immune dysfunction and autoimmune diseases. It is now becoming apparent that many of the non-antigen-presenting HLA genes make significant contributions to autoimmune diseases. Interestingly, it has been reported that autism subjects often have associations with HLA genes/haplotypes, suggesting an underlying dysregulation of the immune system mediated by HLA genes. Genetic studies have only succeeded in identifying autism-causing genes in a small number of subjects suggesting that the genome has not been adequately interrogated. Close examination of the HLA region in autism has been relatively ignored, largely due to extraordinary genetic complexity. It is our proposition that genetic polymorphisms in the HLA region, especially in the non-antigen-presenting regions, may be important in the etiology of autism in certain subjects.

1. Autism the prenatal period or infancy stage of life. Exposure to mercury, maternal viral infections, autoimmune disorders, Leo Kanner first described autism in 1943 [1] after finding 11 and the inheritance of certain gene combinations have been children with common symptoms of obsessiveness, stereo- implicated in the etiology. Unfortunately, none of these areas typy, and echolalia at Johns Hopkins University. Autism have given clear answers as to the etiology. Fortunately, remained an esoteric disorder for several decades until psychologists have made significant strides in treating chil- physicians and parents connected these symptoms with an dren and it appears the earlier behavioral treatment starts, increasing number of patients. It is important to note that the better the outcome. Nevertheless, medical researchers the diagnostic criteria have been modified over the years continue to search for the cause(s) of ASD. This paper to include a broader category of symptoms, thus increasing discusses a possible role for the immune system, and in the number of children diagnosed with the disorder, now particular immune function genes in the human leukocyte referred to as Autism Spectrum Disorder (ASD) [2]. Current- antigen region (HLA), as a research area that should be more ly, the Centers for Disease Control and Prevention (CDC) closely investigated. states that the incidence of ASD is 1 out of 110 children in the United States [3]. The severity of ASD varies greatly with the most severe forms, much like Kanner autism, displaying 2. Infections language regression, seizures, and lower IQ. Altevogt et al. [4] have suggested that autism, or more properly ASD, is not One of the first areas of interest in the 1960s and 1970s was a single disorder, but a collection of similar disorders each the search for an infectious agent that might be involved in with different characteristics and perhaps etiologies. the etiology of ASD [5]. During this time, there were many Even after several decades of research, there is much case reports in the literature that suggested an association debate around the world on the etiology of ASD. It is clear between congenital rubella infection and resultant autistic that ASD results from abnormal brain development in either behaviors. However, after decades of research no definite role 2 Autism Research and Treatment for infectious agents in autism etiology has been confirmed. study of ASD. These newer approaches have identified CNV On the other hand, these endeavors have led to observations mutation differences in genes involved in neuronal cell that perhaps the immune system was involved in autism, and adhesion and ubiquitin degradation as being associated with evidence continues to mount that immune abnormalities are ASD [18]; however, these results have yet to be replicated by indeed associated with ASD. other researchers. The neurexin-1 gene has been associated with a variety of developmental disorders including ASD [19]. The neurexin- 3. Familial Studies 1 gene (2p16.3) is one of the largest genes in the human Studies indicating familial clustering and the increases of genome with 24 exons in 1.1 Mb. With two independent promotors there can be over 1,000 neurexin isoforms gener- ASD in twins have been interpreted by many as an indication ff of genetic predisposition. Twin studies show the concordance ated from the 24 exons in di erent cells or tissue. Another rates of monozygotic twins at 36–96%, whereas dizygotic gene of interest is the contactin-associated protein-like 2 twins are 0–24% concordant, resulting in an estimated herit- (CNTNAP2) gene that was shown to be associated in ability of autism at >90% [6–8]. Additionally, family studies Old-Order Amish subjects with intractable epilepsy and have shown autism to have familial aggregation with 3–8% of ASD [20].Threeothergroupshavenowconfirmedthe subsequently born siblings either being autistic or showing involvement of CNTNAP2 in ASD [21–23]. some form of pervasive developmental disorder (PDD) [9, Both the neurexin-1 and the CNTNAP2 genes are in- 10]. This is a 3- to 8-fold increase in risk for siblings over the volved in synaptic function. Although these approaches have general population. A more recent study gave an estimate of shown a strong association of certain genes with ASD, 18.7% sibling recurrence risk for ASD, a 20-fold increase over only a small percentage of subjects with ASD have these the general population [11]. It is important to note, however, mutations. For example, the neurexin-1gene is found in only that family data should be looked at with great caution, as about 0.5% of autism cases and 0.2% of controls. The 90% individuals living in the same household will have similar missing inheritance may be largely due to marked genetic ff exposures to microorganisms and environmental chemicals. heterogeneity, suggesting that di erent ASD phenotypes Taking this into account, a recent paper based on data from should be examined separately [24–26]. Recent genetic re- twin pairs estimates the genetic heritability for ASD to be search has also associated numerous immune function genes 14–67% [12]. However, this idea is somewhat controversial with autism [27–30]. A large study that examined SNP data as many in the research community continue to feel that the from several genomewide scans on 3,130 subjects with schiz- results from family studies are indicative of a strong genetic ophrenia found that the 5 most significant SNP markers are etiology [13, 14]. found across the HLA region [31]. It appears that some of missing inheritance, at least in schizophrenia, was uncovered in the HLA region and we suggest a similar finding will be 4. Genetics confirmed/uncovered in autism. Many of the early genetic studies involved the examination of microsatellites throughout the human genome in an attempt 5. Immune Abnormalities in Autism to find genomic regions that would associate with autism. Overall, this approach was not very fruitful and researchers It is no surprise to see immune gene associations in ASD, as quickly started to examine single-nucleotide polymorphisms numerous researchers have reported immune abnormalities (SNPs) as techniques advanced [15]. With this approach in autism for over 20 years. It has become increasingly obvi- many researchers proposed that multiple candidate genes ous that inflammatory processes are associated with autism. were associated with ASD. Unfortunately, studies proposing Blood levels of the inflammatory cytokines IL-6, INF-γ,and candidate genes were often contradictory and proved to be TNF-α were shown to be elevated in autistic individuals com- unreliable [15]. One of the most interesting genetic findings pared to controls [32, 33]. Later, seminal work by Vargas et in ASD is the association of autism with the MET receptor al. [34] utilized direct morphological analysis and immuno- tyrosine kinase gene located on chromosome 7q31 as MET histologic techniques to show an active neuroinflammatory signaling participates in gastrointestinal repair, immune process in the cerebral cortex, white matter, and in particular function, neocortical, and cerebellar growth [16]. It is impor- the cerebellum of ASD patients that was dependent on tant to mention that the autism MET associations have activation of microglia and astroglia. Cytokine profiling been replicated by other research groups [17]. This autism demonstrated that neuroinflammation was accompanied by association showed a relative risk of 2.27 which is much upregulation of the macrophage chemoattractant protein lower than the relative risk for HLA gene/allele associations (MCP-1) and TGF-beta in brain tissue, and that MCP-1 was discussed below. also upregulated in cerebral spinal fluid [34]. More recent It is very reasonable to believe that deletions or dupli- work has directly demonstrated that aside from blood, IL- cations of genetic regions, which can cause lower or higher 6, TNF-α,andINF-γ are elevated in ASD brains, along with levels of gene expression, could produce pathological phe- the other inflammatory cytokines GM-CSF and IL-8 [35]. notypes. Consequently, newer approaches examining copy Most recently, it has been reported that the important number variation (CNV) and microarray analyses of 500,000 inflammatory-associated transcription factor, nuclear factor SNPs or more have been in vogue for several years in the kappa-light-chain-enhancer of activated B cells (NF-κB) is Autism Research and Treatment 3

Table 1: A list of proteins against which autoantibodies have been fairly recently, it was believed that most immature lym- found in the serum of subjects with autism. phocytes recognizing self-antigens (autoimmune repertoire) were normally neutralized by virtue of central tolerance Protein Reference before they could mature, and that peripheral tolerance Transglutaminase 2 [39] would insure the removal of any self-reactive lymphocyte 45 and 62 kDa proteins in cerebellum [40, 41] escaping central tolerance. Peripheral tolerance refers to the Voltage dependent anion channel (VDAC) [42] process whereby mature self-reactive lymphocytes which Hexokinase-1 [42] have escaped central tolerance are eliminated, largely by Mitochondrial protein [43] CD95-mediated apoptosis [62]. Thus responses to foreign Antimitochondrial DNA auto-antibodies [43] antigens were viewed as normal, while anti-self responses Nuclear proteins [44] were considered necessarily pathologic. However, with the 52 kDa protein in hypothalamus and thalamus [45] realization that many self-reactive lymphocytes survive cen- 43–48 kDa protein in the hypothalamus [45] tral and peripheral tolerance, this view has had to be modified [49]. Limited immune responses to self-antigens Folate receptor [46] (autoimmunity) are now understood to be normal and not Brain-derived neuro trophic factor [47] necessarily pathologic [63]. HSP90 [48] Upon stimulation of the system with a pathogen, cognate Myelin basic protein (MBP) [38, 49] lymphocyte clones representative of the antiforeign reper- Myelin-associated glycoprotein (MAG) [50] toire normally expand and mature, providing a protective Myelin oligodendrocyte glycoprotein (MOG) [50] immune response. On the other hand, it is the expansion and Neuron-axon filament protein [51] maturation of those clones representative of the autoimmune Glial fibrillary acidic protein [51] repertoire that leads to autoimmune disease. In other words, the developing immune system can be characterized as bal- ancing production between antiforeign (protective) and anti- self (autoimmune) repertoires. While a beneficial function of upregulated in both blood [36] and brain tissue [37]of naturally occurring, low level autoimmune antibodies, also autistic individuals. Other immune abnormalities such as referred to as natural antibodies (NAs), remains a matter of autoantibodies started to be reported in 1993 [38](Table 1). debate, it appears as if the repertoire of NA is reflective of Autoantibodies to myelin basic protein have been noted the susceptibility to develop specific autoimmune diseases by at least a couple of researchers [38, 52]. An increase [64, 65]. Because central tolerance of T cells depends to a in autoantibody reactivity has been reported against other large extent upon the strength of the TCR interaction with brain proteins in ASD including nerve growth factor [53], an autoantigen or an HLA class I or II complex [59], the brain endothelium [54], cerebellar proteins [40], and sero- NA repertoire will to a large extent depend upon the HLA tonin 5-HT receptors [55] and transglutaminase-2, a protein haplotype, with some haplotypes favoring autoantibodies important in synaptic stabilization [39]. Croonenberghs et targeting one antigen over another. For instance, in the case al. [56] noted a significant increase in gamma globulin of ASD we have found an association with low level antibody especially of the IgG2 and IgG4 subclasses in children with responses to tissue transglutaminase, and that this response autism over a control population. Autoantibodies to several appears linked to the HLA-DR3/DQ2 and DR7/DQ2 hap- uncharacterized brain-specific proteins have been reported lotypes [39]. It is likely that the other autoantibodies noted in the plasma of autistic individuals. In particular, western above as being associated with ASD may be linked to different blot analysis has shown the presence of IgG autoantibodies haplotypes. targeting a protein of approximately 52 kDA located in the Aside from autoantibodies and altered cytokine levels hypothalamus and thalamus of adult brain [45]. Other there appear to be other immune abnormalities associated autoantibodies targeting three brain proteins of 43–48 kDA with ASD. It was noted that there were decreased numbers located in the hypothalamus have also been observed in the of T-lymphocytes and an altered ratio of suppressor T- serum of autistic individuals [45]. Autoantibodies targeting lymphocytes to helper T-lymphocytes [66, 67]andalteredT- cerebellar proteins of 45 and 62 kDA have been associated lymphocytes responses in children with autism [68]. Warren with ASD [40]. These autoantibodies may be specific to et al. [69]reportedthatsubjectswithautismhadreduced cerebellar Golgi cells, which are GABAergic interneurons NK cell killing in the standard K562 target cell cytotoxicity [41]. Autoantibodies reactive to human brain proteins in the assay. This observation of decreased NK cell killing has been 36–39 and 61 kDA range have also been found in the sera of repeated by at least a couple of other research teams [28, 70]. mothers of autistic children. [57, 58]. One research group [71] observed that monocyte counts and It is important to note that while autoantibodies associ- neopterin levels were increased in autistic children compared ated with ASD may be biomarkers, they may not necessarily to gender and age-matched healthy controls suggesting that be pathologic in and of themselves. Central tolerance refers the immune system was overactivated in the ASD group. to the process whereby immature lymphocytes are negatively Another elegant set of experiments involved the stimulation selected based on the ability of their antigen receptors (the of cultured monocytes with several toll-like-receptor (TLR) B cell receptor or BCR for B cells and the T-cell receptor ligands. The monocytes from subjects with ASD had signif- or TCR for T cells) to recognize self-antigens [59–61]. Until icant increases or decreases in proinflammatory cytokines 4 Autism Research and Treatment

Table 2: Genes and alleles in the HLA region.

HLA genes Number HLA alleles Number HLA class I genes 6 HLA class I alleles 2215 HLA class II genes 12 HLA class II alleles 986 HLA class I-like genes 2 HLA class I-like alleles 94 Non-HLA genes 112 Total genes 132 HLA class I genes A/B/C/E/F/G HLA class I-like genes MICA/MICB HLA class II genes DRA/DRB/DQ/DP/DM/DO depending on the particular TLR ligand added to the cell cul- specialization, central nervous system (CNS) development ture media [72]. [80–84], and even neurological disorders [85]. The genes One large-scale study found that the frequency of of the HLA region are shown in Figure 1. Shiina et al. autoimmune disorders in the families with autistic chil- [86, 87] have published two excellent reviews on the HLA dren was found to be higher than in control subjects, super-locus. Not only is there extraordinary complexity in especially mothers of autistic children [73]. Another group the HLA genes, there are extensive haplotype-related tran- demonstrated that autoimmune diseases were increased scriptional differences [88]. It has been shown that genetic significantly in families with ASD compared with those of mechanisms outside of the non-antigen-binding HLA genes healthy control subjects [74], suggesting a link between the in the ancestral haplotype 8.1 (also referred to as COX) are disorders. Croen et al. [75] showed that maternal psoriasis associated with susceptibility to many autoimmune diseases diagnosed around the time of pregnancy is significantly [89]. It is our proposition that HLA genes/proteins should associated with a subsequent diagnosis of autism in the child. be more carefully examined due to increasing evidence of Additionally, they showed a 2-fold increase in risk for a child autoimmune type associations in autism. having ASD if the mother was diagnosed and with asthma or allergies during pregnancy. An association between a family history of type 1 diabetes mellitus (T1DM) and infantile 7. HLA Associations in ASD autism as well as a significant association between maternal It was suggested over 30 years ago by Stubbs and Magenis histories of either rheumatoid arthritis (RA) or celiac disease [90] that the HLA region might be important in autism. ´ and ASDs was noted by Atladottir et al. [76]. Warren et al. [91] first reported that the HLA ancestral haplotype 44.1 (B44-SC30-DR4) was associated with autism 6. Autism HLA Genetics with a relative risk of 7.9. That result was confirmed in a separate case/control population [92]. Interestingly, the HLA is the name for the major histocompatibility complex individual components of AH 44.1 (A2-B44-SC30-DR4) (MHC) in humans and HLA and MHC are often used include a deleted C4B gene and DRβ1∗0401, both of which interchangeably in the literature. The HLA region on chro- have been shown independently to be significantly associated 6 mosome 6p21 (about 4 × 10 bp) is of major interest in basic with ASD [93, 94]. Examination with different genetic research as well as medicine as genes/proteins in this region markers than those used by Warren suggested that certain are involved in many biological processes such as histocom- HLA haplotypes are associated with autism in Sardinian and patibility, inflammation, ligands for immune cell receptors, Italian families [95, 96]. and the complement cascade. The HLA region has 20 typical Warren et al. [97] reported that the shared epitope- HLA genes and 112 nontypical HLA genes (Table 2) that are binding pocket (DRβ1∗0401, ∗0404, and ∗0101) in the third inherited together as frozen blocks of DNA called ancestral hypervariable region of DRβ1 has a strong association with or extended haplotypes. Complete DNA sequences have autism. A relative risk of 19.8 for autism was reported for been published for 8 of the more common ancestral hap- subjects with one of the two extended HLA haplotypes. Both lotypes in an effort to expedite basic and disease research of these haplotypes have many allelic similarities especially [77]. It should be mentioned that smaller haplotypes can the DRβ1∗0401. also be constructed for genes that are linked. HLA genes AH 44.1 (HLA-A2, Cw5, B44, Bf∗S, C2∗C, C4A3, also play a role in reproduction, pregnancy maintenance, C4BQ0, DRβ1∗0401, DQB1∗0301). mate selection, and even kin recognition [78, 79]andhave AH 62.1 (HLA-A2, Cw3, B15, Bf∗S, C2∗C, C4A3, C4B3, been associated with over 100 diseases/disorders including DRβ1∗0401, DQB1∗0302). autism. The proteins encoded by HLA genes are ligands, The shared epitope has been associated with several receptors, cytokines, signaling factors, heat shock proteins, autoimmune diseases such as rheumatoid arthritis, psoriatic transcription regulators, and so forth. Current research is arthritis, and systemic lupus erythematosus [98]. Torres et increasingly demonstrating a role for HLA proteins in neural al. [93] confirmed the association of the HLA-DR4 allele and cell interactions, synaptic function, cerebral hemispheric also found that the DR13 and DR14 alleles occurred less Autism Research and Treatment 5

Telomeric side Chr. 6 Telomere block κ Short arm block α Class II region Centromere Class I region Class III region Class I region Long arm Extended class II region Extended

Centromeric side block β Telomere

Figure 1: Gene map of the human leukocyte antigen (HLA) region. The major histocompatibility complex (MHC) gene map corresponds to the genomic coordinates of 29677984 (GABBR I) to 33485635 (KIFC1) in the human genome build 36.3 of the National Center for Biotechnology Information (NCBI) map viewer. The regions separated by arrows show the HLA subregions such as extended class I, classical class I, class III, classical class II, and extended class II regions from telomere (left and top side) to centromere (right and bottom side). White, gray, striped and black boxes show expressed genes, gene candidates, noncoding genes and pseudogenes, respectively. The location of the alpha, beta, and kappa blocks containing the cluster of duplicated HLA class I genes in the class I region are indicated. (Reprinted with permission from the Journal of Human Genetics [87].) often in subjects with autism, suggesting a possible protective three additional research groups. Lee et al. [99] demonstrated mechanism. Interestingly, the DR13 allele was inherited less that boys with autism and their mothers had a significantly frequently than expected from the mothers. Associations higher frequency of DR4 than normal control subjects (odds with autism and the DR4 allele have since been confirmed in ratios 4.20 and 5.54, resp.), suggesting that a maternal-fetal 6 Autism Research and Treatment immune interaction could be involved in autism. Johnson In the CNS, TNF-α is made in microglia cells, astrocytes, and et al. [100] reported significant transmission disequilibrium neurons [141]. In the normal state, TNF-α drives acute and for HLA-DR4 (odds ratio 4.67) from maternal grandparents chronic inflammatory responses that leads to the removal of to mothers of children with autism which also suggests a injurious stimuli and the restoration of homeostasis. TNF- maternal-fetal interaction for HLA-DR4. It has been recently α is necessary for neural cell differentiation and neuron shown in Han Chinese that the HLA-DRβ1allelefrequencies maturation and there is evidence that it is critical in the including DR4 are different in subjects with autism versus normal brain for proper synaptic function. control subjects [101]. There is extensive research that implicates TNF-α as a key Itwasreported20yearsagothatsubjectswithautism mediator in disease progression, inflammation, blood-brain- had a significant increase in the C4B null allele (C4B barrier deterioration, and even cell death [142]. Elevated lev- gene deletion) compared to control subjects [91]. After this els are present in numerous neurological disorders including observation, it was also noted that subjects with autism had multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, a significant deficiency in the plasma C4B protein [102]. ischemia, traumatic brain injury, and as mentioned above, These initial findings of an increase in the deletion of the ASD [141]. It is unclear if TNF-α contributes to the disease C4B gene was supported by examining a new population of state or the higher concentrations limit neuronal injury. subjects with autism [94]. Mostafa and Shehab [103]have There are three adjacent genes: lymphotoxin alpha and beta recently reported a significant increase in the deletion of the (LTA and LTB) and leukocyte-specific transcript-1 (LST1) C4B gene in the Egyptian population. They also reported an in the TNF-α block. LTA and LTB are proinflammatory increased risk when there was a family history of autoim- cytokines like TNF-α, and LST1 plays a role in inflammatory mune diseases in the autism population. Descriptions of and infectious diseases. It has been suggested that vaccine- several non-antigen-binding HLA genes that are associated induced immunity changes with certain haplotypes in these with autoimmune diseases are discussed below and listed genes [143]. Two other genes important in immune function in Table 3. are adjacent to the TNF-α block. Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor-like 1(NF- κBIL1) on the telomeric side of TNA-α is an inhibitor of NF- 8. Non-Antigen-Binding HLA Genes in Class I κB, an important pleiotropic immune system transcription factor that is upregulated in ASD [144, 145]. In addition, NF- Although MICA/MICB genes are structurally similar to clas- κ sical antigen-binding HLA class I genes, they encode proteins BIL1 has been associated with several autoimmune diseases that interact with different T-cell receptors in response to (Table 3). The natural cytotoxicity trigger receptor three stress as infection or neoplastic transformation. The two (NCR3) gene on the centromeric side encodes proteins that genes are located on the centromeric end of the class I region are ligands for activating receptors on NK cells that recognize near HLA-B at the border of the class III region (Figure 1) tumor cells [146]. Typical HLA-B and -C proteins are also [87]. MICA/MICB proteins are ligands for NKG2D recep- ligands for KIR receptors on NK cells. tors on NK cell and gamma/delta T-cell receptors. Adap- There are numerous heat shock proteins (HSPs), also tive immunity involves three lymphocyte populations (B referred to as chaperones, that assist the folding of newly cells, alpha/beta T cells, and gamma/delta T cells). Gam- synthesized proteins as well as those that have been unfolded ma/delta T cells represent a small population of T cells that or denatured [147]. HSPs are also important in cell protective possess a T-cell receptor that is distinct from the typical functions and in inhibiting the apoptosis cascade. They are alpha/beta T cell. They are concentrated in the intestinal mu- named by their molecular weights (HSP100, HSP90, HSP70, cosa and appear to have a prominent role in recognizing HSP60, and smaller HSPs). Although the proteins appear small bacterial phosphoantigens and undefined antigens in normal cellular functions, they are induced to higher presented by MICA/MICB proteins. Gamma/delta T cells levels in trauma, epilepsy, neurodegenerative diseases, and have potent cytotoxic activity and have been considered a other injuries. An underlying feature among AD, Parkinson’s link between innate and adaptive immunity. Polymorphisms disease, spinocerebellar ataxia, and other neurodegenerative in MICA/MICB genes have been associated with T1DM, AD, diseases is the accumulation of misfolded proteins and and SLE independent of DRβ1 alleles. There are several genes HSPs are being studied because of their role in folding associated with psoriasis (PSORSI locus). It is unclear how and refolding proteins [147]. There is intense interest in the risk is spread among these genes as they are closely linked. HSPs as they appear to protect neurons [148]andonemust Two genes RNF39 and TRIM39 in the class I region have been remember that postmitotic neurons are unable to dilute associated with Behc¸et’s disease. misfolded or aggregated proteins through division. There are three HSP70 genes (HSPA1L, HSPA1A, and 9. Non-Antigen-Binding HLA Genes in Class III HSPA1B) in the class III HLA region that are adjacent but separate genes. HSP70 proteins have been demonstrated to Tumor necrosis factor-alpha (TNF-α) is a proinflammatory, stimulate IL-6 and TNF-α production, activate microglial multifunctional cytokine that plays important roles in cell cells, and stimulate phagocytosis [148]. HSP70 proteins are physiology. It is synthesized in numerous cells including also important in autoimmunity by enhancing antigen pre- macrophages, NK cells, T cells, mast cells, osteoblasts, gran- sentation in both HLA class I and HLA class II systems [147, ulocytes, smooth muscle cells, fibroblasts, and keratinocytes. 149]. Peptides that are associated with HSP70 at the time of Autism Research and Treatment 7

Table 3: Non-classical HLA genes associated with autoimmune diseases.

Gene HGNC gene number Autoimmune disease References Extended class I OR2H2 8253 SLE [104] RNF39 18064 Behc¸et’s disease [105] TRIM39 10065 Behc¸et’s disease [105] Class I PSORS1 locus 9573 Systemic sclerosis, Psoriasis [106–111] MICA 7090 T1DM, AD, SLE [112–114] MICB 7091 SLE [104] BAT1–BAT5 13917–21 Alzheimer’s, AIDS [115, 116] NFKBIL1 7800 Sjogren’s¨ syndrome, SLE, RA [117, 118] Alzheimer’s, Psoriasis, Autoimmune TNF Block 11892 [115, 119–121] hepatitis, Sarcoidosis AIF1 352 T1DM [122] Class III HSP70 genes 5232–4 MS [123] Complement genes 1248, 1324, 1323 SLE, myasthenia gravis, T1DM [124–126] SKIV2L 10898 SLE [127] ATF6B (CREBL1) 2349 SLE [104] NOTCH4 7884 Systemic sclerosis [128] C6orf10 13922 SLE [104] BTNL2 1142 Ulcerative colitis, Sarcoidosis [129, 130] TAP2 44 Psoriasis [131] PSMB8 9545 Hypersensitivity pneumonitis [132] Psoriasis [133] Class II TAP1 43 Vitiligo [134] PSMB9 9546 Psoriasis, Vitiligo [133, 134] Psoriasis, Antiphospholipid HLA-DM 4934, 4935 [131, 135–138] auto-antibodies, RA, SLE, T1DM HLA-DO 4936, 4937 common variable immunodeficiency [139] HSD17B8 3554 SLE [104] Class II Extended DAXX 2681 MS [140] Abbreviations: (HGNC) HUGO Gene Nomenclature Committee (http://www.genenames.org/); (SLE) systemic lupus erythematosus; (AIDS) acquired immune deficiency syndrome; (T1DM) type 1 diabetes mellitus; (AD) Addison’s disease; (RA) rheumatoid arthritis; (MS) multiple sclerosis, PSORS1 psoriasis locus genes (CDSN, PSORS1C1, PSORS1C2, CCHCR1, POU5F1, PSORS1C3), TNF Block genes (LTA, TNF, LTB, LST1), HSP70 genes (HSPA1L, HSPA1A, HSPA1B), Complement genes (C2, C4B, C4A).

T-cell presentation have been shown to be more antigenic HLA proteins. DM stabilizes and edits the peptide repertoire and therefore involved in autoimmunity [149, 150]. presented by DQ proteins by catalyzing CLIP release. The Another gene that is independently associated with associations of DQ2 with T1DM and celiac disease have been autoimmune diseases outside of non-antigen-binding HLA known for several decades; however, the biochemistry behind alleles is allograft inflammatory factor-1 (AIF1). The SNP these associations has not been elucidated. It is now known rs2269475 C > T in AIF1 has been associated with RA. that DQ2 is a poor substrate for DM and it has been proposed The T allele was significantly higher in the RA patients and that antigen presentation in the thymus and periphery can be ff there was no significant linkage disequilibria between the a ectedbyimpairedDQ-DMinteractionssoastopromote AIF1 SNP and DRβ1 alleles. Anticyclic citrullinated peptide autoimmune disease [152]. HLA-DO is another nonclassical antibodies commonly used to monitor RA were significantly class II HLA protein that is involved in the loading of peptides increased in carriers with the T allele [151] but not the C to HLA-DR proteins by modulating the function of HLA- allele. DM [153]. There are two genes in the class II region that encode pro- teins involved in the transport of antigen from the cytoplasm 10. Non-Antigen-Binding HLA Genes in Class II to the endoplasmic reticulum for binding to class I HLA proteins: transporter associated with antigen-processing 1 Although HLA-DM and -DQ proteins have structures like and 2 (TAP1 and TAP2). Several research groups have classical antigen-binding HLA proteins, they work in the reported associations of TAP2 genes with SLE independent of cytoplasm and not at the cell surface antigen-presenting classical HLA alleles [154]. This interaction is very interesting 8 Autism Research and Treatment as it means that class II genes can affect class I antigen References binding. [1] L. Kanner, “Autistic disturbances of affective contact,” The There are two other genes in the extended class II Nervous Child, vol. 2, no. 2, pp. 217–250, 1943. region that have been associated with autoimmune dis- [2] F. R. Volkmar, M. State, and A. Klin, “Autism and autism eases (Table 3). Hydroxysteroid 17-beta dehydrogenase 8 spectrum disorders: diagnostic issues for the coming decade,” β (HSD17 8) is important in regulating the concentrations of Journal of Child Psychology and Psychiatry and Allied Disci- active estrogens and androgens and high levels of estrogen plines, vol. 50, no. 1-2, pp. 108–115, 2009. are well known to be associated with SLE and other autoim- [3] Centers for Disease Control and Prevention (CDC), “Preva- mune diseases. The second gene, death-associated protein 6 lence of autism spectrum disorders-Autism and develop- (DAXX), is a very important protein that interacts with a mental disabilities monitoring network, United States, 2006,” variety of proteins in the nucleus and cytoplasm. Perhaps CDC—Morbidity and Mortality Weekly Report, vol. 58, no. most importantly, it interacts with the death receptor Fas. 10, pp. 1–20, 2009. Engstrom et al. [155] published a paper describing decreased [4] B. M. Altevogt, S. L. Hanson, and A. I. Leshner, “Autism and the environment: challenges and opportunities for research,” expression of Fas on CD4 + T lymphocytes but higher serum Pediatrics, vol. 121, no. 6, pp. 1225–1229, 2008. levels of soluble Fas in ASD. [5] S. Chess, “Autism in children with congenital rubella,” Journal of Autism and Childhood Schizophrenia, vol. 1, no. 1, 11. Summary pp. 33–47, 1971. [6] S. Folstein and M. Rutter, “Infantile autism: a genetic study There is mounting evidence that the immune system plays of 21 twin pairs,” Journal of Child Psychology and Psychiatry a role in the pathogenesis of ASD in certain individuals. and Allied Disciplines, vol. 18, no. 4, pp. 297–321, 1977. This evidence comes from several research areas including [7] S. Steffenburg, C. Gillberg, L. Hellgren et al., “A twin an increase in proinflammatory cytokines in blood and study of autism in Denmark, Finland, Iceland, Norway and Sweden,” Journal of Child Psychology and Psychiatry and brain, autoantibodies to numerous antigens, and HLA Allied Disciplines, vol. 30, no. 3, pp. 405–416, 1989. associations. [8] A. Bailey, A. le Couteur, I. Gottesman et al., “Autism as a Autism HLA associations have been observed across the strongly genetic disorder: evidence from a British twin study,” entire HLA region. For example, in the class I region HLA- Psychological Medicine, vol. 25, no. 1, pp. 63–77, 1995. A2 has been associated with autism by at least two research [9] P. Bolton, H. Macdonald, A. Pickles et al., “A case-control groups [156, 157]. In the class II region several researchers family history study of autism,” Journal of Child Psychology have reported autism associations with the DRβ1∗04 allele and Psychiatry and Allied Disciplines, vol. 35, no. 5, pp. 877– [93, 99, 100]. Strong associations also appear in the class 900, 1994. [10] E. R. Ritvo, L. B. Jorde, A. Mason-Brothers et al., “The III region where the C4B null allele has been associated UCLA-University of Utah epidemiologic survey of autism: with autism with relative risks of 4.3 [94]and4.6[97], recurrence risk estimates and genetic counseling,” American and an odds ratio of 6.3 [103]. The HLA-associated risk is Journal of Psychiatry, vol. 146, no. 8, pp. 1032–1036, 1989. the highest for autism (19.8) when combining two ancestral [11] S. Ozonoff, G. S. Young, A. Carter et al., “Recurrence risk haplotypes (44.1 and 62.1). Both of these haplotypes have for autism spectrum disorders: a baby siblings research ∗ HLA-A2 and DRβ1 0401 as well as other genetic similarities; consortium study,” Pediatrics, vol. 128, no. 3, pp. e488–e495, however, these two alleles cannot account for all of the 19.8 2011. risk. Compared to other genetic associations with autism, the [12] J. Hallmayer, S. Cleveland, A. Torres et al., “Genetic heritabil- HLA associations may be more important than realized, as ity and shared environmental factors among twin pairs with they have the highest genetically associated risk, that we are autism,” Archives of General Psychiatry, vol. 68, no. 11, pp. aware of for autism. For example the MET gene, one of the 1095–1102, 2011. most studies genetic regions in autism, has a relative risk of [13] C. M. Freitag, “The genetics of autistic disorders and its 2.27 [17]. clinical relevance: a review of the literature,” Molecular Psychiatry, vol. 12, no. 1, pp. 2–22, 2007. It is our premise that some of the autism missing [14] M. Rutter, “Genetic studies of autism: from the 1970s into inheritance may be hidden in the HLA region, both in the millennium,” Journal of Abnormal Child Psychology, vol. classical HLA alleles and nonclassical HLA genes, as seen in 28, no. 1, pp. 3–14, 2000. schizophrenia [31]. For example, the HLA class III region [15] M. W. State, “The genetics of child psychiatric disorders: contains clusters of genes such as TNF-α, HSP70, C4A/C4B, focus on autism and tourette syndrome,” Neuron, vol. 68, no. and NF-κBIL1 that are seminal in cellular function and 2, pp. 254–269, 2010. are also associated with numerous autoimmune diseases [16]D.B.Campbell,J.S.Sutcliffe, P. J. Ebert et al., “A genetic (Table 3). variant that disrupts MET transcription is associated with autism,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 45, pp. 16834–16839, Acknowledgment 2006. [17] M. C. Judson, K. L. Eagleson, and P. Levitt, “A new synaptic This report was supported in part by NIH Grant player leading to autism risk: met receptor tyrosine kinase,” (RO1ES016669) and the Utah Autism Foundation (Salt Journal of Neurodevelopmental Disorders,vol.3,no.3,pp. Lake City, Utah). 282–292, 2011. Autism Research and Treatment 9

[18] J. T. Glessner, K. Wang, G. Cai et al., “Autism genome- [36] U. S. Naik, C. Gangadharan, K. Abbagani, B. Nagalla, N. wide copy number variation reveals ubiquitin and neuronal Dasari, and S. K. Manna, “A study of nuclear transcription genes,” Nature, vol. 459, no. 7246, pp. 569–573, 2009. factor-κ B in childhood autism,” PLoS ONE,vol.6,no.5,p. [19] M. S. Ching, Y. Shen, W. H. Tan et al., “Deletions of NRXN1 e19488, 2011. (neurexin-1) predispose to a wide spectrum of developmen- [37] A. M. Young, E. Campbell, S. Lynch, J. Suckling, and S. tal disorders,” American Journal of Medical Genetics B, vol. J. Powis, “Aberrant NF-κB expression in autism spectrum 153, no. 4, pp. 937–947, 2010. condition: a mechanism for neuroinflammation,” Frontiers in [20] K. A. Strauss, E. G. Puffenberger, M. J. Huentelman et al., Psychiatry, vol. 2, no. 27, pp. 1–8, 2011. “Recessive symptomatic focal epilepsy and mutant contactin- [38]V.K.Singh,R.P.Warren,J.D.Odell,W.L.Warren,andP. associated protein-like 2,” The New England Journal of Cole, “Antibodies to myelin basic protein in children with Medicine, vol. 354, no. 13, pp. 1370–1377, 2006. autistic behavior,” Brain, Behavior, and Immunity, vol. 7, no. [21] M. Alarcon,´ B. S. Abrahams, J. L. Stone et al., “Linkage, asso- 1, pp. 97–103, 1993. ciation, and gene-expression analyses identify CNTNAP2 as [39] A. Rosenspire, W. Yoo, S. Menard, and A. R. Torres, an autism-susceptibility gene,” American Journal of Human “Autism spectrum disorders are associated with an elevated Genetics, vol. 82, no. 1, pp. 150–159, 2008. autoantibody response to tissue transglutaminase-2,” Autism [22] D. E. Arking, D. J. Cutler, C. W. Brune et al., “A common Research, vol. 4, no. 4, pp. 242–249, 2011. genetic variant in the neurexin superfamily member CNT- [40] P. Goines, L. Haapanen, R. Boyce et al., “Autoantibodies to NAP2 increases familial risk of autism,” American Journal of cerebellum in children with autism associate with behavior,” Human Genetics, vol. 82, no. 1, pp. 160–164, 2008. Brain, Behavior, and Immunity, vol. 25, no. 3, pp. 514–523, [23] B. Bakkaloglu, B. J. O’Roak, A. Louvi et al., “Molecular 2011. cytogenetic analysis and resequencing of contactin associated [41] S. Wills, C. C. Rossi, J. Bennett et al., “Further charac- protein-like 2 in autism spectrum disorders,” American terization of autoantibodies to GABAergic neurons in the Journal of Human Genetics, vol. 82, no. 1, pp. 165–173, 2008. central nervous system produced by a subset of children with [24] T. A. Manolio, F. S. Collins, N. J. Cox et al., “Finding the autism,” Molecular Autism, vol. 2, p. 5, 2011. missing heritability of complex diseases,” Nature, vol. 461, no. [42] M. Gonzalez-Gronow, M. Cuchacovich, R. Francos et al., 7265, pp. 747–753, 2009. “Antibodies against the voltage-dependent anion channel [25] D. B. Goldstein, “Common genetic variation and human (VDAC) and its protective ligand hexokinase-I in children traits,” The New England Journal of Medicine, vol. 360, no. 17, with autism,” Journal of Neuroimmunology, vol. 227, no. 1-2, pp. 1696–1698, 2009. pp. 153–161, 2010. [26] J. McClellan and M. C. King, “Genetic heterogeneity in [43] B. Zhang, A. Angelidou, K. D. Alysandratos et al., “Mito- human disease,” Cell, vol. 141, no. 2, pp. 210–217, 2010. chondrial DNA and anti-mitochondrial antibodies in serum [27] J.P.Gregg,L.Lit,C.A.Baronetal.,“Geneexpressionchanges of autistic children,” Journal of Neuroinflammation, vol. 7, p. in children with autism,” Genomics, vol. 91, no. 1, pp. 22–29, 80, 2010. 2008. [44] G. A. Mostafa and N. Kitchener, “Serum anti-nuclear [28] A. M. Enstrom, L. Lit, C. E. Onore et al., “Altered gene antibodies as a marker of autoimmunity in Egyptian autistic expression and function of peripheral blood natural killer children,” Pediatric Neurology, vol. 40, no. 2, pp. 107–112, cells in children with autism,” Brain, Behavior, and Immunity, 2009. vol. 23, no. 1, pp. 124–133, 2009. [29] J. T. Morgan, G. Chana, C. A. Pardo et al., “Microglial [45] M. Cabanlit, S. Wills, P. Goines, P. Ashwood, and J. van activation and increased microglial density observed in the de Water, “Brain-specific autoantibodies in the plasma of dorsolateral prefrontal cortex in autism,” Biological Psychia- subjects with autistic spectrum disorder,” Annals of the New try, vol. 68, no. 4, pp. 368–376, 2010. York Academy of Sciences, vol. 1107, pp. 92–103, 2007. [30] I. Voineagu, X. Wang, P. Johnston et al., “Transcriptomic [46] V. T. Ramaekers, N. Blau, J. M. Sequeira, M. C. Nassogne, and analysis of autistic brain reveals convergent molecular E. V. Quadros, “Folate receptor autoimmunity and cerebral pathology,” Nature, vol. 474, no. 7351, pp. 380–384, 2011. folate deficiency in low-functioning autism with neurological [31] H. Stefansson, R. A. Ophoff,S.Steinbergetal.,“Common deficits,” Neuropediatrics, vol. 38, no. 6, pp. 276–281, 2007. variants conferring risk of schizophrenia,” Nature, vol. 460, [47] A. M. Connolly, M. Chez, E. M. Streif et al., “Brain-derived no. 7256, pp. 744–747, 2009. neurotrophic factor and autoantibodies to neural antigens in [32] J. Croonenberghs, E. Bosmans, D. Deboutte, G. Kenis, and sera of children with autistic spectrum disorders, Landau- ff M. Maes, “Activation of the inflammatory response system in Kle ner syndrome, and epilepsy,” Biological Psychiatry, vol. autism,” Neuropsychobiology, vol. 45, no. 1, pp. 1–6, 2002. 59, no. 4, pp. 354–363, 2006. [33] P.Ashwood,P.Krakowiak,I.Hertz-Picciotto,R.Hansen,I.N. [48] M. Evers, C. Cunningham-Rundles, and E. Hollander, “Heat Pessah, and J. van de Water, “Associations of impaired behav- shock protein 90 antibodies in autism,” Molecular Psychiatry, iors with elevated plasma chemokines in autism spectrum vol. 7, supplement 2, pp. S26–S28, 2002. disorders,” Journal of Neuroimmunology, vol. 232, no. 1-2, pp. [49] C. M. Brickman and Y. Shoenfeld, “The mosaic of autoim- 196–199, 2011. munity,” Scandinavian Journal of Clinical and Laboratory [34] D. L. Vargas, C. Nascimbene, C. Krishnan, A. W. Zim- Investigation, vol. 61, no. 235, pp. 3–15, 2001. merman, and C. A. Pardo, “Neuroglial activation and [50] A. Vojdani, A. W. Campbell, E. Anyanwu, A. Kashanian, neuroinflammation in the brain of patients with autism,” K. Bock, and E. Vojdani, “Antibodies to neuron-specific Annals of Neurology, vol. 57, no. 1, pp. 67–81, 2005. antigens in children with autism: possible cross-reaction with [35] X. Li, A. Chauhan, A. M. Sheikh et al., “Elevated immune encephalitogenic proteins from milk, Chlamydia pneumo- response in the brain of autistic patients,” Journal of Neuroim- niae and Streptococcus group A,” Journal of Neuroimmunol- munology, vol. 207, no. 1-2, pp. 111–116, 2009. ogy, vol. 129, no. 1-2, pp. 168–177, 2002. 10 Autism Research and Treatment

[51] V. K. Singh, R. Warren, R. Averett, and M. Ghaziuddin, Journal of Autism and Developmental Disorders, vol. 26, no. “Circulating autoantibodies to neuronal and glial filament 1, pp. 87–97, 1996. proteins in autism,” Pediatric Neurology,vol.17,no.1,pp. [68]P.Ashwood,P.Krakowiak,I.Hertz-Picciotto,R.Hansen,I. 88–90, 1997. N. Pessah, and J. van de Water, “Altered T cell responses in [52] H. Jyonouchi, S. Sun, and H. Le, “Proinflammatory and children with autism,” Brain, Behavior, and Immunity, vol. regulatory cytokine production associated with innate and 25, no. 5, pp. 840–849, 2011. adaptive immune responses in children with autism spec- [69]R.P.Warren,A.Foster,andN.C.Margaretten,“Reduced trum disorders and developmental regression,” Journal of natural killer cell activity in autism,” Journal of the American Neuroimmunology, vol. 120, no. 1-2, pp. 170–179, 2001. Academy of Child and Adolescent Psychiatry,vol.26,no.3,pp. [53] V. M. Bashina, I. A. Kozlova, and T. P. Kliushnik, “An 333–335, 1987. elevation in the level of autoantibodies to nerve-growth [70] A. Vojdani, E. Mumper, D. Granpeesheh et al., “Low natural factor in the blood serum of schizophrenic children,” Zhurnal killer cell cytotoxic activity in autism: the role of glutathione, Nevrologii i Psikhiatrii Imeni S.S. Korsakova,vol.97,no.1,pp. IL-2 and IL-15,” Journal of Neuroimmunology, vol. 205, no. 47–51, 1997. 1-2, pp. 148–154, 2008. [54] A. M. Connolly, M. G. Chez, A. Pestronk, S. T. Arnold, [71] T. L. Sweeten, D. J. Posey, and C. J. McDougle, “High S. Mehta, and R. K. Deul, “Serum autoantibodies to brain blood monocyte counts and neopterin levels in children with in Landau-Kleffner variant, autism, and other neurologic autistic disorder,” American Journal of Psychiatry, vol. 160, no. disorders,” The Journal of Pediatrics, vol. 134, no. 5, pp. 607– 9, pp. 1691–1693, 2003. 613, 1999. [72] A. M. Enstrom, C. E. Onore, J. A. van de Water, and P. [55] R. D. Todd and R. D. Ciaranello, “Demonstration of inter- Ashwood, “Differential monocyte responses to TLR ligands and intraspecies differences in serotonin binding sites by in children with autism spectrum disorders,” Brain, Behavior, antibodies from an autistic child,” Proceedings of the National and Immunity, vol. 24, no. 1, pp. 64–71, 2010. Academy of Sciences of the United States of America, vol. 82, [73]A.M.Comi,A.W.Zimmerman,V.H.Frye,P.A.Law,and no. 2, pp. 612–616, 1985. J. N. Peeden, “Familial clustering of autoimmune disorders [56] J. Croonenberghs, A. Wauters, K. Devreese et al., “Increased and evaluation of medical risk factors in autism,” Journal of serum albumin, γ globulin, immunoglobulin IgG, and IgG2 Child Neurology, vol. 14, no. 6, pp. 388–394, 1999. and IgG4 in autism,” Psychological Medicine, vol. 32, no. 8, [74] T. L. Sweeten, S. L. Bowyer, D. J. Posey, G. M. Halberstadt, pp. 1457–1463, 2002. and C. J. McDougle, “Increased prevalence of familial [57] H. S. Singer, C. M. Morris, C. D. Gause, P. K. Gillin, S. autoimmunity in probands with pervasive developmental Crawford, and A. W. Zimmerman, “Antibodies against fetal disorders,” Pediatrics, vol. 112, no. 5, pp. e420–e424, 2003. brain in sera of mothers with autistic children,” Journal of [75] L. A. Croen, J. K. Grether, C. K. Yoshida, R. Odouli, and J. V. Neuroimmunology, vol. 194, no. 1-2, pp. 165–172, 2008. van de Water, “Maternal autoimmune diseases, asthma and [58] L. A. Croen, D. Braunschweig, L. Haapanen et al., “Maternal allergies, and childhood autism spectrum disorders: a case- mid-pregnancy autoantibodies to fetal brain protein: the control study,” Archives of Pediatrics and Adolescent Medicine, early markers for autism study,” Biological Psychiatry, vol. 64, vol. 159, no. 2, pp. 151–157, 2005. no. 7, pp. 583–588, 2008. [76] H. O. Atladottir,´ M. G. Pedersen, P. Thorsen et al., “Associ- [59] K. A. Hogquist, T. A. Baldwin, and S. C. Jameson, “Cen- ation of family history of autoimmune diseases and autism tral tolerance: learning self-control in the thymus,” Nature spectrum disorders,” Pediatrics, vol. 124, no. 2, pp. 687–694, Reviews Immunology, vol. 5, no. 10, pp. 772–782, 2005. 2009. [60] J. Sprent, “Central tolerance of T cells,” International Reviews [77] R. Horton, R. Gibson, P. Coggill et al., “Variation analysis of Immunology, vol. 13, no. 2, pp. 95–105, 1996. and gene annotation of eight MHC haplotypes: the MHC [61] A. Basten, R. Brink, P.Peake et al., “Self tolerance in the B-cell haplotype project,” Immunogenetics, vol. 60, no. 1, pp. 1–18, repertoire,” Immunological Reviews, no. 122, pp. 5–19, 1991. 2008. [62] S. Baumann, A. Krueger, S. Kirchhoff, and P. H. Kram- [78] L. A. Knapp, “The ABCs of MHC,” Evolutionary Anthropol- mer, “Regulation of T cell apoptosis during the immune ogy, vol. 14, no. 1, pp. 28–37, 2005. response,” Current Molecular Medicine, vol. 2, no. 3, pp. 257– [79] A. Ziegler, H. Kentenich, and B. Uchanska-Ziegler, “Female 272, 2002. choice and the MHC,” Trends in Immunology, vol. 26, no. 9, [63] Q. Leng and Z. Bentwich, “Beyond self and nonself: fuzzy pp. 496–502, 2005. recognition of the immune system,” Scandinavian Journal of [80] B. G. Xiao and H. Link, “Immune regulation within the Immunology, vol. 56, no. 3, pp. 224–232, 2002. central nervous system,” Journal of the Neurological Sciences, [64] E. Meffre and J. E. Salmon, “Autoantibody selection and pro- vol. 157, no. 1, pp. 1–12, 1998. ductioninearlyhumanlife,”Journal of Clinical Investigation, [81] G. S. Huh, L. M. Boulanger, H. Du, P. A. Riquelme, T. M. vol. 117, no. 3, pp. 598–601, 2007. Brotz, and C. J. Shatz, “Functional requirement for class I [65] F. J. Quintana and H. L. Weiner, “Understanding natural and MHC in CNS development and plasticity,” Science, vol. 290, pathological autoimmunity,” Journal of Neuroimmunology, no. 5499, pp. 2155–2159, 2000. vol. 174, no. 1-2, pp. 1–2, 2006. [82] L. M. Boulanger and C. J. Shatz, “Immune signalling in [66]R.P.Warren,N.C.Margaretten,N.C.Pace,andA.Foster, neural development, synaptic plasticity and disease,” Nature “Immune abnormalities in patients with autism,” Journal of Reviews Neuroscience, vol. 5, no. 7, pp. 521–531, 2004. Autism and Developmental Disorders, vol. 16, no. 2, pp. 189– [83] S. Cullheim and S. Thams, “The microglial networks of the 197, 1986. brain and their role in neuronal network plasticity after [67] D. R. Denney, B. W. Frei, and G. R. Gaffney, “Lymphocyte lesion,” Brain Research Reviews, vol. 55, no. 1, pp. 89–96, subsets and interleukin-2 receptors in autistic children,” 2007. Autism Research and Treatment 11

[84] M. Ohtsuka, H. Inoko, J. K. Kulski, and S. Yoshimura, during pregnancy,” Archives of Pediatrics and Adolescent “Major histocompatibility complex (Mhc) class Ib gene Medicine, vol. 163, no. 6, pp. 542–546, 2009. duplications, organization and expression patterns in mouse [101] Y. L. Chien, Y. Y. Wu, C. H. Chen et al., “Association of HLA- strain C57BL/6,” BMC Genomics, vol. 9, p. 178, 2008. DRB1 alleles and neuropsychological function in autism,” [85] S. L. Bailey, P. A. Carpentier, E. J. McMahon, W. S. Begolka, Psychiatric Genetics, vol. 22, no. 1, pp. 46–49, 2012. and S. D. Miller, “Innate and adaptive immune responses of [102] R. P. Warren, R. A. Burger, D. Odell, A. R. Torres, and W. the central nervous system,” Critical Reviews in Immunology, L. Warren, “Decreased plasma concentrations of the C4B vol. 26, no. 2, pp. 149–188, 2006. complement protein in autism,” Archives of Pediatrics and [86] T. Shiina, H. Inoko, and J. K. Kulski, “An update of the HLA Adolescent Medicine, vol. 148, no. 2, pp. 180–183, 1994. genomic region, locus information and disease associations,” [103] G. A. Mostafa and A. A. Shehab, “The link of C4B null Tissue Antigens, vol. 64, no. 6, pp. 631–649, 2004. allele to autism and to a family history of autoimmunity in [87] T. Shiina, K. Hosomichi, H. Inoko, and J. K. Kulski, “The Egyptian autistic children,” Journal of Neuroimmunology, vol. HLA genomic loci map: expression, interaction, diversity and 223, no. 1-2, pp. 115–119, 2010. disease,” Journal of Human Genetics, vol. 54, no. 1, pp. 15–39, [104] L. F. Barcellos, S. L. May, P. P. Ramsay et al., “High-density 2009. SNP screening of the major histocompatibility complex in systemic lupus erythematosus demonstrates strong evidence [88] C. Vandiedonck, M. S. Taylor, H. E. Lockstone et al., for independent susceptibility regions,” PLoS Genetics, vol. 5, “Pervasive haplotypic variation in the spliceo-transcriptome no. 10, Article ID e1000696, 2009. of the human major histocompatibility complex,” Genome [105] R. Kurata, H. Nakaoka, A. Tajima et al., “TRIM39 and RNF39 Research, vol. 21, no. 7, pp. 1042–1054, 2011. are associated with Behc¸et’s disease independently of HLA- [89] G. Candore, I. C. Campagna, I. Cuppari, D. di Carlo, C. B∗51 and -A∗26,” Biochemical and Biophysical Research Mineo, and C. Caruso, “Genetic control of immune response Communications, vol. 401, no. 4, pp. 533–537, 2010. in carriers of the 8.1 ancestral haplotype,” Annals of the New [106] Y. Allanore, M. Saad, P. Dieude´ et al., “Genome-wide scan York Academy of Sciences, vol. 1110, pp. 151–158, 2007. identifies TNIP1, PSORS1C1, and RHOB as novel risk loci [90] E. G. Stubbs and R. E. Magenis, “HLA and autism,” Journal for systemic sclerosis,” PLoS Genetics, vol. 7, no. 7, Article ID of Autism and Developmental Disorders, vol. 10, no. 1, pp. 15– e1002091, 2011. 19, 1980. [107] S. Gonzalez, J. Martinez-Borra, J. S. del R´ıo et al., “The [91] R. P. Warren, V. K. Singh, P. Cole et al., “Increased frequency OTF3 gene polymorphism confers susceptibility to psoriasis of the null allele at the complement C4b locus in autism,” independent of the association of HLA-Cw∗0602,” Journal of Clinical and Experimental Immunology,vol.83,no.3,pp. Investigative Dermatology, vol. 115, no. 5, pp. 824–828, 2000. 438–440, 1991. [108] K. Asumalahti, C. Veal, T. Laitinen et al., “Coding haplotype [92] W. W. Daniels, R. P. Warren, J. D. Odell et al., “Increased analysis supports HCR as the putative susceptibility gene frequency of the extended or ancestral haplotype B44-SC30- for psoriasis at the MHC PSORS1 locus,” Human Molecular DR4 in autism,” Neuropsychobiology, vol. 32, no. 3, pp. 120– Genetics, vol. 11, no. 5, pp. 589–597, 2002. 123, 1995. [109] S. J. Holm, L. M. Carlen,´ L. Mallbris, M. Stahle-B˚ ackdahl,¨ [93] A. R. Torres, A. Maciulis, E. G. Stubbs, A. Cutler, and and K. P. O’Brien, “Polymorphisms in the SEEK1 and SPR1 D. Odell, “The transmission disequilibrium test suggests genes on 6p21.3 associate with psoriasis in the Swedish that HLA-DR4 and DR13 are linked to autism spectrum population,” Experimental Dermatology, vol. 12, no. 4, pp. disorder,” Human Immunology, vol. 63, no. 4, pp. 311–316, 435–444, 2003. 2002. [110] S. J. Holm, F. Sanchez,´ L. M. Carlen,´ L. Mallbris, M. Stahle,˚ ∗ [94] D. Odell, A. Maciulis, A. Cutler et al., “Confirmation of andK.P.O’Brien,“HLA-Cw 0602 associates more strongly the association of the C4B null allelle in autism,” Human to psoriasis in the Swedish population than variants of the Immunology, vol. 66, no. 2, pp. 140–145, 2005. novel 6p21.3 gene PSORS1C3,” Acta Dermato-Venereologica, [95] F. R. Guerini, E. Bolognesi, S. Manca et al., “Family- vol. 85, no. 1, pp. 2–8, 2005. based transmission analysis of HLA genetic markers in [111] S. Orru,` E. Giuressi, C. Carcassi, M. Casula, and L. Sardinian children with autistic spectrum disorders,” Human Contu, “Mapping of the major psoriasis-susceptibility locus Immunology, vol. 70, no. 3, pp. 184–190, 2009. (PSORS1) in a 70-Kb interval around the corneodesmosin gene (CDSN),” American Journal of Human Genetics, vol. 76, [96] F. R. Guerini, E. Bolognesi, M. Chiappedi et al., “HLA no. 1, pp. 164–171, 2005. polymorphisms in Italian children with autism spectrum [112] G. Gambelunghe, A. Brozzetti, M. Ghaderi, P. Candeloro, disorders: results of a family based linkage study,” Journal of C. Tortoioli, and A. Falorni, “MICA gene polymorphism in Neuroimmunology, vol. 230, no. 1-2, pp. 135–142, 2011. the pathogenesis of type 1 diabetes,” Annals of the New York [97]R.P.Warren,J.D.Odell,W.L.Warrenetal.,“Strong β Academy of Sciences, vol. 1110, pp. 92–98, 2007. association of the third hypervariable region of HLA-DR 1 [113] A. Falorni, A. Brozzetti, D. L. Torre, C. Tortoioli, and G. with autism,” Journal of Neuroimmunology, vol. 67, no. 2, pp. Gambelunghe, “Association of genetic polymorphisms and 97–102, 1996. autoimmune Addison’s disease,” Expert Review of Clinical [98] D. E. de Almeida, S. Ling, and J. Holoshitz, “New insights Immunology, vol. 4, no. 4, pp. 441–456, 2008. into the functional role of the rheumatoid arthritis shared [114] G. Gambelunghe, R. Gerli, E. B. Bocci et al., “Contribution epitope,” FEBS Letters, vol. 585, no. 23, pp. 3619–3626, 2011. of MHC class I chain-related A (MICA) gene polymorphism [99] L. C. Lee, A. A. Zachary, M. S. Leffell et al., “HLA-DR4 in to genetic susceptibility for systemic lupus erythematosus,” families with autism,” Pediatric Neurology,vol.35,no.5,pp. Rheumatology, vol. 44, no. 3, pp. 287–292, 2005. 303–307, 2006. [115] A. Gnjec, K. L. D’Costa, S. M. Laws et al., “Association of [100] W. G. Johnson, S. Buyske, A. E. Mars et al., “HLA-DR4 as a alleles carried at TNFA -850 and BAT1 -22 with Alzheimer’s risk allele for autism acting in mothers of probands possibly disease,” Journal of Neuroinflammation, vol. 5, p. 36, 2008. 12 Autism Research and Treatment

[116] S. Limou, S. le Clerc, C. Coulonges et al., “Genomewide patients with sarcoidosis,” Clinical Respiratory Journal, vol. 5, association study of an AIDS-nonprogression cohort empha- no. 2, pp. 105–111, 2011. sizes the role played by HLA genes (ANRS genomewide [131] C. W. Pyo, S. S. Hur, Y. K. Kim, T. Y. Kim, and T. G. Kim, association study 02),” Journal of Infectious Diseases, vol. 199, “Association of TAP and HLA-DM genes with psoriasis in no. 3, pp. 419–426, 2009. Koreans,” Journal of Investigative Dermatology, vol. 120, no. [117] J. Castiblanco and J. M. Anaya, “The IκBL gene polymor- 4, pp. 616–622, 2003. phism influences risk of acquiring systemic lupus erythe- [132] A.´ Camarena, A. Aquino-Galvez, R. Falfan-Valencia´ et al., matosus and Sjogren’s¨ syndrome,” Human Immunology, vol. “PSMB8 (LMP7) but not PSMB9 (LMP2) gene polymor- 69, no. 1, pp. 45–51, 2008. phisms are associated to pigeon breeder’s hypersensitivity [118] G. Tamiya, M. Shinya, T. Imanishi et al., “Whole genome pneumonitis,” Respiratory Medicine, vol. 104, no. 6, pp. 889– association study of rheumatoid arthritis using 27039 894, 2010. [133] U. Kramer,T.Illig,T.Grune,J.Krutmann,andC.Esser,¨ microsatellites,” Human Molecular Genetics, vol. 14, no. 16, “Strong associations of psoriasis with antigen processing pp. 2305–2321, 2005. LMP and transport genes TAP differ by gender and pheno- ff [119] K. Reich, U. Hu¨ meier, I. R. Konig¨ et al., “TNF polymor- type,” Genes and Immunity, vol. 8, no. 6, pp. 513–517, 2007. phisms in psoriasis: association of psoriatic arthritis with ∗ [134] C. B. Casp, J. X. She, and W. T. McCormack, “Genes the promoter polymorphism TNF -857 independent of the of the LMP/TAP cluster are associated with the human PSORS1 risk allele,” Arthritis and Rheumatism,vol.56,no.6, autoimmune disease vitiligo,” Genes and Immunity, vol. 4, no. pp. 2056–2064, 2007. 7, pp. 492–499, 2003. [120] L. C. Oliveira, G. Porta, M. L. C. Marin, P. L. Bittencourt, J. [135] M. L. Sanchez, K. Katsumata, T. Atsumi et al., “Association of Kalil, and A. C. Goldberg, “Autoimmune hepatitis, HLA and HLA-DM polymorphism with the production of antiphos- extended haplotypes,” Autoimmunity Reviews, vol. 10, no. 4, pholipid antibodies,” Annals of the Rheumatic Diseases, vol. pp. 189–193, 2011. 63, no. 12, pp. 1645–1648, 2004. [121] R. Kieszko, P. Krawczyk, S. Chocholska, A. Dmoszynska,´ and [136] J. Morel, F. Roch-Bras, N. Molinari, J. Sany, J. F. Eliaou, and J. Milanowski, “TNF-α and TNF-β gene polymorphisms in B. Combe, “HLA-DMA∗0103 and HLA-DMB∗0104 alleles as Polish patients with sarcoidosis. Connection with the suscep- novel prognostic factors in rheumatoid arthritis,” Annals of tibility and prognosis,” Sarcoidosis Vasculitis and Diffuse Lung the Rheumatic Diseases, vol. 63, no. 12, pp. 1581–1586, 2004. Diseases, vol. 27, no. 2, pp. 131–137, 2010. [137] J. Morel, C. S. Cda, O. Avinens, J. Sany, B. Combe, and J. [122] M. C. Eike, M. Olsson, D. E. Undlien et al., “Genetic variants F. Eliaou, “Polymorphism of HLA-DMA and DMB alleles of the HLA-A, HLA-B and AIF1 loci show independent in patients with systemic lupus erythematosus,” Journal of associations with type 1 diabetes in Norwegian families,” Rheumatology, vol. 30, no. 7, pp. 1485–1490, 2003. Genes and Immunity, vol. 10, no. 2, pp. 141–150, 2009. [138] Y. M. Sang, C. Yan, C. Zhu, G. C. Ni, and Y. M. Hu, “Association of human leukocyte antigen non-classical genes [123] H. Cwiklinska, M. P. Mycko, B. Szymanska, M. Matysiak, and with type 1 diabetes,” Chinese Journal of Pediatrics, vol. 41, K. W. Selmaj, “Aberrant stress-induced Hsp70 expression in no. 4, pp. 260–263, 2003. immune cells in multiple sclerosis,” Journal of Neuroscience [139] X. Chen and P. E. Jensen, “MHC class II antigen presentation Research, vol. 88, no. 14, pp. 3102–3110, 2010. and immunological abnormalities due to deficiency of MHC [124] M. C. Pickering and M. J. Walport, “Links between com- class II and its associated genes,” Experimental and Molecular plement abnormalities and systemic lupus erythematosus,” Pathology, vol. 85, no. 1, pp. 40–44, 2008. Rheumatology, vol. 39, no. 2, pp. 133–141, 2000. [140] J. I. Satoh, M. Nakanishi, F. Koike et al., “Microarray analysis [125] D. Franciotta, M. Cuccia, E. Dondi, G. Piccolo, and V. Cosi, identifies an aberrant expression of apoptosis and DNA “Polymorphic markers in MHC class II/III region: a study damage-regulatory genes in multiple sclerosis,” Neurobiology on Italian patients with myasthenia gravis,” Journal of the of Disease, vol. 18, no. 3, pp. 537–550, 2005. Neurological Sciences, vol. 190, no. 1-2, pp. 11–16, 2001. [141] S. L. Montgomery and W. J. Bowers, “Tumor necrosis factor- [126] F. Jenhani, R. Bardi, Y. Gorgi, K. Ayed, and M. Jeddi, “C4 α and the roles it plays in homeostatic and degenerative polymorphism in multiplex families with insulin dependent processes within the central nervous system,” Journal of diabetes in the Tunisian population: standard C4 typing Neuroimmune Pharmacology. In press. methods and RFLP analysis,” Journal of Autoimmunity, vol. [142] M. K. McCoy and M. G. Tansey, “TNF signaling inhibition 5, no. 2, pp. 149–160, 1992. in the CNS: implications for normal brain function and [127] M. M. Fernando, C. R. Stevens, P. C. Sabeti et al., “Identifi- neurodegenerative disease,” Journal of Neuroinflammation, cation of two independent risk factors for lupus within the vol. 5, article 45, 2008. MHC in United Kingdom families,” PLoS Genetics, vol. 3, no. [143] I. G. Ovsyannikova, R. A. Vierkant, V. S. Pankratz, R. M. 11, p. e192, 2007. Jacobson, and G. A. Poland, “Extended LTA, TNF, LST1 and hla gene haplotypes and their association with rubella [128] O. Gorlova, J. E. Martin, B. Rueda et al., “Identification of vaccine-induced immunity,” PLoS ONE, vol. 5, no. 7, Article novel genetic markers associated with clinical phenotypes ID e11806, 2010. of systemic sclerosis through a genome-wide association [144] U. S. Naik, C. Gangadharan, K. Abbagani, B. Nagalla, N. strategy,” PLoS Genetics, vol. 7, no. 7, Article ID e1002178, Dasari, and S. K. Manna, “A study of nuclear transcription 2011. factor-κ B in childhood autism,” PLoS ONE, vol. 6, no. 5, [129] S. Pathan, R. E. Gowdy, R. Cooney et al., “Confirmation Article ID e19488, 2011. of the novel association at the BTNL2 locus with ulcerative [145] A. M. Young, E. Campbell, S. Lynch, J. Suckling, and S. colitis,” Tissue Antigens, vol. 74, no. 4, pp. 322–329, 2009. J. Powis, “Aberrant NF-κB expression in autism spectrum [130] N. Milman, C. B. Svendsen, F. C. Nielsen, and T. V. O. condition: a mechanism for neuroinflammation,” Front Hansen, “The BTNL2 A allele variant is frequent in Danish Psychiatry, vol. 2, no. 27, pp. 1–8, 2011. Autism Research and Treatment 13

[146] C. E. Foster, M. Colonna, and P. D. Sun, “Crystal structure of the human natural killer (NK) cell activating receptor NKp46 reveals structural relationship to other leukocyte receptor complex immunoreceptors,” Journal of Biological Chemistry, vol. 278, no. 46, pp. 46081–46086, 2003. [147] G. Turturici, G. Sconzo, and F. Geraci, “Hsp70 and its molecular role in nervous system diseases,” Biochemistry Research International, vol. 2011, Article ID 618127, 18 pages, 2011. [148] J. I. Kakimura, Y. Kitamura, K. Takata et al., “Microglial activation and amyloid-β clearance induced by exogenous heat-shock proteins,” The FASEB Journal,vol.16,no.6,pp. 601–603, 2002. [149] M. P. Mycko, H. Cwiklinska, A. Walczak, C. Libert, C. S. Raine, and K. W. Selmaj, “A heat shock protein gene (Hsp70.1) is critically involved in the generation of the immune response to myelin antigen,” European Journal of Immunology, vol. 38, no. 7, pp. 1999–2013, 2008. [150] D. G. Millar, K. M. Garza, B. Odermatt et al., “Hsp70 promotes antigen-presenting cell function and converts T- cell tolerance to autoimmunity in vivo,” Nature Medicine, vol. 9, no. 12, pp. 1469–1476, 2003. [151] A. Pawlik, M. Kurzawski, T. Szczepanik et al., “Association of allograft inflammatory factor-1 gene polymorphism with rheumatoid arthritis,” Tissue Antigens, vol. 72, no. 2, pp. 171– 175, 2008. [152] T. Hou, H. Macmillan, Z. Chen et al., “An insertion mutant in DQA1∗0501 restore susceptibility to HLA-DM: implications for disease associations,” Journal of Immunology, vol. 187, no. 5, pp. 2442–2452, 2011. [153] F. Deshaies, D. A. Diallo, J. S. Fortin et al., “Evidence for a human leucocyte antigen-DM-induced structural change in human leucocyte antigen-DOβ,” Immunology, vol. 127, no. 3, pp. 408–417, 2009. [154] P. S. Ramos, C. D. Langefeld, L. A. Bera, P. M. Gaffney, J. A. Noble, and K. L. Moser, “Variation in the ATP-binding cassette transporter 2 gene is a separate risk factor for systemic lupus erythematosus within the MHC,” Genes and Immunity, vol. 10, no. 4, pp. 350–355, 2009. [155] H. A. Engstrom, S. Ohlson, E. G. Stubbs et al., “Decreased expression of CD95 (FAS/APO-1) on CD4+ T-lymphocytes from participants with autism,” Journal of Developmental and Physical Disabilities, vol. 15, no. 2, pp. 155–163, 2003. [156] P. Ferrante, M. Saresella, F. R. Guerini, M. Marzorati, M. C. Musetti, and A. G. Cazzullo, “Significant association of HLA A2-DR11 with CD4 naive decrease in autistic children,” Biomedicine and Pharmacotherapy, vol. 57, no. 8, pp. 372– 374, 2003. [157] A. R. Torres, T. L. Sweeten, A. Cutler et al., “The association and linkage of the HLA-A2 class I allele with autism,” Human Immunology, vol. 67, no. 4-5, pp. 346–351, 2006. Hindawi Publishing Corporation Autism Research and Treatment Volume 2012, Article ID 986519, 10 pages doi:10.1155/2012/986519

Research Article Intracellular and Extracellular Redox Status and Free Radical Generation in Primary Immune Cells from Children with Autism

Shannon Rose, Stepan Melnyk, Timothy A. Trusty, Oleksandra Pavliv, Lisa Seidel, Jingyun Li, Todd Nick, and S. Jill James

Department of Pediatrics, Arkansas Children’s Hospital Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA

Correspondence should be addressed to S. Jill James, [email protected]

Received 30 July 2011; Revised 12 August 2011; Accepted 12 September 2011

Academic Editor: Antonio M. Persico

Copyright © 2012 Shannon Rose et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

The modulation of the redox microenvironment is an important regulator of immune cell activation and proliferation. To investigate immune cell redox status in autism we quantified the intracellular glutathione redox couple (GSH/GSSG) in resting peripheral blood mononuclear cells (PBMCs), activated monocytes and CD4 T cells and the extracellular cysteine/cystine redox couple in the plasma from 43 children with autism and 41 age-matched control children. Resting PBMCs and activated monocytes from children with autism exhibited significantly higher oxidized glutathione (GSSG) and percent oxidized glutathione equivalents and decreased glutathione redox status (GSH/GSSG). In activated CD4 T cells from children with autism, the percent oxidized glutathione equivalents were similarly increased, and GSH and GSH/GSSG were decreased. In the plasma, both glutathione and cysteine redox ratios were decreased in autistic compared to control children. Consistent with decreased intracellular and extracellular redox status, generation of free radicals was significantly elevated in lymphocytes from the autistic children. These data indicate primary immune cells from autistic children have a more oxidized intracellular and extracellular microenvironment and a deficit in glutathione-mediated redox/antioxidant capacity compared to control children. These results suggest that the loss of glutathione redox homeostasis and chronic oxidative stress may contribute to immune dysregulation in autism.

1. Introduction (GSH) and several of its metabolic precursors, an increase in oxidized glutathione disulfide (GSSG), and a decrease in Autism is a behaviorally defined neurodevelopmental disor- glutathione redox ratio (GSH/GSSG) in case-control evalua- der that usually presents in early childhood and is charac- tions of plasma and lymphoblastoid cell lines derived from terized by significant impairments in social interaction and children with autism [13–16]. Recently, several interactive communication and by abnormal repetitive hyper-focused polymorphisms in enzymes regulating glutathione synthesis behaviors. The prevalence of autism spectrum disorders has were found to be more prevalent in children with autism increased more than 10-fold in the last two decades, now suggesting that the glutathione deficit and predisposition to affecting one in 110 US children, yet the etiology of these oxidative stress may be genetically based in some children disorders remains elusive [1]. Glutathione depletion and oxi- [17]. dative stress have been implicated in the pathology of numer- Oxidative stress occurs when cellular antioxidant defense ous neurobehavioral disorders including schizophrenia [2], mechanisms fail to counterbalance endogenous ROS produc- bipolar disorder [3], and Alzheimer’s disease [4]. Accumu- tion and/or exogenous prooxidant environmental exposures. lating evidence suggests that redox imbalance and oxidative Glutathione (γ-L-glutamyl-L-cysteinylglycine) is a tripeptide stress may also contribute to autism pathophysiology. Mul- that functions as the major intracellular antioxidant and tiple biomarkers of oxidative stress have been identified in redox buffer against macromolecular oxidative damage. The blood samples from children with autism [5–12]. Our group glutathione thiol/disulfide redox couple (GSH/GSSG) is the has reported a decrease in concentrations of glutathione predominant mechanism for maintaining the intracellular 2 Autism Research and Treatment microenvironment in a highly reduced state that is essential with autism have been shown to have abnormal responses for antioxidant/detoxification capacity, redox enzyme regula- to stimulation, we also elected to challenge the PBMCs with tion, cell cycle progression, and transcription of antioxidant immune activators known to promote oxidative stress and response elements (ARE) [18–23]. Subtle variation in the measure the resulting intracellular glutathione redox status relative concentrations of reduced and oxidized glutathione in activated isolated monocytes and T cells. provides a dynamic redox signaling mechanism that regu- lates these vital cellular processes [24–27]. For example, in both CNS precursor cells and na¨ıve immune cells, intracel- 2. Subjects and Methods lular glutathione redox status is the primary determinant 2.1. Participants. This investigation was conducted on a modulating the cellular decision to undergo cell cycle arrest, subset of children from the autism IMAGE (Integrated differentiation, or proliferation [27]. A reducing intracellular Metabolic and Genomic Endeavor) study at Arkansas Chil- environment is required for proliferation, while a more dren’s Hospital Research Institute (ACHRI) that has recruit- oxidized microenvironment favors cell cycle arrest and dif- ed over 162 case and control families to date. The IMAGE ferentiation. A chronic deficit in the GSH/GSSG redox ratio cohort for this study consisted of 43 children diagnosed is considered to be a reliable indicator of oxidative stress and with autistic disorder and 41 unaffected control children increased vulnerability to oxidative damage from prooxidant (16 of which were unaffected siblings). The autism case environmental exposures [28, 29]. families were recruited locally after referral to the University In the extracellular plasma compartment, the cysteine/ of Arkansas for Medical Sciences (UAMS), Dennis Devel- cystine (thiol/disulfide) redox couple independently provides opmental Center and diagnosed by trained developmental the ambient redox environment for circulating immune cells. pediatricians. Children aged 3 to 10 with a diagnosis of The ambient extracellular cysteine/cystine redox potential autistic disorder as defined by the Diagnostic and Statistical has been shown to be more oxidized than the intracellular Manual of Mental Disorders, Fourth Edition (DSM-IV 299.0), GSH/GSSG redox potential and is independently regulated the Autism Diagnostic Observation Schedule (ADOS), [30]. Dynamic shifts in the plasma cysteine/cystine redox and/or the Childhood Autism Rating Scales (CARS >30) potential alter the redox status of cysteine moieties in cell were enrolled. Children diagnosed with other conditions on surface proteins to induce conformational changes in protein the autism spectrum or rare genetic diseases associated with structure that can reversibly alter function [31, 32]. For ex- symptoms of autism were excluded from the study. Children ample, under oxidizing extracellular conditions, redox-sensi- with chronic seizure disorders, recent infection, and high- tive cysteine residues in the catalytic core of protein tyrosine dose vitamin or mineral supplements exceeding the RDA phosphatases become oxidized and reversibly inactivate were also excluded because these conditions are potential enzyme activity depending on the ambient cysteine/cystine confounders that could affect redox status. Unaffected sib- redox potential [31, 33, 34]. Extracellular cysteine/cystine lings and unrelated, neurotypical children aged 3 to 10 with redox status is emerging as an important new signal trans- no medical history of behavioral or neurologic abnormalities duction mechanism that can induce posttranslational alter- by parent report made up the comparison group. The ations in downstream redox-sensitive proteins including a protocol was approved by the Institutional Review Board at variety of enzymes, transcription factors, receptors, adhesion UAMS, and all parents signed informed consent. molecules, and membrane signaling proteins resulting in the dynamic modulation of their activity and function [32, 35, 36]. 2.2. Materials. Culture flasks, plates, and pipettes were ob- Recent studies have revealed numerous immunologic tained from Corning Life Sciences (Lowell, Mass, USA). abnormalities among children with autism including alter- RPMI 1640, penicillin/streptomycin, Dulbecco’s phosphate- ff ations in immune cell proportions [37–40] and shifts in bu ered saline (PBS), fetal bovine serum (FBS), and glu- tamine were purchased from Life Technologies (Carlsbad, helper T-cell subpopulations after mitogenic stimulation [41, 42]. Peripheral blood mononuclear cells (PBMCs) from Calif, USA). Carboxy-H2DCFDA (6-carboxy-2 ,7 -dichlo- individuals with autism have been shown to produce higher rodihydrofluorescein diacetate, diacetoxymethyl ester) was levels of proinflammatory cytokines and abnormal levels of obtained from Molecular Probes (Carlsbad, Calif, USA). regulatory cytokines compared to control PBMCs at baseline Human Monocyte Isolation Kit II and Human CD4 T and upon mitogenic stimulation [43–46]. Taken together, cell Isolation Kit II were purchased from Miltenyi Biotec the immunological studies suggest a role for a dysregulated (Bergisch-Gladbach, Germany). Histopaque-1077 and all immune system in autism that potentially could be related to other chemicals were obtained from Sigma-Aldrich (St. a deficit in glutathione-mediated antioxidant capacity and an Louis, Mo, USA). oxidized microenvironment in immune cells. To investigate this possibility, we examined whether primary immune cells 2.3. Isolation of PBMCs and Stimulation of Monocytes and (PBMCs) from children with autism exhibit decreased in- CD4 T Cells. Fasting blood samples (≤20 mL) were collected tracellular glutathione redox capacity compared to PBMCs before 9:00 AM into EDTA-Vacutainer tubes and immedi- from age-matched control children and whether a more ately chilled on ice before centrifuging at 1300 ×gfor10min oxidized intracellular and extracellular microenvironment at 4◦C. Aliquots of plasma were stored at −80◦C in cryostat is associated with increased production of oxidizing intra- tubes until extraction and HPLC quantification. PBMCs cellular free radicals. Because immune cells from children were isolated by centrifugation over Histopaque-1077. Red Autism Research and Treatment 3 blood cells were lysed using a brief (15 s) incubation with Table 1: Demographics of study population. 1 mL ice-cold water. Approximately, 30 × 106 PBMCs were resuspended in RPMI 1640 medium (supplemented with Case children Control children n = 43 n = 41 10% FBS, 1% penicillin/streptomycin, and 2 mM glutamine) at a density of 106 cells/mL. Note that because we were unable Age; mean (SD) 5.42 (1.98) 6.16 (2.29) n to obtain 20 mL blood volume from every child, it was not Male; (%) 36 (84) 20 (49) possible to isolate and analyze monocytes and CD4 T cells White; n (%) 38 (88.4) 31 (75.6) for all participants. For monocyte stimulation, PBMCs were Asian; n (%) 2 (4.65) 0 (0) treated with 0.1 μg/mL lipopolysaccharide (LPS); for T-cell African American; n (%) 2 (4.65) 8 (19.5) stimulation, PBMCs were treated with 10 ng/mL phorbol 12- Hispanic; n (%) 1 (2.3) 2 (4.9) μ myristate 13-acetate (PMA) and 1 g/mL ionomycin. Cells OTC multivitamin use; n (%) ◦ 17 (39.5) 8 (19.5) were placed in a humidified 5% CO2 incubator at 37 C for 4 hr. Stimulated monocytes and CD4 T cells were then isolated by negative selection using magnetic cell labeling as fluorescence normalized to DCF fluorescence of a standard described by the manufacturer (Miltenyi Biotec, Bergisch- PBMC preparation. As an internal control, the standard Gladbach, Germany). Using flow cytometry, we determined PBMC preparation was isolated from a 100 mL blood sample ≥ that 75% of isolated monocytes are positive for CD14 and from an unaffected healthy adult volunteer, aliquoted and ≥ that 87% of isolated CD4 T cells are positive for CD4. For frozen at −180◦C in 90% FBS/10% DMSO. An aliquot of HPLC quantification of GSH and GSSG, approximately 2 × 6 the standard PBMC preparation was stained and analyzed 10 unstimulated (resting) PBMCs, stimulated monocytes, with each subject sample. Evaluation of oxidizing free radical or stimulated CD4 T cells were pelleted, snap frozen on dry production was possible only in those case and unrelated − ◦ ice,andstoredat 80 C. control samples for which sufficient (∼20 mL) blood volume was obtained. 2.4. Cell Extraction and HPLC Quantification of Intracellular Glutathione and Plasma Cysteine Redox Status. The storage 2.6. Statistical Analysis. Within the control group, 16 of the ◦ interval at −80 C before extraction was consistently between 41 unaffected control children were case siblings. There were 1-2 weeks after blood draw and cell isolation to minimize 27 additional case children without a sibling and 25 addi- potential metabolite interconversion. The methodological tional unrelated control children comprising the total case- details for intracellular and extracellular GSH extraction control cohort of 84 children. To down-weight the impact of and HPLC elution and electrochemical detection have been outliers, three metabolites observations were curtailed at the described previously [15, 16], and metabolite detection does extremes of the distributions for PBMC GSH, PBMC GSSG, not require derivatization. Although most GSSG is present and Monocytes GSH/GSSG (see footnote in Table 2). The as a mixed disulfide with other thiols including cysteine, sibling data are correlated resulting in a combined sample our measurements detect only the free GSSG in plasma. of correlated and uncorrelated data; thus, the assumption of Glutathione and cysteine concentrations were calculated all data being independent is not satisfied for the standard from peak areas of standard calibration curves using HPLC two-sample t-test. To make use of all data from dependent software. Intracellular results are expressed as nanomoles and independent observations, we used the corrected Z-test per milligram of protein using the BCA Protein Assay Kit proposed by Looney and Jones [47]. This statistical approach (Pierce, Rockford, Ill, USA), and plasma results are expressed provides adequate control of Type 1 errors and has more as micromoles per liter. power than a standard Student’s t-test. Because the DCF data compared cases and unrelated controls (without siblings) 2.5. Measurement of Intracellular Free Radicals. Carboxy- the standard Student’s t-test was used with significance set H2DCFDA (DCF) is a membrane-permeable ROS/RNS-sen- at 0.05. Nonparametric intercorrelations (Spearman correla- sitive probe that remains nonfluorescent until oxidized by tion coefficients) between age and gender and the 7 outcome intracellular free radicals. The intensity of DCF fluorescence variables, GSH, GSSG, GSH/GSSG, % oxidized glutathione, is directly proportional to the level of free radical oxidation. cysteine, cystine, and cysteine/cystine were determined with Approximately, 106 PBMCs were resuspended in 1 mL RPMI the significance level set at 0.05. Data was analyzed using SAS 1640 medium supplemented with 10% FBS, 1% penicil- 9.2 software (SAS Institute Inc, Cary, NC, USA). lin/streptomycin, and 2 mM glutamine and stained in the ◦ dark for 20 min with 1 μMDCFat37C. Stained cells were 3. Results washed and resuspended in PBS and analyzed immediately onaPartecCyFlowflowcytometer(Gorlitz,¨ Germany) using 3.1. Demographics of Study Population. Table 1 lists the 488 nm excitation wavelength with 530/30 nm (FL1) emis- demographics of the study population. The only major sion filter. For each analysis, the fluorescence properties of differences between cases and controls are that the control 10000 cells were collected, and the data were analyzed using group was composed of a greater proportion of females and the FCS Express software (De Novo Software, Los Angeles, African Americans, whereas the case group had a greater pro- Calif, USA). Intracellular free radical levels are expressed as portion of Asian subjects. Over-the-counter multivitamin median fluorescence intensity (MFI) of subject sample DCF supplement use was higher among cases (39.5%) compared 4 Autism Research and Treatment

Table 2: Intracellular glutathione redox status in resting PBMCs and activated monocytes and CD4 T cells.

Case children Control children Corrected Z-test Metabolite Difference n Mean ± SD n Mean ± SD P value (95% CI) Resting PBMCs GSH (nmol/mg protein) 43 25.45 ± 8.16 41 23.35 ± 6.38 2.09 (−1.09, 5.29) 0.19 GSSG (nmol/mg protein) 43 0.90 ± 0.3410.66 ± 0.23 0.24 (0.13, 0.35) <0.001 GSH/GSSG 43 29.58 ± 9.04 41 37.58 ± 10.89 −7.99 (−12.51, −3.48) <0.001 Oxidized GSH (%) 43 0.07 ± 0.02 41 0.05 ± 0.01 0.02 (0.0075, 0.024) <0.001 Activated monocytes GSH (nmol/mg protein) 18 7.73 ± 3.16 20 8.55 ± 2.5 −0.82 (−2.02, 0.38) 0.18 GSSG (nmol/mg protein) 18 0.62 ± 0.24 20 0.47 ± 0.17 0.14 (0.03, 0.25) 0.01 GSH/GSSG 18 13.31 ± 7.26 20 19.30 ± 6.35 −5.98 (−9.99, −1.97) 0.003 Oxidized GSH (%) 18 0.14 ± 0.05 20 0.10 ± 0.03 0.04 (0.02, 0.07) <0.001 Activated CD4 T cells GSH (nmol/mg protein) 18 6.82 ± 3.01910.16 ± 3.74 −3.33 (−5.24, −1.42) <0.001 GSSG (nmol/mg protein) 18 0.68 ± 0.29 19 0.63 ± 0.24 0.05 (−0.11, 0.22) 0.51 GSH/GSSG 18 10.47 ± 4.19 19 17.49 ± 6.95 −7.02 (−10.17, −3.87) <0.001 Oxidized GSH (%) 18 0.17 ± 0.05 19 0.11 ± 0.05 0.05 (0.03, 0.08) <0.001 GSH: glutathione; GSSG: oxidized glutathione disulfide; oxidized GSH: (%)2GSSG/(GSH+2GSSG); curtailment: PBMC GSH >45 set = 45 (n = 1); PBMC GSSG >1.75 set = 1.75 (n = 1); Monocytes GSH/GSSG >35 set = 35 (n = 1).

to controls (19.5%); however, the glutathione redox status of the outcome measures. The protein content per 106 cells was statistically unaffected by vitamin use (data not shown). did not differ between case and control children (data not shown). 3.2. Decreased Intracellular Glutathione Redox Status in Autism. Table 2 presents the relative intracellular concen- 3.3. Decreased Extracellular Glutathione and Cysteine Redox trations of GSH, GSSG, the glutathione redox ratio, and Status in Autism. Table 3 presents the relative concentrations the percentage of oxidized glutathione equivalents in resting of GSH, GSSG, GSH/GSSG, % oxidized GSH, cysteine, (unstim-ulated) PBMCs and in isolated stimulated mon- cystine, and the cysteine/cystine redox ratio in the extracel- ocytes and CD4 T cells from children with autism and lular plasma compartment. Children with autism exhibited age-matched control children. The percent oxidized gluta- a significantly decreased extracellular concentration of GSH thione is expressed in absolute glutathione equivalents as (∼21%) and GSH/GSSG (∼54%) and increased concentra- 2GSSG/(GSH+2GSSG). Relative to controls, the intracellular tion of GSSG and the percent oxidized glutathione (52% concentration of GSSG and the percent oxidized glutathione and 82%, resp., P<0.001). Figures 1(a) and 1(b) com- were significantly increased (∼40%), and the GSH/GSSG pare GSH/GSSG and % oxidized glutathione equivalents, ratio decreased (∼21%) in PBMCs from children with respectively, in plasma, T cells, and monocytes from case and autism (P<0.001). After stimulation with LPS, mono- control children and graphically demonstrates the consistent cytes from children with autism also exhibited significantly decrease in both extracellular and intracellular glutathione decreased GSH/GSSG (∼31%, P = 0.003), increased GSSG redox status among the case children. concentration (∼32%, P = 0.01), and 40% higher percent The dynamic balance between the reduced and oxidized oxidized glutathione (P<0.001). In mitogen-stimulated forms of glutathione can also be expressed as the redox CD4 T cells from children with autism, the intracellular GSH potential or reducing power of the GSH/GSSG redox couple concentration was ∼33% lower, the GSH/GSSG was ∼40% (Eh) and can be calculated from the Nernst equation, Eh = lower (P<0.001), and the percent oxidized glutathione E0 + RT/nF ln[disulfide]/([thiol 1] ∗ [thiol 2]), where E0 was ∼55% higher than in stimulated CD4 T cells from is the standard potential for the glutathione redox couple control children (<0.001). As expected, activation with LPS (−264 mV), R is the gas constant (8.314 J/◦Kmol), T is the and PMA both resulted in decreased intracellular GSH absolute temperature of analytical measurement (25◦C = levels and GSH/GSSG in isolated monocytes and CD4 T 298◦K), n is 2 for the number of electrons transferred, cells compared to resting (unstimulated) PBMCs. Upon and F is Faraday’s constant (96,485 coulomb/mol) [48]. stimulation, there was a greater decrease in intracellular The calculated Eh value for the GSH pool in the children GSH and GSH/GSSG in both CD4 T cells and monocytes with autism is −116 mV, which is 12 mV more oxidized from children with autism compared to control children. than in the control children, with an Eh value of −128 mV Neither age nor gender was significantly correlated with any (Table 3). Autism Research and Treatment 5

Table 3: Extracellular (plasma) glutathione and cysteine redox status.

Case children Control children Corrected Z-test Metabolite Difference n Mean ± SD n Mean ± SD P value (95% CI) Plasma GSH (μM) 38 1.58 ± 0.23 41 1.99 ± 0.22 −0.41 (−0.50, −0.31) <0.001 GSSG (μM) 38 0.20 ± 0.06 41 0.13 ± 0.04 0.07 (0.05, 0.09) <0.001 GSH/GSSG 38 8.24 ± 2.20 41 17.14 ± 5.54 −8.73 (−10.52, −6.94) <0.001 Oxidized GSH (%) 38 0.20 ± 0.05 41 0.11 ± 0.03 0.09 (0.07, 0.10) <0.001 Eh for GSH −116 mV −128 mV Cysteine (μM) 41 21.7 ± 4.88 41 21.43 ± 4.08 0.13 (−1.88, 2.14) 0.90 Cystine (μM) 41 29.2 ± 10.64119.26 ± 4.8 9.73 (6.25, 13.2) <0.001 Cysteine/Cystine 41 0.79 ± 0.18 41 1.14 ± 0.18 −0.33 (−0.41, −0.26) <0.001 Eh for Cysteine −106 mV −111 mV 2 GSH: glutathione; GSSG: oxidized glutathione disulfide; Eh: steady-state redox potential; Eh for GSH: −264 mV +(30 mV) ∗ log([GSSG]/[GSH] ); Eh for cysteine: −250 mV + (30 mV) ∗ log([CySSCy]/[Cys]2).

30 0.3 ∗ 25 0.25 ∗ ∗ ∗ 20 0.2 ∗ 15 0.15 ∗

GSH/GSSG 10 0.1

5 Oxidized GSH (%) 0.05

0 0 Plasma Activated monocytes Activated T cells Plasma Activated monocytes Activated T cells

Control Control Case Case (a) (b)

Figure 1: Intracellular and extracellular glutathione redox imbalance in autism. (a) presents the GSH/GSSG in plasma, isolated activated monocytes, and CD4 T cells from case and control children; (b) presents the % oxidized glutathione equivalents. Both extracellular and intracellular glutathione redox status are consistently significantly decreased among the case children (∗P<0.01).

The concentration of cystine, the oxidized form of and unaffected control children (normalized to MFI of the cysteine, was significantly elevated (∼52%), while the cys- standard PBMC preparation). Gated lymphocytes from chil- teine/cystine redox ratio was significantly decreased (∼31%) dren with autism exhibited a significantly higher mean level in plasma from children with autism (P<0.001). The Eh of intracellular free radicals compared to lymphocytes from value for the cysteine pool can also be calculated from the control children (P<0.05). No differences in free radical Nernst equation (see above) where the E0 for cysteine is equal production were observed in gated monocytes from case and to −250 mV [30]. The calculated Eh value for the cysteine control children. Intracellular free radical production was pool in children with autism is −106 mV, or 5 mV more not correlated with age or gender in this cohort. oxidized than the control Eh value of −111 mV (Table 3). 4. Discussion 3.4. Elevated Free Radical Production in Autism. The level of intracellular free radicals was measured in available resting Oxidative stress is generally defined as an imbalance between PBMCs from children with autism (n = 15) and unaffected oxidant production and endogenous antioxidant defense control children (n = 16) using DCF, an ROS/RNS-sensitive mechanisms and can be clinically defined in humans by a fluorescent probe. Monocytes and lymphocytes were gated decrease in the redox status of GSH/GSSG and cysteine/cys- based on light scatter properties (size and density) and ana- tine thiol/disulfide redox couples [49].Therelativeequi- lyzed separately. Figure 2 presents the median fluorescence librium between reduced and oxidized sulfhydryl groups intensity (MFI) of lymphocytes from children with autism defines the ambient redox state. Low glutathione redox status 6 Autism Research and Treatment has been associated with the pathophysiology of several neu- 1.5 ∗ robehavioral disorders including schizophrenia [2, 50], bipo- lar disorder [3], alcoholism [51], HIV [52], and Alzheimer’s disease [53]. This is the first study to evaluate intracellular glutathione-mediated antioxidant/redox capacity in primary 1 cells from children with autism as well as the extracellular plasma cysteine/cystine redox status. Because these two redox systems are compartmentalized and independently regulat- ed, evaluation of both redox couples provides a complete 0.5 picture of the primary immune cell microenvironment in Intracellular free radicals children with autism. Supporting and extending our previ- (normalized to standard) ous findings of decreased plasma and lymphoblastoid cell GSH/GSSG, we now report that both primary immune cell 0 GSH/GSSG and plasma cysteine/cystine redox couples are Control Case similarly compromised resulting in a more oxidized immune Figure 2: Intracellular Free Radicals are Elevated in Lymphocytes cell microenvironment in children with autism compared to from Children with Autism. Intracellular free radicals were mea- control children. sured in freshly isolated PBMC from children with autism and Recent evidence supports the notion that subtle fluctu- unaffected control children using 1 uM DCF. Presented is median ations in ambient redox status may provide an important fluorescent intensity (MFI) of the gated lymphocyte population regulatory mechanism that can dynamically modulate im- from subject samples normalized to MFI of a standard PBMC mune cell function. Activation and proliferation of T cells preparation also treated with 1 uM DCF and analyzed with each require a reducing intracellular microenvironment, whereas subject sample. Lymphocytes from children with autism exhibited a significantly higher mean level of intracellular free radicals than a more oxidized environment promotes cell cycle arrest and controls (P = 0.04). Control median (95% CI) = 0.576 (0.551– blunted responsiveness to immune stimulation [54–57]. For 0.640); case median (95% CI) = 0.689 (0.561–1.086). example, a mechanism involving extracellular redox mod- ulation by regulatory T cells (Tregs) was recently elucidated by Yan et al. [35]. Tregs were shown to inhibit the release of cysteine into the immune synapse between dendritic cells to be associated with decreased proliferative response after and na¨ıve T cells, which effectively reduces GSH levels in T mitogen stimulation in children with autism [58]. cells by eliminating the rate-limiting amino acid for GSH The more oxidized GSH/GSSG redox status in plasma synthesis. A high ratio of reduced to oxidized glutathione is and primary immune cells in children with autism (Figure 1) required for cell cycle progression from G1 to S phase and may offer a mechanistic explanation for the abnormal adap- induction of the T-cell proliferative response [55]. Thus, the tive immune response previously reported in these children. more oxidized GSH/GSSG redox state of the intracellular When intracellular oxidative stress exceeds glutathione redox glutathione pool in PBMCs and in activated CD4 T cells capacity, cells export GSSG into the plasma as a mechanism observed in children with autism (Table 2) would suggest to restore internal redox homeostasis [49, 63]. The elevated a hyporesponsive phenotype that is less conducive to T-cell GSSG concentrations in PBMCs (Table 2) suggest that activation and proliferation. Consistent with this hypothesis, the GSSG export mechanism and intracellular antioxidant several recent studies have documented abnormalities in capacity were not sufficient to maintain intracellular redox the adaptive immune response in children with autism homeostasis and that redox imbalance was chronic in these [44, 58]. children. The association between a more oxidized immune A glutathione deficit in T cells has been shown to nega- cell microenvironment and an abnormal adaptive immune tively affect the adaptive immune response and T-cell pro- response warrants continued investigation especially in light liferation by reducing IL-2 receptor turnover and IL-2- of the potential reversibility of immune dysfunction with dependent DNA synthesis [59, 60]. In monocytes, an oxi- targeted treatment to restore redox homeostasis [15]. dized intracellular environment has been shown to alter the The calculated Eh values for the extracellular GSH and cytokine profile and skew the Th1 and Th2 balance [61, 62]. cysteine pools (Table 3) in our control population differ Studies in mice have demonstrated that the intracellular GSH somewhat from previously published values. In adults, the content of antigen presenting cells (APCs) reversibly alters plasma glutathione Eh is more reduced at around −137 mV, the Th1 and Th2 cytokine response pattern [61]. Specifically, and the plasma cysteine redox couple is more oxidized at a GSH deficit reduced Th1-associated IFN-γ production and −80 mV [30, 48]. These discrepancies may reflect method- exaggerated Th2-associated IL-4 production. Restoration of ological differences in sample preparation in that our electro- GSH restored the Th1 cytokine response and normalized chemical detection does not require derivatization for detec- the Th2 response. Consistent with these observations, two tion. It is also possible that children (age 3–10 years) may independent studies have reported that helper T-cell sub- have less reducing capacity than previously reported in adults populations in PBMCs from children with autism are shifted (age 25–35 years) [48]. Nonetheless, our calculated Eh values towards T helper 2 (Th2) dominance [41, 42]. Further, a are consistent with previous reports that plasma cysteine Eh decrease in T-cell IL-2 receptor expression has been reported (−111 mV) is more oxidized than that of GSH (−128 mv). Autism Research and Treatment 7

Mean intracellular free radical production was higher in cysteine/cystine-dependent signaling for mitochondrial ROS primary lymphocytes from children with autism relative to production is not yet clear; the authors provide evidence of lymphocytes from age-matched control children (Figure 2) a possible link to changes in the redox state of cytoskeletal and was driven by a subset of 5 (33.3%) children whose proteins that could be functionally linked to the mitochon- lymphocytes exhibited especially high levels of free radicals. drial membrane. Other studies have demonstrated that an Mitochondria are the primary producers and targets of oxidized plasma cysteine/cystine redox potential is associated intracellular free radicals, and mitochondrial dysfunction has with proinflammatory conditions [78, 79]andcanbemod- been postulated to be a contributing factor in the pathogen- ulated by diet [80, 81]. These observations support the pos- esis of autism and numerous other neurological disorders sibility that the oxidized plasma cysteine/cystine in children [64–67]. In a lymphoblastoid cell model, we previously dem- with autism may be functionally related to the increase in onstrated that the GSH/GSSG redox ratio in mitochondria lymphocyte free radical production observed and contribute was significantly lower in autism compared to control cells to immune cell abnormalities in these children. and was associated with a significantly lower mitochondrial In summary, we show for the first time that both the membrane potential after nitrosative stress [16]. It is well extracellular and intracellular immune cell compartments established that mitochondria are highly concentrated in are more oxidized in children with autism compared to age- presynaptic terminals and that loss of redox control can neg- matched unaffected control children. Randomized clinical atively affect the efficiency of neurotransmission and synap- trials will be needed to determine whether treatment to tic plasticity [68, 69]. Similarly, mitochondrial localization normalize plasma and intracellular redox status will improve and redox signaling at the immunological synapse between immune cell function and possibly the health and behavior lymphocytes and antigen presenting cells are required for in children with autism. immune activation, and excessive ROS can interrupt these signaling pathways [70–72]. A recent study of mitochondrial Conflict of Interests defects in lymphocytes from children with autism found decreased complex I activity and overreplication of and de- The authors declare no conflict of interests. letions in mitochondrial DNA compared to control lym- phocytes [73]. Based on this evidence, it is plausible to hy- Acknowledgments pothesize that mitochondria may be the source of the increased levels of lymphocyte free radicals observed in the The authors would like to express their gratitude to the subset of autistic children presented in Figure 2. Consistent families in Arkansas affected by autism whose participa- with this hypothesis, a recent meta-analysis estimated that tion made this study possible. They also acknowledge the mitochondria dysfunction may affect up to 30% of children invaluable help of the nurses and clinicians at the Dennis with autism [64]. Based on this evidence, further study of Developmental Center for referral and evaluation. This mitochondrial function and redox status in lymphocytes research was supported, in part, with funding from the from children with autism is warranted. National Institute of Child Health and Development (RO1 Relevant to our observations, two recent papers have HD051873; SJJ), the Department of Defense (AS073218P1; revealed that an oxidized extracellular cysteine/cystine redox SJJ), and by grants from the Arkansas Children’s Hospital status can initiate a redox signaling cascade that stimulates and Arkansas Biosciences Institute (SJJ). intracellular mitochondrial ROS production as a mechanism to initiate an inflammatory immune response [74, 75]. The signal transduction from the extracellular to intracellular References compartments occurs through oxidative modification of [1] CDC, “Prevalence of autism spectrum disorders—autism and redox-reactive cysteines on cell surface proteins. Exposed developmental disabilities monitoring network, United States, cysteine sulfhydryl groups on proteins can be reversibly oxi- 2006,” Morbidity and Mortality Weekly Report, vol. 58, no. 10, dized to sulfenic acid or disulfide bonds resulting in altered pp. 1–20, 2009. protein structure and function that initiate downstream [2] J. K. Yao, S. Leonard, and R. Reddy, “Altered glutathione redox signaling cascades [33, 76, 77]. In an elegant series redox state in schizophrenia,” Disease Markers,vol.22,no.1-2, of experiments, Imhoff and Hansen demonstrated that mi- pp. 83–93, 2006. tochondrial ROS production was significantly increased in [3]A.C.Andreazza,M.Kauer-Sant’Anna,B.N.Freyetal., “Oxidative stress markers in bipolar disorder: a meta-analysis,” cells incubated under extracellular oxidized cysteine/cystine ff redox conditions [74]. The stimulated intracellular ROS Journal of A ective Disorders, vol. 111, no. 2-3, pp. 135–144, 2008. production resulted in the expression of Nrf-2, the transcrip- [4] W. R. Markesbery, “Oxidative stress hypothesis in Alzheimer’s tion factor responsible for initiation of the inflammatory disease,” Free Radical Biology and Medicine, vol. 23, no. 1, response. Treatment to block the availability of cell surface pp. 134–147, 1997. cysteine thiol groups abrogated mitochondrial ROS produc- [5] G. A. Mostafa, E. S. El-Hadidi, D. H. Hewedi, and M. M. tion and Nrf-2 expression. Go et al. confirmed and extended Abdou, “Oxidative stress in Egyptian children with autism: these observations by demonstrating that treatment to main- relation to autoimmunity,” Journal of Neuroimmunology, tain mitochondrial redox status abrogated ROS production vol. 219, no. 1-2, pp. 114–118, 2010. in the presence of oxidized extracellular cysteine/cystine [6] N. A. Meguid, A. A. Dardir, E. R. Abdel-Raouf, and A. Hashish, [75]. Although the precise mechanism for the oxidative “Evaluation of oxidative stress in autism: defective antioxidant 8 Autism Research and Treatment

enzymes and increased lipid peroxidation,” Biological Trace [22] Y. W. Kwon, H. Masutani, H. Nakamura, Y. Ishii, and J. Yodoi, Element Research, vol. 143, no. 1, pp. 58–65, 2011. “Redox regulation of cell growth and cell death,” Biological [7] A. Chauhan, V. Chauhan, W. T. Brown, and I. Cohen, Chemistry, vol. 384, no. 7, pp. 991–996, 2003. “Oxidative stress in autism: increased lipid peroxidation and [23] F. Q. Schafer and G. R. Buettner, “Redox environment of the reduced serum levels of ceruloplasmin and transferrin—the cell as viewed through the redox state of the glutathione disul- antioxidant proteins,” Life Sciences, vol. 75, no. 21, pp. 2539– fide/glutathione couple,” Free Radical Biology and Medicine, 2549, 2004. vol. 30, no. 11, pp. 1191–1212, 2001. [8]S.S.Zoroglu,F.Armutcu,S.Ozenetal.,“Increasedoxidative [24] H. M. Lander,ˇ A. J. Mllbank, J. M. Tauras et al., “Redox regula- stress and altered activities of erythrocyte free radical scaveng- tion of cell signalling,” Nature, vol. 381, no. 6581, pp. 380–381, ing enzymes in autism,” European Archives of Psychiatry and 1996. Clinical Neuroscience, vol. 254, no. 3, pp. 143–147, 2004. [25] P. Ghezzi, “Regulation of protein function by glutathionyla- [9] S. So¨gˇut,S.S.Zoro¨ glu,ˇ H. Ozyurt¨ et al., “Changes in nitric tion,” Free Radical Research, vol. 39, no. 6, pp. 573–580, 2005. oxide levels and antioxidant enzyme activities may have a role [26] D. A. Dickinson and H. J. Forman, “Glutathione in defense in the pathophysiological mechanisms involved in autism,” and signaling: lessons from a small thiol,” Annals of the New Clinica Chimica Acta, vol. 331, no. 1-2, pp. 111–117, 2003. York Academy of Sciences, vol. 973, pp. 488–504, 2002. [10] T. L. Sweeten, D. J. Posey, S. Shankar, and C. J. McDougle, [27] M. Noble, J. Smith, J. Power, and M. Mayer-Proschel,¨ “Redox “High nitric oxide production in autistic disorder: a possible state as a central modulator of precursor cell function,” Annals role for interferon-γ,” Biological Psychiatry,vol.55,no.4, of the New York Academy of Sciences, vol. 991, pp. 251–271, pp. 434–437, 2004. 2003. [11] M. A. Junaid, D. Kowal, M. Barua, P. S. Pullarkat, S. S. Brooks, [28] D. P.Jones, “Extracellular redox state: refining the definition of and R. K. Pullarkat, “Proteomic studies identified a single oxidative stress in aging,” Rejuvenation Research, vol. 9, no. 2, nucleotide polymorphism in glyoxalase I as autism suscepti- pp. 169–181, 2006. bility factor,” American Journal of Medical Genetics, vol. 131, [29] M. Asensi, J. Sastre, F. V. Pallardo et al., “Ratio of reduced to no. 1, pp. 11–17, 2004. oxidized glutathione as indicator of oxidative stress status and [12] M. Boso, E. Emanuele, P. Minoretti et al., “Alterations of DNA damage,” Methods in Enzymology, vol. 299, pp. 267–276, circulating endogenous secretory RAGE and S100A9 levels 1999. indicating dysfunction of the AGE-RAGE axis in autism,” Neu- [30] S. S. Iyer, C. J. Accardi, T. R. Ziegler et al., “Cysteine redox roscience Letters, vol. 410, no. 3, pp. 169–173, 2006. potential determines pro-inflammatory IL-1β levels,” PLoS [13] S. J. James, P.Cutler, S. Melnyk et al., “Metabolic biomarkers of One, vol. 4, no. 3, Article ID e5017, 2009. increased oxidative stress and impaired methylation capacity [31] D. Barford, “The role of cysteine residues as redox-sensitive in children with autism,” American Journal of Clinical Nutri- regulatory switches,” Current Opinion in Structural Biology, tion, vol. 80, no. 6, pp. 1611–1617, 2004. vol. 14, no. 6, pp. 679–686, 2004. [14] S. J. James, S. Melnyk, S. Jernigan et al., “Metabolic endophe- [32] D. P. Jones, Y. M. Go, C. L. Anderson, T. R. Ziegler, J. M. notype and related genotypes are associated with oxidative Kinkade Jr., and W. G. Kirlin, “Cysteine/cystine couple is a stress in children with autism,” American Journal of Medical newly recognized node in the circuitry for biologic redox sig- Genetics, Part B, vol. 141, no. 8, pp. 947–956, 2006. naling and control,” FASEB Journal, vol. 18, no. 11, pp. 1246– [15] S. J. James, S. Melnyk, G. Fuchs et al., “Efficacy of methylcobal- 1248, 2004. amin and folinic acid treatment on glutathione redox status in [33] D. Spadaro, B. W. Yun, S. H. Spoel, C. Chu, Y. Q. Wang, and children with autism,” American Journal of Clinical Nutrition, G. J. Loake, “The redox switch: dynamic regulation of protein vol. 89, no. 1, pp. 425–430, 2009. function by cysteine modifications,” Physiologia Plantarum, [16] S. J. James, S. Rose, S. Melnyk et al., “Cellular and mito- vol. 138, no. 4, pp. 360–371, 2010. chondrial glutathione redox imbalance in lymphoblastoid cells [34] J. Den Hertog, A. Groen, and T. van der Wijk, “Redox regu- derived from children with autism,” FASEB Journal, vol. 23, lation of protein-tyrosine phosphatases,” Archives of Biochem- no. 8, pp. 2374–2383, 2009. istry and Biophysics, vol. 434, no. 1, pp. 11–15, 2005. [17]K.Bowers,Q.Li,J.Bressler,D.Avramopoulos,C.Newschaffer, [35] Z. Yan, S. K. Garg, J. Kipnis, and R. Banerjee, “Extracellular and M. D. Fallin, “Glutathione pathway gene variation and redox modulation by regulatory T cells,” Nature Chemical risk of autism spectrum disorders,” Journal of Neurodevelop- Biology, vol. 5, no. 10, pp. 721–723, 2009. mental Disorders, vol. 3, no. 2, pp. 132–143, 2011. [36]C.E.Cooper,R.P.Patel,P.S.Brookes,andV.M.Darley- [18] S. Biswas, A. S. Chida, and I. Rahman, “Redox modifications Usmar, “Nanotransducers in cellular redox signaling: mod- of protein-thiols: emerging roles in cell signaling,” Biochemical ification of thiols by reactive oxygen and nitrogen species,” Pharmacology, vol. 71, no. 5, pp. 551–564, 2006. Trends in Biochemical Sciences, vol. 27, no. 10, pp. 489–492, [19] G. Filomeni, G. Rotilio, and M. R. Ciriolo, “Cell signalling 2002. and the glutathione redox system,” Biochemical Pharmacology, [37] R. P. Warren, L. J. Yonk, R. A. Burger et al., “Deficiency of vol. 64, no. 5-6, pp. 1057–1064, 2002. suppressor-inducer (CD4+CD45RA+) T cells in autism,” Im- [20] M. Fratelli, L. O. Goodwin, U. A. Ørom et al., “Gene expres- munological Investigations, vol. 19, no. 3, pp. 245–251, 1990. sion profiling reveals a signaling role of glutathione in redox [38] L. J. Yonk, R. P. Warren, R. A. Burger et al., “CD4+ helper T regulation,” Proceedings of the National Academy of Sciences of cell depression in autism,” Immunology Letters, vol. 25, no. 4, the United States of America, vol. 102, no. 39, pp. 13998–14003, pp. 341–346, 1990. 2005. [39] D. R. Denney, B. W. Frei, and G. R. Gaffney, “Lymphocyte [21] A. Pastore, G. Federici, E. Bertini, and F. Piemonte, “Analysis of subsets and interleukin-2 receptors in autistic children,” glutathione: implication in redox and detoxification,” Clinica Journal of Autism and Developmental Disorders,vol.26,no.1, Chimica Acta, vol. 333, no. 1-2, pp. 19–39, 2003. pp. 87–97, 1996. Autism Research and Treatment 9

[40] T. L. Sweeten, D. J. Posey, and C. J. McDougle, “High blood [56] M. Klemke, G. H. Wabnitz, F. Funke, B. Funk, H. Kirchgessner, monocyte counts and neopterin levels in children with autistic and Y. Samstag, “Oxidation of cofilin mediates T cell hypore- disorder,” American Journal of Psychiatry, vol. 160, no. 9, sponsiveness under oxidative stress conditions,” Immunity, pp. 1691–1693, 2003. vol. 29, no. 3, pp. 404–413, 2008. [41] S. Gupta, S. Aggarwal, B. Rashanravan, and T. Lee, “Th1- [57] T. Ando, K. Mimura, C. C. Johansson et al., “Transduction and Th2-like cytokines in CD4+ and CD8+ Tcellsinautism,” with the antioxidant enzyme catalase protects human T cells Journal of Neuroimmunology, vol. 85, no. 1, pp. 106–109, 1998. against oxidative stress,” Journal of Immunology, vol. 181, [42]C.A.Molloy,A.L.Morrow,J.Meinzen-Derretal.,“Elevated no. 12, pp. 8382–8390, 2008. cytokine levels in children with autism spectrum disorder,” [58]P.Ashwood,P.Krakowiak,I.Hertz-Picciotto,R.Hansen,I. Journal of Neuroimmunology, vol. 172, no. 1-2, pp. 198–205, N. Pessah, and J. Van de Water, “Altered T cell responses in 2006. children with autism,” Brain, Behavior, and Immunity, vol. 25, [43] P. Ashwood and A. J. Wakefield, “Immune activation of pe- no. 5, pp. 840–849, 2011. ripheral blood and mucosal CD3+ lymphocyte cytokine pro- [59] C. M. Liang, N. Lee, D. Cattell, and S. M. Liang, “Glutathione files in children with autism and gastrointestinal symptoms,” regulates interleukin-2 activity on cytotoxic T-cells,” Journal of Journal of Neuroimmunology, vol. 173, no. 1-2, pp. 126–134, Biological Chemistry, vol. 264, no. 23, pp. 13519–13523, 1989. 2006. [60] H. Gmunder, S. Roth, H. P. Eck, H. Gallas, S. Mihm, and [44] H. Jyonouchi, S. Sun, and H. Le, “Proinflammatory and W. Droge, “Interleukin-2 mRNA expression, lymphokine pro- regulatory cytokine production associated with innate and duction and DNA synthesis in glutathione-depleted T cells,” adaptive immune responses in children with autism spectrum Cellular Immunology, vol. 130, no. 2, pp. 520–528, 1990. disorders and developmental regression,” Journal of Neuroim- [61]J.D.Peterson,L.A.Herzenberg,K.Vasquez,andC. munology, vol. 120, no. 1-2, pp. 170–179, 2001. Waltenbaugh, “Glutathione levels in antigen-presenting cells [45] P. Ashwood, A. Enstrom, P. Krakowiak et al., “Decreased modulate Th1 versus Th2 response patterns,” Proceedings of transforming growth factor beta1 in autism: a potential link the National Academy of Sciences of the United States of Ameri- between immune dysregulation and impairment in clinical ca, vol. 95, no. 6, pp. 3071–3076, 1998. behavioral outcomes,” Journal of Neuroimmunology, vol. 204, [62] Y. Murata, T. Shimamura, and J. Hamuro, “The polarization of no. 1-2, pp. 149–153, 2008. Th1/Th2 balance is dependent on the intracellular thiol redox [46] K. Okada, K. Hashimoto, Y. Iwata et al., “Decreased serum status of macrophages due to the distinctive cytokine produc- levels of transforming growth factor-β1 in patients with tion,” International Immunology, vol. 14, no. 2, pp. 201–212, autism,” Progress in Neuro-Psychopharmacology and Biological 2002. Psychiatry, vol. 31, no. 1, pp. 187–190, 2007. [63] L. Eklow,H.Thor,andS.Orrenius,“Formationande¨ fflux of [47] S. W. Looney and P. W. Jones, “A method for comparing glutathione disulfide studied in isolated rat hepatocytes,” FEBS two normal means using combined samples of correlated and Letters, vol. 127, no. 1, pp. 125–128, 1981. uncorrelated data,” Statistics in Medicine,vol.22,no.9, [64] D. A. Rossignol and R. E. Frye, “Mitochondrial dysfunction pp. 1601–1610, 2003. in autism spectrum disorders: a systematic review and meta- [48] D. P. Jones, J. L. Carlson, V. C. Mody, J. Cai, M. J. Lynn, and analysis,” Molecular Psychiatry. In press. P. Sternberg, “Redox state of glutathione in human plasma,” [65] A. H. Schapira, J. M. Cooper, D. Dexter, P. Jenner, J. B. Clark, Free Radical Biology and Medicine, vol. 28, no. 4, pp. 625–635, and C. D. Marsden, “Mitochondrial complex I deficiency in 2000. Parkinson’s disease,” The Lancet, vol. 1, no. 8649, p. 1269, 1989. [49] Y. M. Go and D. P. Jones, “Redox compartmentalization in [66] A. Maruszak and C. Zekanowski, “Mitochondrial dysfunction eukaryotic cells,” Biochimica et Biophysica Acta, vol. 1780, and Alzheimer’s disease,” Progress in NeuroPsychopharmacolo- no. 11, pp. 1273–1290, 2008. gy and Biological Psychiatry, vol. 35, no. 2, pp. 320–330, 2011. [50] R. Gysin, R. Kraftsik, J. Sandell et al., “Impaired glutathione [67] H. B. Clay, S. Sillivan, and C. Konradi, “Mitochondrial dys- synthesis in schizophrenia: convergent genetic and functional function and pathology in bipolar disorder and schizophre- evidence,” Proceedings of the National Academy of Sciences of nia,” International Journal of Developmental Neuroscience, the United States of America, vol. 104, no. 42, pp. 16621–16626, vol. 29, no. 3, pp. 311–324, 2011. 2007. [68] D. J. Keating, “Mitochondrial dysfunction, oxidative stress, [51] M. Y. Yeh, E. L. Burnham, M. Moss, and L. A. Brown, “Chronic regulation of exocytosis and their relevance to neurodegen- alcoholism alters systemic and pulmonary glutathione redox erative diseases,” Journal of Neurochemistry, vol. 104, no. 2, status,” American Journal of Respiratory and Critical Care Med- pp. 298–305, 2008. icine, vol. 176, no. 3, pp. 270–276, 2007. [69] Z. Li, K. I. Okamoto, Y. Hayashi, and M. Sheng, “The [52] H. Nakamura, H. Masutani, and J. Yodoi, “Redox imbalance importance of dendritic mitochondria in the morphogenesis and its control in HIV infection,” Antioxidants and Redox and plasticity of spines and synapses,” Cell, vol. 119, no. 6, Signaling, vol. 4, no. 3, pp. 455–464, 2002. pp. 873–887, 2004. [53] C. Cecchi, S. Latorraca, S. Sorbi et al., “Gluthatione level is [70] A. Quintana, C. Schwindling, A. S. Wenning et al., “T cell ac- altered in lymphoblasts from patients with familial Alzheim- tivation requires mitochondrial translocation to the immuno- er’s disease,” Neuroscience Letters, vol. 275, no. 2, pp. 152–154, logical synapse,” Proceedings of the National Academy of Scien- 1999. ces of the United States of America, vol. 104, no. 36, pp. 14418– [54] A. Larbi, J. Kempf, and G. Pawelec, “Oxidative stress modula- 14423, 2007. tion and T cell activation,” Experimental Gerontology, vol. 42, [71] G. Pani, R. Colavitti, S. Borrello, and T. Galeotti, “Redox reg- no. 9, pp. 852–858, 2007. ulation of lymphocyte signaling,” IUBMB Life,vol.49,no.5, [55]J.P.MessinaandD.A.Lawrence,“Cellcycleprogressionof pp. 381–389, 2000. glutathione-depleted human peripheral blood mononuclear [72] T. M. Buttke and P. A. Sandstrom, “Redox regulation of pro- cells is inhibited at S phase,” Journal of Immunology, vol. 143, grammed cell death in lymphocytes,” Free Radical Research, no. 6, pp. 1974–1981, 1989. vol. 22, no. 5, pp. 389–397, 1995. 10 Autism Research and Treatment

[73] C. Giulivi, Y. F. Zhang, A. Omanska-Klusek et al., “Mitochon- drial dysfunction in autism,” Journal of the American Medical Association, vol. 304, no. 21, pp. 2389–2396, 2010. [74] B. R. Imhoff and J. M. Hansen, “Extracellular redox status regulates Nrf2 activation through mitochondrial reactive oxy- gen species,” Biochemical Journal, vol. 424, no. 3, pp. 491–500, 2009. [75] Y. M. Go, H. Park, M. Koval et al., “A key role for mitochondria in endothelial signaling by plasma cysteine/cystine redox potential,” Free Radical Biology and Medicine, vol. 48, no. 2, pp. 275–283, 2010. [76] L. I. Leichert and U. Jakob, “Protein thiol modifications visualized in vivo,” PLoS Biology, vol. 2, no. 11, Article ID e333, 2004. [77] P. Ghezzi, “Oxidoreduction of protein thiols in redox regula- tion,” Biochemical Society Transactions, vol. 33, no. 6, pp. 1378– 1381, 2005. [78]S.S.Iyer,D.P.Jones,K.L.Brigham,andM.Rojas,“Oxidation of plasma cysteine/cystine redox state in endotoxin-induced lung injury,” American Journal of Respiratory Cell and Molecu- lar Biology, vol. 40, no. 1, pp. 90–98, 2009. [79] S. K. Garg, V. Vitvitsky, R. Albin, and R. Banerjee, “Astrocytic redox remodeling by amyloid beta peptide,” Antioxidants and Redox Signaling, vol. 14, no. 12, pp. 2385–2397, 2011. [80] S. E. Moriarty-Craige, J. Adkison, M. Lynn et al., “Antioxidant supplements prevent oxidation of cysteine/cystine redox in patients with age-related macular degeneration,” American Journal of Ophthalmology, vol. 140, no. 6, pp. 1020–1026, 2005. [81] D. P. Jones, Y. Park, N. Gletsu-Miller et al., “Dietary sulfur amino acid effects on fasting plasma cysteine/cystine redox potential in humans,” Nutrition, vol. 27, no. 2, pp. 199–205, 2011. Hindawi Publishing Corporation Autism Research and Treatment Volume 2012, Article ID 868576, 6 pages doi:10.1155/2012/868576

Clinical Study High Complement Factor I Activity in the Plasma of Children with Autism Spectrum Disorders

Naghi Momeni,1 Lars Brudin,2 Fatemeh Behnia,3 Berit Nordstrom,¨ 4 Ali Yosefi-Oudarji,5 Bengt Sivberg,4 Mohammad T. Joghataei,5 and Bengt L. Persson1

1 School of Natural Sciences, Linnaeus University, 39182 Kalmar, Sweden 2 Department of Clinical Physiology, Kalmar County Hospital, 39185 Kalmar, Sweden 3 Department of Occupational Therapy, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran 4 Department of Health Sciences, Autism Research, Faculty of Medicine, Lund University, Box 157, 22100 Lund, Sweden 5 Cellular and Molecular Research Centre, Tehran University of Medical Sciences (TUMS), Tehran, Iran

Correspondence should be addressed to Bengt Sivberg, [email protected]

Received 17 June 2011; Revised 22 August 2011; Accepted 22 August 2011

Academic Editor: Judy Van de Water

Copyright © 2012 Naghi Momeni et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Autism spectrum disorders (ASDs) are neurodevelopmental and behavioural syndromes affecting social orientation, behaviour, and communication that can be classified as developmental disorders. ASD is also associated with immune system abnormality. Im- mune system abnormalities may be caused partly by complement system factor I deficiency. Complement factor I is a serine pro- tease present in human plasma that is involved in the degradation of complement protein C3b, which is a major opsonin of the complement system. Deficiency in factor I activity is associated with an increased incidence of infections in humans. In this paper, we show that the mean level of factor I activity in the ASD group is significantly higher than in the control group of typically de- veloped and healthy children, suggesting that high activity of complement factor I might have an impact on the development of ASD.

1. Introduction The complement system comprises a group of proteins which, when activated, provide one of the first lines of Autism spectrum disorders (ASDs) are characterized by im- defence by promoting lysis and the removal of invading pairments in social interaction, communication, and repeti- microbes. Activation of the complement system in response tive or restricted patterns of interests, or behaviours, and are to an infection or foreign antigen is achieved via three com- classified as developmental disorders in DSM-IV [1]. ASD is plement pathways, the classical pathway, which is activated about 4-5 times more prevalent in boys than in girls. The by antigen-antibody complexes, the lectin pathway, which is ratio is estimated to range from 5.5 : 1.4 to 16.8 : 4.0 [2]. activated by the interaction of microbial carbohydrates with Recent research clearly indicates that the underlying causes mannose-binding proteins in the plasma and tissue fluids, of autism are neurobiological disorders and combinations of ff and the alternative complement pathway, which is activated di erent factors, such as environmental and genetic factors, by C3b binding to microbial surfaces and to antibody mole- and abnormality in the communication between neurons, cules. All of the three pathways converge with the activation probably associated with an abnormal set of neuropeptides of the central C3 component. This leads to a final common in the brain [3–9]. The symptoms of ASD have been linked with elevated pathway with assembly of the C5–C9 components to form plasma levels of serotonin [10, 11] and opioid [12], abnormal a cell surface transmembrane pore (membrane attack com- levels of melatonin [13], altered levels of activity of the serine plex) [22, 23]. It has been shown by comparison with healthy protease prolyl endopeptidase [14], and infectious and im- control children that several differentially expressed proteins munological factors such as abnormalities of T cells, B cells, are related to the complement system in children with ASD natural killer (NK) cells, and of the complement system [15– [22]. The alternative pathway consists of six proteins: C3, fac- 21]. tor B, factor D, factor H, factor I, and properdin. The plasma 2 Autism Research and Treatment glycoprotein factor I (C3b/C4b inactivator) is a serine pro- of Pediatrics in its Embargo from 2007 [35]. The control tease that acts as a regulator of the complement C3 cascade. group consisted of typically developed and healthy children Factor I has a molecular weight of about 88 kDa, consists of showing no signs of neurodevelopmental disorders who were two disulfide-linked polypeptide chains (50 kDa and 38 kDa, recruited from the same area as the children with ASD. resp.), and is synthesized as a single-chain precursor in the Children who had any kind of infection/infectious disease liver [24, 25]. Factor I cleaves C3b and C4b in a reaction, within two weeks prior to the time of examination were wherefIisdependentonvariouscofactors,suchasfactor excluded from this study. H, C4b-binding protein CR1 and membrane cofactor protein The study was approved (MT/1247) by the ethics com- (MCP) [26]. Factor I-mediated cleavage of the α chain of C3b mittee of the Iran University of Medical Sciences, Tehran. liberates 3 fragments with molecular weights of 68 kDa, 43 kDa, and 2 kDa. Degradation of C3b by fI abrogates the 2.2. Procedure action of this protein in the C3 pathway [27]. Complement C3b is the major opsonin of the complement system which 2.2.1. Blood Sample Collection. Blood samples were collected facilitates the phagocytosis process by coating antigens (each by a paediatric nurse, and those from the children diagnosed of the phagocytes expresses a complement receptor such as with autism were collected under the supervision of a child CR1, CR3, or CR4 that binds C3b, C4b, or C3bi) [28, 29]. psychiatrist with special training in the field of childhood Factor I deficiency can be conferred by a C3 deficiency, since psychosis. Venous blood was collected into 3 mL EDTA this also increases susceptibility to pyogenic infections by tubes (Vacutainer System; Becton-Dickinson Inc., Plymouth, UK), and plasma was separated immediately thereafter by Neisseria meningitides, Haemophilus influenza, and Strep- ◦ tococcus pneumonia and increases the incidence of immune centrifugation at 1,300 g for 10 min at 4 C. Thereafter, an μ complex diseases due to impaired complement-mediated inhibitors cocktail (30 L per 1 mL plasma) was added to the function [30]. Immune system abnormalities have been asso- resultant plasma sample (cocktail inhibitor solution: 2.0 M μ ciated with autism [15–20], and it has been suggested that Tris, 0.9 M Na-EDTA, 0.2 M Benzamidine, 92 M E-64, and 48 μM Pepstatin; Sigma, St. Louis, Mich, USA). The plasma children with ASD might have an increased incidence of bac- ◦ terial inflammation [31]. Immunological aspects of the early was stored at −80 C. onset of autism have recently highlighted the fact that im- mune dysfunction may occur in some children with autism 2.2.2. Assay. Methods based on the hydrolysis of fluorogenic [31, 32]. substrates have previously been described by Tsiftoglou and Having previously discovered altered levels of the serine Sim [36] and Gupta et al. [15]. The following assay procedure was found to be optimal for assaying fI activity in the protease prolyl endopeptidase in children with ASD [14], the μ μ ff aim of this study was to investigate if an association exists plasma. 20 L of plasma was incubated with 80 Lofbu er (100 mM phosphate buffer, pH 7.5, containing 1 mM EDTA, between serine protease fI deficiency and the development of ◦ ASD. 1 mM DTT and 1 mM sodium azide) for 10 min at 37 Cto reach thermal equilibrium. 100 μL of substrate solution (200 μM Boc-Asp(OBz)-Pro-Arg-7-amino-4-methylcouma- rin; Bachem, Bubendorf, Switzerland) in 25 mM phosphate 2. Materials and Methods buffer, pH 7.4, was then added, and the mixture was incu- bated at 37◦C for 60 min (see Figure 1). The reaction was 2.1. Participants. Thirty children with ASD and thirty typical inhibited by the addition of 1 mL of 1.5 M acetic acid, and the control children participated in this study. The ASD group release of 7-amino-4-methylcoumarin was measured by comprised 23 boys and 7 girls with a mean age of 4.5 years spectrofluorometer (Hitachi-f 2000; λ : 360 nm; λ : (age range 3–9 years). The control group comprised 13 boys ex em 440 nm; slit width: 2.5). All measurements were carried out and 17 girls, mean age 6.0 years (age range 3–12 years), randomized and in duplicate. Background fluorescence in (Table 1). the assay was monitored by the use of plasma in the absence Children in the ASD group were recruited from the Au- of substrate and was subtracted from values obtained in the tism Rehabilitation Centre at the University of Social Welfare presence of substrate. and Rehabilitation Sciences in Tehran, Iran. After having obtained informed consent from the parents, blood samples 2.3. Data Analysis and Statistics. Plasma fI activity was log- were collected. All children with ASD were examined by cli- normally distributed, and logarithmic values were, therefore, nical specialists on autism. A child psychiatrist and a child used when analysing differences between the ASD group and neurologist or child psychologist examined all of the chil- the control group. To adjust for age (dichotomized using the dren. All consultants agreed on the diagnosis of autism ac- median value, 5 years) and gender, factorial ANOVA was cording to the DSM-IV criteria [1]. However, diagnostic pro- used. A P value < 0.05 was considered statistically significant.  cedures applied in Europe and in the US/Canada using the Statistica 8.0 (StatSoft c , Tulsa, Okla, USA) was used. Intra- autism diagnostic observation schedule (ADOS) [33] and the and interassay variability of the plasma fI activity was expres- Autism Diagnostic Interview—Revised [34]werenotusedin sed as the standard error of a single determination (Smethod), the diagnostic process applied in Iran. This shortcoming was using the formula first proposed by Dahlberg [37]    compensated for by the extensive clinical experience by the √ d 2 child neurologist/child psychiatrist who was familiar with the S = i method n ,(1) core behaviours in autism stated by the American Academy (2 ) Autism Research and Treatment 3

Table 1: Age/y, gender, and medication of the participants.

Parameter ASD (n = 30) Controls (n = 30) P value∗ Mean (SD) 4.8 (1.7) 6.1 (2.3) Median (range) 4.5 (3–9) 6 (3–12) 0.033 Age ≤5 years 21 14 >5 years 9 16 0.115 Males 23 13 Gender Females 7 17 0.017 No specific medication 8 30 — Medication Risperdal alone or in combination 18 0 — Ritalin alone or in combination 4 0 — ∗ Difference between ASD and controls. Mann-Whitney U-test for age and Fisher’s exact test for age category and gender.

Factor I activity and substrate stability medication with Risperdal to reduce hyperactivity and vio- 3000 lent behavior, and a few were under medication with Ritalin to improve attention (Table 1). It would have been ethically questionable to discontinue medication with the purpose 2500 of controlling the experimental design. We have correlated the data shown in Figure 2(a) with the type of medication 2000 the children in the ASD group were receiving. Although we did not see any clear correlation between medication and distribution for the scatter plot data, it cannot be excluded 1500 that some differences in the pattern may be influenced by medication, as has been previously discussed [22]. The values were statistically significantly higher in the 1000 children with ASD, and there was a weak association with Fluorescence intensity unit gender. No statistically significant differences were found, however, between the age groups. 500 The methodological intra-assay error was small, 0.5%. The interassay methodological error was 13%. We found a significantly higher complement factor I enzyme activity 0 123in children with ASD compared to the control group of Time (hours) around the same age. This is, as far as we know, the first report regarding dysfunction of fI activity in children with Figure 1: Fluorescence intensity of release of 7-amino-4-methyl ASD. Although not statistically significant, males tended to coumarin as a function of plasma incubation period (mean ± SD; exhibit higher fI activity than females, and the difference 1h(974 ± 44.2), 2h (1995 ± 45.2) and 3 h (2374 ± 1.2)). between the control group and the ASD group was more convincing amongst the younger children, as shown in Figure 2. Due to fI’s role as a regulating factor in the comple– ment system pathway, an fI abnormality could play a role where di is the difference between the i : th paired measure- in the onset of ASD. A defect in this pathway makes the- ment and n is the number of differences. The Smethod was ex- individual more vulnerable to various inflammations. Some pressed as the coefficient of variation (%). reports [21, 24–26] provide increasing evidence of a con- nection between immunological abnormality and human disease. ASD numbers among the types of disorders that 3. Results are associated with immune system abnormalities [28, 38]. Our results are consistent with a recent proteomic study of There was significantly higher activity of plasma fI in the serum from ASD children, where a significant differential children with ASD (geometric mean (95% confidence limit): expression was shown for proteins related to the complement 523 (154–1776) when compared with the control group: 361 system [22]. (135–967; ANOVA P = 0.015, adjusted for age and gender; Figure 2). No statistically significant interactions were found with 4. Discussion regard to gender and age, and no significant associations were found between fI activity and age or gender (ANOVA; P = The aetiology of ASD is still largely unknown despite the fact 0.25 for gender and 0.42 for the two age groups, Figure 2). In that many factors such as genetic, environmental, immuno- the ASD group, some children with severe autism were under logical, and neurological aspects are thought to influence 4 Autism Research and Treatment

3000

Age ≤ 5 years Age > 5years 7 2000

6.6 1000 Males

Fluorescence intensity Males 6.2

0 Females Control ASD 5.8

Participants log (fluorescence intensity) Females

Non medicated Combination (Risperdal and Ritalin) 5.4 Risperdal Control ASD Control ASD Others Ritalin Participants Participants (a) (b)

Figure 2: (a) Complement factor I activity in EDTA plasma from children with ASD (n = 30) and healthy control children (n = 30). A scatter plot of factor I activity for each individual is shown. Samples from ASD children who were not under medication at the time of the investigation, those under medication with Risperdal, Ritalin, a combination thereof, or other medications such as antipsychotics (thiorida- zine) or anticonvulsants (fenobarbital and sodium valproate) are shown. (b) Mean values and standard errors of the complement factor I activity in EDTA plasma are shown for the different age groups and genders for both children with ASD and the healthy control group. In the ASD group; age ≤ 5 years: males (n = 17), females (n = 4) and age > 5 years: males (n = 6), females (n = 3). In the healthy control group; age ≤ 5 years: males (n = 4), females (n = 10) and age > 5 years: males (n = 9), females (n = 7). the development of ASD [39]. A dysregulated immune res- a functioning C3b protein. C3b is degraded by fI, and abnor- ponse has been reported among children with ASD. Pro- mal fI activity might cause an abnormal and uncontrolled tein products of immune activation, such as cytokines, can degradation of C3b protein, resulting in the loss of the be linked to core features of ASD, such as difficulties to reg- phagocytosis function for this particular protein (a function ulate affect, sleep, nutritional uptake, and can also affect which partly protects the body from invasion of foreign behaviour and social communication [39]. This study high- organisms). Altered levels of other serine protease activities, lights an elevated level of factor I that may contribute to a such as that of proline endopeptidase (PEP), have also been dysregulation of the immune response associated with the found in a group of children with ASD when compared to a complement system. Recent research proposes an extensive control group [14]. The results of the present study, together communication between the immune and nervous systems, with our previous findings on altered levels of PEP activity, affecting both the development of the central nervous system may indicate a connection between the onset of ASD and [32] in the promotion of health as well as disease. Early serine protease dysfunction. The higher plasma fI activity damage during critical periods in neurodevelopment of the observed in the male group as compared to that of the foetus has in a mouse model study been shown to affect female group (Figure 2) is paralleled by a higher occurrence cognitive development. It is reasonable to suggest that of ASD in male children. Also, the higher plasma fI activity in complement factor I might contribute to ASD, while changes children younger than six years of age may indicate that the in the complement system may predispose the mother or inflammatory process is more active in younger ages or that foetus to infections during development, possibly causing we may be dealing with two subgroups of children with ASD resultant abnormalities in brain development [22]. Also, the with different onsets of the disease. frequency of autoimmune diseases has been reported to be significantly higher in families with a child with ASD [40] 5. Conclusions than in families with children with typical development. The pathophysiology of ASD is poorly understood. Chil- The preliminary findings of this study together with our dren with ASD are prone to recurrent viral and bacterial in- previous report [14] suggest that there may be an associa- flammations. There are also some reports of immune sys- tion between abnormal serine protease activity and the deve- tem abnormalities in children with ASD [15–20]. An associa- lopment of ASD. Further research is needed, however, to tion between ASD and immune system abnormalities toge- establish a possible role of serine proteases in the aetiology ther with the vulnerability of the ASD group with regard to of ASD. inflammatory processes may indicate an impaired mecha- nism in this system. It is known that phagocytosis is an Conflict of Interests important part of the body’s defence mechanism. This mechanism requires an active complement system and The authors declare that they have no conflict of interest. Autism Research and Treatment 5

Acknowledgments of Autism and Developmental Disorders, vol. 26, no. 4, pp. 439– 452, 1996. The authors wish to thank Dr. Mohammad A. Karbasian and [16] G. Delgiudice-Asch and E. Hollander, “Altered immune fun- all of the staff at the Robat Karim Medical Diagnostic Labora- ction in autism,” CNS Spectrums, vol. 2, no. 5, pp. 61–68, 1997. tory and the staff at the Cellular and Molecular Research [17] R. P. Warren and A. N. C. Foster Margaretten, “Reduced nat- Centre, TUMS, Tehran, for their assistance in this study. ural killer cell activity in autism,” Journal of the American Aca- demy of Child and Adolescent Psychiatry, vol. 26, no. 3, pp. 333– 335, 1987. References [18] R. P. Warren, L. J. Yonk, R. A. Burger et al., “Deficiency of sup- pressor-inducer (CD4+CD45RA+) T cells in autism,” Immun- [1] American Psychiatric Association, “Disorders usually first ological Investigations, vol. 19, no. 3, pp. 245–251, 1990. diagnosed in infancy, childhood, or adolescence,” in Diagnostic [19] L. J. Yonk, R. P.Warren, R. A. Burger et al., “CD4+ helper T cell and Statistical Manual of Mental Disorders, American Psychi- depression in autism,” Immunology Letters, vol. 25, no. 4, pp. atric Association, Arlington, Va, USA, 4th edition, 1994. 341–346, 1990. [2] “Prevalence of autism spectrum disorders—Autism and [20]R.P.Warren,N.C.Margaretten,N.C.Pace,andA.Foster, Developmental Disabilities Monitoring Network Surveillance “Immune abnormalities in patients with autism,” Journal of Year 2006 Principal Investigators, Prevalence of autism spec- Autism and Developmental Disorders, vol. 16, no. 2, pp. 189– trum disorders—autism and developmental disabilities mon- 197, 1986. itoring network, United States, 2006,” MMWR Surveillance [21]G.Goldberger,M.A.Arnaout,D.Aden,R.Kay,M.Rits,and Summaries, vol. 58, no. 10, pp. 1–20, 2009. H. R. Colten, “Biosynthesis and postsynthetic processing of [3] M. L. Bauman and T. L. Kemper, “Neuropathology of infantile human C3b/C4b inactivator (factor I) in three hepatoma cell autism,” Molecular Psychiatry, vol. 7, no. 2, pp. S12–S13, 2002. lines,” Journal of Biological Chemistry, vol. 259, no. 10, pp. [4] K. B. Nelson, J. K. Grether, L. A. Croen et al., “Neuropeptides 6492–6497, 1984. [22] B. A. Corbett, A. B. Kantor, H. Schulman et al., “A proteomic and neurotrophins in neonatal blood of children with autism study of serum from children with autism showing differential or mental retardation,” Annals of Neurology,vol.49,no.5,pp. expression of apolipoproteins and complement proteins,” 597–606, 2001. Molecular Psychiatry, vol. 12, no. 3, pp. 292–306, 2007. [5] S. H. Fatemi, J. M. Stary, A. R. Halt, and G. R. Realmuto, “Dys- [23] J. Parkin and B. Cohen, “An overview of the immune system,” regulation of Reelin and Bcl-2 Proteins in Autistic Cerebel- Lancet, vol. 357, no. 9270, pp. 1777–1789, 2001. lum,” Journal of Autism and Developmental Disorders, vol. 31, [24]G.Goldberger,G.A.P.Bruns,M.Rits,M.D.Edge,and no. 6, pp. 529–535, 2001. D. J. Kwiatkowski, “Human complement factor I: analysis of [6] M. L. Bauman, “Microscopic neuroanatomic abnormalities in cDNA-derived primary structure and assignment of its gene autism,” Pediatrics, vol. 87, no. 5, pp. 791–796, 1991. to chromosome 4,” Journal of Biological Chemistry, vol. 262, [7]G.J.Blatt,C.M.Fitzgerald,J.T.Guptill,A.B.Booker,T.L. no. 21, pp. 10065–10071, 1987. Kemper, and M. L. Bauman, “Density and distribution of hip- [25] R. B. Sim, A. J. Day, B. E. Moffatt, and M. Fontaine, “Com- pocampal neurotransmitter receptors in autism: An Autorad- plement factor I and cofactors in control of complement sys- iographic Study,” Journal of Autism and Developmental Disor- tem convertase enzymes,” Methods in Enzymology, vol. 223, pp. ders, vol. 31, no. 6, pp. 537–543, 2001. 13–35, 1993. [8] L. Y. Tsai, R. R. Crowe, S. R. Patil, J. Murray, and J. Quinn, [26] C. J. Soames and R. B. Sim, “Interactions between human “Search for DNA markers in two autistic males with the fragile complement components factor H, factor I and C3b,” Bio- X syndrome,” Journal of Autism and Developmental Disorders, chemical Journal, vol. 326, no. 2, pp. 553–561, 1997. vol. 18, no. 4, pp. 681–685, 1988. [27] B. P. Morgan and M. J. Walport, “Complement deficiency and [9] E. London, “The environment as an etiologic factor in autism: disease,” Immunology Today, vol. 12, no. 9, pp. 301–306, 1991. a new direction for research,” Environmental Health Perspec- [28] M. M. Frank and L. F. Fries, “The role of complement in tives, vol. 108, no. 3, pp. 401–404, 2000. inflammation and phagocytosis,” Immunology Today, vol. 12, [10] S. Tordjman, L. Gutknecht, M. Carlier et al., “Role of the sero- no. 9, pp. 322–326, 1991. tonin transporter gene in the behavioral expression of autism,” [29] J. Kuby, Immunology,W.H.FreemanandCompany,New Molecular Psychiatry, vol. 6, no. 4, pp. 434–439, 2001. York, NY, USA, 3rd edition, 1997. [11] V. K. Singh, E. A. Singh, and R. P. Warren, “Hyperserotonine- [30]T.J.Vyse,P.J.Spath,K.A.Daviesetal.,“Hereditarycomple- mia and serotonin receptor antibodies in children with autism ment factor I deficiency,” Quarterly Journal of Medicine, vol. but not mental retardation,” Biological Psychiatry, vol. 41, no. 87, no. 7, pp. 385–401, 1994. 6, pp. 753–755, 1997. [31] P. Ashwood, B. A. Cobett, A. Kantor, H. Schulman, J. Van [12] S. I. Deutsch, “Rationale for the administration of opiate anta- de Water, and D. G. Amaral, “In search of cellular immuno- gonists in treating infantile autism,” American Journal of Men- phenotypes in the blood of children with autism,” Public Libr- tal Deficiency, vol. 90, no. 6, pp. 631–635, 1986. ary of Science One, vol. 6, no. 5, Article ID e19299, 2011. [13] J. Melke, H. Goubran Botros, P.Chaste et al., “Abnormal mela- [32] P. A. Garay and A. K. McAllister, “Novel roles for immune tonin synthesis in autism spectrum disorders,” Molecular Psy- molecules in neural development: implications for neurode- chiatry, vol. 13, no. 1, pp. 90–98, 2008. velopmental disorders,” Frontiers in Synaptic Neuroscience, vol. [14] N. Momeni, B. M. Nordstrom,¨ V. Horstmann, H. Avarsaji, and 2, pp. 1–16, 2010. B. V. Sivberg, “Alterations of prolyl endopeptidase activity in [33] C. Lord, M. Rutter, P. C. Dilavore, and S. Risi, Autism Diagnos- the plasma of children with autistic spectrum disorders,” BMC tic Observation Schedule (ADOS), Western Psychological Ser- Psychiatry, vol. 5, no. 27, article 27, pp. 1–6, 2005. vices, Los Angeles, Calif, USA, WPS edition, 1999. [15] S. Gupta, S. Aggarwal, and C. Heads, “Dysregulated immune [34] M. Rutter, A. Lecouteur, and C. Lord, Autism Diagnostic system in children with autism—beneficial effects of intra- Interview—Revised (ADI-R), Western Psychological Services, venous immune globulin on autistic characteristics,” Journal Los Angeles, Calif, USA, WPS edition, 2003. 6 Autism Research and Treatment

[35] Embargo Policy, “Identification and evaluation of children with autism spectrum disorders,” The American Academy of Pediatrics, vol. 120, no. 5, pp. 1183–1215, 2007. [36] S. A. Tsiftsoglou and R. B. Sim, “Human complement factor I does not require cofactors for cleavage of synthetic substrates,” Journal of Immunology, vol. 173, no. 1, pp. 367–375, 2004. [37] G. Dahlberg, Statistical Methods for Medical and Biological Students, George Allen and Unwin, London, UK, 2nd edition, 1940. [38] E. Korvatska, J. Van de Water, T. F. Anders, and M. E. Gersh- win, “Genetic and immunologic considerations in autism,” Neurobiology of Disease, vol. 9, no. 2, pp. 107–125, 2002. [39] P. Ashwood, S. Wills, and J. Van De Water, “The immune response in autism: a new frontier for autism research,” Journal of Leukocyte Biology, vol. 80, no. 1, pp. 1–15, 2006. [40] G. A. Mostafa and A. A. Shehab, “The link of C4B null allele to autism and to a family history of autoimmunity in Egyptian autistic children,” Journal of Neuroimmunology, vol. 223, no. 1-2, pp. 115–119, 2010.