Neurotox Res DOI 10.1007/s12640-014-9473-0

REVIEW

Neurodegenerative Aspects in Vulnerability to Schizophrenia Spectrum Disorders Trevor Archer • Serafino Ricci • Danilo Garcia Max Rapp Ricciardi



Received: 8 January 2014 / Revised: 21 April 2014 / Accepted: 21 April 2014 Ó Springer Science+Business Media New York 2014

Abstract The neurodegenerative and neurotoxic aspects of schizophrenia and/or psychosis involve genetic, epigenetic, and neurotoxic propensities that impinge upon both the symptom domains and the biomarkers of the disorder, involving cellular apoptosis/excitotoxicity, increased reactive oxygen species formation, viral and bacterial infections, anoxic birth injury, maternal starvation, drugs of abuse, particularly cannabis, metabolic accidents, and other chemical agents that disrupt normal brain development or the integrity of brain tissues. Evidence for premorbid and prodromal psychotic phases, aspects of neuroimaging, dopamine, and psychosis vulnerability, and perinatal aspects provide substance for neurodegenerative influences. Not least, the agencies of antipsychotic contribute to the destructive spiral that disrupts normal structure and function. The etiopathogenesis of psychosis is distinguished also by disruptions of the normal functioning of the neurotrophins, in particular brain-derived neurotrophic factor, dyskinesic aspects, immune system disturbances, and metabolic aspects. Whether detrimental to neurodevelopment or tissue-destructive, or an acceleration of

T. Archer (&)  D. Garcia  M. R. Ricciardi Department of Psychology, University of Gothenburg, Box 500, 40530 Go¨thenburg, Sweden e-mail: [email protected] T. Archer  D. Garcia  M. R. Ricciardi Network for Empowerment and Well-Being, Lund, Sweden S. Ricci Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University, Rome, Italy D. Garcia Center for Ethics, Law and Mental Health (CELAM), University of Gothenburg, Go¨thenburg, Sweden

neurotoxic pathways, the notion of neurodegeneration in the pathophysiology of schizophrenia spectrum and psychotic disorders continues to gather momentum. Keywords Apoptosis  Infection  Neurotoxin  Neurogenesis  Dopamine  Cannabis  Neurotrophins  Metabolism  Immune system  Neurodevelopment

Introduction The status of the neurodegenerative influence in the development of schizophrenic psychoses has not been considered straightforward and perhaps may even seem tenuous. Nevertheless, the plausibility of evidence from laboratory animal models studies, neuroimaging studies, and postmortem assays together with a neurodevelopmental viewpoint of disorder etiopathogenesis does imply that neurodegenerative processes may contribute to heightened predisposition and susceptibility to the precipitation of symptom domains through a staging notion (Archer et al. 2010a, b). For example, in a five-day repeated ketamine administration animal model of schizophrenia, Faizi et al. (2014) observed increased reactive oxygen species production, mitochondrial membrane potential collapse, mitochondrial swelling, and cytochrome c release in mitochondria of schizophrenia group. Genetic risk factors may impact upon or disrupt developmental trajectories (Dauvermann et al. 2012, 2013; Sullivan et al. 2003). There is much support for the notion that schizophrenia spectrum disorders have their origin during gestation and/or in early infancy (Graff et al. 2011; Sawa and Snyder 2002). Insults, during the early infancy or perinatal period, or even during the course of early-to-late childhood and adolescence, are implicated in the etiopathogenesis of the marked structural

123

Neurotox Res

and functional abnormalities that distinguish the symptoms of disorder (Cannon et al. 2002a, b; Fontes et al. 2011; Geddes et al. 1999). The eventual contribution of neurodegenerative processes affecting the disorder must take into account the plethora of interactive factors and agents that include inherited and acquired attributes (Elia et al. 2011; Jablonka and Raz 2009). The disorder pathophysiology may involve the influences of several contributory process, including cellular apoptosis/excitotoxicity, viral and bacterial infections, anoxic birth injury, maternal starvation, drug abuse in particular cannabis, trauma, and other conditions exerting detrimental effects on neurogenesis. Both neurogenetic and epigenetic forces arising from gene-environment interactions affecting developmental trajectories impart levels of predisposing vulnerability that may or may not lead to the manifestation of schizophrenic symptoms (Gratacos et al. 2007; Hoenicka et al. 2010), or those of related conditions (Bergman et al. 2011; Jacob et al. 2010). Whether or not the neurodegenerative aspect of the disorder may be neurogenetic or epigenetic, there compelling evidence exists for expressions of the disruptive influences affecting normal development. In the present treatise, the available evidence supporting the notion of neurodegenerative impacts of liability to psychosis is reviewed critically. Recent studies showing that psychosis is usually predated by a prodromal phase characterized by dramatic neurobiological and psychopathological changes are reviewed. Then, the imaging findings supporting neurodegenerative impacts in the development of psychosis with some specific insight into molecular and structural studies are examined. Following this, dopamine and psychosis vulnerability, perinatal agents, cannabis abuse, neurotrophin disruption, dyskinetic movement, brain insulin dysregulation, and cytokine imbalance aspects are discussed. Throughout, it ought to be considered that (i) histopathological features are suggestive of apoptotic rather than necrotic processes, and (ii) loss of structure and function in disorder progression, and (iii) the presence of infections and/or adverse environmental conditions that perturb the course of normal development resulting in unsuitable developmental trajectories.

The Evidence for a Prodromal Psychotic Phases Over the recent two decades, accumulating evidence has showed that psychosis onset is usually preceded by a prodromal phase (Fusar-Poli et al. 2013a). This period is spanning childhood and adolescence and is characterized by profound neurodevelopmental changes. Subjects experiencing putative prodromal signs or symptoms of psychosis have a greater risk of developing the disease (from

123

Table 1 Putative neurodegenerative factors influencing vulnerability and altered developmental trajectory Imaging Volumetric loss Loss of connectivity Catecholamine metabolite alterations Tissue loss Thalamic gray matter Glutamate-transmission integrity Temporal lobe and Inferior frontal gyrus Dopamine Presynaptic alterations Synthesis and turnover Perinatal Ventral HPL lesion MAM neurotoxicity Adverse childhood experience Infection Neurotrophin BDNF loss/reduction Insulin Brain insulin dysregulation Immune system disturbance Cytokine imbalance HPF hippocampus, MAM methylazoxymethanol, BDNF brainderived neurotrophic factor

18 % at 6 months up to 36 % after 3 years) over time (Fusar-Poli et al. 2012a). The majority of the subjects who will develop a psychotic disorder later on will transit toward schizophrenic psychoses rather than to affective psychoses (Fusar-Poli et al. 2013b). Furthermore, the presence of putative prodromal symptoms per se is associated with significant psychosocial impairment, decreased quality of life, and subtle cognitive deficits (Borgwardt et al. 2006, 2010, 2011; Bu¨schlen et al. 2011; Fusar-Poli et al. 2011a, b, c, 2012b; Haller et al. 2009; Koutsouleris et al. 2012; Mechelli et al. 2011; Rothlisberger et al. 2012; Shenton et al. 2001; Smieskova et al. 2012a, b). At a neurobiological level, the prodromal psychotic phase is characterized by significant alterations in the structure (Buehlmann et al. 2010; Crossley et al. 2009; Fusar-Poli et al. 2007, 2011a, b, c; Smieskova et al. 2010a, b, 2012c), function (Fusar-Poli et al. 2010, 2011a, b, c; Fusar-Poli and Meyer-Lindenberg 2013a), connectivity (Fusar-Poli and Meyer-Lindenberg 2013b), and neurochemistry (Howes et al. 2007; Lyon et al. 2011; McGuire et al. 2008; MeyerLindenberg et al. 2002; Miyake et al. 2011) of the brain (Laruelle et al. 1996). The presence of a subtle prepsychotic phase is the clinical ground on which neurodegenerative factors can be tested. As evident from genetic risk

Neurotox Res

factors and risk factors linked to childhood ill health/ adversity, the developmental trajectories of the afflicted individuals are not optimal (Arango and Fraguas 2013). Some of the stronger pieces of evidence supporting the notion of neurodegenerative aspects in liability to psychosis (cf. Pasternak et al. 2012) are considered below (Table 1).

Imaging Aspects At first sight, the accumulated evidence for progressive volumetric loss, e.g., in cortical volume/thickness (Goldman et al. 2009; Honea et al. 2008) and loss of functional connectivity (Meyer-Lindenberg et al. 2005; Yu et al. 2011), argues for the aspect of neurodegeneration in schizophrenia; yet, the possible impacts of peristaltic and environmental agents must needs to be understood prior to a reliable conclusion (Meyer-Lindenberg 2011). For example, Goto et al. (2010) compared 18 patients (nine males, nine females; age range: 13–52 years) with 18 healthy volunteers (nine males, nine females; age range: 15–49 years) with no current or past psychiatric history, using magnetic resonance spectroscopy (MRS). They found that levels of N-acetylaspartate/Cr in the left basal ganglia and parieto-occipital lobe, but not in the frontal lobe, were significantly lower in patients with first-episode schizophrenia psychosis than in control subjects. No differences were observed between the serum brain-derived neurotrophic factor (BDNF) levels of patients with firstepisode schizophrenia psychosis and the healthy control subjects. The plasma levels of catecholamine metabolites, plasma 3-methoxy-4-hydroxyphenylglycol, but not homovanillic acid, were significantly lower in the patients with first-episode psychosis than in control subjects. In addition, a significantly positive correlation was observed between the levels of N-acetylaspartate/Cr of the left basal ganglia and plasma MHPG in all the subjects. The authors concluded that brain N-acetylaspartate levels in the left basal ganglia and plasma 3-methoxy-4-hydroxyphenylglycol levels were significantly reduced at the first-episode of schizophrenia psychosis, indicating that neurodegeneration via noradrenergic neurons might be associated with the initial progression of the disease. Increased prefrontal gyrification (cortical folding) and increased local short range prefrontal connectivity and reduced long range connectivity have been observed in highgenetic risk subjects (Dauvermann et al. 2012); gyrification and connectivity are disrupted in Schizopsychotic disorders. Dauvermann et al. (2013) have found that Bayesian Model Selection analysis has demonstrated significantly lower connection strength of the thalamocortical connection with nonlinear modulation from the mediodorsal thalamus in

high-risk subjects with psychotic symptoms, and those individuals who subsequently developed schizophrenia.

Aspects of Tissue Loss Regional loss of glutamate and glutamine in schizophrenic patients (Tayoshi et al. 2009) may offer evidence of an excitotoxic process in the disorder pathophysiology. The loss of parvalbumin-containing cells, an observation that has been reported consistently in postmortem analysis of the brains of schizophrenic patients) following NMDA receptor blockade offers support for this notion (Adell et al. 2012; Dean et al. 2011). Aoyama et al. (2011) have demonstrated that the loss of thalamic glutamate in the brain of schizophrenia patients correlated significantly with loss a gray matter in the middle and inferior frontal gyrus and temporal pole. Thalamic gray matter and glutamate losses were observed in individuals at risk for developing schizophrenia (see also Aoyama et al. 2013; Stone et al. 2009). Correlations between glutamate and glutamine ratios in the frontal cortices of adolescents at risk for the disorder and performance scores on the Global Assessment of Functioning Scale have been observed (Tibbo et al. 2004). It has been suggested that the elevated glutamine levels in never-treated patients followed by the decreased level of thalamic glutamine and gray-matter loss in connected regions may be indicative of either neurodegeneration or a plastic response to reduced subcortical activity (The´berge et al. 2007). There appears to be a consensus of sorts for an excitotoxic contribution primarily in earlystage schizophrenia (Bustillo et al. 2010). Glutamatergic alterations and loss of cortical gray matter originating from basal ganglia-corticothalamic circuits may be the expressions of excitotoxicity through these regions. First-episode schizophrenic patients have exhibited increased membrane breakdown in a magnetic resonance spectroscopy by applying labeled phosphorous (Miller et al. 2009). Takei et al. (2013) have demonstrated reduced functional performance activation in both the temporal lobes and the right inferior frontal gyrus during task performance (measured by mean [oxyhemoglobin] changes). Reduced activation in the left temporal lobe was negatively correlated with the Positive and Negative Syndrome Scale disorganization and negative symptoms sub-scores; right inferior frontal gyrus activation was negatively correlated with illness duration, Positive and Negative Syndrome Scale disorganization, and negative symptom sub-scores. Their results imply that brain dysfunction in schizophrenia during a conversation task is related to functional deficits in both the temporal lobes and the right inferior frontal gyrus with manifestation primarily in the form of disorganized thinking and negative symptomatology.

123

Neurotox Res

Dopamine and Psychosis Vulnerability Over the past two decades, new neurochemical imaging techniques have enabled the subcortical dopaminergic system to be carefully investigated in vivo (McGuire et al. 2008). In particular, the development of new radiotracers combined with single-photon emission computed tomography (SPECT) or positron emission tomography (PET) techniques has allowed accurate investigations of striatal presynaptic dopaminergic alterations in schizophrenia (for reviews see Howes et al. 2007; Lyon et al. 2011; Miyake et al. 2011). Different indexes of presynaptic dopamine neurotransmission such as dopamine synthesis capacity studies (Meyer-Lindenberg et al. 2002) and dopamine release (Laruelle et al. 1996), or depletion (Kegeles et al. 2010) studies have suggested significant alterations in the striatal presynaptic dopaminergic function in schizophrenia. For the sake of the present discussions, it ought to be noted, however, that elevated dopamine synthesis and release is also seen in subjects at risk for schizophrenia (Huttunen et al. 2008) and in the prodromal state (Mechelli et al. 2011), suggesting that they are part of the risk architecture of schizophrenia. The original neurodegenerative model of schizophrenia proposed that functional alterations in dopamine neurotransmission are secondary to a change in the number or density of striatal dopamine terminals over the course of the illness (Lieberman et al. 1990, 1997). Two recent meta-analyses have recently tested this hypothesis (Fusar-Poli and Meyer-Lindenberg 2013a, b). The authors found that dopamine synthesis capacity was increased of 14 % in patients with schizophrenia as compared to controls, while there were no differences in structural indexes of neuronal integrity (such as measures of Dopamine Active Transporter). The functional alterations observed during the psychosis onset can be related to a number of environmental factors such as perinatal insults, reviewed below here.

Perinatal Aspects Several perinatal insults have been shown to induce schizophrenia-like symptoms in laboratory setting (Archer 2010; Powell et al. 2012). Bilateral excitotoxin-induced lesions, administering ibotenic acid to the ventral hippocampus typically to 7-day-old rat pups, induce disruptions of prepulse inhibition (PPI) to the acoustic startle response, a model of sensory gating in rodents, hyper-responsiveness to stressful stimuli, supersensitivity to dopamine (DA) agonists, and N-methyl-D-aspartic acid (NMDA) antagonists (Lipska et al. 1992, 1995, 2001); additionally, persistent deficits in working memory and spatial navigational learning (O’Donnell et al. 2002), together with hyper-

123

responsiveness to stimulation of the ventral tegmental area (Lillrank et al. 1999; Lipska and Weinberger 1994) have all been observed in neonatal ventral hippocampus-lesioned rats. Prenatal administration of methylazoxymethanol (MAM) disrupted early brain development with structural and functional abnormalities in the cortex and hippocampus, many of which bear similarities to schizophrenia, expressed in the adult rats (Flagstad et al. 2005; Lodge and Grace 2008; Mohammed et al. 1986a, b; Moore et al. 2006). The utility of prenatal MAM as an animal model of the disorder highlights the neurodegenerative aspect (Chen and Hillman 1986), particularly with regard to the neurocognitive deficits (Fiore et al. 2004; Gourevitch et al. 2004; Lee et al. 2011; Leng et al. 2005). Similar to patients presenting cognitive deficit profiles in schizophrenia spectrum disorder (Maat et al. 2012), MAM-treated marked deficits on an attentional-shift task (analogous to the Wisconsin Card Sorting Task), and on a differential reinforcement of low rate of responding (DRL-20 performance, analogous to continuous performance) task (Featherstone et al. 2007). Prenatal MAM administration disrupts early cortical development causing deficits in medial prefrontal functions at adult ages (Goto and Grace 2006). These effects of the neurotoxin interrupt normal neuronal development, stress and immune response systems, and signal transduction mechanisms (Ciani et al. 2003; Ferrer et al. 1997; Lafarga et al. 1997; Rice and Barone 2000). Traumatic/adverse experiences during infancy and early childhood compromise healthy brain development in several domains, including cognitive, emotional, and motor. Traumatic environmental confrontation during early neurodevelopment, such as parental loss (maternal separation), that induces a marked provocation of the hypothalamic– pituitary–adrenal (HPA) axis was found to cause schizophrenia-like, e.g., impairments of sensory-gating, symptoms in the rats as adults (Ellenbroek et al. 1998), as well as other expression of the disorder (Furukawa et al. 1998). Rosenberg et al. (2007) surveyed 569 adults presenting schizophrenia with regard adverse childhood events (including physical abuse, sexual abuse, parental mental illnesses, loss of a parent, parental separation or divorce, witnessing domestic violence, and foster or kinship care), evaluating the relationships between cumulative exposure to these events and psychiatric, physical, and functional outcomes. They found that increased exposure to adverse childhood events was strongly related to psychiatric problems in both cognitive and emotional domains (suicidal thinking, hospitalizations, distress, and posttraumatic stress disorder), substance abuse, physical health problems (HIV infection), medical service utilization (physician visits), and poor social and daily care functioning (homelessness or criminal justice involvement). Ellman et al. (2009) observed genetic and/or environmental factors linked to

Neurotox Res

psychosis that caused fetal brains to be particularly vulnerable to the effects of influenza B, leading to poorer cognitive performance even before symptom onset. Similarly but related to sorrow, Morgan et al. (2007) observed that separation from, and/or the death of, one parent prior to the age of 16 were both strongly associated with a twoto three-fold increases in the risk of psychosis. It was shown that the strength of these associations were similar for White British and Black Caribbean (but not for Black African) subjects, although separation from (but not death of) a parent was more common among Black Caribbean controls than White British controls. Finally, a 10-year cohort of high secure hospital patients who had either a personality disorder or schizophrenia found a rate of childparent separation of 178/289 (62 %) in the schizophrenia group compared with a rate of 119/147 (81 %) in the personality disorder group of patients that had been separated from one or both parents before the age of 16 years (Pert et al. 2004). Chew et al. (2013) imply that cellular abnormalities extend to glial cells since oligodendroglial, microglial, and astrocyte activity alternations in SSDs are associated with neonatal brain injury.

Neurotrophin Disruption Brain-derived neurotrophic factor is essential for the maintenance of functional neurons, regulating growth, differentiation, synaptic connectivity, neurorepair, and longevity (Altar et al. 1997; Niu and Yip 2011). BDNF, central for the survival and differentiation of midbrain dopamine neurons (Hyman et al. 1991), and phenotypic differentiation of locus coeruleus noradrenergic neurons (Traver et al. 2006), has emerged as an important biomarker for schizophrenia spectrum disorder (Favalli et al. 2012; Pae et al. 2012; Zhang et al. 2012). In 63 patients presenting schizophrenia compared to 52 age- and sex-matched healthy controls were examined with neuropsychological tests, Niitsu et al. (2011) found that although there were no significant differences in serum BDNF levels between normal controls and schizophrenic patients, serum BDNF levels for normal controls, but not schizophrenic patients, showed negative correlations with verbal working memory. On the other hand, serum BDNF levels of schizophrenic patients indicated positive correlations with the scores of the Scale for the Assessment of Negative Symptoms (SANS) and the Information subtest scores of Wechsler Adult Intelligence Scale Revised (WAIS-R). Serum BDNF levels were related with the impairment of verbal working memory and negative symptoms in patients with schizophrenia. In 22 acute schizo-

phrenic patients and 22 age-matched healthy volunteers, Lee et al. (2011) observed significantly reduced serum levels in the unmedicated schizophrenic patients (n = 22; 4.38 ± 2.1 ng/mL) compared to the age-matched healthy volunteers. The percentage change of BDNF (increase, 173 % ± 110) correlated negatively with the percentage change of PANSS score with BDNF increase during psychotic treatment, as shown by several others (Pedrini et al. 2011; Yoshimura et al. 2007, 2010). Reduced BDNF concentrations have been reported in chronic antipsychotictreated patients (Grillo et al. 2007; Ikeda et al. 2008; Rizos et al. 2010), neuroleptic-free (Palomino et al. 2006), and neuroleptic-naı¨ve (Chen and Huang 2011; Rizos et al. 2008) patients.

Dyskinesic Aspects Long-term administration of antipsychotic agents induces tardive dyskinesias (TDs), a syndrome composed of involuntary, hyperkinetic, and abnormal movements often expressed through excessive chewing or dancing/foot shuffling behaviors (Jafari et al. 2012; Correll and Schenk 2008). Lower serum concentrations of BDNF, inversely correlated with Abnormal Involuntary Movement Scale (AIMS) scores, have been observed in patients presenting TDs (Tan et al. 2005). Additionally, associations between TD and BDNF polymorphisms have accumulated (Park et al. 2009; Zai et al. 2009). Yang et al. (2011) compared serum BDNF levels in schizophrenic patients with (n = 129) and without (n = 235) TDs with healthy controls (n = 323), with concurrent assessment of AIMS and the Positive and Negative Symptoms Scale (PANSS). Schizophrenic patients presenting TDs showed lower BDNF concentrations than those without and healthy controls. Lower BDNF serum concentrations correlated with higher PANSS negative sub-scores, but not with AIMS scores. The authors concluded that reduced BDNF concentrations may be associated with greater TD pathophysiology and negative symptoms in schizophrenia. Zhang et al. (2010) have indicated that schizophrenic patients with TD presented higher serum levels of S100B, a calciumbinding protein, than normal, healthy controls, and those patients without TD. The serum S100B levels were positively correlated with AIMS scores in patients with TD. The authors concluded from these results that increased S100B levels may be related to the neurodegenerative aspect of disorder, associated with TD pathophysiology. TDs are generally indicative of neurodegenerative pathologies (Miller and Chouinard 1993).

123

Neurotox Res

Cannabis Abuse Growing research evidence suggests that cannabis use leads to more frequent psychotic relapses, impairs cognitive performance and is associated generally with a poor prognosis in vulnerable individuals (Borgwardt et al. 2006). A long-term association between treatment adherence, type of first admission, and long cannabis use by firstepisode patients and high-risk juveniles (Barbeito et al. 2013; Gill et al. 2013). Additionally, there is a great interest in the hypothesis that cannabis use plays a causal role in the development of psychosis (Borgwardt et al. 2007; Haller et al. 2009). Analyses of prospective birth cohort and population studies suggest that cannabis use is indeed associated with an earlier age at onset of psychotic disorders, particularly schizophrenia (Smieskova et al. 2012a, b, c; Borgwardt et al. 2011; Bu¨schlen et al. 2011; Rothlisberger et al. 2012). The potential role as risk factor played by cannabis in the development of psychotic disorders has been recently investigated during the early prepsychotic phases of the illness. Cannabis abuse can impact the neurobiological alterations observed in potentially prodromal individuals (Smieskova et al. 2012a, b, c; Borgwardt et al. 2010; Bossong et al. 2013; Garakani et al. 2013; Morgan et al. 2013), with daily use, especially of high-potency cannabis, drives the earlier onset of psychosis in cannabis users (Di Forti et al. 2013). Imaging research in the healthy brain has consistently indicated cannabis abuse can modulate the neuroanatomical and neurofunctional areas which are implicated in psychosis onset (Buehlmann et al. 2010; Crossley et al. 2009; Fusar-Poli et al. 2007, 2010, 2011a, b, c; Smieskova et al. 2010a, b, 2012a, b, c). Finally, in a first population-based study mapping the association between lifetime cannabis use, psychosis, and schizotypal personality traits, Davis et al. (2013) have observed marked relationships between these factors. Stefanis et al. (2013) have shown that the use of cannabis may induce some cumulative toxic effects on those individuals abusing the drug, thereby placing them on the road to developing psychosis, the manifestation and debut of this condition are delayed for *7–8 years, independent of the age at which cannabis use was initiated.

Metabolic Aspects The glycogen synthase kinase-3s (GSK, a and b), involved in glycogen metabolism, are expressed ubiquitously over various tissues and are abundant in brain tissue (Jaworski et al. 2011). Kaidanovich-Beilin and Woodgett (2011) have discussed the fundamental roles for these protein kinases in memory, behavior, and neuronal fate determination. It has been argued that disrupted in Schizophrenia-1 (DISC1)

123

provides a candidate gene for neuropsychiatric disorders and an influential involvement during brain structural and functional development. Singh et al. (2011) have observed that DISC1 variants are implicated in the loss of function in Wnt/GSK3b signaling, and thereby interrupt the course of brain development. Common DISC1 polymorphisms (variants) are associated with neuropsychiatric phenotypes including altered cognition, brain structure, and function. The contribution of the DISC1 gene to neural development and the eventual neurodevelopment of schizophrenia risk has been studied (Wexler and Geschwind 2011). GSK-3, a regulator of a wide range of cellular processes, plays a key dual role in apoptosis with putative contribution to schizophrenia neuropathology (Emamian et al. 2004) and other neurodegenerative disorders (Avila et al. 2004; Berger et al. 2005; Onishi et al. 2011). Its role in the apoptotic signaling underlying excessive cell death may imply a neurodegenerative involvement in schizophrenia spectrum disorders, at least during the early stages of development (Gomez-Sintes et al. 2011). Astrocytic glycogenolysis and glycogen mobilization are implicated in normal brain function (Brown and Ransom 2007; Brown et al. 2003; Swanson 1992), and neuronal energy requirements (Brown et al. 2003; Wender et al. 2000), and not least in the necessities of cognitive function (Gibbs et al. 2006; Hertz et al. 2003; Suzuki et al. Suzuki et al. 2011). The links between glycogen metabolism and the glutathione (GSH) system have been studied: Shinohara et al. (2010) have shown that activation of GSK-3beta is a key mediator of the initial phase of acetaminophen-induced liver injury through modulating glutamate cysteine ligase (GCL) and myeloid cell leukemia sequence-1 degradation. GSH deficits occur both in neurodegenerative conditions and schizophrenia (Ballatori et al. 2009; Do et al. 2009; Dringen and Hirrlinger 2003), and a GCL gene polymorphism, affecting GSH synthesis, is associated with the disorder (Tosic et al. 2006). In this regard, Lavoie et al. (2011) have found that glucose metabolism and glycogen utilization are dysregulated in astrocytes showing chronic deficit in GSH implying dysfunctional brain energy metabolism in schizophrenia. It ought to be noted that Saitohin, an intronless gene nested within the human tau gene, that contains a single nucleotide polymorphism (A/G) may be involved in the pathophysiology of neurodegenerative disorders (Combarros et al. 2003). In a sample of 48 schizophrenic patients and 47 healthy controls, Bosia et al. (2011) tested the role of saitohin polymorphism as a concurring factor of cognitive decline among these patients using the Wisconsin Card Sorting Test for executive functioning. They observed a significantly greater frequency of G allele among patients with frontotemporal dementia and schizophrenic patients presenting impaired Wisconsin Card Sorting Test

Neurotox Res

performance. They concluded that Saitohin gene products affected core frontal executive function deterioration likely through mechanisms occurring through neurodevelopment with neurodegenerative outcome (Bosia et al. 2011). Finally, there exists shared susceptibility for type 2 diabetes and schizophrenia (Lin and Shuldiner 2010).

Brain Insulin Dysregulation There are several indications that risk factors for psychosis are associated with cardiometabolic disease risk factors, and vice versa (Galletly et al. 2012). Insulin, a major factor in cardiometabolic disease risk, is involved in several physiological function of the brain such as food intake and weight control, reproduction, learning and memory, neuromodulation and neuroprotection development, and progression of neurodegenerative and neuropsychiatric disorders (Bloemer et al. 2014; de la Monte and Tong 2013). In individuals afflicted by dysregulation of insulin the risk of brain plasticity derailment emerges (Deutsch et al. 2006; Ghasemi et al. 2012; Huang and Lee 2010). Graham et al. (2008) have observed in a group of patients presenting first-episode psychosis, 6-month treatment with second generation antipsychotics was associated with the exacerbation of pre-existing and emergence of new CVD and diabetes risk factors. Foley et al. (2013) have observed cardiometabolic risk factors/indicators distinguished those individuals presenting psychosis from the general population, through a number of factors including age, gender, obesity, etc., in a population of 1,642 psychosis-diagnosed Australians (aged 18–64 years) compared with a national comparator sample of 8,866 controls (aged 25–64 years) from the general population. From age 25, psychotic individuals presented a significantly higher mean BMI, waist circumference, triglycerides, glucose [women only], and diastolic blood pressure, and significantly lower HDL-cholesterol than control individuals. With the exception of triglycerides at age 60? in men, and glucose in women at various ages, these differences were present at every age. Differences in BMI and waist circumference between samples, although dramatic, could not explain all differences in diastolic blood pressure, HDL-cholesterol, or triglycerides, but did explain differences in glucose. They have postulated that psychosis shows the hallmarks of insulin-resistance by at least in the age of 25. Similarly, Chen et al. (2013) have shown that schizophreniarelated psychopathology was associated with insulin-resistance and/or dyslipidemia in Chinese patients with antipsychotic-naı¨ve first-episode schizophrenia patients were more prone to insulin-resistance and dyslipidemia as compared to the healthy population, which was correlated negatively to positive symptoms.

Immune System Imbalance Finally, there exists much evidence implicating cytokine imbalance in schizophrenia spectrum/schizopsychotic disorders. For example, Song et al. (2013) showed that drug naı¨ve, first-episode schizophrenic patients presenting normal weight indicated up-regulated inflammatory status associated with elevated levels of the cytokines, IL-1b, IL6, and TNF-a. Furthermore, inflammatory cytokine markers, such as IL-1Ra and sTNF-R1, are associated with both general disease severity and psychotic features (Hope et al. 2013; Kirkpatrick and Miller 2013). Dimitrov et al. (2013) observed that levels of GRO, MCP-1, MDC, and sCD40L were elevated significantly and that levels of IFN-c, IL-2, IL-12p70, and IL-17 were reduced significantly in schizophrenia patients compared to controls; positive correlations between cytokine levels and disorder severity, assessed with Positive and Negative Symptoms Scale (PANSS) scores in schizophrenic subjects, for G-CSF, IL-1b, IL1ra, IL-3, IL-6, IL-9, IL-10, sCD40L, and TNF-b. Bergink et al. (2013a) observed that postpartum psychosis patients failed to show the postpartum T cell level elevations of healthy women, but rather marked elevations of monocyte levels, and up-regulation of several immune-related monocyte genes. Notably, Di Nichola et al. (2013) found markedly higher serum levels of IL-1a, IL-1b, IL-8, and TNF-a as well as the tendency for higher IL-6 serum levels in comparison with controls; Leukocyte m-RNA levels of IL1a, IL-6, and TNF-a, but not IL-1b and IL-8, were significantly higher in patients also. Childhood trauma was associated with greater TNF-a levels with recent stressful life events linked to greater TNF-a m-RNA levels in leukocytes. Bergink et al. (2013b) have described a scenario through which both infection and environmental stressors interact during gestation/early life period to activate microglia thereby disturbing the fine balance neuronal and/or regional development; this concatenation of adverse events sets the stage for the burgeoning vulnerability for later psychotic disorders. As the adverse scenario unfolds, endocrine changes, stress, or infection, may activate microglia alterations further, leading to functional abnormalities of the neuronal circuitry in the brain and the expressions of psychosis. Borovcanin et al. (2012) have shown reduced levels of pro-inflammatory markers, IL-17 and IL-17/TGFb ratio, in patients presenting psychosis. They indicate that the presence of enhanced anti-inflammatory/immunosuppressive activity in schizophrenic patients express attempts of the patients’ immune system to counteract or limit ongoing pro-inflammatory processes and down-regulating chronic inflammatory processes (see also Borovcanin et al. 2013).

123

Neurotox Res

Conclusions Neurodegenerative aspects contributing to the pathophysiology of psychotic disorders appear, by and large, to exert their influences through interruption of the normal structural–functional trajectories of brain development and/or by neurotoxic destruction of tissue integrity. Not least is structural-functional integrity compromised by the presence of motor deficits (Singh et al. 2014). Observations of neuropathological structural/functional abnormalities in the postmortem brains of schizophrenic patients have been shown to occur over a wide range of brain areas (as shown, for example, by reduction in the volume of cerebral cortex and/or thalamus, and an increase in the volume of the ventricles) and nevertheless there are more reports describing the temporal lobe and frontal lobe compared to those describing other areas of the brain; all these suggesting degeneration of developmental origin. Intrauterine infection and inflammation are known risk factors for brain damage in the neonate irrespective of the gestational age. Infection-induced maternal immune activation leads to a fetal inflammatory response mediated by cytokines that has been implicated in the development of not only periventricular leukomalacia and cerebral palsy but also a spectrum of neurodevelopmental disorders, such as autism and schizophrenia (Burd et al. 2012; Ricci et al. 2013). The specific timing of the immune challenge with respect to the gestational age and neurologic development of the fetus may be crucial in the elicited response, which are the neuropsychiatric disorders associated with intrauterine inflammation appears to be the evidence for immune dysregulation in the developing brain. Laboratory model studies of maternal gestational inflammation provide critical roles for the elucidation of mechanisms involved in fetal brain injury associated with exposure to the maternal milieu. These animal models present different expressions of fetal microglial activation, neurotoxicity in combination with motor deficits and behavioral abnormalities in the offspring. The vulnerability bestowed through genetic and epigenetic (i.e., gene-environment interactions) forces exacerbate the deficits expressed through symptom profiles and biomarkers. In this respect, Kirkbride et al. (2012) have proposed that maternal prenatal nutrition can influence offspring schizophrenia risk via epigenetic effects. It appears that the prenatal nutrition may be linked to epigenetic outcomes in offspring and schizophrenia in offspring, and that schizophrenia is associated with these observed epigenetic changes. Disorder progression in the brains of patients was found to be characterized by progressive gray-matter volume decreases and lateral ventricular volume increases (Fusar-Poli et al. 2013a, b, c), possibly related to treatment. Further, increased levels of dopamine synthesis capacity in the dorsal striatum region

123

present a robust feature of individuals at ultra-high risk for psychosis (Egerton et al. 2013). Finally, the prevailing and potential risks for disorder, that influence developmental trajectories, yet, render distinctions within the spectrum vague, provoke activation of the immune system, and acting in parallel with underlying genetic liability, are linked with imperfect regulation of the genome mediating these prenatal or early postnatal environmental events (Jenkins 2013). Acknowledgments The development of this manuscript was supported by the Bliwa Stiftelsen. Conflict of interests peting interests.

The authors declare that they have no com-

References Adell A, Jime´nez-Sa´nchez L, Lo´pez-Gil X, Romo´n T (2012) Is the acute NMDA receptor hypofunction a valid model of schizophrenia? Schizophr Bull 38:9–14 Altar CA, Cai N, Bliven T, Juhasz M, Conner JM et al (1997) Anterograde transport of brain-derived neurotrophic factor and its role in the brain. Nature 389:856–860 Aoyama N, The´berge J, Drost DJ, Manchanda R, Northcott S, Neufeld RW, Menon RS, Rajakumar N, Pavlosky WF, Densmore M, Schaefer B, Williamson PC (2011) Grey matter and social functioning correlates of glutamatergic metabolite loss in schizophrenia. Br J Psychiatry 198(6):448–456. doi:10.1192/bjp. bp.110.079608 Aoyama Y, Mouri A, Toriumi K, Koseki T, Narusawa S, Ikawa N, Mamiya T, Nagai T, Yamada K, Nabeshima T (2013) Clozapine ameliorates epigenetic and behavioral abnormalities induced by phencyclidine through activation of dopamine D1 receptor. Int J Neuropsychopharmacol 17:1–15 Arango C, Fraguas D, Parellada M (2013) Differential neurodevelopmental trajectories in patients with early-onset bipolar and schizophrenia disorders. Schizophr Bull 40(Suppl 2):S138–S146 Archer T (2010) Neurodegeneration in schizophrenia. Expert Rev Neurother 10:1131–1141 Archer T, Kostrzewa RM, Beninger RJ, Palomo T (2010a) Staging perspectives in neurodevelopmental aspects of neuropsychiatry: agents, phases and ages at expression. Neurotox Res 18:287–305 Archer T, Kostrzewa RM, Palomo T, Beninger RJ (2010b) Clinical staging in the pathophysiology of psychotic and affective disorders: facilitation of prognosis and treatment. Neurotox Res 18:211–228 Avila J, Lucas JJ, Perez M, Hernandez F (2004) Role of tau protein in both physiological and pathological conditions. Physiol Rev 84:361–384 Ballatori N, Krance SM, Notenboom S, Shi S, Tieu K et al (2009) Glutathione dysregulation and the etiology and progression of human diseases. Biol Chem 390:191–214 Barbeito S, Vega P, Ruiz de Azu´a S, Saenz M, Martinez-Cengotitabengoa M, Gonza´lez-Ortega I, Bermudez C, Hernanz M, Corres BF, Gonza´lez-Pinto A (2013) Cannabis use and involuntary admission may mediate long-term adherence in firstepisode psychosis patients: a prospective longitudinal study. BMC Psychiatry 13(1):326. doi:10.1186/1471-244X-13-326 Berger Z, Ttofi EK, Michel CH, Pasco MY, Tenant S et al (2005) Lithium rescues toxicity of aggregate-prone proteins in

Neurotox Res Drosophila by perturbing Wnt pathway. Hum Mol Genet 14:3003–3011 Bergink V, Burgerhout KM, Weigelt K, Pop VJ, de Wit H, Drexhage RC, Kushner SA, Drexhage HA (2013a) Immune system dysregulation in first-onset postpartum psychosis. Biol Psychiatry 73(10):1000–1007. doi:10.1016/j.biopsych.2012.11.006 Bergink V, Gibney SM, Drexhage HA (2013b) Autoimmunity, inflammation and psychosis: a search for peripheral markers. Biol Psychiatry. doi:10.1016/j.biopsych.2013.09.037 Bergman O, Westberg L, Lichtenstein P, Eriksson E, Larsson H (2011) Study on the possible association of brain-derived neurotrophic factor polymorphism with the developmental course of symptoms of attention deficit and hyperactivity. Int J Neuropsychopharmacol 14:1367–1376 Bloemer J, Bhattacharya S, Amin R, Suppiramaniam V (2014) Impaired insulin signaling and mechanisms of memory loss. Prog Mol Biol Transl Sci 121:413–449. doi:10.1016/B978-0-12800101-1.00013-2 Borgwardt SJ, Radue EW, Gotz K, Aston J, Drewe M, Gschwandtner U, Haller S, Pfluger M, Stieglitz RD, McGuire PK, RiecherRossler A (2006) Radiological findings in individuals at high risk of psychosis. J Neurol Neurosurg Psychiatry 77(2):229–233 Borgwardt SJ, Riecher-Rossler A, Dazzan P, Chitnis X, Aston J, Drewe M, Gschwandtner U, Haller S, Pfluger M, Rechsteiner E, D’Souza M, Stieglitz RD, Radu EW, McGuire PK (2007) Regional gray matter volume abnormalities in the at risk mental state. Biol Psychiatry 61(10):1148–1156 Borgwardt S, Smieskova R, Bendfeldt K, Buhlmann E, Berger G, Aston J, Gschwandtner U, Pflueger M, Stieglitz RD, RiecherRossler A, Radue EW (2010) Hippocampal volume reduction specific for later transition to psychosis or substance-associated effects? J Psychiatry Neurosci 35(3):214–215 author reply 215 Borgwardt S, McGuire P, Fusar-Poli P (2011) Gray matters!–mapping the transition to psychosis. Schizophr Res 133(1–3):63–67 Borovcanin M, Jovanovic I, Radosavljevic G, Djukic Dejanovic S, Bankovic D, Arsenijevic N, Lukic ML (2012) Elevated serum level of type-2 cytokine and low IL-17 in first episode psychosis and schizophrenia in relapse. J Psychiatr Res 46(11):1421–1426. doi:10.1016/j.jpsychires.2012.08.016 Borovcanin M, Jovanovic I, Radosavljevic G, Djukic Dejanovic S, Stefanovic V, Arsenijevic N, Lukic ML (2013) Antipsychotics can modulate the cytokine profile in schizophrenia: attenuation of the type-2 inflammatory response. Schizophr Res 147(1): 103–109. doi:10.1016/j.schres.2013.03.027 Bosia M, Buonocure M, Guglielmino C, Pirovana A, Lorenzi C et al (2011) Saitohin polymorphism and executive dysfunction in schizophrenia. Neurol Sci. doi:10.1007/s10072-0893-9 Bossong MG, Jansma JM, Bhattacharyya S, Ramsey NF (2013) Role of the endocannabinoid system in brain functions relevant for schizophrenia: an overview of human challenge studies with cannabis or D9-tetrahydrocannabinol (THC). Prog Neuropsychopharmacol Biol Psychiatry. doi:10.1016/j.pnpbp.2013.11.017 Brown AM, Ransom BR (2007) Astrocyte glycogen and brain energy metabolism. Glia 55:1263–1271 Brown AM, Baltan Tekkok S, Ransom BR (2003) Glycogen regulation and functional role in mouse white matter. J Physiol 549:501–512 Buehlmann E, Berger GE, Aston J, Gschwandtner U, Pflueger MO, Borgwardt SJ, Radue EW, Riecher-Ro¨ssler A (2010) Hippocampus abnormalities in at risk mental states for psychosis? A cross-sectional high resolution region of interest magnetic resonance imaging study. J Psychiatr Res 44(7):447–453. doi:10.1016/j.jpsychires.2009.10.008 Burd I, Balakrishnan B, Kannan S (2012) Models of fetal brain injury, intrauterine inflammation, and preterm birth. Am J Reprod Immunol 67:287–294. doi:10.1111/j.1600-0897.2012.01110.x

Bu¨schlen J, Berger GE, Borgwardt SJ, Aston J, Gschwandtner U, Pflueger MO, Kuster P, Radu¨ EW, Stieglitz RD, Riecher-Ro¨ssler A (2011) Pituitary volume increase during emerging psychosis. Schizophr Res 125(1):41–48. doi:10.1016/j.schres.2010.09.022 Bustillo JR, Rowland LM, Mullins P, Jung R, Chen H, Qualls C, Hammond R, Brooks WM, Lauriello J (2010) 1H-MRS at 4 tesla in minimally treated early schizophrenia. Mol Psychiatry 15:629–636 Cannon M, Caspi A, Moffitt TE (2002a) Evidence for early, specific, pandevelopment impairment in schizophreniform disorder: results from a longitudinal birth cohort. Arch Gen Psychiatry 59:449–456 Cannon M, Murray RM, Jones P (2002b) Obstetric complications and schizophrenia: past, present and future. Am J Psychiatry 159:1080–1092 Chen S, Hillman DE (1986) Selective ablation of neurons by methylazoxymethanol during pre- and postnatal brain development. Exp Neurol 94:103–119 Chen CC, Huang TL (2011) Effects of antipsychotics on the serum BDNF levels in schizophrenia. Psychiatry Res 189:327–330 Chen S, Broqueres-You D, Yang G, Wang Z, Li Y, Wang N, Zhang X, Yang F, Tan Y (2013) Relationship between insulin resistance, dyslipidemia and positive symptom in Chinese antipsychotic-naive first-episode patients with schizophrenia. Psychiatry Res 210(3):825–829. doi:10.1016/j.psychres.2013.08. 056 Chew LJ, Fusar-Poli P, Schmitz T (2013) Oligodendroglial alterations and the role of microglia in white matter injury: relevance to schizophrenia. Dev Neurosci 35(2–3):102–129. doi:10.1159/ 000346157 Ciani E, Frenquelli M, Contestabile A (2003) Developmental expression of the cell cycle and apoptosis controlling gene, Lotl, in the rat cerebellum and in cerebellar granule cells. Brain Res Dev Brain Res 142:193–202 Combarros O, Rodero L, Infante J, Palacio E, Llorca J, Ferna´ndezViadero C, Pen˜a N, Berciano J (2003) Age-dependent association between the Q7R polymorphism in the Saitohin gene and sporadic Alzheimer’s disease. Dement Geriatr Cogn Disord 16:132–135 Correll CU, Schenk EM (2008) Tardive dyskinesias and new antipsychotics. Curr Opin Psychiatry 21:151–156 Crossley NA, Mechelli A, Fusar-Poli P, Broome MR, Matthiasson P, Johns LC, Bramon E, Valmaggia L, Williams SC, McGuire PK (2009) Superior temporal lobe dysfunction and frontotemporal dysconnectivity in subjects at risk of psychosis and in firstepisode psychosis. Hum Brain Mapp 30(12):4129–4137 Dauvermann MR, Mukherjee P, Moorhead WT, Stanfield AC, FusarPoli P, Lawrie SM, Whalley HC (2012) Relationship between gyrification and functional connectivity of the prefrontal cortex in subjects at high genetic risk of schizophrenia. Curr Pharm Des 18(4):434–442 Dauvermann MR, Whalley HC, Romaniuk L, Valton V, Owens DG, Johnstone EC, Lawrie SM, Moorhead TW (2013) The application of nonlinear Dynamic Causal Modelling for fMRI in subjects at high genetic risk of schizophrenia. Neuroimage 73:16–29. doi:10.1016/j.neuroimage.2013.01.063 Davis GP, Compton MT, Wang S, Levin FR, Blanco C (2013) Association between cannabis use, psychosis, and schizotypal personality disorder: findings from the National Epidemiologic Survey on Alcohol and Related Conditions. Schizophr Res 151(1–3):197–202. doi:10.1016/j.schres.2013.10.018 de la Monte SM, Tong M (2013) Brain metabolic dysfunction at the core of Alzheimer’s disease. Biochem Pharmacol. doi:10.1016/j. bcp.2013.12.012 Dean OM, van den Buuse M, Berk M, Copolov DL, Mavros C, Bush AI (2011) N-acetyl cysteine restores brain glutathione loss in

123

Neurotox Res combined 2-cyclohexene-1-one and D-amphetamine-treated rats: relevance to schizophrenia and bipolar disorder. Neurosci Lett 499:149–153 Deutsch SI, Rosse RB, Lakshman RM (2006) Dysregulation Prog Neuropsychopharmacol Biol Psychiatry. 30(8):1369–80. of tau phosphorylation is a hypothesized point of convergence in the pathogenesis of alzheimer’s disease, frontotemporal dementia and schizophrenia with therapeutic implications Di Forti M, Sallis H, Allegri F, Trotta A, Ferraro L, Stilo SA, Marconi A, La Cascia C, Reis Marques T, Pariante C, Dazzan P, Mondelli V, Paparelli A, Kolliakou A, Prata D, Gaugrhan F, David AS, Morgan C, Sthal D, Khondoker M, Maccabe JH, Murray RM (2013) Daily use, especially of high-potency cannabis, drives the earlier onset of psychosis in cannabisUsers. Schizophr Bull Di Nicola M, Cattaneo A, Hepgul N, Di Forti M, Aitchison KJ, Janiri L, Murray RM, Dazzan P, Pariante CM, Mondelli V (2013) Serum and gene expression profile of cytokines in first-episode psychosis. Brain Behav Immun 31:90–95. doi:10.1016/j.bbi. 2012.06.010 Dimitrov DH, Lee S, Yantis J, Valdez C, Paredes RM, Braida N, Velligan D, Walss-Bass C (2013) Differential correlations between inflammatory cytokines and psychopathology in veterans with schizophrenia: potential role for IL-17 pathway. Schizophr Res 151(1–3):29–35. doi:10.1016/j.schres.2013.10. 019 Do KQ, Cabungal JH, Frank A, Steullet P, Cuenod M (2009) Redox dysregulation, neurodevelopment and schizophrenia. Curr Opin Neurobiol 19:220–230 Dringen R, Hirrlinger J (2003) Glutathione pathways in the brain. Biol Chem 384:505–516 Egerton A, Chaddock CA, Winton-Brown TT, Bloomfield MA, Bhattacharyya S, Allen P, McGuire PK, Howes OD (2013) Presynaptic striatal dopamine dysfunction in people at ultra-high risk for psychosis: findings in a second cohort. Biol Psychiatry 74(2):106–112. doi:10.1016/j.biopsych.2012.11.017 Elia J, Laracy S, Allen J, Nissley-Tsiopinis J, Borgmann-Winter K (2011) Epigenetics: genetics versus life experiences. Curr Topics Behav Neurosci. doi:10.1007/7854_2011_144 Ellenbroek BA, van den Kroonenberg PT, Cools AR (1998) The effects of an early stressful life event on sensorimotor gating in adult rats. Schizophr Res 30:251–260 Ellman LM, Yolken RH, Buka SL, Torrey EF, Cannon TD (2009) Cognitive functioning prior to the onset of psychosis: the role of fetal exposure to serologically determined influenza infection. Biol Psychiatry 65(12):1040–1047. doi:10.1016/j.biopsych.2008. 12.015 Emamian ES, Hall D, Birnbaum MJ, Karayiorgou M, Gogos JA (2004) Convergent evidence for impaired AKT1-GSK3 signaling in schizophrenia. Nat Genet 36:131–137 Faizi M, Salimi A, Rasoulzadeh M, Naserzadeh P, Pourahmad J (2014) Schizophrenia induces oxidative stress and cytochrome C release in isolated rat brain mitochondria: a possible pathway for induction of apoptosis and neurodegeneration. Iran J Pharm Res 13(Suppl):93–100 Favalli G, Li J, Belmonte-de-Abreu P, Wong AH, Daskalakis ZJ (2012) The role of BDNF in the pathophysiology and treatment of schizophrenia. J Psychiatr Res 46:1–11 Featherstone RE, Rizos Z, Nobrega JN, Kapur S, Fletcher PJ (2007) Gestational methylazoxymethanol acetate treatment impairs select cognitive functions: parallels to schizophrenia. Neuropsychopharmacology 32:483–492 Ferrer I, Pozas E, Marti M, Blanco R, Planas AM (1997) Methylazoxymethanol-induced apoptosis in the external granule cell layer of the developing cerebellum of the rat is associated with strong c-jun expression and formation of high molecular weight c-jun complexes. J Neuropathol Exp Neurol 56:1–9

123

Fiore M, Grace AA, Korf J, Stampachiacchiere B, Aloe L (2004) Impaired brain development in the rat following prenatal exposure to methylazoxymethanol acetate at gestational day 17 and neurotrophin distribution. NeuroReport 15(11):1791–1795 Flagstad P, Glenthoj BY, Didriksen M (2005) Cognitive deficits caused by late gestational disruption of neurogenesis in rats: a preclinical model of schizophrenia. Neuropsychopharmacology 30:250–260 Foley DL, Mackinnon A, Watts GF, Shaw JE, Magliano DJ, Castle DJ, McGrath JJ, Waterreus A, Morgan VA, Galletly CA (2013) Cardiometabolic risk indicators that distinguish adults with psychosis from the general population, by age and gender. PLoS ONE 8(12):e82606. doi:10.1371/journal.pone.0082606 Fontes MA, Bolla KI, Cunha PJ, Almeida PP, Jungerman F, Laranjeira RR, Bressan RA, Lacerda AL (2011) Cannabis use before age 15 and subsequent executive functioning. Br J Psychiatry 198:442–447 Furukawa T, Mizukawa R, Hirai T, Fujihara S, Kitamura T, Takahashi K (1998) Childhood parental loss and schizophrenia: evidence against pathogens but for some pathoplastic effects. Psychiatry Res 81:353–362 Fusar-Poli P, Meyer-Lindenberg A (2013a) Striatal presynaptic dopamine in schizophrenia, part I: meta-analysis of dopamine active transporter (DAT) density. Schizophr Bull 39(1):22–32. doi:10.1093/schbul/sbr111 Fusar-Poli P, Meyer-Lindenberg A (2013b) Striatal presynaptic dopamine in schizophrenia, part II: meta-analysis of [18F]/ [11C] DOPA PET studies. Schizophr Bull 39(1):33–42. doi:10. 1093/schbul/sbr180 Fusar-Poli P, Perez J, Broome M, Borgwardt S, Placentino A, Caverzasi E, Cortesi M, Veggiotti P, Politi P, Barale F, McGuire P (2007) Neurofunctional correlates of vulnerability to psychosis: a systematic review and meta-analysis. Neurosci Biobehav Rev 31(4):465–484 Fusar-Poli P, Howes OD, Allen P, Broome M, Valli I, Asselin MC, Grasby PM, McGuire PK (2010) Abnormal frontostriatal interactions in people with prodromal signs of psychosis: a multimodal imaging study. Arch Gen Psychiatry 67(7):683–691. doi:10.1001/archgenpsychiatry.2010.77 Fusar-Poli P, Borgwardt S, Crescini A, D’Este G, Kempton M, Lawrie S, Guire PM, Sacchetti E (2011a) Neuroanatomy of vulnerability to psychosis: a voxel-based meta-analysis. Neurosci Biobehav Rev 35(5):1175–1185 Fusar-Poli P, Stone JM, Broome MR, Valli I, Mechelli A, McLean MA, Lythgoe DJ, O’Gorman RL, Barker GJ, McGuire PK (2011b) Thalamic glutamate levels as a predictor of cortical response during executive functioning in subjects at high risk for psychosis. Arch Gen Psychiatry 68(9):881–890. doi:10.1001/ archgenpsychiatry.2011.46 Fusar-Poli P, Howes OD, Allen P, Broome M, Valli I, Asselin MC, Montgomery AJ, Grasby PM, McGuire P (2011c) Abnormal prefrontal activation directly related to pre-synaptic striatal dopamine dysfunction in people at clinical high risk for psychosis. Mol Psychiatry 16(1):67–75 Fusar-Poli P, Bonoldi I, Yung AR, Borgwardt S, Kempton M, Barale F, Caverzasi E, McGuire P (2012a) Predicting psychosis: a metaanalysis of transition outcomes in individuals at high clinical risk. Arch Gen Psychiatry 69(3):1–10 Fusar-Poli P, Deste G, Smieskova R, Barlati G, Yung AR, Howes O, Stieglitz R, McGuire P, Borgwardt S (2012b) Cognitive functioning in prodromal psychosis: a meta-analysis. Arch Gen Psychiatry 69(6):562–571. doi:10.1001/archgenpsychiatry.2011. 1592 Fusar-Poli P, Borgwardt S, Bechdolf A, Addington J, Riecher-Ro¨ssler A, Schultze-Lutter F, Keshavan M, Wood S, Ruhrmann S, Seidman L, Valmaggia L, Cannon T, Velthorst E, De Haan L,

Neurotox Res Cornblatt B, Birchwood M, McGlashan T, Carpenter W, McGorry P, Klosterko¨tter J, McGuire P, Yung A (2013a) The psychosis high risk state: a comprehensive state of the art review. Arch Gen Psychiatry 70(1):107–120. doi:10.1001/jamapsychia try.2013.269 Fusar-Poli P, Smieskova R, Kempton MJ, Ho BC, Andreasen NC, Borgwardt S (2013b) Progressive brain changes in schizophrenia related to antipsychotic treatment? A meta-analysis of longitudinal MRI studies. Neurosci Biobehav Rev. doi:10.1016/j. neubiorev.2013.06.001 Fusar-Poli P, Bechdolf A, Taylor M, Bonoldi I, Carpenter W, Yung A, McGuire P (2013c) At risk for schizophrenic or affective psychosis? A meta-analysis of ICD/DSM diagnostic outcomes in individuals at high clinical risk. Schizophr Bull 39(4):923–932. doi:10.1093/schbul/sbs060 Galletly CA, Foley DL, Waterreus A, Watts GF, Castle DJ, McGrath JJ, Mackinnon A, Morgan VA (2012) Cardiometabolic risk factors in people with psychotic disorders: the second Australian national survey of psychosis. Aust N Z J Psychiatry 46(8):753–761. doi:10.1177/0004867412453089 Garakani A, Welch AK, Jaffe RJ, Protin CA, McDowell DM (2013) Psychosis and low cyanocobalamin in a patient abusing nitrous oxide and cannabis. Psychosomatics. doi:10.1016/j.psym.2013. 11.001 Geddes JR, Verdoux H, Takei N (1999) Schizophrenia and complications of pregnancy and labour: an individual patient data metaanalysis. Schizophrenia Bull 25:413–423 Ghasemi R, Dargahi L, Haeri A, Moosavi M, Mohamed Z, Ahmadiani A (2012) Brain insulin dysregulation: implication for neurological and neuropsychiatric disorders. Mol Neurobiol 47(3):1045–1065. doi:10.1007/s12035-013-8404-z Gibbs ME, Anderson DG, Hertz L (2006) Inhibition of glycogenolysis in astrocytes interrupts memory consolidation in young chickens. Glia 54:214–222 Gill KE, Poe L, Azimov N, Ben-David S, Vadhan NP, Girgis R, Moore H, Cressman V, Corcoran CM (2013) Reasons for cannabis use among youths at ultra high risk for psychosis. Early Interv Psychiatry. doi:10.1111/eip.12112 Goldman AL, Pezawas L, Mattay VS, Fischl B, Verchinski BA, Chen Q, Weinberger DR, Meyer-Lindenberg A (2009) Widespread reductions of cortical thickness in schizophrenia and spectrum disorders and evidence of heritability. Arch Gen Psychiatry 66:467–477 Gomez-Sintes R, Hernandez F, Lucas JJ, Avila J (2011) GSK-3 mouse models to study neuronal apoptosis and neurodegeneration. Fron Mol Neurosci 4:1–11. doi:10.3389/fnmol.2011.00045 Goto Y, Grace AA (2006) Alterations in medial prefrontal cortical activity and plasticity in rats with disruption of cortical development. Boil psychiatry 60:1259–1267 Goto N, Yoshimura R, Kakeda S, Moriya J, Hayashi K, IkenouchiSugita A, Umene-Nakano W, Hori H, Ueda N, Korogi Y, Nakamura J (2010) Comparison of brain N-acetylaspartate levels and serum brain-derived neurotrophic factor (BDNF) levels between patients with first-episode schizophrenia psychosis and healthy controls. Eur Psychiatry 26:57–63 Gourevitch R, Rocher C, Le Pen G, Krebs MO, Jay TM (2004) Working memory deficits in adult rats after prenatal disruption of neurogenesis. Behav Pharmacol 15:287–292 Graff J, Dohoon K, Dobbin MM, Tsai LH (2011) Epigenetic regulation of gene expression in physiological and pathological brain processes. Physiol Rev 91:603–649 Graham KA, Cho H, Brownley KA, Harp JB (2008) Early treatmentrelated changes in diabetes and cardiovascular disease risk markers in first episode psychosis subjects. Schizophr Res 101(1–3):287–294. doi:10.1016/j.schres.2007.12.476

Gratacos M, Gonzalez JR, Mercader JM, de Cid R, Urretavizcaya M, Estivill X (2007) Brain-derived neurotrophic factor Val66Met and psychiatric disorders: meta-analysis of case-control studies confirm association to substance-related disorders, eating disorders and schizophrenia. Biol Psychiatry 61:911–922 Grillo RW, Ottoni GL, Leke R, Souza DO, Portela LV, Lara DR (2007) Reduced serum BDNF levels in schizophrenic patients on clozapine or typical antipsychotics. J Psychiatry Res 41:31–35 Haller S, Borgwardt SJ, Schindler C, Aston J, Radue EW, RiecherRo¨ssler A (2009) Can cortical thickness asymmetry analysis contribute to detection of at-risk mental state and first-episode psychosis? A pilot study. Radiology 250(1):212–221. doi:10. 1148/radiol.2501072153 Hertz L, O’Dowd BS, Ng KT, Gibbs ME (2003) Reciprocal changes in forebrain contents of glycogen and glutamate/glutamate during early memory consolidation in the day-old chick. Brain Res 994:226–233 Hoenicka J, Garrido E, Martı´nez I, Ponce G, Aragu¨e´s M, Rodrı´guezJime´nez R, Espan˜a-Serrano L, Alvira-Botero X, Santos JL, Rubio G, Jime´nez-Arriero MA, Palomo T, PARGPARG (2010) Gender-specific COMT Val158Met polymorphism association in Spanish schizophrenic patients. Am J Med Genet B Neuropsychiatr Genet 153B:79–85 Honea RA, Meyer-Lindenberg A, Hobbs KB, Pezawas L, Mattay VS, Egan MF, Verchinski B, Passingham RE, Weinberger DR, Callicott JH (2008) Is grey matter volume an intermediate phenotype for schizophrenia? A voxel-based morphometric study of patients with schizophrenia and their healthy siblings. Biol Psychiatry 63:465–474 Hope S, Ueland T, Steen NE, Dieset I, Lorentzen S, Berg AO, Agartz I, Aukrust P, Andreassen OA (2013) Interleukin 1 receptor antagonist and soluble tumor necrosis factor receptor 1 are associated with general severity and psychotic symptoms in schizophrenia and bipolar disorder. Schizophr Res 145(1–3):36–42. doi:10.1016/j.schres.2012.12.023 Howes OD, Montgomery AJ, Asselin M, Murray R, Grasby P, McGuire P (2007) Molecular imaging studies of the striatal dopaminergic system in psychosis and predictions for the prodromal phase of psychosis. Br J Psychiatry s51:s13–s18 Huang CC, Lee CC (2010) Hsu KS (2010) The role of insulin receptor signaling in synaptic plasticity and cognitive function. Chang Gung Med J 33(2):115–125 Huttunen J, Heinimaa M, Svirskis T, Nyman M, Kajander J, Forsback S, Solin O, Ilonen T, Korkeila J, Ristkari T, McGlashan T, Salokangas RK, Hietala J (2008) Striatal dopamine synthesis in first-degree relatives of patients with schizophrenia. Biol Psychiatry 63(1):114–117 Hyman C, Hofer M, Barde YA, Juhasz M, Yancopoulos GD, Squinto SP, Lindsay RM (1991) BDNF is a neurotrophic factor for dopaminergic neurons of the substantia nigra. Nature 350:230–232 Ikeda Y, Yahata N, Ito I, Nagano M, Toyota T et al (2008) Low serum levels of brain derived neurotrophic factor and epidermal growth factor in patients with chronic schizophrenia. Schizophr Res 101:58–66 Jablonka E, Raz G (2009) Transgenerational epigenetic inheritance: prevalence, mechanisms, and implications for the study of heredity and evolution. Q Rev Biol 84:131–176 Jacob CP, Nguyen TT, Dempfle A, Heine M, Windemuth-Kieselbach C, Baumann K, Jacob F, Prechtl J, Wittlich M, Herrmann MJ, Gross-Lesch S, Lesch KP, Reif A (2010) A gene-environment investigation on personality traits in two independent clinical sets of adult patients with personality disorder and attention deficit/hyperactivity disorder. Eur Arch Psychiatr Clin Neurosci 260:317–326

123

Neurotox Res Jafari S, Fernandez-Enright F, Huang XF (2012) Structural contributions of antipsychotic drugs to their therapeutic profiles and metabolic side effects. J Neurochem 120:371–384 Jaworski T, Dewachter I, Lechat B, Gees M, Kremer A, Demedts D, Borghgraef P, Devijver H, Ku¨gler S, Patel S, Woodgett JR, Van Leuven F (2011) GSK-3a/b kinases and amyloid production in vivo. Nature 480(7376):E4–E5 Jenkins TA (2013) Perinatal complications and schizophrenia: involvement of the immune system. Front Neurosci 7:110. doi:10.3389/fnins.2013.00110 Kaidanovich-Beilin O, Woodgett JR (2011) GSK-3: functional insights from cell biology and animal models. Front Mol Neurosci 4:40 Kegeles LS, Abi-Dargham A, Frankle WG, Gil R, Cooper TB, Slifstein M, Hwang DR, Huang Y, Haber SN, Laruelle M (2010) Increased synaptic dopamine function in associative regions of the striatum in schizophrenia. Arch Gen Psychiatry 67(3): 231–239 Kirkbride JB, Susser E, Kundakovic M, Kresovich JK, Davey Smith G, Relton CL (2012) Prenatal nutrition, epigenetics and schizophrenia risk: can we test causal effects? Epigenomics 4:303–315 Kirkpatrick B, Miller BJ (2013) Inflammation and schizophrenia. Schizophr Bull 39(6):1174–1179. doi:10.1093/schbul/sbt141 Koutsouleris N, Borgwardt S, Meisenzahl EM, Bottlender R, Moller HJ, Riecher-Rossler A (2012) Disease prediction in the At-risk mental State for psychosis using neuroanatomical biomarkers: results from the FePsy Study. Schizophr Bull 38(6):1234–1246. doi:10.1093/schbul/sbr145 Lafarga M, Lerga A, Andres MA, Polanco JL, Calle E, Berciano MT (1997) Apoptosis induced by methylazoxymethanol in developing rat cerebellum: organization of the cell nucleus and its relationship to DNA and tRNA degradation. Cell Tissue Res 289:25–38 Laruelle M, Abi-Dargham A, van Dyck CH, Gil R, D’Souza CD, Erdos J, McCance E, Rosenblatt W, Fingado C, Zoghbi SS, Baldwin RM, Seibyl JP, Krystal JH, Charney DS, Innis RB (1996) Single photon emission computerized tomography imaging of amphetamine-induced dopamine release in drug-free schizophrenic subjects. Proc Natl Acad Sci USA 93(17): 9235–9240 Lavoie S, Allaman I, Petit JM, Do KQ, Magistretti PJ (2011) Altered glycogen metabolism in cultured astrocytes from mice with chronic glutathione deficit: relevance for neuroenergetics in schizophrenia. PLoS ONE 6:e22875 Lee AH, Lange C, Ricken R, Hellweg R, Lang UE (2011) Reduced brain-derived neurotrophic factor serum concentrations in acute schizophrenic patients increase during antipsychotic treatment. J Clin Psychopharmacol 31:334–336 Leng A, Jongen-Reˆlo AL, Pothuizen HH, Feldon J (2005) Effects of prenatal methylazoxymethanol acetate (MAM) treatment in rats on water maze performance. Behav Brain Res 161(2):291–298 Lieberman JA, Kinon BJ, Loebel AD (1990) Dopaminergic mechanisms in idiopathic and drug-induced psychoses. Schizophr Bull 16(1):97–110 Lieberman JA, Sheitman BB, Kinon BJ (1997) Neurochemical sensitization in the pathophysiology of schizophrenia: deficits and dysfunction in neuronal regulation and plasticity. Neuropsychopharmacology 17(4):205–229 Lillrank SM, Lipska BK, Kolachana BS, Weinberger DR (1999) Attenuated extracellular dopamine levels after stress and amphetamine in the nucleus accumbens of rats with neonatal ventral hippocampal damage. J Neural Transm 106:183–196 Lin PL, Shuldiner AR (2010) Rethinking the genetic basis of comorbidity of schizophrenia and type 2 diabetes. Schizophrenia Res 123:234–243. doi:10.1016/j.schres.2010.08.022

123

Lipska BK, Weinberger DR (1994) Subchronic treatment with haloperidol and clozapine in rats with neonatal excitotoxic hippocampal damage. Neuropsychopharmacology 10:199–205 Lipska BK, Jaskiw GE, Chrapusta S, Karoum F, Weinberger DR (1992) Ibotenic acid lesions of the ventral hippocampus differentially affects dopamine and its metabolites in the nucleus accumbens and prefrontal cortex in the rat. Brain Res 585:1–6 Lipska BK, Swerdlow NR, Geyer MA, Jaskiw GE, Braff DL, Weinberger DR (1995) Neonatal excitotoxic lesions hippocampal damage in rats causes post pubertal changes in prepulse inhibition of startle and its disruption by apomorphine. Psychopharmacology 122:35–43 Lipska BK, Kolb B, Halim N, Weinberger DR (2001) Synaptic abnormalities in prefrontal cortex and nucleus accumbens of adult rats with neonatal hippocampal damage. Schizophr Res 49:47–51 Lodge DJ, Grace AA (2008) Hippocampal dysfunction and disruption of dopamine system regulation in an animal model of schizophrenia. Neurotox Res 14:97–104 Lyon GJ, Abi-Dargham A, Moore H, Lieberman JA, Javitch JA, Sulzer D (2011) Presynaptic regulation of dopamine transmission in schizophrenia. Schizophr Bull 37(1):108–117 Maat A, Fett AK, Derks E, GROUP Investigators (2012) Social cognition and quality of life in schizophrenia. Schizophr Res 137(1–3):212–218. doi:10.1016/j.schres.2012.02.017 McGuire P, Howes OD, Stone J, Fusar-Poli P (2008) Functional neuroimaging in schizophrenia: diagnosis and drug discovery. Trends Pharmacol Sci. 29(2):91–98 Mechelli A, Riecher-Rossler A, Meisenzahl EM, Tognin S, Wood SJ, Borgwardt SJ, Koutsouleris N, Yung AR, Stone JM, Phillips LJ, McGorry PD, Valli I, Velakoulis D, Woolley J, Pantelis C, McGuire P (2011) Neuroanatomical abnormalities that predate the onset of psychosis: a multicenter study. Arch Gen Psychiatry. 68(5):489–495 Meyer-Lindenberg A (2011) Neuroimaging and the question of neurodegeneration in schizophrenia. Prog Neurobiol 95:514–516 Meyer-Lindenberg A, Miletich RS, Kohn PD, Esposito G, Carson RE, Quarantelli M, Weinberger DR, Berman KF (2002) Reduced prefrontal activity predicts exaggerated striatal dopaminergic function in schizophrenia. Nat Neurosci 5(3):267–271 Meyer-Lindenberg A, Olsen RK, Kohn PD, Brown T, Egan MF, Weinberger DR, Berman KF (2005) Regionally specific disturbance of dorsolateral prefrontal-hippocampal functional connectivity in Schizophrenia. Arch Gen Psychiatry 62:379–386 Miller R, Chouinard G (1993) Loss of striatal cholinergic neurons as a basis for tardive and L-dopa-induced dyskinesias, neurolepticinduced supersensitivity psychosis and refractory schizophrenia. Biol Psychiatry 34:713–738 Miller J, Williamson P, Jensen JE, Manchanda R, Menon R et al (2009) Longitudinal 4.0 Testa (31)P magnetic resonance spectroscopy changes in the anterior cingulate and left thalamus in first episode schizophrenia. Psychiatr Res Neuroimaging 173:155–157 Miyake N, Thompson J, Skinbjerg M, Abi-Dargham A (2011) Presynaptic dopamine in schizophrenia. CNS Neurosci Ther 17(2):104–109. doi:10.1111/j.1755-5949.2010.00230.x Mohammed AK, Jonsson G, So¨derberg U, Archer T (1986a) Impaired selective attention in methylazoxymethanol-induced microencephalic rats. Pharmacol Biochem Behav 24:975–981 Mohammed AK, Jonsson G, Sundstro¨m E, Minor BG, So¨derberg U, Archer T (1986b) Selective attention and place navigation in rats treated prenatally with methylazoxymethanol. Brain Res Dev Brain Res 30:145–155 Moore H, Jentsch JD, Ghajarnia M, Geyer MA, Grace AA (2006) A neurobehavioral systems analysis of adult rats exposed to

Neurotox Res methylazoxymethanol acetate on E17: implications for the neuropathology of schizophrenia. Biol Psychiatry 60:253–264 Morgan C, Kirkbride J, Leff J, Craig T, Hutchinson G, McKenzie K, Morgan K, Dazzan P, Doody GA, Jones P, Murray R, Fearon P (2007) Parental separation, loss and psychosis in different ethnic groups: a case-control study. Psychol Med 37:495–503 Morgan VA, McGrath JJ, Jablensky A, Badcock JC, Waterreus A, Bush R, Carr V, Castle D, Cohen M, Galletly C, Harvey C, Hocking B, McGorry P, Neil AL, Saw S, Shah S, Stain HJ, Mackinnon A (2013) Psychosis prevalence and physical, metabolic and cognitive co-morbidity: data from the second Australian national survey of psychosis. Psychol Med 23:1–14 Niitsu T, Shirayama Y, Matsuzawa D, Hasegawa T, Kanahara N, Hashimoto T, Shiraishi T, Shiina A, Fukami G, Fujisaki M, Watanabe H, Nakazato M, Asano M, Kimura S, Hashimoto K, Iyo M (2011) Associations of serum brain-derived neurotrophic factor with cognitive impairments and negative symptoms in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 35:1836–1840 Niu C, Yip HK (2011) Neuroprotective signaling mechanisms of telomerase are regulated by brain-derived neurotrophic factor in rat spinal cord motor neurons. J Neuropathol Exp Neurol 70(7):634–652 O’Donnell P, Lewis BL, Weinberger DR, Lipska BK (2002) Neonatal hippocampal damage alters electrophysiological properties of prefrontal cortical neurons in adult rats. Cereb Cortex 12:975–982 Onishi T, Iwashita H, Uno Y, Kunitomo J, Saitoh M, Kimura E, Fujita H, Uchiyama N, Kori M, Takizawa M (2011) A novel glycogen synthase kinase-3 inhibitor 2-methyl-5-(3-{4-[(S )-methylsulfinyl]phenyl}-1-benzofuran-5-yl)-1,3,4-oxadiazole decreases tau phosphorylation and ameliorates cognitive deficits in a transgenic model of Alzheimer’s disease. J Neurochem 119: 1330–1340 Pae CU, Chiesa A, Porcelli S, Han C, Patkar AA, Lee SJ, Park MH, Serretti A, De Ronchi D (2012) Influence of BDNF variants on diagnosis and response to treatment in patients with major depression, bipolar disorder and schizophrenia. Neuropsychobiology 65(1):1–11 Palomino A, Vallejo-Illarramendi A, Gonzalez-Pinto A, Aldama A, Gonzalez-Gomez C, Mosquera F, Gonzalez-Garcia G, Matute C (2006) Decreased levels of plasma BDNF in first-episode schizophrenia and bipolar disorder patients. Schizophr Res 6:321–322 Park SW, Lee JG, Kong BG, Lee SJ, Lee CH, Kim JI, Kim YH (2009) Genetic association of BDNF val66met and GSK-3beta-50T/C polymorphisms with tardive dyskinesia. Psychiatry Clin Neurosci 63:433–439 Pasternak O, Westin CF, Bouix S, Seidman LJ, Goldstein JM, Woo TU, Petryshen TL, Mesholam-Gately RI, McCarley RW, Kikinis R, Shenton ME, Kubicki M (2012) Excessive extracellular volume reveals a neurodegenerative pattern in schizophrenia onset. J Neurosci 32(48):17365–17372. doi:10.1523/JNEUR OSCI.2904-12.2012 Pedrini M, Chendo I, Grande I, Lobato MI, Belmonte-de-Abreu PS et al (2011) Serum brain-derived neurotrophic factor and clozapine daily dose in patients with schizophrenia, a positive correlation. Neurosci Lett 491:207–210 Pert L, Ferriter M, Saul C (2004) Parental loss before the age of 16 years: a comparative study of patients with personality disorder and patients with schizophrenia in a high secure hospital’s population. Psychol Psychother 77(Pt 3):403–407 Powell SB, Sejnowski TJ, Behrens MM (2012) Behavioral and neurochemical consequences of cortical oxidative stress on parvalbumin-interneuron maturation in rodent models of

schizophrenia. Neuropharmacology 62(3):1322–1331. doi:10. 1016/j.neuropharm.2011.01.049 Ricci S, Businaro R, Massoni F, Ippoliti F, Troili GM, Pontecorvi V, Morelli M, Rapp Ricciardi M, Archer T (2013) Altered cytokine and BDNF levels in autism spectrum disorder. Neurotox Res. doi:10.1007/s12640-013-9393-4 Rice D, Barone S (2000) Critical periods of vulnerability for the developing nervous system: evidence from human and animal models. Environment Health Perspect 108(Suppl. 3):511–533 Rizos EN, Rontos I, Laskos E, Arsenis G, Michalopoulos PG et al (2008) Investigation of serum BDNF levels in drug-naive patients with schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 32:1308–1311 Rizos EN, Papadopoulou A, Laskos E, Michalopoulos PG, Kastania D et al (2010) Reduced serum BDNF levels in patients with chronic schizophrenic disorder in relapse, who were treated with typical or atypical antipsychotics. World J Biol Psychiatry 1:251–255 Rosenberg SD, Lu W, Mueser KT, Jankowski MK, Cournos F (2007) Correlates of adverse childhood events among adults with schizophrenia spectrum disorders. Psychiatr Serv 58:245–253 Rothlisberger M, Riecher-Rossler A, Aston J, Fusar-Poli P, Radu EW, Borgwardt S (2012) Cingulate volume abnormalities in emerging psychosis. Curr Pharm Des 18(4):495–504 Sawa A, Snyder SH (2002) Schizophrenia: diverse approaches to a complex disease. Science 296:692–695 Shenton ME, Dickey CC, Frumin M, McCarley RW (2001) A review of MRI findings in schizophrenia. Schizophr Res 49(1–2):1–52 Shinohara M, Ybanez MD, Win S, Than TA, Jain S, Gaarde WA, Han D, Kaplowitz N (2010) Silencing glycogen synthase kinase3beta inhibits acetaminophen hepatotoxicity and attenuates JNK activation and loss of glutamate cysteine ligase and myeloid cell leukemia sequence 1. J Biol Chem 285:8244–8255 Singh KK, De Rienzo G, Drane L, Mao Y, Flood Z, Madison J, Ferreira M, Bergen S, King C, Sklar P, Sive H, Tsai LH (2011) Common DISC1 polymorphisms disrupt Wnt/GSK3B signaling and brain development. Neuron 72:545–558 Singh S, Goyal S, Modi S, Kumar P, Singh N, Bhatia T, Deshpanda SN, Khushu S (2014) Motor function deficits in Schizophrenia: an fMRI and VBM study. Neuroradiology. doi:10.1007/s00234014-1325-3 Smieskova R, Fusar-Poli P, Allen P, Bendfeldt K, Stieglitz RD, Drewe J, Radue EW, McGuire PK, Riecher-Ro¨ssler A, Borgwardt SJ (2010a) Neuroimaging predictors of transition to psychosis–a systematic review and meta-analysis. Neurosci Biobehav Rev 34(8):1207–1222. doi:10.1016/j.neubiorev.2010. 01.016 Smieskova R, Fusar-Poli P, Allen P, Bendfeldt K, Stieglitz R, Drewe J, Radue E, McGuire P, Riecher-Ro¨ssler A, Borgwardt S (2010b) Neuroimaging predictors of transition to psychosis—a systematic review and meta-analysis. Neurosci Biobehav Rev 34(8):1207–1222 Smieskova R, Fusar-Poli P, Aston J, Simon A, Bendfeldt K, Lenz C, Stieglitz RD, McGuire P, Riecher-Rossler A, Borgwardt SJ (2012a) Insular volume abnormalities associated with different transition probabilities to psychosis. Psychol Med 42(8): 1613–1625. doi:10.1017/S0033291711002716 Smieskova R, Fusar-Poli P, Riecher-Rossler A, Borgwardt S (2012b) Neuroimaging and resilience factors - staging of the at-risk mental state? Curr Pharm Des 18(4):416–421 Smieskova R, Allen P, Simon A, Aston J, Bendfeldt K, Drewe J, Gruber K, Gschwandtner U, Klarhoefer M, Lenz C, Scheffler K, Stieglitz RD, Radue EW, McGuire P, Riecher-Rossler A, Borgwardt SJ (2012c) Different duration of at-risk mental state associated with neurofunctional abnormalities. A multimodal

123

Neurotox Res imaging study. Hum Brain Mapp 33(10):2281–2294. doi:10. 1002/hbm.21360 Song X, Fan X, Song X, Zhang J, Zhang W, Li X, Gao J, Harrington A, Ziedonis D, Lv L (2013) Elevated levels of adiponectin and other cytokines in drug naı¨ve, first episode schizophrenia patients with normal weight. Schizophr Res 150(1):269–273. doi:10.1016/j.schres.2013.07.044 Stefanis NC, Dragovic M, Power BD, Jablensky A, Castle D, Morgan VA (2013) Age at initiation of cannabis use predicts age at onset of psychosis: the 7- to 8-year trend. Schizophr Bull 39(2): 251–254. doi:10.1093/schbul/sbs188 Stone JM, Day F, Tsagaraki H, Valli McLean MA et al (2009) Glutamate dysfunction in people with prodromal symptoms of psychosis: relationship to grey matter volume. Biol Psychiatry 66:533–539 Sullivan PF, Kendler KS, Neale MC (2003) Schizophrenia as a complex trait: evidence from a meta-analysis of twin studies. Arch Gen Psychiatry 60:1187–1192 Suzuki A, Stern SA, Bozdagi O, Huntley GW, Walker RH, Magistretti PJ, Alberini CM (2011) Astrocyte-neuron lactate transport is required for long-term memory formation. Cell 144:810–823 Swanson RA (1992) Physiologic coupling of glial glycogen metabolism to neuronal activity in brain. Can J Physiol Pharmacol 70(Suppl):S138–S144 Takei Y, Suda M, Aoyama Y, Yamaguchi M, Sakurai N, Narita K, Fukuda M, Mikuni M (2013) Temporal lobe and inferior frontal gyrus dysfunction in patients with schizophrenia during face-toface conversation: a near-infrared spectroscopy study. J Psychiatr Res 47(11):1581–1589. doi:10.1016/j.jpsychires.2013.07.029 Tan YL, Zhou DF, Zhang XY (2005) Decreased plasma brain-derived neurotrophic factor levels in schizophrenic patients with tardive dyskinesia: association with dyskinetic movements. Schizophr Res 74:263–270 Tayoshi S, Sumitani S, Taniguchi K, Shibuya-Tayoshi S, Numata S et al (2009) Metabolite changes and gender differences in schizophrenia using 3-Testa proton magnetic resonance spectroscopy (1H-MRS). Schizophr Res 108:69–77 The´berge J, Williamson KE, Aoyama N, Drost DJ, Manchanda R, Malla AK, Northcott S, Menon RS, Neufeld RW, Rajakumar N, Pavlosky W, Densmore M, Schaefer B, Williamson PC (2007) Longitudinal grey-matter and glutamatergic losses in firstepisode schizophrenia. Br J Psychiatry 191:325–334 Tibbo P, Hanstock C, Valiakalayil A, Allen P (2004) 3-T proton MRS investigation of glutamate and glutamine in adolescents at high genetic risk for schizophrenia. Am J Psychiatry 161:1116–1118

123

Tosic M, Ott J, Barral S, Bovet P, Deppen P et al (2006) Schizophrenia and oxidative stress: glutamate cysteine ligase modifier as a susceptibility gene. Am J Hum Genet 79:586–592 Traver S, Marien M, Martin E, Hirsch EC, Michel PP (2006) The phenotypic differentiation of locus coeruleus noradrenergic neurons mediated by brain-derived neurotrophic factor is enhanced by corticotrophin releasing factor through the activation of a cAMP-dependent signaling pathway. Mol Pharmacol 70:30–40 Wender R, Brown AM, Fern R, Swanson RA, Farrell K et al (2000) Astrocytic glycogen influences axon function and survival during glucose deprivation in central white matter. J Neurosci 20:6804–6810 Wexler EM, Geschwind DH (2011) DISC1: a schizophrenia gene with multiple personalities. Neuron 72:501–503 Yang YQ, Sun S, Yu YQ, Li WJ, Zhang X, Xiu MH (2011) Decreased serum brain-derived neurotrophic factor levels in schizophrenic patients with tardive dyskinesia. Neurosci Lett 502:37–40 Yoshimura R, Hori H, Sugita N, Ueda N, Kakihara S (2007) Treatment with risperidone for 4 weeks increased plasma 3-methoxy-4-hydroxyphenylglycol (MHPG) levels, but did not alter plasma brain-derived neurotrophic factor (BDNF) levels in schizophrenic patients. Prog Neuropsychopharmacol Biol Psychiatry 31:1072–1077 Yoshimura R, Ueda N, Hori H, Ikenouchi-Sugita A, Umene-Nakano W et al (2010) Different patterns of longitudinal changes in plasma levels of catecholamine metabolites and brain-derived neurotrophic factor after administration of atypical antipsychotics in first episode untreated schizophrenic patients. World J Biol Psychiatry 11:256–261 Yu Q, Plis SM, Erhardt EB, Allen EA, Sui J, Kiehl KA, Pearlson G, Calhoun VD (2011) Modular organization of functional network connectivity in healthy controls and patients with schizophrenia during the resting State. Front Syst Neurosci 5:103 Zai CC, Tiwari AK, De Luca V, Muller DJ, Bulgin N et al (2009) Genetic study of BDNF, DRD3, and their interaction in tardive dyskinesia. Eur Neuropsychopharmacol 19:317–328 Zhang XY, Xiu MH, da Chen C, Zhu FY, Wu GY, Haile CN, Lu L, Kosten TA, Kosten TR (2010) Increased S100B serum levels in schizophrenic patients with tardive dyskinesia: association with dyskinetic movements. J Psychiatr Res 44:429–433 Zhang XY, Liang J, Chen DC, Xiu MH, De Yang F, Kosten TA, Kosten TR (2012) Low BDNF is associated with cognitive impairment in chronic patients with schizophrenia. Psychopharmacology 222(2):277–284. doi:10.1007/s00213-012-2643-y

Neurodegenerative aspects in vulnerability to schizophrenia spectrum disorders.

The neurodegenerative and neurotoxic aspects of schizophrenia and/or psychosis involve genetic, epigenetic, and neurotoxic propensities that impinge u...
334KB Sizes 1 Downloads 3 Views