You are on page 1of 9

Schizophrenia Research 176 (2016) 14–22

Contents lists available at ScienceDirect

Schizophrenia Research

journal homepage: www.elsevier.com/locate/schres

Clinical studies of neuroinflammatory mechanisms in schizophrenia


Crystal C. Watkins a,b,⁎, Sarah Ramsay Andrews b
a
Memory Center in Neuropsychiatry, Sheppard Pratt Health Systems, Baltimore, MD, United States
b
Department of Psychiatry, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, United States

a r t i c l e i n f o a b s t r a c t

Article history: Schizophrenia is a pervasive neurodevelopmental disorder that appears to result from genetic and environmen-
Received 10 April 2014 tal factors. Although the dopamine hypothesis is the driving theory behind the majority of translation research in
Received in revised form 8 June 2015 schizophrenia, emerging evidence suggests that aberrant immune mechanisms in the peripheral and central ner-
Accepted 8 July 2015
vous system influence the etiology of schizophrenia and the pathophysiology of psychotic symptoms that define
Available online 30 July 2015
the illness. The initial interest in inflammatory processes comes from epidemiological data and historical obser-
Keywords:
vations, dating back several decades. A growing body of research on developmental exposure to infection, stress-
Schizophrenia induced inflammatory response, glial cell signaling, structural and functional brain changes and therapeutic trials
Inflammation demonstrates the impact that inflammation has on the onset and progression of schizophrenia. Research in an-
Microglia imal models of psychosis has helped to advance clinical and basic science investigations of the immune mecha-
Glutamate nisms disrupted in schizophrenia. However, they are limited by the inability to recapitulate the human
Stress response experience of hallucinations, delusions and thought disorder that define psychosis. To date, translational studies
Psychosis of inflammatory mechanisms in human subjects have not been reviewed in great detail. Here, we critically re-
view clinical studies that focus on inflammatory mechanisms in schizophrenia. Understanding the
neuroinflammatory mechanisms involved in schizophrenia may be essential in identifying potential therapeutic
targets to minimize the morbidity and mortality of schizophrenia by interrupting disease development.
© 2015 Elsevier B.V. All rights reserved.

1. Introduction Schizophrenia is likely the most researched of the neuropsychiatric


diseases with an advanced scientific understanding of the genetic, envi-
Schizophrenia is a disabling psychiatric disorder that affects an esti- ronmental, molecular and physiologic contributing factors (Insel, 2011;
mated 250 million people across the world at some point in their life Jaaro-Peled et al., 2009a; Muller and Schwarz, 2010; van Os and Kapur,
(van Os and Kapur, 2009). Delusions, hallucinations, disorganized 2009). However, there remains insufficient knowledge regarding the
thinking and cognitive impairment are hallmarks of schizophrenia. primary origin and subsequent progression of the disease. This may be
Schizophrenia is a disease with no cure and long-term individual, family due in part to the complexity of the illness and logistical challenges of
and societal costs. Suicide rates of those suffering from schizophrenia studying psychotic symptoms in human subjects. Given the ability to
approach 10–15% (Rossler et al., 2005). Significant discoveries related manipulate neurotransmitter signals, animal models have been critical
to early identification, early treatment and stabilization with medica- in determining that inflammatory signatures and cytokine signaling
tions have led to more extensive research and a better long-term prog- are precursors to psychosis. The collective literature in animal models
nosis for schizophrenic patients. appears to mimic the progressive nature of the clinical syndrome of
schizophrenia, with exposure to pathogens in-utero and then develop-
Abbreviations: CNS, central nervous system; COX, cyclooxygenase; CSF, cerebrospinal
ment of aberrant pathology and clinical/behavioral symptoms emerging
fluid; EAAT2, excitatory amino acid transporter 2; GWAS, genome-wide association study; in adolescence or early adulthood. However, animal models have limita-
HLA, human leukocyte antigen; IFN, interferon; IL, interleukin; KYP, kynurenine pathway; tions in being able to recapitulate positive and negative symptoms of
LPS, lipopolysaccharide; MRS, magnetic resonance spectroscopy; MHC, major histocom- psychosis. The need for translational approaches in clinical schizophre-
patibility complex; NAC, N-acetylcysteine; iNOS, inducible nitric oxide synthase; NK, nat-
nia investigations, the difficult nature of experimental design, and the
ural killer; NMDA, N-methyl D-aspartate; NSAID, non-steroidal anti-inflammatory agents;
PET, positron emission tomography; Polyl:C, polyriboinosinic–polyribo-cytidylic acid; RA, clinical morbidity and mortality of the disease have generated research
rheumatoid arthritis; ROS, reactive oxygen species; SLE, systemic lupus erythematosus; interest in new hypotheses that address the etiological process of
SNP, single nucleotide polymorphism; TGF, transforming growth factor; Th, T helper; schizophrenia in human subjects, cells and tissue and subsequently
TNF, tumor necrosis factor; TSPO, translocator protein. help lay the foundation for new therapies.
⁎ Corresponding author at: Department of Neuropsychiatry, Sheppard Pratt Health
Systems, Gibson Building, Suite 100, 6501 N. Charles St., Baltimore, MD 21204, United
Historically, the pathophysiology of schizophrenia has been
States. linked to abnormal neurodevelopment and deficits in dopamine.
E-mail address: cwatkins@jhmi.edu (C.C. Watkins). While the dopamine hypothesis has defined schizophrenia for

http://dx.doi.org/10.1016/j.schres.2015.07.018
0920-9964/© 2015 Elsevier B.V. All rights reserved.

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
C.C. Watkins, S.R. Andrews / Schizophrenia Research 176 (2016) 14–22 15

many years, a growing number of research investigations and scien- studies, an increased release of cytokines is suggested to mediate
tific curiosity have developed around the immune system and the a cascade that “desensitizes” the immune system. This leads to
role of neuroinflammation in precipitating psychotic symptoms in changes in cellular proliferation, which further increases proin-
a subset of patients with psychosis (Drexhage et al., 2010; Miller flammatory cytokines downstream. Cytokines are also important
et al., 2011; Potvin et al., 2008; Upthegrove et al., 2014). These stud- in the immune mechanisms of schizophrenia, as they activate the
ies provide a detailed review of the theories and mechanisms that kynurenine pathway — an alternate route for tryptophan metabo-
support a role for inflammation in schizophrenia. Since the 1980s lism that leads to long-term changes in glutamatergic function, tro-
the immune hypothesis of schizophrenia has emerged as a theory phic support and synaptic function (Fig. 1). Glia cells in the form of
unified by data from human developmental, molecular imaging astrocytes and microglia further support the role of the immune
and therapeutic trial techniques. The consensus is that alterations system. Microglia in particular act as cytokine sensors and serve
in the immune system and neuroinflammation lead to progressive as the key regulatory cells of the immune system in the central ner-
brain changes in schizophrenia (Fudenberg et al., 1983; Stevens, vous system (CNS). Structural, molecular and functional changes in
1983). Epidemiologic, developmental, neuropathological and neu- microglia were noted in post-mortem schizophrenia patients who
roimaging observations have further advanced clinical and neuro- completed suicide (Radewicz et al., 2000; Steiner et al., 2008b,
science investigations in support of neuroinflammatory pathways 2011b; Wierzba-Bobrowicz et al., 2005). However, investigations
in psychiatric illness. using immunohistochemical markers have not consistently report-
Immune system dysfunction may result in part from prenatal expo- ed glial cell changes across all studies (Matthews and Harrison,
sure to a maternal infection of cerebral insult from Toxoplasma gondii, 2011). Inflammatory mediators, through microglia and kynurenine
Cytomegalovirus, Chlamydia, influenza or other infectious agents that metabolism, provide a related link to glutamate, dopamine and
generate an immune response (Ellman et al., 2009; Khandaker et al., downstream reactive oxygen species as markers of oxidative stress
2014b; Smesny et al., 2010). The subsequent cytokine cascade is in the pathophysiology of schizophrenia (Flatow et al., 2013;
thought to alter neuronal development before the illness is clinically Kohen and Nyska, 2002; Muller, 2014; Swerdlow et al., 2009).
expressed (Chew et al., 2013; Hagberg et al., 2012; Jaaro-Peled et al., We conducted a critical review of the literature for articles on
2009b). More recent developmental studies using schizophrenia pa- PubMed involving clinical studies and the search terms, schizophrenia
tients and tissues have clarified this initial observation and shown and inflammation. We then assessed the data related to immune mod-
that the inflammatory response, not necessarily the disease pathogen, ulation of schizophrenia in either human tissue, cells or patients with
alters the developmental trajectory of neurons (Meyer et al., 2010; schizophrenia. We selected articles where the primary language was
Miller et al., 2011). Immune-related genes have also been linked to ab- English and then critically reviewed the multidisciplinary, translational
errant immune signaling in schizophrenia (Jia et al., 2010; Stefansson research in schizophrenia subjects and subjects with non-affective psy-
et al., 2009). chosis. We discuss and analyze these investigations and identify poten-
In response to infection, stress-induced inflammation appears tial therapeutic targets that may mitigate the debilitating effects of the
to lead to psychopathological symptoms. In animal behavioral disease and lead to a better-long term prognosis for patients.

Fig. 1. Neuroinflammatory mechanism involved in schizophrenia and linked through kynurenine pathway (CRP — C-reactive protein; IDO — indoleamine 2,3-dioxygenase; 5-HT — serotonin;
RNS — reactive nitrogen species; ROS — reactive oxygen species).

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
16 C.C. Watkins, S.R. Andrews / Schizophrenia Research 176 (2016) 14–22

2. Systemic infections and neurodevelopmental mechanisms of Gressens, 2007). During the developmental insult, cytokines such as IL-
immune system activation in schizophrenia 8 (Brown et al., 2004) and TNF-alpha (Buka et al., 2001b) appear to cir-
culate at higher blood levels in the maternal blood of individuals later
2.1. Systemic infections and neurodevelopmental mechanisms at risk for psychotic disorders. Even infections in later stages of develop-
ment, particularly the second and third trimesters cause higher IL-8
Schizophrenia is thought of as a multifactorial disorder with contri- levels that correlate with structural changes in the progeny with schizo-
butions from the environment, genetics and other pathophysiological phrenia in the left entorhinal cortex and right posterior cingulated
processes that form a cluster of clinical symptoms. Given the strong in- (Ellman et al., 2010; Meyer, 2011). Although promising, specific mater-
teractions between genetics and the environment, more research has nal–fetal expression studies in schizophrenia are limited to these few in-
focused on the immune system in early exposure to infection as well vestigations. In addition, these retrospective and observational studies
as genes that are common in infection and psychosis. The perception are not able to identify the causal mechanism behind psychotic symp-
of an infectious cause of schizophrenia emerged from anecdotal toms. More recent, prospective cohort studies show a positive correla-
evidence over 150 years ago. Modern epidemiological evidence from tion between elevated systemic inflammatory marker interleukin 6 (IL-
national registries describes a cohort of individuals with genetic vulner- 6) and C-reactive protein (CRP) in childhood (measured at 9 years of
abilities for immune disorders and psychosis (Eaton et al., 2010; age) and development of future psychosis and depression in early life
Severance et al., 2014a, 2014b). Severe infections and autoimmune dis- (after age 18) (Khandaker et al., 2014a, 2014b). Childhood atopic disor-
orders over a lifetime appeared to confer additive risk for schizophrenia ders such as asthma and eczema were also suggested to increase the
and schizophrenia spectrum disorders (Benros et al., 2013b; Meyer, risk of psychotic experiences in adolescence (Khandaker et al., 2013).
2011). In contrast, parental infection alone did not appear to be a defin- Translational approaches in mouse and rat models with lipo-
itive risk factor (Benros et al., 2013a, 2013b). These data suggest that polysaccharide (LPS) and polyriboinosinic–polyribo-cytidylic acid
systemic infections might impact the developing fetal brain, making it (Polyl:C) have also examined the role of chronic versus acute inflamma-
vulnerable to brain illnesses later in life (Ellman et al., 2009). tion in neurodevelopment. The collective literature in animals appears
Epidemiologic research, post-mortem research, and imaging research to mimic the progressive nature of the clinical syndrome of schizophre-
in human subjects, consistently support the relationship between the im- nia. Although exposure to numerous pathogens occurs in-utero, the ab-
mune system and psychosis, specifically the neurodevelopmental expo- errant pathology and clinical/behavioral symptoms are not evident
sure to infection (Brown and Derkits, 2010; Cannon et al., 2002; Meda until adolescence or early adulthood.
et al., 2014; Rapoport et al., 2012). Pregnancy, a period where the mother
is more vulnerable to infections and elevations in proinflammatory
cytokines, may reflect a deviation from the normal immune processing. 2.2. Genetic polymorphisms
During influenza epidemics and heightened infection exposure from the
winter to spring months, pregnant women are at higher risk for giving In addition to the environmental contribution from prenatal infec-
birth to offspring that develop schizophrenia (Brown and Derkits, 2010; tion, immune related genes and alterations in the immune response
Meyer et al., 2010). Respiratory infections (Sorensen et al., 2009; Selten have been found in schizophrenia patients (Consortium, 2014; Jia
et al., 2009) and reproductive tract infections (Babulas et al., 2006; et al., 2010; Schwarcz et al., 2001). These and other studies have looked
Sorensen et al., 2009) during pregnancy have also been linked to an in- at the collective involvement of inflammatory genes in schizophrenia
creased risk for schizophrenia. Other pathogens linked to schizophrenia and the details and number of genes involved are beyond the scope of
include maternal infection with T. gondii (Brown et al., 2005), rubella our review. Genome-wide association studies allow for the identifica-
(Brown et al., 2000), measles (Torrey et al., 1997), polio (Suvisaari et al., tion of genetic polymorphisms or divergent genes and altered immune
1999) and herpes simplex type 2 (Buka et al., 2001a). With the epidemi- signaling that may confer a genetic susceptibility to infection (Drago
ologic data that infections before birth increase the risk of developing et al., 2014). Genetic studies from large, multiple cohorts suggest that
schizophrenia (Brown and Derkits, 2010; Buka et al., 2001a, 2001b; the polymorphisms vulnerable to schizophrenia are positioned on dif-
Dalman et al., 2008; Gattaz et al., 2004; Yudofsky, 2009), one would also ferent gene loci located on chromosome 6p22 (Drago et al., 2014;
expect a corresponding increase in antibody titers against the virus. How- Purcell et al., 2009; Stefansson et al., 2009) (Consortium, 2014; Muller,
ever, measurements of maternal antibody titers against viruses during 2014). Several immune-related genes share this region. The most signif-
pregnancy have yielded inconsistent results between different studies icant gene associations were with HLA complex genes that moderate
(Blomstrom et al., 2012; Brown et al., 2004; Leweke et al., 2004). A the inflammatory response (Fellerhoff et al., 2007), histone genes that
case–control study of 198 individuals born in Sweden from 1975 to focus on antimicrobial defense (Kawasaki and Iwamuro, 2008) and his-
1985 and diagnosed with schizophrenia showed elevated levels of IgG di- tone proteins potentially involved in DNA repair and methylation (Costa
rected at T. gondii. CMV was also elevated as markers of maternal expo- et al., 2009; Kundakovic et al., 2007). A meta-analysis complied many of
sure and associated with subsequent development of schizophrenia the genetic studies to date and was able to pinpoint specific variants of
(Blomstrom et al., 2012). There were no associations between any of immune-related genes in schizophrenia (Ripke et al., 2013). However,
the infectious agents and other non-affective psychoses. However, epide- the specific polymorphisms were expressed in other psychiatric disor-
miologic studies are often limited because of their observational nature; ders and were not exclusive to psychosis or schizophrenia (Ripke
they are unable to show the role of inflammation as a cause or an effect et al., 2013; Consortium, 2014).
of schizophrenia. Some suggest that the discrepancy is because a genetic disposition
Most infections do not cross the placenta, with the exception of for immune activation in mental illness may only be expressed in a
parasitic infections like T. gondii. Therefore, damaging effects to the subgroup of people (Pathmanandavel et al., 2013). Others argue that ex-
fetal brain are likely through maternal or fetal responses to infections pression of inflammatory genes may be brain region specific or have a
through the placenta (Patterson, 2009; Shi et al., 2005). Cognitive and temporal relationship to a specific brain insult. One microarray and
neurological impairments have been observed after in-utero exposure RNA sequence study shows abnormal immune/inflammatory responses
to infection and appear to correlate with changes in white matter around limited to the hippocampus (Hwang et al., 2013). Another study looking
the ventricles, termed periventricular leukomalacia (Dammann and at mRNA expression of inflammatory genes in a cohort of schizophrenia
Leviton, 1997; Favrais et al., 2011; Sorensen et al., 2006; Chew et al., subjects showed a difference in psychotic patients older 40 years of age
2013; Hagberg et al., 2012). The subsequent pro-inflammatory changes compared to those under 40 (Tang et al., 2012). The specific location of
and microglial activation in periventricular leukomalacia seem to act as gene expression may be directly related to the long-term neurocognitive
mediators of CNS inflammation (Dammann et al., 2008; Leviton and deficits associated with schizophrenia. There may also be a temporal

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
C.C. Watkins, S.R. Andrews / Schizophrenia Research 176 (2016) 14–22 17

relationship, with immune genes more prominently expressed with du- (Nathan and Shiloh, 2000). Activation of pro-inflammatory cyto-
ration of the illness and/or age. kines such as IL-6 or TNF-alpha might induce the activation of apo-
ptosis through microglial activation and subsequent production of
3. Immune response and inflammatory mediators reactive oxygen species (ROS) and increase reactive nitrogen species
(Fig. 2). An additional pathway that promotes oxidative stress in
3.1. Innate immune pathways, cytokine signaling and downstream schizophrenia is the activation of NMDA receptor through glutamate.
inflammation Glutamate is actively taken up into astrocytes and is converted into
glutamine. Alterations in glutamine increase calcium influx into neu-
Although much of the developmental literature stems from models rons, which may contribute to excitotoxicity, NMDA antagonism in
of peripheral infection acting directly to contribute to neuroinflamma- schizophrenia and altered neurotransmission. The imbalance in the
tion, many initially doubted the theory because the blood–brain barrier glutamatergic neurotransmission leads to further production of
protects the CNS from systemic infection. More recent research has ROS and RNS, which leads to nitrosative damage to DNA, proteins
been able to distinguish immune signals in the innate immune system, and lipids, in a situation where oxidative defenses are already
predominated by monocytes and macrophages that defend the blood– vulnerable.
brain barrier, and the adaptive immune system, which is comprised of The anti-oxidant defense system appears to be altered in schizophre-
cytokines and chemokine signals secreted from these cells (Miller nia (Flatow et al., 2013). A recent meta-analysis demonstrated decreased
et al., 2011; Muller, 2014; Upthegrove et al., 2014). These cytokines serum and plasma antioxidant markers in first episode schizophrenia pa-
and their receptors appear to be fundamental to fetal neuronal growth, tients that increased longitudinally with anti-psychotic treatment (Flatow
migration and survival (Bakhiet et al., 2002; Mehler and Kessler, 1998; et al., 2013). Another study showed a reduction in superoxide dismutase
Meyer et al., 2010; Mousa et al., 1999). In the adult brain, in addition enzyme (SOD-1) in the serum and CSF of individuals experiencing their
to their immune function, cytokines are signaling molecules within first episode of schizophrenia (Coughlin et al., 2012). Increasing evi-
the brain and important in astrocyte and glial cell communication dence from the fields of neurophysiology and neuropathology has un-
(Muller, 2014). The individual pro- and anti-inflammatory markers in- covered the role of polyunsaturated fatty acids (PUFA) in protecting
volved in schizophrenia and inflammation are reviewed elsewhere in neuronal cells from oxidative damage, controlling inflammation, regu-
detail (Tomasik et al., in press). Previous studies have shown heteroge- lating neurogenesis, and preserving neuronal function (Hashimoto
neous results with peripheral cytokine measures, in part due to con- et al., 2014). Based on a recent review of oxidative stress and schizo-
founding variables including medications and duration of illness. phrenia, understanding this relationship through both human and ani-
However, a number of studies in both chronic and first episode schizo- mal studies could be the key to treatment options for patients with
phrenia show increased expression of interleukin (IL)-1beta, IL-6, IL-8, schizophrenia (Emiliani et al., 2014).
tumor necrosis factor (TNF) and C-reactive protein in both peripheral
blood and cerebral spinal fluid (CSF) (Hayes et al., 2014; Kunz et al., 3.3. COX inhibition and anti-inflammatory mechanisms
2011; Miller et al., 2011; Schwieler et al., 2014). Although many im-
mune modulators have been studied, the most consistent research iden- Cyclooxygenase-2 (COX-2), an influential regulator of pain and in-
tifies elevated levels of IL-6 in both the cerebral spinal fluid (CSF) and flammation, is the key enzyme in the arachidonic acid pathway respon-
serum of individuals with schizophrenia. In particular, drug-naïve, first sive to cytokines. Inflammatory cytokines such as IL-1 beta and tumor
episode schizophrenia patients had elevated IL-6, IL-10 and TNF-α at necrosis factor (TNF)-alpha have been shown to enhance expression
baseline. Treatment with risperidone over time leads to and repeated of COX-2 mRNA (Muller and Schwarz, 2010; Sommer et al., 2013).
measurements showed a decrease in all three cytokines and IL-4. Fur- Therefore, inhibition of COX-2 is important in dampening the cytokine
thermore, there was no difference in cytokine levels in healthy controls response and minimizing inflammation.
compared to the post-treatment group (Noto et al., 2014). This is of in- Like the inflammatory cytokines, COX is also influenced by the
terest because IL-6 is directly secreted from monocytes and is a major kynurenine pathway. Inhibition of COX-2 by celecoxib decreases levels
component of the adaptive immune system (Hayes et al., 2014; Miller of kynurenic acid, while inhibition of COX-1 increases levels of
et al., 2011; Muller, 2014). The change after treatment suggests that in- kynurenic acid (Muller and Schwarz, 2010; Schwieler et al., 2014).
flammatory cytokines are related to state changes in schizophrenia. Expanding on this anti-inflammatory mechanism, double-blind ran-
Translational techniques that combine peripheral blood and CSF mea- domized trials in acute schizophrenia reported symptom improvement
surements with gene microarray date and longitudinal measurements when risperidone was augmented with celecoxib (Akhondzadeh et al.,
will be necessary in order to further advance the field. We may also be 2007; Muller et al., 2004, 2010). Although the results have not been rep-
able to gain more information from other potential state markers of in- licated in subsequent studies, Muller and colleagues concluded that the
flammation, like methylation of phosphatase SHP-1, a key negative reg- duration of schizophrenia impacts the timing and effectiveness of COX-
ulator of the inflammatory process. SHP-1 is easily measured in 2 inhibitors (Muller et al., 2004). Prospective, randomized, double-blind
peripheral blood monocytes and appears to be impaired in schizophre- clinical trials of first episode schizophrenia (Fond et al., 2013) and
nia (Pesce et al., 2014). chronic schizophrenia (Akhondzadeh et al., 2007), demonstrated that
augmentation of atypical antipsychotics with celecoxib had a therapeu-
3.2. Oxidative stress and reactive oxygen species tic effect. Individuals with long-standing psychotic symptoms did not
appear to respond to the COX-2 inhibitor. As we mentioned with cyto-
In addition to activating cytokines, induction of the immune re- kine expression earlier, there also appears to be an age effect in arachi-
sponse and activation of the inflammatory components appear to donic acid related inflammatory pathways with PTGS-1 (a measure of
have downstream effects on glutamate, oxidative stress, and the ar- COX-1) and PTGS-2 (a measure of COX-2) decreased in older subjects
achidonic acid pathway (Muller, 2014; Muller and Schwarz, 2010). with schizophrenia compared to those younger than 40 (Tang et al.,
Oxidative stress is defined as the “imbalance between the systemic 2012). Therefore, age may be a factor in therapeutic response. While
manifestation of reactive oxygen species and the body's ability to promising, interpretation of the celecoxib trials is limited due to few
eliminate the reactive oxygen species (ROS)” (Kohen and Nyska, studies on mono-treatment with celecoxib and the brief duration of
2002; Muller and Schwarz, 2010). Oxidative stress may also affect these trials.
lipid and glucose metabolism in psychosis (Aleksovska et al., 2014). The benefits of anti-inflammatory therapy are not limited to
The innate immune system generates ROS and reactive nitrogen celecoxib. Nonsteroidal anti-inflammatory drugs (NSAIDs), reviewed
species (RNS) to assist with the destruction of foreign pathogens in a recent meta-analysis, added to either risperidone or olanzapine

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
18 C.C. Watkins, S.R. Andrews / Schizophrenia Research 176 (2016) 14–22

Fig. 2. Activation of pro-inflammatory cytokines such as IL-6 or TNF-alpha might induce activation of apoptosis through microglial activation and subsequent production of reactive oxygen
species (ROS) and increase reactive nitrogen species. An additional pathway that promotes oxidative stress in schizophrenia is the activation of NMDA receptor through glutamate. Glu-
tamate is actively taken up into astrocytes and is converted into glutamine. Alterations in glutamine increase calcium influx into neurons, which may contribute to excitotoxicity, NMDA
antagonism in schizophrenia and altered neurotransmission. The imbalance in the glutamatergic neurotransmission leads to further production of ROS and RNS, which leads to damage to
DNA, proteins and lipids, in a situation where oxidative defenses are already vulnerable. These differences are the following: CRP-C — reactive protein; IDO — indoleamine 2,3-
dioxygenase; 5-HT — serotonin; RNS — reactive nitrogen species; ROS — reactive oxygen species; and GLU — glutamate.

resulted in a decline in psychotic symptomatology in patients with Postmortem brain studies of schizophrenia were some of the first to
schizophrenia (Meyer, 2011; Sommer et al., 2013). Adjuvant aspirin demonstrate preservation of the number of neurons with diminished
therapy reduces the symptoms of schizophrenia measured by the Posi- glia cell numbers and altered morphology (Monji et al., 2009). Further
tive and Negative Symptoms Score (PANSS) scale, with a more pro- research has led disagreements about which glial-subtype is most im-
nounced reduction in those with the more altered immune function portant in schizophrenia and the neuroinflammatory hypothesis.
(Laan et al., 2010). Compared to antipsychotic monotherapy, Changes in white matter intensities have been successively reported
minocycline (a broad-spectrum antibiotic), when added to an antipsy- in schizophrenia patients and post-mortem studies and appear to be
chotic, showed a significant reduction in symptoms and additive cogni- linked to oligodendrocyte gene polymorphism, resulting in a decrease
tive benefit in double-blind, randomized trials of patients with recent of the overall number of oligodendrocytes (Bernstein et al., 2012;
onset schizophrenia (Levkovitz et al., 2010). Several studies have also Hercher et al., 2014). More recent studies have examined the comple-
reported that antipsychotic drugs themselves counteract the inflamma- ment system and increased risk for schizophrenia in adult offspring
tory effects of cytokine networks, although the exact mechanism is not by focusing on complement regulatory proteins in glial signaling
known (Kroken et al., 2014; Sommer et al., 2013). (Severance et al., 2014a, 2014b; Mayilyan et al., 2006). However, most
studies have focused on the role of astrocytes and microglia in schizo-
4. Structural and functional mediators of neuroinflammation phrenia because of their roles in glutamate metabolism and cytokine
signaling, respectively (Le Hellard et al., 2008).
4.1. Glial signaling and the immune response

Glia cells, the connecting tissue in the brain, also appear to bridge ge- 4.2. Astrocytes and microglia
netic susceptibility with inflammation and neurotransmitter signaling
in schizophrenia (Steiner et al., 2011a). In addition to changes in im- Replication of both astrocyte and microglia pathology using immu-
mune cell activation and response, there is accumulating evidence of nohistochemical methods in post-mortem brains of schizophrenic
abnormalities in the homeostatic support and protection in the brain patients has led to inconsistent results and conclusions. This is often ex-
that is preserved by glia cells (oligodendrocytes, astrocytes and microg- plained by age-related brain changes, differences in tissue processing
lia) in individuals with schizophrenia. Oligodendrocytes produce and differences in immunostaining technique. However, the consensus
myelin and provide insulation and support for neurons. Astrocytes are is that glial damage is suspected given the increase in astrocyte concen-
star-shaped cells that provide biochemical support to neurons, secrete tration in patients with schizophrenia. Schizophrenia patients also ap-
IL-10 and inhibit the production of IL-12. Microglia are transformed pe- pear to have hyper-functioning astrocytes, measured by increases in
ripheral macrophages in the CNS that sense inflammation and secrete the astrocyte marker S100B and glial cell activation (Rothermundt
IL-12. The anatomic and functional changes of glial cells have emerged et al., 2007). The strongest supporting evidence comes from the modu-
as a mechanism of interest in multiple psychiatric disorders (Bernstein lation of glutamate metabolism by astrocytes (Steiner et al., 2008a,
et al., 2009; Monji et al., 2009; Muller, 2014; Watkins et al., 2014). 2011b). Defective astrocyte signaling is thought to result in low levels

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
C.C. Watkins, S.R. Andrews / Schizophrenia Research 176 (2016) 14–22 19

of glutamate and increased NMDA receptor activity. We will discuss the to be important cells in glutamate metabolism, the KYP and the immune
role of glutamate in glial cells in detail in the next section. system.
Microglia, upon sensing a change in the brain environment, undergo
a conformational change and become “activated.” When activated they 5. Structural and functional brain imaging and inflammation
produce pro-inflammatory cytokines and express protein markers.
Several post-mortem investigations of completed suicides by schizophrenic 5.1. Magnetic resonance imaging
patients show increased microglial activation (Matthews and Harrison,
2011; Radewicz et al., 2000; Wierzba-Bobrowicz et al., 2005; Bernstein Molecular imaging methods that study glia as markers of neuroin-
et al., 2012). There are also signs of altered expression of microglial protein flammation may lend insight into the pathogenesis of schizophrenia.
markers in peripheral blood, monocytes and cultured microglia from Structural imaging studies using magnetic resonance imaging (MRI)
schizophrenic patients (Zheng et al., 2008). Recently one group affiliated demonstrate reduced CNS brain volume in recent onset schizophrenia
with the Sydney Tissue Resource Center discovered increased expression (Dieset et al., 2015; Steen et al., 2006). Chronic disease also shows a pro-
of cytokine mRNA levels in the post-mortem brains of people with gressive deterioration in brain volume that is related to more clinically
schizophrenia that corresponded with alterations in microglial density severe disease symptoms (Gogtay et al., 2008; Steen et al., 2006).
(Fillman et al., 2013). In contrast, other studies did not observe a significant Changes in the cellular architecture of the brain are associated with in-
difference in the number or appearance of microglia (Arnold et al., 1996; creased levels of IL-1 and IL-6 in acute exacerbation episodes of schizo-
Steiner et al., 2008b; Takano et al., 2010). phrenia (Garver et al., 2003; Meisenzahl et al., 2001). Other studies of
Antipsychotic drugs have been shown to inhibit microglial activa- the Edinburgh High Risk cohort showed reduction in brain volume in
tion, astrocyte signaling and cytokine production, with the majority of the temporal gyrus with disease progression that was apparent prior
studies in rodent models (Zheng et al., 2008) and a limited few in to the worsening of clinical symptoms (Chakos et al., 2005; Job et al.,
human cultured cells (Chew et al., 2013; Monji et al., 2009; Radomska 2005, 2006). Diffusion tensor imaging (DTI), a newer MRI technique,
et al., 2013) or postmortem studies (Kim et al., 2012) (Busse et al., uses water and other diffusion patterns to map to understand brain
2012). Some propose that the activation of microglia leads to increased structure and function at the microscopic level. A recent study com-
nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase and bined DTI and measurements of IL-6 and C-reactive protein in patients
inducible (i)-NOS expression that contributes to increased oxidative with early psychosis. IL-6 levels were inversely correlated with func-
stress. The acute on chronic activation damages the supporting cells tional anisotropy (an estimate of fiber density, axonal diameter, and
and may contribute to the clinical symptoms of schizophrenia. Taken myelination in white matter) and positively correlated with radial diffu-
together, astrocytes and glia appear to have both complementary and sivity (Prasad et al., 2014). C-reactive protein also showed a similar
contradictory roles in the neuroinflammatory response. Further re- pattern in psychosis compared to healthy controls. These results suggest
search into the mechanism of glial dysfunction and inflammation may a vulnerability of selected neural pathways to immune signals in
give rise to new therapeutic approaches. schizophrenia.

4.3. Downstream effects of inflammation on neuro-circuitry 5.2. Positron emission tomography and spectroscopy studies

Like astrocytes, microglia have a functional role in neurotransmitter When microglia are activated, they express an 18-kDa protein, TSPO.
signaling through cytokine-induced mechanisms that ultimately affect Positron emission tomography studies have used TSPO ligands as
neurogenesis, glutamate production and excitotoxicity in the brain markers for neuroinflammation in psychiatric disorders to estimate
(Muller, 2014; Steiner et al., 2011a). Microglia appear to play a the rate of microglia activity (Watkins et al., 2014). The first generation
major role in mood and serotonin metabolism through the enzyme TSPO ligand, PK1195 showed increased expression of TSPO in the hippo-
indoleamine 2,3-dioxygenase (IDO) in the kynurenine pathway (KYP). campus of schizophrenia patients (Doorduin et al., 2009). Second gener-
Schizophrenia patients tend to have co-morbid depression symptoms ation TSPO ligands like 11C-PBR28, 11C[DAA1106], [(18)F]-FEPPA and
11
that may be related to changes in serotonin (Steiner et al., 2011b). C[DPA-713] are being studied in both first episode and chronic schizo-
Working through common enzymes and cytokine signals in the KYP, as- phrenia (Kreisl et al., 2012) {Sawa et al., unpublished data}. In chronic
trocytes are responsible for the release and re-uptake of glutamate. Clin- schizophrenia, 11C[DAA1106] PET showed no significant difference be-
ical observations using ketamine and phencyclidine treatment show tween binding in normal controls (Takano et al., 2010). However, partic-
that alterations in glutamate receptors and the ultimate antagonism of ipants with schizophrenia and a predominance of positive symptoms did
NMDA receptors produce a constellation of symptoms comparable to show increased binding. Another PET ligand, [(18)F]-FEPPA examined
the psychotic, behavioral and cognitive disturbances in schizophrenia white and gray matter regional binding in patients with active psychosis.
(Coyle, 2006). This study failed to show a significant difference between symptomatic
Glutamate is also linked to schizophrenia and neuroinflammation patients and healthy controls (Kenk et al., 2014). However, the partici-
through microglia and the kynurenine–tryptophan signaling mecha- pants were actively taking medications. Therefore, either the change in
nism (Fig. 2). Glutamatergic hypofunction may proceed or be the result microglia occurs earlier in the process and/or antipsychotic treatment
of dopaminergic dysfunction, in the limbic system and frontal cortex of may have negated any significant difference between the groups.
the brain. Dopamine has long been accepted as one of the main neuro- Magnetic resonance spectroscopy (MRS) is another imaging modal-
transmitters disturbed in schizophrenia. Inflammatory mediators, ity that is able to look at the underlying brain chemistry related to in-
through microglia and kynurenine metabolism, provide a related link flammation. 13C-MRS performed using a 1.5 Tesla magnetic resonance
to glutamate, dopamine and downstream reactive oxygen species as scanner was able to demonstrate abnormalities in cerebral I-13C glu-
markers of oxidative stress in the pathophysiology of schizophrenia cose, the principal nutrient for glia along with blunted glial metabolism
(Erhardt et al., 2001; Muller, 2014; Sathyasaikumar et al., 2010; (Chang et al., 2007). With assistance from the newer 3-Tesla and 7-Tesla
Schwarcz et al., 2001; Swerdlow et al., 2009). scanners, proton magnetic resonance spectroscopy (1H MRS) and posi-
Genetic defects in the glutamate receptor gene GRM3 are also relat- tron emission tomography investigations, researchers now have the
ed to schizophrenia-type symptoms (Bustillo et al., 2014). Immunohis- ability to indirectly measure glutamate by quantitating levels of gluta-
tochemical data confirms GRM3 localization to astrocytes (Spangaro thione and glycine, as markers of neuroinflammation (Bustillo et al.,
et al., 2012). Molecular biology studies also show that astrocytes help 2014; Poels et al., 2013). (1H)-MRS was used to measure glutamate,
regulate glutamate transport that involves the excitatory amino acid GABA, and NAAG levels in the anterior cingulate (AC) and centrum
transporter 2 (EAAT2) (Huerta et al., 2006). In summary, glia appear semiovale (CSO) regions (Rowland et al., 2012). Younger schizophrenia

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
20 C.C. Watkins, S.R. Andrews / Schizophrenia Research 176 (2016) 14–22

subjects had higher NAAG in the CSO when compared with younger Contribution statement
Both Dr. Watkins and Dr. Andrews contributed to this manuscript. Dr. Watkins con-
control subjects. Older groups had the reverse finding. Independent of
tributed to the conception and design, interpretation of data, drafting and revising the ar-
age, glutamate was reduced in the schizophrenia group. In another ticle and funding for the project. Dr. Andrews contributed to the data collection, writing
(1H)-MRS study, schizophrenic patients showed increased glutamine the article and revision of the manuscript for publication.
and glutamine to glutamate ratios, but no changes in glutamate levels
in the brain (Bustillo et al., 2014). A recent study using the 7-T (1H)- Conflict of interest
MRS was able to non-invasively distinguish glutamate from GABA in We have no conflicts of interest to disclose.
the brain. The group showed decreased GABA/Cr ratios in the prefrontal
cortex in schizophrenia as compared to healthy controls, with GABA/Cr Acknowledgments
ratios inversely correlated with cognitive functioning in the patients. This work was supported by the Johns Hopkins University Mosaic Initiative Award
They did not observe any significant change in the GABA/Cr ratio, gluta- and a Sheppard Pratt Neuropsychiatry grant from the Women's Hospital Foundation
(CCW).
mate, NAA, creatine, or choline in the parieto-occipital cortex region
when patients were compared to controls.
Another study looked at human subjects' in vivo, postmortem brain References
and mouse models (Stan et al., 2014). The data demonstrated signifi- Akhondzadeh, S., Tabatabaee, M., Amini, H., Ahmadi Abhari, S.A., Abbasi, S.H., Behnam, B., 2007.
cantly lower glutamate levels in the hippocampus in patients, which Celecoxib as adjunctive therapy in schizophrenia: a double-blind, randomized and placebo-
also correlated with lower GluN1 protein levels selectively in the den- controlled trial. Schizophr Res. 90 (1-3), 179–185.
Aleksovska, K., Leoncini, E., Bonassi, S., Cesario, A., Boccia, S., Frustaci, A., 2014. Systematic review
tate gyrus (DG) in postmortem brains. Interestingly, targeted deletion and meta-analysis of circulating S100B blood levels in schizophrenia. PLoS One 9 (9),
of the GRIN1 in the mouse model also confirmed a region specific de- e106342.
Arnold, S.E., Franz, B.R., Trojanowski, J.Q., Moberg, P.J., Gur, R.E., 1996. Glial fibrillary acidic
crease in the glutamate concentrations in the whole hippocampus
protein-immunoreactive astrocytosis in elderly patients with schizophrenia and dementia.
(Stan et al., 2014). More translation studies like these will be needed Acta Neuropathol. 91 (3), 269–277.
to fully understand neurotransmitter signaling in schizophrenia. Emerg- Babulas, V., Factor-Litvak, P., Goetz, R., Schaefer, C.A., Brown, A.S., 2006. Prenatal exposure to ma-
ternal genital and reproductive infections and adult schizophrenia. Am. J. Psychiatry 163
ing investigations may provide potential translational techniques to (5), 927–929.
look at the relationships between brain architecture, brain chemistry Bakhiet, M., Mousa, A., Seiger, A., Andersson, J., 2002. Constitutive and inflammatory induction
and the inflammatory pathways involved in schizophrenia. of alpha and beta chemokines in human first trimester forebrain astrocytes and neurons.
Mol. Immunol. 38 (12–13), 921–929.
Benros, M.E., Laursen, T.M., Dalton, S.O., Nordentoft, M., Mortensen, P.B., 2013a. The risk of
schizophrenia and child psychiatric disorders in offspring of mothers with lung cancer
and other types of cancer: a Danish nationwide register study. PLoS One 8 (11), e79031.
6. Conclusion
Benros, M.E., Pedersen, M.G., Rasmussen, H., Eaton, W.W., Nordentoft, M., Mortensen, P.B.,
2013b. A nationwide study on the risk of autoimmune diseases in individuals with a per-
The immune system appears to be important in schizophrenia at sonal or a family history of schizophrenia and related psychosis. Am. J. Psychiatry 171
(2), 218–226.
multiple-neurobiological levels. The study of inflammatory mechanisms Bernstein, H.G., Steiner, J., Bogerts, B., 2009. Glial cells in schizophrenia: pathophysiological sig-
in psychosis has evolved over the past decades, but further investigations nificance and possible consequences for therapy. Expert. Rev. Neurother. 9 (7), 1059–1071.
directed at clinical models and cell cultures from human subjects are Bernstein, H.G., Smalla, K.H., Durrschmidt, D., Keilhoff, G., Dobrowolny, H., Steiner, J., et al., 2012.
Increased density of prohibitin-immunoreactive oligodendrocytes in the dorsolateral pre-
needed. Translational approaches that combine epidemiology, genetics, frontal white matter of subjects with schizophrenia suggests extraneuronal roles for the
oxidative stress, glutamatergic transmission and clinical treatment have protein in the disease. Neuromol. Med. 14 (4), 270–280.
Blomstrom, A., Karlsson, H., Wicks, S., Yang, S., Yolken, R.H., Dalman, C., 2012. Maternal antibod-
advanced our understanding of the complex neuroinflammatory media- ies to infectious agents and risk for non-affective psychoses in the offspring — a matched
tors involved in the onset and progress of schizophrenia. The current re- case–control study. Schizophr. Res. 140 (1–3), 25–30.
search supports neurodevelopment, oxidative stress and glial pathology Brown, A.S., Derkits, E.J., 2010. Prenatal infection and schizophrenia: a review of epidemiologic
and translational studies. Am. J. Psychiatry 167 (3), 261–280.
as relevant immune mechanisms and important targets for therapeutic Brown, A.S., Cohen, P., Greenwald, S., Susser, E., 2000. Nonaffective psychosis after prenatal ex-
intervention in schizophrenia. Still, little is known about the temporal re- posure to rubella. Am. J. Psychiatry 157 (3), 438–443.
lationship between inflammatory changes and onset of illness. Some of Brown, A.S., Hooton, J., Schaefer, C.A., Zhang, H., Petkova, E., Babulas, V., et al., 2004. Elevated ma-
ternal interleukin-8 levels and risk of schizophrenia in adult offspring. Am. J. Psychiatry 161
the research in human subjects still shows heterogeneous results or no (5), 889–895.
difference between schizophrenia patients and controls. The validity Brown, A.S., Schaefer, C.A., Quesenberry Jr., C.P., Liu, L., Babulas, V.P., Susser, E.S., 2005. Maternal
exposure to toxoplasmosis and risk of schizophrenia in adult offspring. Am. J. Psychiatry
and reliability of these studies are limited by co-morbid factors like 162 (4), 767–773.
smoking, cannabis use, medication effects and life stressors, all of which Buka, S.L., Tsuang, M.T., Torrey, E.F., Klebanoff, M.A., Bernstein, D., Yolken, R.H., 2001a. Maternal
are difficult to regulate in a longitudinal clinical study of psychosis. With infections and subsequent psychosis among offspring. Arch. Gen. Psychiatry 58 (11),
1032–1037.
ubiquitous confounds like stress and inflammation, it is difficult to identi- Buka, S.L., Tsuang, M.T., Torrey, E.F., Klebanoff, M.A., Wagner, R.L., Yolken, R.H., 2001b. Maternal cy-
fy a causal relationship between the initial infection and other mediating tokine levels during pregnancy and adult psychosis. Brain Behav. Immun. 15 (4), 411–420.
Busse, S., Busse, M., Schiltz, K., Bielau, H., Gos, T., Brisch, R., et al., 2012. Different distribution pat-
effects. There may be another affiliated mechanism in schizophrenia that
terns of lymphocytes and microglia in the hippocampus of patients with residual versus
distinguishes the “prodromal phase” and general psychosis from clinical paranoid schizophrenia: further evidence for disease course-related immune alterations?
schizophrenia, which has yet to be identified. Advances in schizophrenia Brain Behav. Immun. 26 (8), 1273–1279.
Bustillo, J.R., Chen, H., Jones, T., Lemke, N., Abbott, C., Qualls, C., et al., 2014. Increased glutamine
will come at the intersection of techniques that combine a specific pheno- in patients undergoing long-term treatment for schizophrenia: a proton magnetic reso-
type of schizophrenia, control for gene polymorphisms, measure periph- nance spectroscopy study at 3 T. JAMA Psychiatry 71 (3), 265–272.
eral markers, incorporated cell culture and/or utilize molecular imaging. Cannon, M., Jones, P.B., Murray, R.M., 2002. Obstetric complications and schizophrenia: histori-
cal and meta-analytic review. Am. J. Psychiatry 159 (7), 1080–1092.
Given that further studies are needed to interpret the role of neuroinflam- Chakos, M.H., Schobel, S.A., Gu, H., Gerig, G., Bradford, D., Charles, C., et al., 2005. Duration of ill-
mation in schizophrenia, research will have to continue to expand across ness and treatment effects on hippocampal volume in male patients with schizophrenia. Br.
J. Psychiatry 186, 26–31.
multiple disciplines and preclinical and clinical modes. Even with incor- Chang, L., Friedman, J., Ernst, T., Zhong, K., Tsopelas, N.D., Davis, K., 2007. Brain metabolite abnor-
poration of diverse experimental approaches, targeted therapeutic ap- malities in the white matter of elderly schizophrenic subjects: implication for glial dysfunc-
proaches will be needed at early stages of development in order to tion. Biol. Psychiatry 62 (12), 1396–1404.
Chew, L.J., Fusar-Poli, P., Schmitz, T., 2013. Oligodendroglial alterations and the role of microglia
ultimately manage schizophrenia. in white matter injury: relevance to schizophrenia. Dev. Neurosci. 35 (2–3), 102–129.
Consortium, S. W. o. t. P. G., 2014. Biological insights from 108 schizophrenia-associated genetic
loci. Nature 511 (7510), 421–427.
Costa, E., Chen, Y., Dong, E., Grayson, D.R., Kundakovic, M., Maloku, E., et al., 2009. GABAergic
Funding source
promoter hypermethylation as a model to study the neurochemistry of schizophrenia vul-
Dr. Watkins' research is supported by the Mosaic Initiative Award from the Johns Hop- nerability. Expert. Rev. Neurother. 9 (1), 87–98.
kins University, a faculty development grant for early career academic physicians. She is Coughlin, J.M., Ishizuka, K., Kano, S.I., Edwards, J.A., Seifuddin, F.T., Shimano, M.A., et al., 2012.
also funded by the Women's Hospital Foundation from a grant through Neuropsychiatry Marked reduction of soluble superoxide dismutase-1 (SOD1) in cerebrospinal fluid of pa-
at Sheppard Pratt. tients with recent-onset schizophrenia. Mol. Psychiatry 18 (1), 10–11.

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
C.C. Watkins, S.R. Andrews / Schizophrenia Research 176 (2016) 14–22 21

Coyle, J.T., 2006. NMDA receptor and schizophrenia: a brief history. Schizophr. Bull. 38 (5), Job, D.E., Whalley, H.C., McIntosh, A.M., Owens, D.G., Johnstone, E.C., Lawrie, S.M., 2006. Grey
920–926. matter changes can improve the prediction of schizophrenia in subjects at high risk. BMC
Dalman, C., Allebeck, P., Gunnell, D., Harrison, G., Kristensson, K., Lewis, G., et al., 2008. Infections Med. 4, 29.
in the CNS during childhood and the risk of subsequent psychotic illness: a cohort study of Kawasaki, H., Iwamuro, S., 2008. Potential roles of histones in host defense as antimicrobial
more than one million Swedish subjects. Am. J. Psychiatry 165 (1), 59–65. agents. Infect. Disord. Drug Targets 8 (3), 195–205.
Dammann, O., Leviton, A., 1997. Maternal intrauterine infection, cytokines, and brain damage in Kenk, M., Selvanathan, T., Rao, N., Suridjan, I., Rusjan, P., Remington, G., et al., 2014. Imaging neu-
the preterm newborn. Pediatr. Res. 42 (1), 1–8. roinflammation in gray and white matter in schizophrenia: an in-vivo PET study with [18F]-
Dammann, O., Bueter, W., Leviton, A., Gressens, P., Dammann, C.E., 2008. Neuregulin-1: a poten- FEPPA. Schizophr. Bull. 41 (1), 85–93.
tial endogenous protector in perinatal brain white matter damage. Neonatology 93 (3), Khandaker, G.M., Zammit, S., Lewis, G., Jones, P.B., 2013. A population-based study of atopic dis-
182–187. orders and inflammatory markers in childhood before psychotic experiences in adoles-
Dieset, I., Haukvik, U.K., Melle, I., Rossberg, J.I., Ueland, T., Hope, S., et al., 2015. Association be- cence. Schizophr. Res. 152 (1), 139–145.
tween altered brain morphology and elevated peripheral endothelial markers — implica- Khandaker, G.M., Pearson, R.M., Zammit, S., Lewis, G., Jones, P.B., 2014a. Association of serum in-
tions for psychotic disorders. Schizophr. Res. 161 (2–3), 222–228. terleukin 6 and C-reactive protein in childhood with depression and psychosis in young
Doorduin, J., de Vries, E.F., Willemsen, A.T., de Groot, J.C., Dierckx, R.A., Klein, H.C., 2009. Neuro- adult life: a population-based longitudinal study. JAMA Psychiatry 71 (10), 1121–1128.
inflammation in schizophrenia-related psychosis: a PET study. J. Nucl. Med. 50 (11), Khandaker, G.M., Stochl, J., Zammit, S., Lewis, G., Jones, P.B., 2014b. Childhood Epstein–Barr Virus
1801–1807. infection and subsequent risk of psychotic experiences in adolescence: a population-based
Drago, A., Giegling, I., Schafer, M., Hartmann, A.M., Konte, B., Friedl, M., et al., 2014. Genome- prospective serological study. Schizophr. Res. 158 (1–3), 19–24.
wide association study supports the role of the immunological system and of the Kim, S., Zavitsanou, K., Gurguis, G., Webster, M.J., 2012. Neuropathology markers and pathways
neurodevelopmental processes in response to haloperidol treatment. Pharmacogenet. Ge- associated with molecular targets for antipsychotic drugs in postmortem brain tissues: ex-
nomics 24 (6), 314–319. ploration of drug targets through the Stanley Neuropathology Integrative Database. Eur.
Drexhage, R.C., Knijff, E.M., Padmos, R.C., Heul-Nieuwenhuijzen, L.v.d., Beumer, W., Versnel, Neuropsychopharmacol. 22 (10), 683–694.
M.A., et al., 2010. The mononuclear phagocyte system and its cytokine inflammatory net- Kohen, R., Nyska, A., 2002. Oxidation of biological systems: oxidative stress phenomena, antiox-
works in schizophrenia and bipolar disorder. Expert. Rev. Neurother. 10 (1), 59–76. idants, redox reactions, and methods for their quantification. Toxicol. Pathol. 30 (6),
Eaton, W.W., Pedersen, M.G., Atladottir, H.O., Gregory, P.E., Rose, N.R., Mortensen, P.B., 2010. The 620–650.
prevalence of 30 ICD-10 autoimmune diseases in Denmark. Immunol. Res. 47 (1–3), Kreisl, W.C., Jenko, K.J., Hines, C.S., Lyoo, C.H., Corona, W., Morse, C.L., et al., 2012. A genetic poly-
228–231. morphism for translocator protein 18 kDa affects both in vitro and in vivo radioligand bind-
Ellman, L.M., Yolken, R.H., Buka, S.L., Torrey, E.F., Cannon, T.D., 2009. Cognitive functioning prior ing in human brain to this putative biomarker of neuroinflammation. J. Cereb. Blood Flow
to the onset of psychosis: the role of fetal exposure to serologically determined influenza Metab. 33 (1), 53–58.
infection. Biol. Psychiatry 65 (12), 1040–1047. Kroken, R.A., Loberg, E.M., Dronen, T., Gruner, R., Hugdahl, K., Kompus, K., et al., 2014. A critical
Ellman, L.M., Deicken, R.F., Vinogradov, S., Kremen, W.S., Poole, J.H., Kern, D.M., et al., 2010. review of pro-cognitive drug targets in psychosis: convergence on myelination and inflam-
Structural brain alterations in schizophrenia following fetal exposure to the inflammatory mation. Front. Psychiatry 5, 11.
cytokine interleukin-8. Schizophr. Res. 121 (1–3), 46–54. Kundakovic, M., Chen, Y., Costa, E., Grayson, D.R., 2007. DNA methyltransferase inhibitors coor-
Emiliani, F.E., Sedlak, T.W., Sawa, A., 2014. Oxidative stress and schizophrenia: recent break- dinately induce expression of the human reelin and glutamic acid decarboxylase 67 genes.
throughs from an old story. Curr. Opin. Psychiatry 27 (3), 185–190. Mol. Pharmacol. 71 (3), 644–653.
Erhardt, S., Blennow, K., Nordin, C., Skogh, E., Lindstrom, L.H., Engberg, G., 2001. Kynurenic acid Kunz, M., Cereser, K.M., Goi, P.D., Fries, G.R., Teixeira, A.L., Fernandes, B.S., et al., 2011. Serum
levels are elevated in the cerebrospinal fluid of patients with schizophrenia. Neurosci. Lett. levels of IL-6, IL-10 and TNF-alpha in patients with bipolar disorder and schizophrenia: dif-
313 (1–2), 96–98. ferences in pro- and anti-inflammatory balance. Rev. Bras. Psiquiatr. 33 (3), 268–274.
Favrais, G., van de Looij, Y., Fleiss, B., Ramanantsoa, N., Bonnin, P., Stoltenburg-Didinger, G., et al., Laan, W., Grobbee, D.E., Selten, J.P., Heijnen, C.J., Kahn, R.S., Burger, H., 2010. Adjuvant aspirin
2011. Systemic inflammation disrupts the developmental program of white matter. Ann. therapy reduces symptoms of schizophrenia spectrum disorders: results from a random-
Neurol. 70 (4), 550–565. ized, double-blind, placebo-controlled trial. J. Clin. Psychiatry 71 (5), 520–527.
Fellerhoff, B., Laumbacher, B., Mueller, N., Gu, S., Wank, R., 2007. Associations between Le Hellard, S., Muhleisen, T.W., Djurovic, S., Ferno, J., Ouriaghi, Z., Mattheisen, M., et al., 2008.
Chlamydophila infections, schizophrenia and risk of HLA-A10. Mol. Psychiatry 12 (3), Polymorphisms in SREBF1 and SREBF2, two antipsychotic-activated transcription factors
264–272. controlling cellular lipogenesis, are associated with schizophrenia in German and Scandina-
Fillman, S.G., Cloonan, N., Miller, L.C., Weickert, C.S., 2013. Markers of inflammation in the pre- vian samples. Mol. Psychiatry 15 (5), 463–472.
frontal cortex of individuals with schizophrenia. Mol. Psychiatry 18 (2), 133. Leviton, A., Gressens, P., 2007. Neuronal damage accompanies perinatal white-matter damage.
Flatow, J., Buckley, P., Miller, B.J., 2013. Meta-analysis of oxidative stress in schizophrenia. Biol. Trends Neurosci. 30 (9), 473–478.
Psychiatry 74 (6), 400–409. Levkovitz, Y., Mendlovich, S., Riwkes, S., Braw, Y., Levkovitch-Verbin, H., Gal, G., et al., 2010. A
Fond, G., Hamdani, N., Kapczinski, F., Boukouaci, W., Drancourt, N., Dargel, A., et al., 2013. Effec- double-blind, randomized study of minocycline for the treatment of negative and cognitive
tiveness and tolerance of anti-inflammatory drugs' add-on therapy in major mental disor- symptoms in early-phase schizophrenia. J. Clin. Psychiatry 71 (2), 138–149.
ders: a systematic qualitative review. Acta Psychiatr. Scand. 129 (3), 163–179. Leweke, F.M., Gerth, C.W., Koethe, D., Klosterkotter, J., Ruslanova, I., Krivogorsky, B., et al., 2004.
Fudenberg, H.H., Whitten, H.D., Merler, E., Farmati, O., 1983. Is schizophrenia an immunologic Antibodies to infectious agents in individuals with recent onset schizophrenia. Eur. Arch.
receptor disorder? Med. Hypotheses 12 (1), 85–93. Psychiatry Clin. Neurosci. 254 (1), 4–8.
Garver, D.L., Tamas, R.L., Holcomb, J.A., 2003. Elevated interleukin-6 in the cerebrospinal fluid of Matthews, P.R., Harrison, P.J., 2011. A morphometric, immunohistochemical, and in situ hybrid-
a previously delineated schizophrenia subtype. Neuropsychopharmacology 28 (8), ization study of the dorsal raphe nucleus in major depression, bipolar disorder, schizophre-
1515–1520. nia, and suicide. J. Affect. Disord. 137 (1–3), 125–134.
Gattaz, W.F., Abrahao, A.L., Foccacia, R., 2004. Childhood meningitis, brain maturation and the Mayilyan, K.R., Arnold, J.N., Presanis, J.S., Soghoyan, A.F., Sim, R.B., 2006. Increased complement
risk of psychosis. Eur. Arch. Psychiatry Clin. Neurosci. 254 (1), 23–26. classical and mannan-binding lectin pathway activities in schizophrenia. Neurosci. Lett. 404
Gogtay, N., Lu, A., Leow, A.D., Klunder, A.D., Lee, A.D., Chavez, A., et al., 2008. Three-dimensional (3), 336–341.
brain growth abnormalities in childhood-onset schizophrenia visualized by using tensor- Meda, S.A., Ruano, G., Windemuth, A., O'Neil, K., Berwise, C., Dunn, S.M., et al., 2014. Multivariate
based morphometry. Proc. Natl. Acad. Sci. U. S. A. 105 (41), 15979–15984. analysis reveals genetic associations of the resting default mode network in psychotic bipo-
Hagberg, H., Gressens, P., Mallard, C., 2012. Inflammation during fetal and neonatal life: implica- lar disorder and schizophrenia. Proc. Natl. Acad. Sci. U. S. A. 111 (19), E2066–E2075.
tions for neurologic and neuropsychiatric disease in children and adults. Ann. Neurol. 71 Mehler, M.F., Kessler, J.A., 1998. Cytokines in brain development and function. Adv. Protein
(4), 444–457. Chem. 52, 223–251.
Hashimoto, M., Maekawa, M., Katakura, M., Hamazaki, K., Matsuoka, Y., 2014. Possibility of poly- Meisenzahl, E.M., Rujescu, D., Kirner, A., Giegling, I., Kathmann, N., Leinsinger, G., et al., 2001. As-
unsaturated fatty acids for the prevention and treatment of neuropsychiatric illnesses. sociation of an interleukin-1beta genetic polymorphism with altered brain structure in pa-
J. Pharmacol. Sci. 124 (3), 294–300. tients with schizophrenia. Am. J. Psychiatry 158 (8), 1316–1319.
Hayes, L.N., Severance, E.G., Leek, J.T., Gressitt, K.L., Rohleder, C., Coughlin, J.M., et al., 2014. In- Meyer, U., 2011. Anti-inflammatory signaling in schizophrenia. Brain Behav. Immun. 25 (8),
flammatory molecular signature associated with infectious agents in psychosis. Schizophr. 1507–1518.
Bull. 40 (5), 963–972. Meyer, U., Weiner, I., McAlonan, G.M., Feldon, J., 2010. The neuropathological contribution of
Hercher, C., Chopra, V., Beasley, C.L., 2014. Evidence for morphological alterations in prefrontal prenatal inflammation to schizophrenia. Expert. Rev. Neurother. 11 (1), 29–32.
white matter glia in schizophrenia and bipolar disorder. J. Psychiatry Neurosci. 39 (6), Miller, B.J., Buckley, P., Seabolt, W., Mellor, A., Kirkpatrick, B., 2011. Meta-analysis of cytokine al-
376–385. terations in schizophrenia: clinical status and antipsychotic effects. Biol. Psychiatry 70 (7),
Huerta, I., McCullumsmith, R.E., Haroutunian, V., Gimenez-Amaya, J.M., Meador-Woodruff, J.H., 663–671.
2006. Expression of excitatory amino acid transporter interacting protein transcripts in Monji, A., Kato, T., Kanba, S., 2009. Cytokines and schizophrenia: microglia hypothesis of schizo-
the thalamus in schizophrenia. Synapse 59 (7), 394–402. phrenia. Psychiatry Clin. Neurosci. 63 (3), 257–265.
Hwang, Y., Kim, J., Shin, J.Y., Kim, J.I., Seo, J.S., Webster, M.J., et al., 2013. Gene expression Mousa, A., Seiger, A., Kjaeldgaard, A., Bakhiet, M., 1999. Human first trimester forebrain cells ex-
profiling by mRNA sequencing reveals increased expression of immune/ press genes for inflammatory and anti-inflammatory cytokines. Cytokine 11 (1), 55–60.
inflammation-related genes in the hippocampus of individuals with schizophrenia. Muller, N., 2014. Immunology of schizophrenia. Neuroimmunomodulation 21 (2–3), 109–116.
Transl Psychiatry 3, e321. Muller, N., Schwarz, M.J., 2010. Immune system and schizophrenia. Curr. Immunol. Rev. 6 (3),
Insel, T.R., 2011. Rethinking schizophrenia. Nature 468 (7321), 187–193. 213–220.
Jaaro-Peled, H., Ayhan, Y., Pletnikov, M.V., Sawa, A., 2009a. Review of pathological hallmarks of Muller, N., Ulmschneider, M., Scheppach, C., Schwarz, M.J., Ackenheil, M., Moller, H.J., et al., 2004.
schizophrenia: comparison of genetic models with patients and nongenetic models. COX-2 inhibition as a treatment approach in schizophrenia: immunological considerations
Schizophr. Bull. 36 (2), 301–313. and clinical effects of celecoxib add-on therapy. Eur. Arch. Psychiatry Clin. Neurosci. 254
Jaaro-Peled, H., Hayashi-Takagi, A., Seshadri, S., Kamiya, A., Brandon, N.J., Sawa, A., 2009b. (1), 14–22.
Neurodevelopmental mechanisms of schizophrenia: understanding disturbed postnatal Muller, N., Krause, D., Dehning, S., Musil, R., Schennach-Wolff, R., Obermeier, M., et al., 2010.
brain maturation through neuregulin-1-ErbB4 and DISC1. Trends Neurosci. 32 (9), Celecoxib treatment in an early stage of schizophrenia: results of a randomized, double-
485–495. blind, placebo-controlled trial of celecoxib augmentation of amisulpride treatment.
Jia, P., Wang, L., Meltzer, H.Y., Zhao, Z., 2010. Common variants conferring risk of schizophrenia: Schizophr. Res. 121 (1–3), 118–124.
a pathway analysis of GWAS data. Schizophr. Res. 122 (1–3), 38–42. Nathan, C., Shiloh, M.U., 2000. Reactive oxygen and nitrogen intermediates in the relationship
Job, D.E., Whalley, H.C., Johnstone, E.C., Lawrie, S.M., 2005. Grey matter changes over time in between mammalian hosts and microbial pathogens. Proc. Natl. Acad. Sci. U. S. A. 97
high risk subjects developing schizophrenia. Neuroimage 25 (4), 1023–1030. (16), 8841–8848.

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
22 C.C. Watkins, S.R. Andrews / Schizophrenia Research 176 (2016) 14–22

Noto, C., Ota, V.K., Gouveia, E.S., Rizzo, L.B., Spindola, L.M., Honda, P.H., et al., 2014. Effects Sommer, I.E., van Westrhenen, R., Begemann, M.J., de Witte, L.D., Leucht, S., Kahn, R.S., 2013. Ef-
of risperidone on cytokine profile in drug naive first episode psychosis. Int. ficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia:
J. Neuropsychopharmacol. 18 (4), 1–8. an update. Schizophr. Bull. 40 (1), 181–191.
Pathmanandavel, K., Starling, J., Dale, R.C., Brilot, F., 2013. Autoantibodies and the immune hy- Sorensen, H.J., Mortensen, E.L., Parnas, J., Mednick, S.A., 2006. Premorbid neurocognitive func-
pothesis in psychotic brain diseases: challenges and perspectives. Clin. Dev. Immunol. tioning in schizophrenia spectrum disorder. Schizophr. Bull. 32 (3), 578–583.
2013, 257184. Sorensen, H.J., Mortensen, E.L., Reinisch, J.M., Mednick, S.A., 2009. Association between prenatal
Patterson, P.H., 2009. Immune involvement in schizophrenia and autism: etiology, pathology exposure to bacterial infection and risk of schizophrenia. Schizophr. Bull. 35 (3), 631–637.
and animal models. Behav. Brain Res. 204 (2), 313–321. Spangaro, M., Bosia, M., Zanoletti, A., Bechi, M., Cocchi, F., Pirovano, A., et al., 2012. Cognitive dys-
Pesce, M., Ferrone, A., Rizzuto, A., Tatangelo, R., Iezzi, I., Ladu, S., et al., 2014. The SHP-1 expres- function and glutamate reuptake: effect of EAAT2 polymorphism in schizophrenia.
sion is associated with cytokines and psychopathological status in unmedicated first epi- Neurosci. Lett. 522 (2), 151–155.
sode schizophrenia patients. Brain Behav. Immun. 41, 251–260. Stan, A.D., Ghose, S., Zhao, C., Hulsey, K., Mihalakos, P., Yanagi, M., et al., 2014. Magnetic reso-
Poels, E.M., Kegeles, L.S., Kantrowitz, J.T., Slifstein, M., Javitt, D.C., Lieberman, J.A., et al., 2013. Im- nance spectroscopy and tissue protein concentrations together suggest lower glutamate
aging glutamate in schizophrenia: review of findings and implications for drug discovery. signaling in dentate gyrus in schizophrenia. Mol. Psychiatry 20 (4), 433–439.
Mol. Psychiatry 19 (1), 20–29. Steen, R.G., Mull, C., McClure, R., Hamer, R.M., Lieberman, J.A., 2006. Brain volume in first-
Potvin, S., Stip, E., Sepehry, A.A., Gendron, A., Bah, R., Kouassi, E., 2008. Inflammatory cytokine episode schizophrenia: systematic review and meta-analysis of magnetic resonance imag-
alterations in schizophrenia: a systematic quantitative review. Biol. Psychiatry 63 (8), ing studies. Br. J. Psychiatry 188, 510–518.
801–808. Stefansson, H., Ophoff, R.A., Steinberg, S., Andreassen, O.A., Cichon, S., Rujescu, D., et al., 2009.
Prasad, K.M., Upton, C.H., Nimgaonkar, V.L., Keshavan, M.S., 2014. Differential susceptibility of Common variants conferring risk of schizophrenia. Nature 460 (7256), 744–747.
white matter tracts to inflammatory mediators in schizophrenia: an integrated DTI study. Steiner, J., Bernstein, H.G., Bielau, H., Farkas, N., Winter, J., Dobrowolny, H., et al., 2008a. S100B-
Schizophr. Res. 161 (1), 119–125. immunopositive glia is elevated in paranoid as compared to residual schizophrenia: a mor-
Purcell, S.M., Wray, N.R., Stone, J.L., Visscher, P.M., O'Donovan, M.C., Sullivan, P.F., et al., 2009. phometric study [Research Support, Non-U.S. Gov't] J. Psychiatr. Res. 42 (10), 868–876.
Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Na- Steiner, J., Bielau, H., Brisch, R., Danos, P., Ullrich, O., Mawrin, C., et al., 2008b. Immunological as-
ture 460 (7256), 748–752. pects in the neurobiology of suicide: elevated microglial density in schizophrenia and de-
Radewicz, K., Garey, L.J., Gentleman, S.M., Reynolds, R., 2000. Increase in HLA-DR immunoreac- pression is associated with suicide. J. Psychiatr. Res. 42 (2), 151–157.
tive microglia in frontal and temporal cortex of chronic schizophrenics. J. Neuropathol. Exp. Steiner, J., Bogerts, B., Sarnyai, Z., Walter, M., Gos, T., Bernstein, H.G., et al., 2011a. Bridging the
Neurol. 59 (2), 137–150. gap between the immune and glutamate hypotheses of schizophrenia and major depres-
Radomska, K.J., Halvardson, J., Reinius, B., Lindholm Carlstrom, E., Emilsson, L., Feuk, L., et al., sion: potential role of glial NMDA receptor modulators and impaired blood–brain barrier
2013. RNA-binding protein QKI regulates Glial fibrillary acidic protein expression in integrity. World J. Biol. Psychiatry 13 (7), 482–492.
human astrocytes. Hum. Mol. Genet. 22 (7), 1373–1382. Steiner, J., Walter, M., Gos, T., Guillemin, G.J., Bernstein, H.G., Sarnyai, Z., et al., 2011b. Severe de-
Rapoport, J.L., Giedd, J.N., Gogtay, N., 2012. Neurodevelopmental model of schizophrenia: update pression is associated with increased microglial quinolinic acid in subregions of the anterior
2012. Mol. Psychiatry 17 (12), 1228–1238. cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission?
Ripke, S., O'Dushlaine, C., Chambert, K., Moran, J.L., Kahler, A.K., Akterin, S., et al., 2013. Genome- J. Neuroinflammation 8, 94.
wide association analysis identifies 13 new risk loci for schizophrenia. Nat. Genet. 45 (10), Stevens, J.R., 1983. Pathophysiology of schizophrenia. Clin. Neuropharmacol. 6 (2), 77–90.
1150–1159. Suvisaari, J.M., Haukka, J.K., Tanskanen, A.J., Lonnqvist, J.K., 1999. Decline in the incidence of
Rossler, W., Salize, H.J., van Os, J., Riecher-Rossler, A., 2005. Size of burden of schizophrenia and schizophrenia in Finnish cohorts born from 1954 to 1965. Arch. Gen. Psychiatry 56 (8),
psychotic disorders. Eur. Neuropsychopharmacol. 15 (4), 399–409. 733–740.
Rothermundt, M., Ohrmann, P., Abel, S., Siegmund, A., Pedersen, A., Ponath, G., et al., 2007. Glial Swerdlow, N.R., van Bergeijk, D.P., Bergsma, F., Weber, E., Talledo, J., 2009. The effects of
cell activation in a subgroup of patients with schizophrenia indicated by increased S100B memantine on prepulse inhibition. Neuropsychopharmacology 34 (7), 1854–1864.
serum concentrations and elevated myo-inositol. Prog. Neuropsychopharmacol. Biol. Psy- Takano, A., Arakawa, R., Ito, H., Tateno, A., Takahashi, H., Matsumoto, R., et al., 2010. Peripheral
chiatry 31 (2), 361–364. benzodiazepine receptors in patients with chronic schizophrenia: a PET study with
Rowland, L.M., Kontson, K., West, J., Edden, R.A., Zhu, H., Wijtenburg, S.A., et al., 2012. In vivo [11C]DAA1106. Int. J. Neuropsychopharmacol. 13 (7), 943–950.
measurements of glutamate, GABA, and NAAG in schizophrenia. Schizophr. Bull. 39 (5), Tang, B., Capitao, C., Dean, B., Thomas, E.A., 2012. Differential age- and disease-related effects on
1096–1104. the expression of genes related to the arachidonic acid signaling pathway in schizophrenia.
Sathyasaikumar, K.V., Stachowski, E.K., Wonodi, I., Roberts, R.C., Rassoulpour, A., McMahon, R.P., Psychiatry Res. 196 (2–3), 201–206.
et al., 2010. Impaired kynurenine pathway metabolism in the prefrontal cortex of individ- Tomasik, J., Rahmoune, H., Guest, P.C., Bahn, S., 2014. Neuroimmune biomarkers in schizophre-
uals with schizophrenia. Schizophr. Bull. 37 (6), 1147–1156. nia. Schizophr. Res. (pii: S0920-9964(14)00382-x. doi: [Epub ahead of print]).
Schwarcz, R., Rassoulpour, A., Wu, H.Q., Medoff, D., Tamminga, C.A., Roberts, R.C., 2001. In- Torrey, E.F., Miller, J., Rawlings, R., Yolken, R.H., 1997. Seasonality of births in schizophrenia and
creased cortical kynurenate content in schizophrenia. Biol. Psychiatry 50 (7), 521–530. bipolar disorder: a review of the literature. Schizophr. Res. 28 (1), 1–38.
Schwieler, L., Larsson, M.K., Skogh, E., Kegel, M.E., Orhan, F., Abdelmoaty, S., et al., 2014. Increased Upthegrove, R., Manzanares-Teson, N., Barnes, N.M., 2014. Cytokine function in medication-
levels of IL-6 in the cerebrospinal fluid of patients with chronic schizophrenia — significance naive first episode psychosis: a systematic review and meta-analysis. Schizophr. Res. 155
for activation of the kynurenine pathway. J. Psychiatry Neurosci. 39 (6), 140126. (1–3), 101–108.
Selten, J.P., Frissen, A., Lensvelt-Mulders, G., Morgan, V.A., 2009. Schizophrenia and 1957 pan- van Os, J., Kapur, S., 2009. Schizophrenia. Lancet 374 (9690), 635–645.
demic of influenza: meta-analysis. Schizophr. Bull. 36 (2), 219–228. Watkins, C.C., Sawa, A., Pomper, M.G., 2014. Glia and immune cell signaling in bipolar disorder:
Severance, E.G., Yolken, R.H., Eaton, W.W., 2014a. Autoimmune diseases, gastrointestinal disor- insights from neuropharmacology and molecular imaging to clinical application. Transl Psy-
ders and the microbiome in schizophrenia: more than a gut feeling. Schizophr. Res. (pii: chiatry 4, e350.
S0920-9964(14)00319-3. doi: [Epub ahead of print]). Wierzba-Bobrowicz, T., Lewandowska, E., Lechowicz, W., Stepien, T., Pasennik, E., 2005. Quanti-
Severance, E.G., Gressitt, K.L., Buka, S.L., Cannon, T.D., Yolken, R.H., 2014b. Maternal complement tative analysis of activated microglia, ramified and damage of processes in the frontal and
C1q and increased odds for psychosis in adult offspring. Schizophr. Res. 159 (1), 14–19. temporal lobes of chronic schizophrenics. Folia Neuropathol. 43 (2), 81–89.
Shi, L., Tu, N., Patterson, P.H., 2005. Maternal influenza infection is likely to alter fetal brain de- Yudofsky, S.C., 2009. Contracting schizophrenia: lessons from the influenza epidemic of
velopment indirectly: the virus is not detected in the fetus. Int. J. Dev. Neurosci. 23 (2–3), 1918–1919. JAMA 301 (3), 324–326.
299–305. Zheng, L.T., Hwang, J., Ock, J., Lee, M.G., Lee, W.H., Suk, K., 2008. The antipsychotic spiperone at-
Smesny, S., Kunstmann, C., Kunstmann, S., Willhardt, I., Lasch, J., Yotter, R.A., et al., 2010. Phos- tenuates inflammatory response in cultured microglia via the reduction of proinflammato-
pholipase A(2) activity in first episode schizophrenia: associations with symptom severity ry cytokine expression and nitric oxide production. J. Neurochem. 107 (5), 1225–1235.
and outcome at week 12. World J. Biol. Psychiatry 12 (8), 598–607.

Downloaded for Perpus FK UNPAD 02 (perpus.fkunpad02@gmail.com) at Padjadjaran University from ClinicalKey.com by Elsevier on May 26, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.

You might also like