Skip to main content

REVIEW article

Front. Psychiatry, 05 December 2018
Sec. Psychological Therapy and Psychosomatics
This article is part of the Research Topic Brain-Metabolic Crossroads in Severe Mental Disorders – Focus on Metabolic Syndrome View all 11 articles

Antipsychotics, Metabolic Adverse Effects, and Cognitive Function in Schizophrenia

  • 1Centre for Addiction and Mental Health, Toronto, ON, Canada
  • 2Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
  • 3Department of Psychiatry, University of Toronto, Toronto, ON, Canada
  • 4Women's College Hospital, Toronto, ON, Canada

Cognitive impairment is a core symptom domain of schizophrenia. The effect of antipsychotics, the cornerstone of treatment in schizophrenia, on this domain is not fully clear. There is some evidence suggesting that antipsychotics may partially improve cognitive function, and that this improvement may vary depending on the specific cognitive domain. However, this research is confounded by various factors, such as age, duration/stage of illness, medication adherence, and extrapyramidal side effects that complicate the relationship between antipsychotics and cognitive improvement. Furthermore, antipsychotics—particularly the second generation, or “atypical” antipsychotics—can induce serious metabolic side effects, such as obesity, dyslipidemia and type 2 diabetes, illnesses which themselves have been linked to impairments in cognition. Thus, the inter-relationships between cognition and metabolic side effects are complex, and this review aims to examine them in the context of schizophrenia and antipsychotic treatment. The review also speculates on potential mechanisms underlying cognitive functioning and metabolic risk in schizophrenia. We conclude that the available literature examining the inter-section of antipsychotics, cognition, and metabolic effects in schizophrenia is sparse, but suggests a relationship between metabolic comorbidity and worse cognitive function in patients with schizophrenia. Further research is required to determine if there is a causal connection between the well-recognized metabolic adverse effects of antipsychotics and cognitive deficits over the course of the illness of schizophrenia, as well as, to determine underlying mechanisms. In addition, findings from this review highlight the importance of monitoring metabolic disturbances in parallel with cognition, as well as, the importance of interventions to minimize metabolic abnormalities for both physical and cognitive health.

Introduction

Schizophrenia is a severe psychiatric disorder characterized by a wide range of symptoms. These include positive symptoms, such as hallucinations and delusions, negative symptoms, such as apathy and amotivation, and impaired cognition (1). From the advent of chlorpromazine in the 1950s, the first antipsychotic (AP) for the treatment of schizophrenia, to today, APs uniformly alleviate positive symptoms with minimal effect on negative symptoms (2). Treatment with APs may be associated with a modest positive impact on cognitive functioning but there are many caveats to this finding (3). Furthermore, most APs, led by clozapine and olanzapine, cause serious metabolic side effects including weight gain, insulin resistance, and dyslipidemia. Independent studies suggest that about 50% of patients treated with APs develop metabolic complications (4, 5). These rates are even higher in young, first episode patients (6, 7). Cognitive impairment and metabolic aberrations have important functional and physical consequences. Schizophrenia continues to be associated with severe disability, owing largely to cognitive impairments (2), while metabolic illness contributes to decreased patient lifespan by about 20 years due to cardiovascular disease (8). In addition, these two critical domains of health and functioning might interact, as metabolic dysregulation is associated with impaired cognition in both patients with schizophrenia (9), as well as, the non-psychiatrically ill (10). The inter-relationships between cognition and metabolic side effects are therefore complex, and this review aims to examine them in the context of schizophrenia and AP treatment.

Cognition and Schizophrenia

Cognition is broadly defined as the ability to accurately perceive, attend to, process and remember information (2). An impairment in cognition is a hallmark symptom experienced by individuals with schizophrenia, and has been postulated to be a core aspect of the disorder (11). Important work over the last couple of decades has informed our understanding of the nature and properties of these deficits and has been extensively reviewed elsewhere (3, 12, 13). The field, however, is characterized by heterogeneous findings and many unanswered questions. This has been fueled, in part, by heterogeneity in the clinical profiles of the participants recruited and the methods used for cognitive testing. However, there are certain broad points of agreement that are briefly summarized below.

Cognitive deficits are observed in the majority of patients with schizophrenia, and this deficit is robust with a large effect size (3, 13). However, inter-subject variability is also an important aspect of this impairment. There is both a generalized impairment of cognition, as well, as impairment in several specific areas of functioning (14). These specific domains include speed of processing, attention, working memory, verbal learning and memory, visual learning and memory, reasoning and problem solving, and social cognition (15). The deficits are present pre-morbidly and tend to persist throughout the course of the illness (12). The course of these deficits is not completely clear, however, but in general have a weak relationship with positive symptoms, and a stronger association with the severity of negative symptoms (16). These impairments are also seen in unaffected relatives, albeit to a lesser degree, suggesting genetic underpinnings and shared risk among family members of schizophrenia patients (17). As will be reviewed, emerging evidence suggests that impairment in cognitive functioning may be exacerbated in the presence of metabolic comorbidity, which may in part be caused by the main-stay treatment for psychosis (i.e., AP medications) (9). Cognitive impairments are of fundamental importance since they substantially change the way individuals interact with their environment and have been linked directly and robustly to social and vocational functioning outcomes (2). This impact on functional outcomes makes it imperative that we understand fully the effect of treatment with AP medications on cognitive functioning. This includes consideration of medication-associated metabolic side effects and whether these may limit treatment efficacy in this critical illness domain.

Antipsychotics and Cognition

Given that antipsychotics are the cornerstone of schizophrenia treatment, the interaction of APs with cognitive impairment is of critical importance when looking at treatment outcomes. In spite of the large number of studies evaluating this relationship, however, the effects of AP medications on cognition remains controversial (13). Historically, the so-called “first generation” or “conventional” class of APs (FGA) was considered to have neutral or even detrimental effects on cognitive functioning. The introduction of so called “atypical,” or second-generation APs (SGA) spawned hope that these newer medications would improve cognitive functioning relative to their “first generation” counterparts. Indeed, soon after the introduction of the newer APs, several studies seemed to suggest that treatment with these compounds could improve cognitive functioning in schizophrenia patients (18, 19). For example, multiple studies have shown that SGAs improved set shifting ability (13), a component of cognitive flexibility. Clozapine has shown large positive effects in attention and verbal fluency with modest improvements in executive functioning (20) and delayed recall (21). Olanzapine has also demonstrated significant improvement in vigilance, selective attention, delayed recall, as well as, verbal learning and memory, verbal fluency, and executive functioning (20, 21). Risperidone has generally shown more modest effects in comparison to the aforementioned medications, demonstrating moderate improvements in working memory, executive functioning, attention (20, 21), and delayed recall (21). Aripiprazole has been shown to improve reaction time with correct responses to stimuli (22), as well as, verbal cognitive functioning (23) while quetiapine has been shown to improve global cognitive functioning in the early stages of treatment (2426) and verbal short-term memory (27).

While there is strong support for the effect of SGAs and their impact on cognition, more recent work has challenged the finding that they are superior to FGAs in this regard. Results from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) study, which included a large sample size of chronic schizophrenia patients and sample characteristics considered reflective of the general schizophrenia population, indicated that APs are very similar in their action across chemical classes, and this similarity extends to the effects APs have on cognition. Additionally, the effect size for improvement in cognition was found to be small (28), with questionable clinical significance (29). The debatable superiority of SGAs on cognitive functioning was also highlighted by a meta-analysis that suggested that “older” drugs did, in fact, have a moderate beneficial effect on cognition (30). Two recent network based meta-analyses have shed further light on this question (31, 32). When nine studies investigating long-term (>6 months) effect of APs were meta-analyzed, olanzapine and quetiapine emerged superior (32), but when all studies longer than 8 weeks were considered, there were no clear differences between antipsychotics (31). Furthermore, short follow-ups and methodologies that do not allow for a confident discounting of practice effects raise questions over the claim that APs significantly improve cognition, as practice effects may contribute to the modest gains seen across treatments (12, 28).

There are several confounders that likely have a bearing on this relationship including general symptom improvement, the stage, and duration of the illness, dose of the AP, adherence to treatment, medication-related sedation, and anticholinergic side effects. Many of these aspects have been investigated and reviewed in detail elsewhere (3337), and are summarized briefly here.

General Symptom Improvement with AP Treatment in Relation to Cognition

The degree to which general symptom improvement contributes to the changes in cognitive outcomes seen with AP treatment is an important consideration (38). More specifically, it is important to think about how different symptom domains interact (i.e., positive, negative, cognitive) and whether improvement of some symptoms relates to improvement in others. Studies differ with respect to their findings in this area. For example, multiple studies have demonstrated that although positive (and to some degree negative) symptoms may improve with AP use, these changes do not interact with executive functioning (27, 3941). This also speaks to the notion that certain aspects of cognitive deficits (i.e., cognitive flexibility) may be inherent to the illness of schizophrenia, occurring independently of positive or negative symptomatology (4042). Conversely, there is also evidence to suggest that improvements in cognition correlate with improvements in symptomology. For example, improvement in processing speed has been found to relate to negative symptom improvement, where improvement in negative symptoms occurred alongside improvements in processing speed after 24 weeks of treatment with a second generation antipsychotic (43). Taken together, the possibility arises that symptoms impact specific facets of cognition, rather than impacting global cognitive function.

Illness Course and Stage of Intervention in Relation to Cognitive Improvement

It may also be important to consider the stage at which treatment begins when looking for interactions between improvement in specific symptom domains and subsequent cognitive improvement. A recent review suggests that the small cognitive improvement seen after AP treatment initiation is possibly due to an improvement in psychotic symptoms, and that there is not much further improvement in cognitive outcomes beyond the first 1–2 years of treatment (12). This is in agreement with studies that have evaluated cognition in patients either over the course of illness or at varied time points. In first-episode patients, significant improvement in cognition has been reported across many studies and small but significant cognitive improvements related to AP treatment have been seen in first-episode patients as early as 3 months into treatment, in correlation with positive symptom alleviation (44). Significant improvements also continue to be seen in this population at three (45) and 5-years follow-up (46), supporting the idea that if treatment is adequate in the early stages of psychosis, greater improvements in cognition are evident without later decline (45). These outcomes are different than what has been observed in chronic schizophrenia. Studies in chronically ill patients often show little improvement in cognitive symptoms with AP treatment; one study which looked at patients with chronic schizophrenia found no cognitive improvement at 6-months follow-up, and found that this lack of improvement was independent of positive symptom alleviation, as rated on the Positive and Negative Syndrome Scale (PANSS) (47). Cognitive functioning in the chronic stage of the illness is sometimes described as static, as even at a minimum of a year follow-up, no significant improvements are evident (48). Therefore, it appears that the potential cognitive gains with AP treatment are most prominent during the early stages of the illness.

Antipsychotic dose and Adherence in Relation to Cognitive Improvement

There are several other factors that may impact the extent to which APs augment cognitive outcomes. The dose of AP medication may have an effect on cognition, with higher AP doses, as well as, AP polypharmacy, reported to be associated with worse cognitive functioning (49, 50). Further to this point, it has been reported that when AP dose is reduced, cognition significantly improves in several domains, including memory, visuospatial, language, attention, and delayed memory (5154). There may also be a relationship between dose and length of time receiving treatment with high vs. low AP dose. One study has shown that with higher doses of APs taken over the long term, verbal learning and recall performance decline significantly over time, independently of age of illness onset or severity of illness. Interestingly, there were no significant differences in cognitive decline observed between low-dose cases and non-psychiatric controls over the 9-years naturalistic follow up (50). Conversely, in a meta-analysis of older schizophrenia patients, medication status or dosing according to chlorpromazine equivalence failed to demonstrate a significant association with cognition over 1–6 years of follow-up (55).

Medication adherence also contributes to the relationship between AP and cognitive functioning. In studies where medication adherence was considered, those who were adherent, not surprisingly, have been reported to show greater improvements in cognition compared to those who did not take their medications regularly (37). However, whether poor adherence leads to a lack of improvement in cognition or inherent cognitive deficits related to the diagnosis of schizophrenia lead to poor medication adherence remains an unanswered question. Further to this point, a recent review has shown that long acting injectable (LAI) agents show superiority in terms of efficacy when compared to oral agents (56), with reported improvement in general cognitive performance in individuals switching from oral to injectable agents (57). In a direct comparison between risperidone in LAI vs. oral form, it was found that white matter and myelination increased with LAI use while it declined in oral risperidone use (58). This was attributed to adherence to medication (58) and suggests that perhaps adherence to treatment may impact the extent to which cognitive benefits are seen.

Extrapyramidal Adverse Effects and Cognition

Extrapyramidal symptoms (EPS) represent one of the more well-known side-effects of AP medications. While these side-effects have been found to be correlated with impaired cognitive abilities, such as set-shifting (59), it appears that cognitive impairments may be associated with EPS even in the absence of active antipsychotic treatment (35). This raises the question of whether the apparent impairment in cognition is a result of the medication or inherently linked to the illness and its premorbid cognitive dysfunction. Interestingly, with the introduction of SGAs less focus has been given to EPS and studies generally do not report significant correlations with cognition (6062).

With this said, in cases where EPS is present, it is also critical to account for the role of anticholinergic treatments adjunct and inherent within antipsychotics when evaluating cognitive outcomes. Recent reviews have found that co-treatment with anticholinergic medications have a negative effect on cognition (36, 63), with individuals with schizophrenia particularly susceptible to these effects (64). The aspects of cognition which are most commonly impaired include memory and attention (64). Of note, cognition improves when anticholinergic treatment is discontinued (36, 63).

While each of the above-mentioned factors are important, another facet of AP effects that has not received adequate attention is the effect of AP-induced metabolic dysfunction on cognition. This presents a potentially modifiable therapeutic target (i.e., metabolic syndrome), the management of which can potentially lead to better clinical outcomes.

Antipsychotics and Metabolic Dysregulation

Although “atypical” or SGAs, as a class were associated with higher metabolic risk than first-generation antipsychotics, a more nuanced understanding of this “class” [which in itself has become a topic of controversy (65, 66)], has shown that there is a differential metabolic liability [i.e., olanzapine = clozapine > sertindole > risperidone > = quetiapine > = amisulpride > ziprasidone, lurasidone, aripiprazole 33, 67] among the different constituent medications classified as an SGA. More recent work, however, suggests that all AP drugs are associated with early significant weight gain (68), particularly in young AP-naïve patients (6972). The effect on weight gain is also of FGAs, which have historically been considered to be metabolically neutral; haloperidol has been found to cause significant weight gain in AP-naïve patients, with an average 3.8 kg weight gain within the first 3 months of treatment (70). Interestingly, while long-term prospective trials examining AP-induced weight gain are lacking, it has been demonstrated that when drug-naïve first episode patients are followed over the long-term (>2 years), the differences between individual antipsychotic agents disappear (70). This may suggest that treatments differ more by pattern of weight gain, rather than amount.

Similar to the trends seen with weight gain, studies published recently show concerning rates of prediabetes (>15%) in first episode patients within 6 months of AP exposure (73), and significantly higher cumulative risk and incidence of type two diabetes (T2D) (74). A recent population-wide national registry estimated that APs, irrespective of whether they were FGAs or SGAs increased the risk of diabetes 3-fold in patients with schizophrenia (75). Moreover, while overweight and obesity represent leading risk factors for diabetes, work in animals and humans consistently demonstrates that APs can directly induce insulin resistance and glucose dysregulation even in absence of weight or adiposity changes (76). Notably, initial concern with metabolic side-effects linked to these agents was focused primarily on cardiometabolic morbidity and mortality (5). These concerns have been extended to quality of life, self-esteem, medication adherence (77), and most recently, as will be reviewed in detail, potentially to cognitive functioning.

Metabolic Dysregulation and Cognition in Non-Psychiatrically ILL Populations

The term metabolic syndrome (MetS) refers to the presence of multiple, interrelated cardiovascular risk factors occurring simultaneously (78, 79). While specific criteria for MetS differ according to different guidelines, in general, three out of the five following criteria must be met: increased waist circumference, elevated triglycerides, reduced high-density lipoprotein (HDL), cholesterol, elevated blood pressure, and elevated fasting glucose (78, 80). There is a large body of data demonstrating impairments in cognition as a result of MetS in psychiatrically non-ill individuals (10, 8185) and a recent literature review found a strong link between MetS and specific deficits in memory, visuospatial abilities, executive function, processing speed, and intellectual function (86). There are also significant correlations between specific components of MetS and cognition. For example, patients with hypertension performed significantly worse on executive functioning tasks, which was correlated with reduced frontal lobe volume and impaired glucocorticoid feedback (87). Moreover, high body mass index (BMI) has been found to correlate with lower cognition scores (12, 95).

An accumulating body of evidence suggests a mechanistic relationship between cognitive decline, obesity, MetS, diabetes, and Alzheimer's disease (88, 89) with shared neuropathological characteristics and insulin resistance, oxidative stress, and a persistent inflammatory state as the core pathology. These lines of evidence suggest a tight link between cognitive and metabolic processes and outcomes, which would be predicted to extend to patients with schizophrenia as well. If this association does hold in patients with schizophrenia, an additional intriguing question is also the role of AP-related metabolic perturbations in the context of AP-related effects (or lack there-of) on cognitive function. Thus, focus must be turned to: (1) the association between MetS and cognitive deficits in schizophrenia, and, (2) whether AP-related metabolic dysfunction could, in part, mediate this association.

Metabolic Dysregulation and Cognition in Schizophrenia

MetS occurs in ~33.5% of patients with schizophrenia (90). There are multiple factors which contribute to the elevated prevalence rate of MetS in the schizophrenia population, including lifestyle factors, such as smoking (91), poor eating habits and sedentary behavior (see Ringen (92) for an in-depth review). Biological and genetic factors inherent to the illness of schizophrenia and treatment response also overlap with genes related to metabolic function (93). For example, genes associated with the illness of schizophrenia have independently been linked to regulation of fat mass, leptin signaling (94), insulin signaling, glucose metabolism, and inflammation (95, 96). In addition, as reviewed here, antipsychotic medications are well-established to contribute to cardiometabolic risk in this already metabolically vulnerable population. Some of these agents are also implicated in cognition. The serotonin receptor gene 5HT2A has been found to influence both lipid levels and glucose intolerance (94), as well as, attention span and cognitive flexibility (97). As well, the methylenetetrahydrofolate reductase (MTHFR) gene is believed to increase the susceptibility of developing MetS in patients with schizophrenia (94), and is also implicated in poorer verbal recall and cognitive flexibility (98). Thus, it appears that the genes implicated in poor metabolic outcomes for individuals with schizophrenia could also be associated with cognitive outcomes.

Taken together, it is perhaps not surprising that the majority of studies in schizophrenia suggest that the relationship between MetS and cognitive dysfunction is similar among schizophrenia patients as it is to non-psychiatrically ill individuals. MetS has a negative impact on cognition, and indeed schizophrenia patients with MetS have been reported to perform worse on measures of cognition than those with schizophrenia in the absence of MetS (9, 99, 100). Performance appears to be negatively impacted on tasks measuring processing speed, memory, attention, and reasoning in those with co-morbid MetS as compared to those with schizophrenia alone (9, 99, 100). Furthermore, this impairment has been shown to develop post-morbidly (101), suggesting that there could be an active mechanism in the developmental course of the disorder that may impair cognition as one's metabolic health declines.

With that said, not all studies have found a relationship between MetS and cognitive impairment in schizophrenia. As reviewed by Bora and colleagues, the CATIE trial stands out as the largest study which failed to find an association (102). Other studies, which may have failed to find an association between MetS and cognitive dysfunction have however found that specific aspects of MetS (i.e., hypertension) were correlated with lower cognitive scores, whereas other factors, such as increased waist circumference and dyslipidemia were less consistently associated with general impairments in cognition in schizophrenia (49, 103). Other factors, which constitute a poor metabolic profile, such as high BMI, have also not consistently been found to predict poor cognitive functioning across several cognitive domains (104). Furthermore, in one study, hyperglycemia was actually found to predict better verbal memory performance (49).

The discrepancy in the schizophrenia literature examining associations between metabolic comorbidity and cognitive deficits could, in part, be attributable to the amalgamation of diverse metabolic aberrations under the common umbrella of MetS (i.e., considering MetS as merely a binary factor) and suggest value in considering cognition alongside individual metabolic symptoms. Indeed, studies that failed to find a relationship between metabolic aberrations and cognitive functioning, such as CATIE study (102), also did not examine effects of individual components of MetS in relation to cognition. This suggests that individual metabolic outcomes might interact independently with cognitive abilities and should be studied as distinct entities to understand the relationship between metabolic and cognitive parameters better (102).

Disentangling Antipsychotic-Induced Metabolic Dysregulation in Relation to Cognition in Schizophrenia

Cognition is a core area of impairment in schizophrenia and the impact of APs on cognition is not fully clear. Also, there appear to be associations between cognition and metabolic parameters that are being gradually understood. In summary, independent relationships exist between APs, metabolic outcomes, and cognition: (i) APs might influence cognition (with benefits more consistently seen early on in illness, possibly in relation to improvements in other domains of psychopathology); (ii) APs cause metabolic dysregulation, and (iii) metabolic dysregulation has been linked to cognitive dysfunction, with the predominance of evidence suggesting this association applies to patients with SCZ. However, the majority of existing studies have looked at individual associations between these factors, leaving more complex interactions between these factors largely unexplored. Here, we attempt to identify and extrapolate associations between these three factors in the presence of schizophrenia.

There have been a small number of studies investigating the interaction of APs, metabolic factors, and cognition in schizophrenia (Supplementary Table 1). A study by Chen and colleagues examined metabolic side effects and cognition in first-episode patients to establish whether a cognitive impairment related to metabolic dysfunction would be observed in the early stages of illness and AP treatment (105). In first-episode, newly medicated patients, there was a generally worse metabolic profile and poorer cognitive performance compared to their healthy counterparts; however, the effect size was small. Chen et al. also demonstrated that an inverse relationship between cognition and metabolic symptoms within the newly medicated patients was present but that it was relatively small (105). The specific contribution of AP medications on metabolic comorbidity in relation to cognition was not examined in this cross-sectional design. In another study by Li and colleagues (99), cognitive performance of two groups of chronic schizophrenia patients, according to presence or absence of MetS was cross-sectionally compared, with secondary analyses considering relationships with disease course and antipsychotic medications. In the MetS group, cognition was significantly worse, as has been replicated in similar studies examining the interaction between APs, cognition, and metabolic outcomes (9). In the study by Li and colleagues, this association was attributed to higher fasting triglycerides and systolic blood pressure. Interestingly, however, the course of disease was significantly longer in the MetS compared to non-MetS group. Both duration of disease and use of FGAs (as compared to SGAs) were independently correlated with lower cognitive scores. The caveat here was that the most commonly prescribed FGA was chlorpromazine, which has been established to have a similar metabolic risk profile to higher metabolic liability SGAs, such as clozapine. Thus, while interpretation of specific contributing effects of APs according to predicted metabolic liability was not possible, duration of illness leading to cumulative effects of metabolic comorbidity appeared to emerge as a key predictive factor for worse cognitive function. Similar results were also seen in a study by Boyer et al, who found that longer disease duration was correlated with increased rates of MetS, and this was subsequently correlated with worse cognitive impairment, which was particularly robust in individuals taking second generation APs (106). Taken together, illness duration appears to be the more consistent factor linked to cognitive impairments.

Furthermore, specific components of MetS also appear to be differentially related to cognition in studies which considered the presence of AP medications. Hypertension has been shown to correlate with lower verbal cognition scores, including memory and fluency (49). Triglycerides have also been shown to correlate with poorer cognition scores (100), however this effect has been shown to cease to exist when cholesterol is controlled for (107). As for waist circumference (i.e., abdominal obesity), there is discord in the literature on whether it has an effect on cognition (100, 104) or if it does not (49).

From the evidence presented above, perhaps as illness course and treatment continues metabolic comorbidities accumulate and become more severe (in association with illness duration which is invariably linked in the majority of cases with longer exposure to medications); we subsequently see cumulative effects play out on cognition more clearly. This may explain why the relationship between cognitive performance and metabolic indicators appears to be more robust in patients with longer duration of illness. The accretion of structural changes in the brain (with illness progression, treatment, and metabolic abnormalities) may also contribute to this relationship, as cognitive performance has been shown to be associated with lower gray matter volume (108, 109).

Potential Mechanisms Underlying Cognitive Functioning and Metabolic Risk in Schizophrenia

While there is a paucity of literature examining the interrelation among APs, cognition, and metabolic disturbances in schizophrenia, the existing literature points to a number of potential mechanistic influences to explain the effects of APs on cognition and metabolic disturbances.

i. Direct receptor action: While all antipsychotics bind to the dopamine, D2 receptor, a majority of them bind to multiple other receptors including the histamine, serotonin, muscarinic and adrenergic receptors. Antagonism of each these receptor systems is independently known to affect cognition, as well as, metabolic outcomes, such as weight gain, insulin and glucose dysregulation, and dyslipidemia. Hence, it is quite plausible that the effects of APs on cognition and metabolic measures are really two sides of the same coin: as binding to a given receptor leads to downstream effects on both domains (110). For instance, acute dopamine depletion in humans and rodents reduces peripheral insulin sensitivity (111, 112), purportedly via central dopamine effects in the striatum. Moreover, reduced insulin sensitivity and obesity are associated with reduced dopamine synthesis capacity and endogenous dopamine levels in the striatum (112114). These neurochemical alterations in turn may be associated with poor cognition. Thus, it is plausible that central dopamine receptor blockade by antipsychotics in schizophrenia may produce both peripheral insulin resistance and poor cognition. Interestingly, evidence suggests that acute antipsychotic exposure does not alter striatal dopamine levels and dopamine synthesis capacity in first-episode patients with schizophrenia (115). While highly speculative, this high dopaminergic reserve in the first-episode may help potentially explain why metabolic side effects have not been related to cognitive deficits in first-episode patients.

ii. Gut microbiome mediated interactions: In addition to the mechanisms of direct receptor action, the role of the gut microbiome (GMB) represents another pathway that may mediate the relationship between cognitive and metabolic dysfunction in schizophrenia (116). The GMB is the collective term for the community of microorganisms residing in the digestive tract. High throughput sequencing is beginning to provide insight into the differences in GMB composition among those with and without schizophrenia. An increased alpha diversity in the GMB has been reported in patients with schizophrenia and has been found to be a significant predictor of schizophrenia status (117). Within the GMB of patients with schizophrenia, most microbial taxa are derived from bacteria and arachae (117). At the phylum level, individuals with schizophrenia appear to have higher proportions of Firmicutes and lower proportions of Bacteroidetes and Actinobacteria in comparison to healthy controls (118). Overall, individuals with schizophrenia have been found to have greater species diversity than those without (119). The severity of different symptom domains of schizophrenia has been found to be significantly correlated with increases in Lactobacillus (120). When looking solely at the interplay between the GMB and schizophrenia, it appears that abnormalities in the GMB contribute to the production of key molecules (such as Brain-derived neurotrophic factor) and/or the promotion of an intestinal immune response (120). Both human and mice studies have confirmed that an altered composition of the GMB influences adverse cognitive and metabolic changes seen in schizophrenia (121123). The relationship between the GMB and schizophrenia pathogenesis becomes even more complicated when antipsychotics are introduced. There are several overlapping sites of action among the GMB and APs (124), and some APs have been shown to modulate the composition of the GMB, which incidentally has been shown to cause weight gain (125). The interactions between schizophrenia, APs, and the GMB have gained increasing attention over the past few years, however to date only one study has investigated the use of antipsychotic and their effect on the GMB in humans (125). This lack of knowledge represents an opportunity to better understand the pathophysiology of schizophrenia concurrently to metabolic comorbidity associated with the GMB and may lead to new therapeutic interventions to target both psychopathological and metabolic indices.

iii. Central insulin resistance: While receptor action and gut microbiome mediated interactions may underlie these relationships, central insulin resistance might be the final common pathway by which metabolic and cognitive outcomes interact. Central insulin has been well-established to regulate cognition in humans. Intranasal insulin, which delivers insulin directly to the CNS, is demonstrated to enhance declarative and working memory (126). Thus, dysregulation or resistance to central insulin action could result in impaired cognition, and indeed, recent work suggests that insulin is the common link between metabolic disorders and disorders of cognition (127). Central insulin resistance has been found to be a central piece of the pathology in cognitive dysfunction in the context obesity and type 2 diabetes, as well as, in aging and dementia. Insulin resistance has been shown to be associated with impaired hippocampal synaptic plasticity and memory (128), as well as, neurogenesis (129). It is possible that this involves the palmitic acid pathway (128). Furthermore, addressing this pathology via administration of insulin intranasally is emerging as a possible treatment strategy in Alzheimer's dementia (130). Interestingly, work from our lab demonstrates that short term use of APs can induce central insulin resistance resulting in glucose dysregulation and changes in feeding independently of weight gain and other longer term metabolic dysfunction (131). The propensity of APs to cause insulin resistance both in the short term, and in the long term via changes in weight and adiposity, might explain, at least in part, worsening of cognition seen in schizophrenia patients with long standing illness who accumulate metabolic dysfunction over time.

iv. Microangiopathy: Another mechanism by which APs, metabolic abnormalities and cognition might interact is microangiopathy or microvascular abnormalities. Microangiopathy is a well-known consequence of metabolic abnormalities including diabetes and metabolic syndrome (132, 133), and is closely related with the development of insulin resistance (134). While classical targets include the eye, the kidney, and the peripheral nervous system, more recent work shows that the effect is more widespread and includes the brain and adipose tissue as well (132). Microangiopathic changes in the brain have been implicated in cognitive decline (135). Furthermore, there is preliminary evidence that APs may affect the neurovascular unit and small vessels in frontal cortex in a post-mortem study (136). Thus, it is possible that microangiopathy could be a potential mechanism by which APs, metabolic abnormalities and cognition interact. The gradual development of microangiopathy would also, in part, explain why the relationship between metabolic abnormalities and cognitive decline is seen later on in the illness course.

Thus, the effects of APs on cognition and metabolic outcomes likely result from a complex relationship between differential receptor binding, changes in glucose metabolism, insulin sensitivity, the gut microbiome, blood vessel related changes, along with other yet to be described mechanisms to manifest in the clinical outcomes seen. This complex interaction, combined with underlying pathophysiological and lifestyle factors, is likely responsible for the heterogeneity in results when addressing the relationship between APs, cognition, and metabolic disturbances.

Concluding Thoughts

There is still a great deal of uncertainty regarding the relationship between cognition, APs, MetS and schizophrenia. First, there are varying results regarding whether APs actually improve cognition, which aspects of cognition are improved, and the degree of improvement. APs are shown to cause metabolic dysregulation that has been shown to negatively impact cognition. It is tempting to simplify the scenario and assume that as APs induce metabolic dysfunction, cognition should therefore be impaired. However, this does not seem to consistently be the case, given that in some cases, though metabolic side effects were observed, this was not associated with impairments in cognition. As a case in point, young, AP-naïve individuals are most vulnerable to obesogenic effects of AP medications, yet, as reviewed here, the association between MetS and cognitive deficits appears to be most prominent in individuals in the chronic stage of illness. Moreover, it might be expected that individual AP agents that are associated with the greatest metabolic risk (i.e., olanzapine and clozapine), should be associated with worse cognitive functioning. Again, this does not appear to be the case. At first glance these observations would appear to argue against a direct association between AP-induced metabolic adverse effects and detrimental cognitive effects. However, several other points warrant consideration. Firstly, early weight gain, glucose, and lipid abnormalities induced by AP medications may not, in themselves, be sufficient to immediately translate to cognitive impairments. It is possible then that adverse metabolic side-effects of APs act as a catalyst for later development of microvascular insults to the brain that may take place several years following onset of individual metabolic comorbidities. It is perhaps also not surprising that a “between agent,” “according to metabolic liability” effect on cognition is not observed. As discussed in earlier sections, the between agent difference in metabolic liability appears to diminish overtime. Thus, it can be argued that individual differences between agents may be relatively less important than the acceleration in metabolic risk that occurs overtime.

To summarize, the relationship between metabolic comorbidity and worse cognitive function appears to be generalizable to patients with schizophrenia. The current literature, however, does not allow assessment and establishment of a causal connection between the well-recognized metabolic adverse effects of APs and cognitive deficits. A proposed threshold effect in the relationship between metabolic dysfunction and cognition could suggest that earlier on in treatment, metabolic adverse effects related to treatment may have a minimal impact on cognition. However, once an individual passes a certain threshold of metabolic dysregulation, they likely become susceptible to cognitive impairment. Thus, the impact of metabolic side effects may begin to override any positive or neutral effects on cognition observed in individual taking APs once this threshold (which may be dependent on many factors) is reached. Brains of schizophrenia patients show evidence of accelerated aging even early on in the illness (123). Thus, metabolic aberrations may interact with and add to the existing deficits, and thereby contribute to cognitive impairment, with brain aging perhaps playing a role. Like many psychiatric disorders, schizophrenia has been hypothesized to be a syndrome of accelerated aging (137, 138). Support for this stems from the shared syndromes and risk factors between schizophrenia and age related cognitive and metabolic chronic diseases, such as Alzheimer's Disease and Type II diabetes, respectively. Patients with schizophrenia have an increased risk of diabetes and experience an earlier onset of this illness (139). While the focus has been on AP-related metabolic dysregulation, we are reminded that lifestyle factors (smoking, dietary habits, activity levels) also play a critical role in accumulation of metabolic risk in severe mental illness, and both the primary disorder, as well as, accumulation of metabolic risk has been shown to accelerate aging related changes (140, 141).

Going forward, there are important therapeutic implications to keep in mind in regards metabolic disturbances and cognition. Lifestyle interventions, such as diet and exercise, to prevent and/or correct metabolic abnormalities (whether due to APs, lifestyle or illness pathophysiology) can have gains above and beyond physical health. In particular, the available literature suggests that improvement of metabolic status may improve or prevent further impairment of cognition in patients with schizophrenia. For example, a recent review by Firth et al. (142) found that aerobic exercise improved cognitive functioning in patients in schizophrenia (104). In addition, reduction/prevention of AP-induced metabolic side effects is particularly important in the AP-naive and youth population, as APs appear to have the greatest potential to improve cognition at the beginning of treatment, yet AP-naive population are at greatest risk of AP-induced metabolic side effects. This review highlights that assessment of metabolic factors should be considered when studying cognition in schizophrenia, as these metabolic disturbances may be influencing cognitive outcomes. Future studies investigating the mechanism behind the impact of metabolic disturbances upon cognition are suggested to allow for more specific treatments. Overall, more research is required to identify the complex interactions between APs, metabolic status, and cognition, in schizophrenia to enable patients to receive the maximum therapies for cognitive improvements.

Author Contributions

NM, CK, SA, and MH contributed to the conception and design of the review. NM, CK, SA, and MH wrote the first draft of the manuscript. KC-D, FC, PG, GR, VT, DM, AG-G, and AC wrote sections of the manuscript. All authors contributed to manuscript revision, read and approved the submitted version.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

SA is supported by the Hugh Sellers Post-doctoral Fellowship by the Banting and Best Diabetes Center, University of Toronto, and the Bebensee Schizophrenia Research Fellowship by the Ian Douglas Bebensee Foundation.

CK is supported by the Banting and Best Diabetes Center (BBDC), University of Toronto, and the CAMH Foundation Discovery Fund.

KC-D is supported by the Cleghorn Fellowship by the University of Toronto.

GR has received research support from the Research Hospital Fund–Canada Foundation, Novartis, and HLS.

VT is a speaker for Shire, Sunovion, Lunbeck, Valiant, and Novo Nordisk.

DM is supported by the Canadian Institutes of Health Research (CIHR Operating Grant MOP 142192), the National Institutes of Health (R01MH085801), the Centre for Addiction and Mental Health Foundation (Joanne Murphy Professorship) and has received a Brain & Behaviour Research (NARSAD) Independent Investigator Award.

AG-G has received support from the United States National Institute of Health, CIHR, OMHF, the Brain & Behavior Research Foundation (Formerly NARSAD), the Ontario Ministry of Health and Long-Term Care, Brain Canada Foundation, Centre for Aging & Brain Health Innovation (CABHI), Spark, and the Ontario Ministry of Research and Innovation Early Research Award.

MH has acted as a consultant for Alkermes. MH has received funding from Canadian Institutes of Health Research (CIHR), American Academy of Child and Adolescent Psychiatry (AACAP), Bating Research Foundation, Banting and Best Diabetes Centre (BBDC), and the Ontario Ministry of Research and Innovation, and the CAMH Foundation.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fpsyt.2018.00622/full#supplementary-material

References

1. Owen MJ, Sawa A, Mortensen PB. Schizophrenia. Lancet (2016) 388:86–97. doi: 10.1016/S0140-6736(15)01121-6

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Green MF. Impact of cognitive and social cognitive impairment on functional outcomes in patients with schizophrenia. J. Clin. Psychiatry (2016) 77(Suppl. 2):8–11. doi: 10.4088/JCP.14074su1c.02

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Tandon R, Nasrallah HA, Keshavan MS. Schizophrenia, “just the facts” 4. Clinical features and conceptualization. Schizophr Res. (2009) 110:1–23. doi: 10.1016/j.schres.2009.03.005

CrossRef Full Text | Google Scholar

4. Cohn T, Prud'homme D, Streiner D, Kameh H, Remington G. Characterizing coronary heart disease risk in chronic schizophrenia: high prevalence of the metabolic syndrome. Can J Psychiatry (2004) 49:753–60. doi: 10.1177/070674370404901106

PubMed Abstract | CrossRef Full Text | Google Scholar

5. McEvoy JP, Meyer JM, Goff DC, Nasrallah HA, Davis SM, Sullivan L, et al. Prevalence of the metabolic syndrome in patients with schizophrenia: baseline results from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) schizophrenia trial and comparison with national estimates from NHANES III. Schizophr Res. (2005) 80:19–32. doi: 10.1016/j.schres.2005.07.014

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Zipursky RB, Gu H, Green AI, Perkins DO, Tohen MF, McEvoy JP, et al. Course and predictors of weight gain in people with first-episode psychosis treated with olanzapine or haloperidol. Br J Psychiatry (2005) 187:537–43. doi: 10.1192/bjp.187.6.537

CrossRef Full Text | Google Scholar

7. Correll CU, Manu P, Olshanskiy V, Napolitano B, Kane JM, Malhotra AK. Cardiometabolic risk of second-generation antipsychotic medications during first-time use in children and adolescents. JAMA (2009) 302:1765–73. doi: 10.1001/jama.2009.1549

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Hennekens CH, Hennekens AR, Hollar D, Casey DE. Schizophrenia and increased risks of cardiovascular disease. Am Heart J. (2005) 150:1115–21. doi: 10.1016/j.ahj.2005.02.007

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Bora E, Akdede BB, Alptekin K. The relationship between cognitive impairment in schizophrenia and metabolic syndrome: a systematic review and meta-analysis. Psychol Med. (2017) 47:1030–40. doi: 10.1017/S0033291716003366

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Gold SM, Dziobek I, Sweat V, Tirsi A, Rogers K, Bruehl H, et al. Hippocampal damage and memory impairments as possible early brain complications of type 2 diabetes. Diabetologia (2007) 50:711–9. doi: 10.1007/s00125-007-0602-7

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Elvevag B, Goldberg TE. Cognitive impairment in schizophrenia is the core of the disorder. Crit Rev Neurobiol. (2000) 14:1–21. doi: 10.1615/CritRevNeurobiol.v14.i1.10

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Keefe RS. The longitudinal course of cognitive impairment in schizophrenia: an examination of data from premorbid through posttreatment phases of illness. J Clin Psychiatry (2014) 75(Suppl. 2):8–13. doi: 10.4088/JCP.13065su1.02

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Keefe RSE, Harvey PD. Cognitive impairment in schizophrenia. In: MA Geyer and G Gross editors. Novel Antischizophrenia Treatments. Berlin; Heidelberg, Springer Berlin Heidelberg (2012). p. 11–37.

Google Scholar

14. Dickinson D, Ragland JD, Gold JM, Gur RC. General and specific cognitive deficits in schizophrenia: Goliath defeats David? Biol Psychiatry (2008) 64:823–7. doi: 10.1016/j.biopsych.2008.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Nuechterlein KH, Barch DM, Gold JM, Goldberg TE, Green MF, Heaton RK. Identification of separable cognitive factors in schizophrenia. Schizophr Res. (2004) 72:29–39. doi: 10.1016/j.schres.2004.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Addington J, Addington D, Maticka-Tyndale E. Cognitive functioning and positive and negative symptoms in schizophrenia. Schizophr Res. (1991) 5:123–34. doi: 10.1016/0920-9964(91)90039-T

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Tandon R, Keshavan MS, Nasrallah HA. Schizophrenia, “Just the Facts”: what we know in 2008 part 1: overview. Schizophr Res. (2008) 100:4–19. doi: 10.1016/j.schres.2008.01.022

CrossRef Full Text | Google Scholar

18. Jann MW. Implications for atypical antipsychotics in the treatment of schizophrenia: neurocognition effects and a neuroprotective hypothesis. Pharmacotherapy (2004) 24:1759–83. doi: 10.1592/phco.24.17.1759.52346

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Harvey PD. Cognitive and functional effects of atypical antipsychotic medications. J Clin Psychiatry (2006) 67:e13. doi: 10.4088/JCP.1006e13

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Meltzer HY, McGurk SR. The effects of clozapine, risperidone, and olanzapine on cognitive function in schizophrenia. Schizophr Bull. (1999) 25:233–55. doi: 10.1093/oxfordjournals.schbul.a033376

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Woodward ND, Purdon SE, Meltzer HY, Zald DH. A meta-analysis of neuropsychological change to clozapine, olanzapine, quetiapine, and risperidone in schizophrenia. Int J Neuropsychopharmacol. (2005) 8:457–72. doi: 10.1017/S146114570500516X

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Goozee R, Reinders AA, Handley R, Marques T, Taylor H, O'Daly O, et al. Effects of aripiprazole and haloperidol on neural activation during the n-back in healthy individuals: a functional MRI study. Schizophr Res. (2016) 173:174–81. doi: 10.1016/j.schres.2015.02.023

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Bervoets C, Morrens M, Vansteelandt K, Kok F, de Patoul A, Halkin V, et al. Effect of aripiprazole on verbal memory and fluency in schizophrenic patients: results from the ESCAPE study. CNS Drugs (2012) 26:975–82. doi: 10.1007/s40263-012-0003-4

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Johnsen E, Jorgensen HA, Kroken RA, Loberg EM. Neurocognitive effectiveness of quetiapine, olanzapine, risperidone, and ziprasidone: a pragmatic, randomized trial. Eur Psychiatry (2013) 28:174–84. doi: 10.1016/j.eurpsy.2011.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Riedel M, Spellmann I, Strassnig M, Douhet A, Dehning S, Opgen-Rhein M, et al. Effects of risperidone and quetiapine on cognition in patients with schizophrenia and predominantly negative symptoms. Eur Arch Psychiatr Clin Neurosci. (2007) 257:360–70. doi: 10.1007/s00406-007-0739-x

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Voruganti LP, Awad AG, Parker G, Forrest C, Usmani Y, Fernando ML, et al. Cognition, functioning and quality of life in schizophrenia treatment: results of a one-year randomized controlled trial of olanzapine and quetiapine. Schizophr Res. (2007) 96:146–55. doi: 10.1016/j.schres.2007.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Urben S, Baumann P, Barcellona S, Hafil M, Preuss U, Peter-Favre C, et al. Cognitive efficacy of quetiapine in early-onset first-episode psychosis: a 12-week open label trial. Psychiatr Q. (2012) 83:311–24. doi: 10.1007/s11126-011-9201-3

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Keefe RS, Bilder RM, Davis SM, Harvey PD, Palmer BW, Gold JM, et al. Neurocognitive effects of antipsychotic medications in patients with chronic schizophrenia in the CATIE Trial. Arch Gen Psychiatry (2007) 64:633–47. doi: 10.1001/archpsyc.64.6.633

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Heinrichs RW. Cognitive improvement in response to antipsychotic drugs: neurocognitive effects of antipsychotic medications in patients with chronic schizophrenia in the CATIE Trial. Arch Gen Psychiatry (2007) 64:631–2. doi: 10.1001/archpsyc.64.6.631

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Mishara AL, Goldberg TE. A meta-analysis and critical review of the effects of conventional neuroleptic treatment on cognition in schizophrenia: opening a closed book. Biol Psychiatry (2004) 55:1013–22. doi: 10.1016/j.biopsych.2004.01.027

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Nielsen RE, Levander S, Kjaersdam Telleus G, Jensen SO, Ostergaard Christensen T, Leucht S. Second-generation antipsychotic effect on cognition in patients with schizophrenia—A meta-analysis of randomized clinical trials. Acta Psychiatr Scand. (2015) 131:185–96. doi: 10.1111/acps.12374

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Desamericq G, Schurhoff F, Meary A, Szoke A, Macquin-Mavier I, Bachoud-Levi AC, et al. Long-term neurocognitive effects of antipsychotics in schizophrenia: a network meta-analysis. Eur J Clin Pharmacol. (2014) 70:127–34. doi: 10.1007/s00228-013-1600-y

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Allison DB, Mentore JL, Heo M, Chandler LP, Cappelleri JC, Infante MC, et al. Antipsychotic-induced weight gain: a comprehensive research synthesis. Am J Psychiatry (1999) 156:1686–96.

PubMed Abstract | Google Scholar

34. Benarroch L, Kowalchuk C, Wilson V, Teo C, Guenette M, Chintoh A, et al. Atypical antipsychotics and effects on feeding: from mice to men. Psychopharmacology (2016) 233:2629–53. doi: 10.1007/s00213-016-4324-8

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Fervaha G, Agid O, Takeuchi H, Lee J, Foussias G, Zakzanis KK, et al. Extrapyramidal symptoms and cognitive test performance in patients with schizophrenia. Schizophr Res. (2015) 161:351–6. doi: 10.1016/j.schres.2014.11.018

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Ogino S, Miyamoto S, Miyake N, Yamaguchi N. Benefits and limits of anticholinergic use in schizophrenia: focusing on its effect on cognitive function. Psychiatr Clin Neurosci. (2014) 68:37–49. doi: 10.1111/pcn.12088

PubMed Abstract | CrossRef Full Text | Google Scholar

37. El-Missiry A, Elbatrawy A, El Missiry M, Moneim DA, Ali R, Essawy H. Comparing cognitive functions in medication adherent and non-adherent patients with schizophrenia. J Psychiatr Res. (2015) 70:106–12. doi: 10.1016/j.jpsychires.2015.09.006

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Kapur S, Remington G. Atypical antipsychotics: new directions and new challenges in the treatment of schizophrenia. Annu Rev Med. (2001) 52:503–17. doi: 10.1146/annurev.med.52.1.503

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Bender S, Dittmann-Balcar A, Schall U, Wolstein J, Klimke A, Riedel M, et al. Influence of atypical neuroleptics on executive functioning in patients with schizophrenia: a randomized, double-blind comparison of olanzapine vs. clozapine. Int J Neuropsychopharmacol. (2006) 9:135–45. doi: 10.1017/S1461145705005924

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Joshua N, Gogos A, Rossell S. Executive functioning in schizophrenia: a thorough examination of performance on the Hayling Sentence Completion Test compared to psychiatric and non-psychiatric controls. Schizophr Res. (2009) 114:84–90. doi: 10.1016/j.schres.2009.05.029

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Spagna A, Dong Y, Mackie MA, Li M, Harvey PD, Tian Y, et al. Clozapine improves the orienting of attention in schizophrenia. Schizophr Res. (2015) 168:285–91. doi: 10.1016/j.schres.2015.08.009

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Remillard S, Pourcher E, Cohen H. The effect of neuroleptic treatments on executive function and symptomatology in schizophrenia: a 1-year follow up study. Schizophr Res. (2005) 80:99–106. doi: 10.1016/j.schres.2005.07.026

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Lipkovich IA, Deberdt W, Csernansky JG, Sabbe B, Keefe RS, Kollack-Walker S. Relationships among neurocognition, symptoms and functioning in patients with schizophrenia: a path-analytic approach for associations at baseline and following 24 weeks of antipsychotic drug therapy. BMC Psychiatry (2009) 9:44. doi: 10.1186/1471-244X-9-44

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Trampush JW, Lencz T, DeRosse P, John M, Gallego JA, Petrides G, et al. Relationship of cognition to clinical response in first-episode schizophrenia spectrum disorders. Schizophr Bull. (2015) 41:1237–47. doi: 10.1093/schbul/sbv120

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Addington J, Saeedi H, Addington D. The course of cognitive functioning in first episode psychosis: changes over time and impact on outcome. Schizophr Res. (2005) 78:35–43. doi: 10.1016/j.schres.2005.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Gold S, Arndt S, Nopoulos P, O'Leary DS, Andreasen NC. Longitudinal study of cognitive function in first-episode and recent-onset schizophrenia. Am J Psychiatry (1999) 156:1342–8.

PubMed Abstract | Google Scholar

47. Hughes C, Kumari V, Soni W, Das M, Binneman B, Drozd S, et al. Longitudinal study of symptoms and cognitive function in chronic schizophrenia. Schizophr Res. (2003) 59:137–46. doi: 10.1016/S0920-9964(01)00393-0

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Rund B. A review of longitudinal studies of cognitive functions in schizophrenia patients. Schizophr Bull. (1998) 24:425–35. doi: 10.1093/oxfordjournals.schbul.a033337

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Goughari AS, Mazhari S, Pourrahimi AM, Sadeghi MM, Nakhaee N. Associations between components of metabolic syndrome and cognition in patients with schizophrenia. J Psychiatr Pract. (2015) 21:190–7. doi: 10.1097/PRA.0000000000000065

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Husa AP, Rannikko I, Moilanen J, Haapea M, Murray GK, Barnett J, et al. Lifetime use of antipsychotic medication and its relation to change of verbal learning and memory in midlife schizophrenia—An observational 9-year follow-up study. Schizophr Res. (2014) 158:134–41. doi: 10.1016/j.schres.2014.06.035

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Takeuchi H. [Optimal antipsychotic dose and dosing interval in the treatment of schizophrenia]. Seishin Shinkeigaku Zasshi (2015) 117:562–7.

PubMed Abstract | Google Scholar

52. Takeuchi H, Suzuki T, Remington G, Bies RR, Abe T, Graff-Guerrero A, et al. Effects of risperidone and olanzapine dose reduction on cognitive function in stable patients with schizophrenia: an open-label, randomized, controlled, pilot study. Schizophr Bull. (2013) 39:993–8. doi: 10.1093/schbul/sbt090

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Rajji TK, Mulsant BH, Nakajima S, Caravaggio F, Suzuki T, Uchida H, et al. Cognition and dopamine D2 receptor availability in the striatum in older patients with schizophrenia. Am J Geriatr Psychiatry (2017) 25:1–10. doi: 10.1016/j.jagp.2016.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Graff-Guerrero A, Rajji TK, Mulsant BH, Nakajima S, Caravaggio F, Suzuki T, et al. Evaluation of antipsychotic dose reduction in late-life schizophrenia: a prospective dopamine D2/3 receptor occupancy study. JAMA Psychiatry (2015) 72:927–34. doi: 10.1001/jamapsychiatry.2015.0891

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Irani F, Kalkstein S, Moberg EA, Moberg PJ. Neuropsychological performance in older patients with schizophrenia: a meta-analysis of cross-sectional and longitudinal studies. Schizophr Bull. (2011) 37:1318–26. doi: 10.1093/schbul/sbq057

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Fagiolini A, Rocca P, De Giorgi S, Spina E, Amodeo G, Amore M. Clinical trial methodology to assess the efficacy/effectiveness of long-acting antipsychotics: randomized controlled trials vs naturalistic studies. Psychiatr Res. (2017) 247:257–64. doi: 10.1016/j.psychres.2016.11.044

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Kim SW, Shin IS, Kim JM, Lee SH, Lee YH, Yang SJ, et al. Effects of switching to long-acting injectable risperidone from oral atypical antipsychotics on cognitive function in patients with schizophrenia. Hum Psychopharmacol. (2009) 24:565–73. doi: 10.1002/hup.1057

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Bartzokis G, Lu PH, Amar CP, Raven EP, Detore NR, Altshuler LL, et al. Long acting injection versus oral risperidone in first-episode schizophrenia: differential impact on white matter myelination trajectory. Schizophr Res. (2011) 132:35–41. doi: 10.1016/j.schres.2011.06.029

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Hwang SS, Kim Y, Yun DY, Kim YS, Jung HY. Exploration of the associations between neurocognitive function and neuroleptics side effects. J Psychiatr Res. (2012) 46:913–9. doi: 10.1016/j.jpsychires.2012.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Ramaekers JG, Louwerens JW, Muntjewerff ND, Milius H, de Bie A, Rosenzweig P, et al. Psychomotor, Cognitive, extrapyramidal, and affective functions of healthy volunteers during treatment with an atypical (amisulpride) and a classic (haloperidol) antipsychotic. J Clin Psychopharmacol. (1999) 19:209–21. doi: 10.1097/00004714-199906000-00003

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Tandon R, Jibson MD. Extrapyramidal side effects of antipsychotic treatment: scope of problem and impact on outcome. Ann Clin Psychiatry (2002) 14:123–9. doi: 10.3109/10401230209149099

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Malla A, Norman R, Scholten D, Townsend L, Manchanda R, Takhar J, et al. A comparison of two novel antipsychotics in first episode non-affective psychosis: one-year outcome on symptoms, motor side effects and cognition. Psychiatr Res. (2004) 129:159–69. doi: 10.1016/j.psychres.2004.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Desmarais JE, Beauclair L, Margolese HC. Anticholinergics in the era of atypical antipsychotics: short-term or long-term treatment? J Psychopharmacol. (2012) 26:1167–74. doi: 10.1177/0269881112447988

CrossRef Full Text | Google Scholar

64. Gerretsen P, Pollock BG. Drugs with anticholinergic properties: a current perspective on use and safety. Expert Opin Drug Saf. (2011) 10:751–65. doi: 10.1517/14740338.2011.579899

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Remington G, Kapur S. Atypical antipsychotics: are some more atypical than others? Psychopharmacology (2000) 148:3–15. doi: 10.1007/s002130050017

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Fischer-Barnicol D, Lanquillon S, Haen E, Zofel P, Koch HJ, Dose M, et al. Typical and atypical antipsychotics—The misleading dichotomy. Results from the Working Group ‘Drugs in Psychiatry' (AGATE). Neuropsychobiology (2008) 57:80–7. doi: 10.1159/000135641

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Rummel-Kluge C, Komossa K, Schwarz S, Hunger H, Schmid F, Lobos CA, et al. Head-to-head comparisons of metabolic side effects of second generation antipsychotics in the treatment of schizophrenia: a systematic review and meta-analysis. Schizophr Res. (2010) 123:225–33. doi: 10.1016/j.schres.2010.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

68. DE Hert M, Correll CU, Bobes J, Cetkovich-Bakmas M, Cohen D, Asai I, et al. Physical illness in patients with severe mental disorders. I. Prevalence, impact of medications and disparities in health care. World Psychiatry (2011) 10:52–77. doi: 10.1002/j.2051-5545.2011.tb00014.x

CrossRef Full Text | Google Scholar

69. Zipursky RB, Christensen BK, Daskalakis Z, Epstein I, Roy P, Furimsky I, et al. Treatment response to olanzapine and haloperidol and its association with dopamine D receptor occupancy in first-episode psychosis. Can J Psychiatry (2005) 50:462–9. doi: 10.1177/070674370505000806

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Perez-Iglesias R, Crespo-Facorro B, Martinez-Garcia O, Ramirez-Bonilla ML, Alvarez-Jimenez M, Pelayo-Teran JM, et al. Weight gain induced by haloperidol, risperidone and olanzapine after 1 year: findings of a randomized clinical trial in a drug-naive population. Schizophr Res. (2008) 99:13–22. doi: 10.1016/j.schres.2007.10.022

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Perez-Iglesias R, Vazquez-Barquero JL, Amado JA, Berja A, Garcia-Unzueta MT, Pelayo-Teran JM, et al. Effect of antipsychotics on peptides involved in energy balance in drug-naive psychotic patients after 1 year of treatment. J Clin Psychopharmacol. (2008) 28:289–95. doi: 10.1097/JCP.0b013e318172b8e6

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Correll CU. Safety and tolerability of antipsychotic treatment in young patients with schizophrenia. J Clin Psychiatry (2011) 72:e26. doi: 10.4088/JCP.9101tx5c

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Correll CU, Robinson DG, Schooler NR, Brunette MF, Mueser KT, Rosenheck RA, et al. Cardiometabolic risk in patients with first-episode schizophrenia spectrum disorders: baseline results from the RAISE-ETP study. JAMA Psychiatry (2014) 71:1350–63. doi: 10.1001/jamapsychiatry.2014.1314

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Galling B, Roldan A, Nielsen RE, Nielsen J, Gerhard T, Carbon M, et al. Type 2 diabetes mellitus in youth exposed to antipsychotics: a systematic review and meta-analysis. JAMA Psychiatry (2016) 73:247–59. doi: 10.1001/jamapsychiatry.2015.2923

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Rajkumar AP, Horsdal HT, Wimberley T, Cohen D, Mors O, Borglum AD, et al. Endogenous and antipsychotic-related risks for diabetes mellitus in young people with schizophrenia: a danish population-based cohort study. Am J Psychiatry (2017) 174:686–94. doi: 10.1176/appi.ajp.2016.16040442

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Kowalchuk C, Castellani L, Chintoh A, Remington G, Giacca A, Hahn M. Antipsychotics and glucose metabolism: how brain and body collide. Am J Physiol Endocrinol Metabol. (2018). doi: 10.1152/ajpendo.00164.2018. [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

77. De Hert M, Peuskens B, van Winkel R, Kalnicka D, Hanssens L, Van Eyck D, et al. Body weight and self-esteem in patients with schizophrenia evaluated with B-WISE. Schizophr Res. (2006) 88:222–6. doi: 10.1016/j.schres.2006.07.025

CrossRef Full Text | Google Scholar

78. Huang PL. A comprehensive definition for metabolic syndrome. Dis Model Mech. (2009) 2:231–7. doi: 10.1242/dmm.001180

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, et al. Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation (2005) 112:2735–52. doi: 10.1161/CIRCULATIONAHA.105.169404

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, Donato KA, et al. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation (2009) 120:1640–5. doi: 10.1161/CIRCULATIONAHA.109.192644

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Strachan MW, Deary IJ, Ewing FM, Frier BM. Is type II diabetes associated with an increased risk of cognitive dysfunction? A critical review of published studies. Diab Care (1997) 20:438–45. doi: 10.2337/diacare.20.3.438

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Pavlik VN, Hyman DJ, Doody R. Cardiovascular risk factors and cognitive function in adults 30–59 years of age (NHANES III). Neuroepidemiology (2005) 24:42–50. doi: 10.1159/000081049

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Bruehl H, Rueger M, Dziobek I, Sweat V, Tirsi A, Javier E, et al. Hypothalamic-pituitary-adrenal axis dysregulation and memory impairments in type 2 diabetes. J Clin Endocrinol Metab. (2007) 92:2439–45. doi: 10.1210/jc.2006-2540

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Ryan JD, Althoff RR, Whitlow S, Cohen NJ. Amnesia is a deficit in relational memory. Psychol Sci. (2000) 11:454–61. doi: 10.1111/1467-9280.00288

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Ryan JD, Cohen NJ. Processing and short-term retention of relational information in amnesia. Neuropsychologia (2004) 42:497–511. doi: 10.1016/j.neuropsychologia.2003.08.011

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Yates KF, Sweat V, Yau PL, Turchiano MM, Convit A. Impact of metabolic syndrome on cognition and brain: a selected review of the literature. Arterioscler Thromb Vasc Biol. (2012) 32:2060–7. doi: 10.1161/ATVBAHA.112.252759

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Gold SM, Dziobek I, Rogers K, Bayoumy A, McHugh PF, Convit A. Hypertension and hypothalamo-pituitary-adrenal axis hyperactivity affect frontal lobe integrity. J Clin Endocrinol Metab. (2005) 90:3262–7. doi: 10.1210/jc.2004-2181

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Frisardi V, Solfrizzi V, Seripa D, Capurso C, Santamato A, Sancarlo D, et al. Metabolic-cognitive syndrome: a cross-talk between metabolic syndrome and Alzheimer's disease. Ageing Res Rev. (2010) 9:399–417. doi: 10.1016/j.arr.2010.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Walker J, Harrison F. Shared neuropathological characteristics of obesity, type 2 diabetes and Alzheimer's disease: impacts on cognitive decline. Nutrients (2015) 7:7332–57. doi: 10.3390/nu7095341

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Mitchell AJ, Vancampfort D, Sweers K, van Winkel R, Yu W, De Hert M. Prevalence of metabolic syndrome and metabolic abnormalities in schizophrenia and related disorders—A systematic review and meta-analysis. Schizophr Bull. (2013) 39:306–18. doi: 10.1093/schbul/sbr148

PubMed Abstract | CrossRef Full Text | Google Scholar

91. de Leon J, Diaz FJ. A meta-analysis of worldwide studies demonstrates an association between schizophrenia and tobacco smoking behaviors. Schizophr Res. (2005) 76:135–57. doi: 10.1016/j.schres.2005.02.010

CrossRef Full Text | Google Scholar

92. Ringen PA, Engh JA, Birkenaes AB, Dieset I, Andreassen OA. Increased mortality in schizophrenia due to cardiovascular disease—A non-systematic review of epidemiology, possible causes, and interventions. Front Psychiatry (2014) 5:137. doi: 10.3389/fpsyt.2014.00137

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Raben AT, Marshe VS, Chintoh A, Gorbovskaya I, Muller DJ, Hahn MK. The complex relationship between antipsychotic-induced weight gain and therapeutic benefits: a systematic review and implications for treatment. Front Neurosci. (2017) 11:741. doi: 10.3389/fnins.2017.00741

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Malan-Müller S, Kilian S, van den Heuvel LL, Bardien S, Asmal L, Warnich L, et al. A systematic review of genetic variants associated with metabolic syndrome in patients with schizophrenia. Schizophr Res. (2016) 170:1–17. doi: 10.1016/j.schres.2015.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Potvin S, Stip E, Sepehry AA, Gendron A, Bah R, Kouassi E. Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry (2008) 63:801–8. doi: 10.1016/j.biopsych.2007.09.024

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Leonard B, Maes M. Mechanistic explanations how cell-mediated immune activation, inflammation and oxidative and nitrosative stress pathways and their sequels and concomitants play a role in the pathophysiology of unipolar depression. Neurosci Biobehav Rev. (2012) 36:764–85. doi: 10.1016/j.neubiorev.2011.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Üçok A, Alpsan H, Çakir S, Saruhan-Direskeneli G. Association of a serotonin receptor 2A gene polymorphism with cognitive functions in patients with schizophrenia. Am J Med Gen Part B Neuropsychiatr Genet. (2007) 144B:704–7. doi: 10.1002/ajmg.b.30463

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Roffman JL, Weiss AP, Deckersbach T, Freudenreich O, Henderson DC, Purcell S, et al. Effects of the methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism on executive function in schizophrenia. Schizophr Res. (2007) 92:181–8. doi: 10.1016/j.schres.2007.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Li C, Zhan G, Rao S, Zhang H. Metabolic syndrome and its factors affect cognitive function in chronic schizophrenia complicated by metabolic syndrome. J Nerv Ment Dis. (2014) 202:313–8. doi: 10.1097/NMD.0000000000000124

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Lindenmayer JP, Khan A, Kaushik S, Thanju A, Praveen R, Hoffman L, et al. Relationship between metabolic syndrome and cognition in patients with schizophrenia. Schizophr Res. (2012) 142:171–6. doi: 10.1016/j.schres.2012.09.019

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Morgan VA, McGrath JJ, Jablensky A, Badcock JC, Waterreus A, Bush R, et al. Psychosis prevalence and physical, metabolic and cognitive co-morbidity: data from the second Australian national survey of psychosis. Psychol Med. (2014) 44:2163–76. doi: 10.1017/S0033291713002973

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Meyer JM, Nasrallah HA, McEvoy JP, Goff DC, Davis SM, Chakos M, et al. The Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Schizophrenia Trial: clinical comparison of subgroups with and without the metabolic syndrome. Schizophr Res. (2005) 80:9–18. doi: 10.1016/j.schres.2005.07.015

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Takayanagi Y, Cascella NG, Sawa A, Eaton WW. Diabetes is associated with lower global cognitive function in schizophrenia. Schizophr Res. (2012) 142:183–7. doi: 10.1016/j.schres.2012.08.034

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Depp CA, Strassnig M, Mausbach BT, Bowie CR, Wolyniec P, Thornquist MH, et al. Association of obesity and treated hypertension and diabetes with cognitive ability in bipolar disorder and schizophrenia. Bipolar Disorders (2014) 16:422–31. doi: 10.1111/bdi.12200

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Chen DC, Du XD, Yin GZ, Yang KB, Nie Y, Wang N, et al. Impaired glucose tolerance in first-episode drug-naive patients with schizophrenia: relationships with clinical phenotypes and cognitive deficits. Psychol Med. (2016) 46:3219–30. doi: 10.1017/S0033291716001902

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Boyer L, Richieri R, Dassa D, Boucekine M, Fernandez J, Vaillant F, et al. Association of metabolic syndrome and inflammation with neurocognition in patients with schizophrenia. Psychiatr Res. (2013) 210:381–6. doi: 10.1016/j.psychres.2013.06.020

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Krakowski M, Czobor P. Cholesterol and cognition in schizophrenia: a double-blind study of patients randomized to clozapine, olanzapine and haloperidol. Schizophr Res. (2011) 130:27–33. doi: 10.1016/j.schres.2011.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Dempster K, Norman R, Theberge J, Densmore M, Schaefer B, Williamson P. Cognitive performance is associated with gray matter decline in first-episode psychosis. Psychiatr Res Neuroimag. (2017) 264:46–51. doi: 10.1016/j.pscychresns.2017.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Czepielewski LS, Wang L, Gama CS, Barch DM. The relationship of intellectual functioning and cognitive performance to brain structure in schizophrenia. Schizophr Bull. (2017) 43:355–64. doi: 10.1093/schbul/sbw090

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Rajji TK, Mulsant BH, Davies S, Kalache SM, Tsoutsoulas C, Pollock BG, et al. Prediction of working memory performance in schizophrenia by plasma ratio of clozapine to N-desmethylclozapine. Am J Psychiatry (2015) 172:579–85. doi: 10.1176/appi.ajp.2015.14050673

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Ter Horst KW, Lammers NM, Trinko R, Opland DM, Figee M, Ackermans MT, et al. Striatal dopamine regulates systemic glucose metabolism in humans and mice. Sci Transl Med. (2018) 10:eaar3752. doi: 10.1126/scitranslmed.aar3752

CrossRef Full Text | Google Scholar

112. Caravaggio F, Borlido C, Hahn M, Feng Z, Fervaha G, Gerretsen P, et al. Reduced insulin sensitivity is related to less endogenous dopamine at D2/3 receptors in the ventral striatum of healthy nonobese humans. Int J Neuropsychopharmacol. (2015) 18:pyv014. doi: 10.1093/ijnp/pyv014

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Lee Y, Kroemer NB, Oehme L, Beuthien-Baumann B, Goschke T, Smolka MN. Lower dopamine tone in the striatum is associated with higher body mass index. Eur Neuropsychopharmacol. (2018) 28:719–31. doi: 10.1016/j.euroneuro.2018.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Wilcox CE, Braskie MN, Kluth JT, Jagust WJ. Overeating behavior and striatal dopamine with 6-[F]-fluoro-L-m-tyrosine PET. J Obes. (2010) 2010:909348. doi: 10.1155/2010/909348

CrossRef Full Text | Google Scholar

115. Caravaggio F, Borlido C, Wilson A, Graff-Guerrero A. Examining endogenous dopamine in treated schizophrenia using [11C]-(+)-PHNO positron emission tomography: a pilot study. Clin Chim Acta (2015) 449:60–2. doi: 10.1016/j.cca.2015.03.020

CrossRef Full Text | Google Scholar

116. Kanji S, Fonseka TM, Marshe VS, Sriretnakumar V, Hahn MK, Muller DJ. The microbiome-gut-brain axis: implications for schizophrenia and antipsychotic induced weight gain. Eur Arch Psychiatr Clin Neurosci. (2018) 268:3–15. doi: 10.1007/s00406-017-0820-z

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Olde Loohuis LM, Mangul S, Ori APS, Jospin G, Koslicki D, Yang HT, et al. Transcriptome analysis in whole blood reveals increased microbial diversity in schizophrenia. Transl Psychiatry (2018) 8:96. doi: 10.1038/s41398-018-0107-9

CrossRef Full Text | Google Scholar

118. Dickerson F, Severance E, Yolken R. The microbiome, immunity, and schizophrenia and bipolar disorder. Brain Behav Immun. (2017) 62:46–52. doi: 10.1016/j.bbi.2016.12.010

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Castro-Nallar E, Bendall ML, Perez-Losada M, Sabuncyan S, Severance EG, Dickerson FB, et al. Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. Peer J. (2015) 3:e1140. doi: 10.7717/peerj.1140

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Lv F, Chen S, Wang L, Jiang R, Tian H, Li J, et al. The role of microbiota in the pathogenesis of schizophrenia and major depressive disorder and the possibility of targeting microbiota as a treatment option. Oncotarget (2017) 8:100899–907. doi: 10.18632/oncotarget.21284

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Dinan TG, Borre YE, Cryan JF. Genomics of schizophrenia: time to consider the gut microbiome? Mol Psychiatry (2014) 19:1252–7. doi: 10.1038/mp.2014.93

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Sherwin E, Sandhu KV, Dinan TG, Cryan JF. May the force be with you: the light and dark sides of the microbiota-gut-brain axis in neuropsychiatry. CNS Drugs (2016) 30:1019–41. doi: 10.1007/s40263-016-0370-3

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Nguyen TT, Kosciolek T, Eyler LT, Knight R, Jeste DV. Overview and systematic review of studies of microbiome in schizophrenia and bipolar disorder. J Psychiatr Res. (2018) 99:50–61. doi: 10.1016/j.jpsychires.2018.01.013

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Diamant M, Blaak EE, de Vos WM. Do nutrient-gut-microbiota interactions play a role in human obesity, insulin resistance and type 2 diabetes? Obes Rev. (2011) 12:272–81. doi: 10.1111/j.1467-789X.2010.00797.x

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Bahr SM, Tyler BC, Wooldridge N, Butcher BD, Burns TL, Teesch LM, et al. Use of the second-generation antipsychotic, risperidone, and secondary weight gain are associated with an altered gut microbiota in children. Transl Psychiatry (2015) 5:e652. doi: 10.1038/tp.2015.135

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Ott V, Benedict C, Schultes B, Born J, Hallschmid M. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diab Obes Metab. (2012) 14:214–21. doi: 10.1111/j.1463-1326.2011.01490.x

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Haring HU. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol Rev. (2016) 96:1169–209. doi: 10.1152/physrev.00032.2015

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Spinelli M, Fusco S, Mainardi M, Scala F, Natale F, Lapenta R, et al. Brain insulin resistance impairs hippocampal synaptic plasticity and memory by increasing GluA1 palmitoylation through FoxO3a. Nat Commun. (2017) 8:2009. doi: 10.1038/s41467-017-02221-9

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Ramos-Rodriguez JJ, Molina-Gil S, Ortiz-Barajas O, Jimenez-Palomares M, Perdomo G, Cozar-Castellano I, et al. Central proliferation and neurogenesis is impaired in type 2 diabetes and prediabetes animal models. PLoS ONE (2014) 9:e89229. doi: 10.1371/journal.pone.0089229

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Morris JK, Burns JM. Insulin: an emerging treatment for Alzheimer's disease dementia? Curr Neurol Neurosci Rep. (2012) 12:520–7. doi: 10.1007/s11910-012-0297-0

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Castellani L, Wilkin J, Ahsan Z, Teo C, Kowalchuk C, Giacca A, et al. O10.6. Olanzapine impairs central insulin action: effects on body fuel preference in rats. Schizophr Bull. (2018) 44:S104–5. doi: 10.1093/schbul/sby015.258

CrossRef Full Text | Google Scholar

132. Barrett EJ, Liu Z, Khamaisi M, King GL, Klein R, Klein BEK, et al. Diabetic microvascular disease: an endocrine society scientific statement. J Clin Endocrinol Metab. (2017) 102:4343–410. doi: 10.1210/jc.2017-01922

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Labazi H, Trask AJ. Coronary microvascular disease as an early culprit in the pathophysiology of diabetes and metabolic syndrome. Pharmacol Res. (2017) 123:114–21. doi: 10.1016/j.phrs.2017.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Suzuki M, Kanazawa A, Shiba M, Kojima H, Harano Y. Insulin resistance in diabetic microangiopathies. J Diab Complications (2000) 14:40–5. doi: 10.1016/S1056-8727(00)00052-0

PubMed Abstract | CrossRef Full Text | Google Scholar

135. De Silva TM, Faraci FM. Microvascular dysfunction and cognitive impairment. Cell Mol Neurobiol. (2016) 36:241–58. doi: 10.1007/s10571-015-0308-1

CrossRef Full Text | Google Scholar

136. Udristoiu I, Marinescu I, Pirlog MC, Militaru F, Udristoiu T, Marinescu D, et al. The microvascular alterations in frontal cortex during treatment with antipsychotics: a post-mortem study. Rom J Morphol Embryol. (2016) 57:501–6.

PubMed Abstract | Google Scholar

137. Kirkpatrick B, Kennedy BK. Accelerated aging in schizophrenia and related disorders: future research. Schizophr Res. (2018) 196:4–8. doi: 10.1016/j.schres.2017.06.034

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Kirkpatrick B, Messias E, Harvey PD, Fernandez-Egea E, Bowie CR. Is schizophrenia a syndrome of accelerated aging? Schizophr Bull. (2008) 34:1024–32. doi: 10.1093/schbul/sbm140

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Strassnig M, Clarke J, Mann S, Remington G, Ganguli R. Body composition, pre-diabetes and cardiovascular disease risk in early schizophrenia. Early Intervent Psychiatry (2017) 11:229–36. doi: 10.1111/eip.12225

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Hajek T, Franke K, Kolenic M, Capkova J, Matejka M, Propper L, et al. Brain age in early stages of bipolar disorders or schizophrenia. Schizophr Bull. (2017). 99:151–8. doi: 10.1093/schbul/sbx172

CrossRef Full Text | Google Scholar

141. Kolenic M, Franke K, Hlinka J, Matejka M, Capkova J, Pausova Z, et al. Obesity, dyslipidemia and brain age in first-episode psychosis. J Psychiatr Res. (2018) 99:151–8. doi: 10.1016/j.jpsychires.2018.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Firth J, Stubbs B, Rosenbaum S, Vancampfort D, Malchow B, Schuch F, et al. Aerobic Exercise Improves Cognitive Functioning in People With Schizophrenia: A Systematic Review and Meta-Analysis. Schizophr Bull. (2017) 43:546–56. doi: 10.1093/schbul/sbw115

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: schizophrenia, antipsychotics, metabolic syndrome, cognitive dysfunction, mechanism, inter-relationship

Citation: MacKenzie NE, Kowalchuk C, Agarwal SM, Costa-Dookhan KA, Caravaggio F, Gerretsen P, Chintoh A, Remington GJ, Taylor VH, Müeller DJ, Graff-Guerrero A and Hahn MK (2018) Antipsychotics, Metabolic Adverse Effects, and Cognitive Function in Schizophrenia. Front. Psychiatry 9:622. doi: 10.3389/fpsyt.2018.00622

Received: 21 August 2018; Accepted: 05 November 2018;
Published: 05 December 2018.

Edited by:

Tomas Hajek, Dalhousie University, Canada

Reviewed by:

Pavel Mohr, National Institute of Mental Health, Czechia
Ellen E. Lee, University of California, San Diego, United States

Copyright © 2018 MacKenzie, Kowalchuk, Agarwal, Costa-Dookhan, Caravaggio, Gerretsen, Chintoh, Remington, Taylor, Müeller, Graff-Guerrero and Hahn. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Sri Mahavir Agarwal, mahavir.agarwal@camh.ca
Margaret K. Hahn, margaret.hahn@camh.ca

These authors share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.