Next Article in Journal
Molecular Interactions of Autophagy with the Immune System and Cancer
Next Article in Special Issue
Multiple Sclerosis and Schizophrenia
Previous Article in Journal
Ghrelin Therapy Decreases Incidents of Intracranial Hemorrhage in Mice after Whole-Body Ionizing Irradiation Combined with Burn Trauma
Previous Article in Special Issue
Abnormally Increased Secretion in Olfactory Neuronal Precursors from a Case of Schizophrenia Is Modulated by Melatonin: A Pilot Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Targets for Schizophrenia Treatment beyond the Dopamine Hypothesis

1
Department of Psychiatry, Taipei Veterans General Hospital, Taipei 112, Taiwan
2
Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
3
Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(8), 1689; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081689
Submission received: 14 July 2017 / Revised: 30 July 2017 / Accepted: 1 August 2017 / Published: 3 August 2017

Abstract

:
Schizophrenia has been primarily associated with dopamine dysfunction, and treatments have been developed that target the dopamine pathway in the central nervous system. However, accumulating evidence has shown that the core pathophysiology of schizophrenia might involve dysfunction in dopaminergic, glutamatergic, serotonergic, and gamma-aminobutyric acid (GABA) signaling, which may lead to aberrant functioning of interneurons that manifest as cognitive, behavioral, and social dysfunction through altered functioning of a broad range of macro- and microcircuits. The interactions between neurotransmitters can be modeled as nodes and edges by using graph theory, and oxidative balance, immune, and glutamatergic systems may represent multiple nodes interlocking at a central hub; imbalance within any of these nodes might affect the entire system. Therefore, this review attempts to address novel treatment targets beyond the dopamine hypothesis, including glutamate, serotonin, acetylcholine, GABA, and inflammatory cytokines. Furthermore, we outline that these treatment targets can be possibly integrated with novel treatment strategies aimed at different symptoms or phases of the illness. We anticipate that reversing anomalous activity in these novel treatment targets or combinations between these strategies might be beneficial in the treatment of schizophrenia.

Graphical Abstract

1. Introduction

Since the discovery of chlorpromazine to treat schizophrenia, studies have been focusing on dopamine dysfunction, particularly in the mesolimbic dopamine pathway, which increases dopamine synthesis and release capacity, and can lead to psychosis [1,2].
Many studies have indicated that schizophrenia is a disorder of the dopamine signal system. Dopamine was initially reported to be related to motor function, but was then found to be associated with reward and motivation in animal studies [3]. Central nervous system stimulants, such as amphetamine, can increase dopamine release and may cause psychotic symptoms. The potency of an antipsychotic is proportional to its ability to antagonize dopamine D2/3 receptors [4]. In early imaging studies based on positron emission tomography (PET), patients with schizophrenia showed increased dopamine activity in the striatum [5] and the midbrain origin of neurons [6] compared with the controls. In addition, this increase was observed in patients having a high risk of schizophreniform psychosis [7] and was specifically linked to those who later developed psychosis [8]. Therefore, dopaminergic dysfunction is proposed as a final common pathway leading to psychosis in schizophrenia [4]. However, the mechanism through which increased dopamine synthesis and release capacity leads to the surfacing of the symptoms and signs of psychosis remains unclear [2,9].
The dopamine hypothesis, which states that the dysregulation of the dopaminergic system is etiologic for schizophrenia, is among the most enduring biological theories in psychiatry. Although variations within genes related to dopaminergic functioning have been associated with schizophrenia, an aggregate test of variations—by using the largest publicly available schizophrenia dataset—has not been conducted. Dopamine dysfunction and its treatment are not sufficient to explain the psychopathology of schizophrenia and its treatment outcomes. In addition to the devastating symptoms of psychosis, many patients with schizophrenia have cognitive impairment. These cognitive symptoms cause substantial dysfunction and can affect their ability to work, to adhere to treatment, and to apply social skills. In addition to the dopamine hypothesis, novel targets have been proposed for schizophrenia treatment. This review intends to address the dopamine hypothesis and the novel targets that have been proposed in recent years (Table 1).

2. Dopamine Hypothesis and Beyond

The dopamine hypothesis of schizophrenia was first proposed in the 1960s when the first antipsychotic chlorpromazine was found to successfully treat the positive symptoms of patients with schizophrenia. Since then, the development of newer antipsychotics has generally been following the dopamine hypothesis that patients with schizophrenia have increased dopaminergic activity, which can be normalized using dopamine antagonists, particularly the dopamine D2 receptor antagonist.
The dopamine D2 receptor is a G protein-coupled receptor, which is a common target for antipsychotic drugs. The antagonism of dopamine D2 receptor in the mesolimbic pathway is thought to be the main mode of action of antipsychotic medication in treating psychotic symptoms. However, a dopamine receptor antagonist is not clinically effective at treating cortical-related symptoms, such as cognitive deficits, in schizophrenia. Although the exact mechanism underlying these cognitive deficits remains largely unknown, factors such as deficits in cortical dopamine function [10], dysfunction in the NMDA receptor [11], or synaptic elimination [12], are likely to play a contributing role.
Molecular imaging studies have supported an association of increased subcortical dopamine transmission with the positive symptoms of schizophrenia, with the limitation that this finding is not pathognomonic, due to the neurochemical heterogeneity of the populations of patients with schizophrenia. Although hyperactivity in subcortical dopaminergic function contributes substantially to aberrant salience (possibly manifesting in positive symptoms), the original dopamine hypothesis must be extended to include contributions of other neurotransmitter systems in the pathophysiology of schizophrenia [13].

2.1. Glutamate

Glutamate is one of the excitatory neurotransmitters and the most abundant neurotransmitter in the brain. Glutamate is mediated by N-methyl-d-aspartate receptors (NMDARs) [14,15], and its pathways link to the cortex, the limbic system, and the thalamus regions, which have been implicated in schizophrenia [16]. The association of glutamate dysfunction with schizophrenia could originate from the observation that the cerebrospinal fluid of patients with schizophrenia had decreased glutamate levels [17]. Furthermore, patients who abuse NMDAR antagonists, such as phencyclidine or ketamine, frequently show psychotic symptoms [18]. NMDARs bind to glutamate and its coagonists glycine or d-serine, both of which offer additional therapeutic targets. Dysfunction of glutamatergic neurotransmission can be a promising treatment target of schizophrenia for its essential role in the pathophysiology of schizophrenia in terms of negative symptoms and cognitive impairment.

2.2. Serotonin

The serotonin (5-hydroxytryptamine; 5-HT) hypothesis of schizophrenia is based on the early studies of interactions between the hallucinogenic drug lysergic acid diethylamide (LSD) and 5-HT [19]. Observations of the psychotogenic effects of LSD and the antipsychotic effects of serotonin-dopamine antagonists, such as clozapine and risperidone, have led to increased interest in the interaction between these two neurotransmitter systems as a possible pathophysiological target in schizophrenia [20].
Serotonin antagonists ameliorate the extrapyramidal effects of antipsychotics. However, direct evidence of serotonergic dysfunction in the pathogenesis of schizophrenia is not yet available; specific serotonin receptors continue to be a focus of interest (particularly 5HT-3 and 5HT-6) in schizophrenia [21].

2.3. Acetylcholine

A significant proportion of patients with schizophrenia are heavy smokers. The high rate and heavy level of smoking observed in this population may be related to the illness or its treatment [22]. Studies have suggested the effect of altered brain muscarinic activity in schizophrenic patients [23,24]. Patients reported that they smoked for sedation to reduce negative symptoms and to counteract medication side effects [25,26]. These observations may reflect the patients’ efforts to overcome the deficit in nicotinic cholinergic receptors.
Patients with schizophrenia were found to have a poor inhibition of P50-evoked responses to repeated auditory stimuli, which may reflect impaired sensory gating. However, the effect of smoking on the reversal of diminished auditory sensory gating in schizophrenia may be attenuated due to the desensitization of the nicotine receptor. This observation has been associated with the chromosome 15q14 locus of the α-7 nicotinic receptor gene [27] and has motivated studies on promuscarinic agents, such as α-7 nicotinic agonists, in treating certain symptoms of schizophrenia [28]. Thus, the nicotinic cholinergic-mediated inhibitory process may represent a potential treatment target in schizophrenia.

2.4. Gamma-Aminobutyric Acid

GABA is the major inhibitory neurotransmitter in the central nervous system. A model has suggested the role of GABA (including GABA–dopamine interactions) in schizophrenia [29,30]. Alterations in the GABA neurotransmitter system have been reported in clinical and basic neuroscience schizophrenia studies as well as in animal models, and may be involved in the pathophysiology of schizophrenia. The chandelier subtype of parvalbumin-positive GABA neurons may be particularly affected by, and specific to schizophrenia [31].
GABA interneurons are central to brain's rhythm-generating networks, and synchrony of neural oscillations is a fundamental mechanism for the memory, perception and consciousness [32]. GABA abnormalities may underlie alterations in neural synchrony [32], abnormal gamma oscillations [33], and working memory impairments, which are observed in schizophrenia [34]. In clinical studies, the adjunctive GABA agonists has been shown to be effective in improving core schizophrenia symptoms [35]. However, it is unclear that how GABA interacts with other neurotransmitter systems, and requires further studies to elucidate the potential therapeutic role of GABA in schizophrenia treatment.

2.5. Inflammation

Inflammation and oxidative stress are other areas of interest in studies on the pathophysiology of schizophrenia [36,37]. Alterations in the complement system, which mediates innate immunity, have been implicated in schizophrenia, with observations of increases in C1, C3 and C4 complement protein activity [38,39]. Complement proteins may “tag” synapses for phagocytosis by activated microglia, leading to accelerated pruning of synapses [40,41].
An example of an inflammatory model of psychotic disorders is the anti-NMDAR encephalitis syndrome [42]. In this syndrome, schizophrenia-like features, including catatonic symptoms and autonomic dysfunction, may be combined with an increase in the level of NMDAR autoantibodies; immunotherapy is helpful in its treatment [43]. Another treatable immune model of schizophrenia is gluten sensitivity, which may involve an increase in the level of transglutaminase antibodies; patients with gluten sensitivity may benefit from gluten-free diets [44].

2.6. Summary of the Hypothesis beyond Dopamine Mechanism

In summary, the core pathophysiology of schizophrenia might primarily involve anomalies of the dopaminergic system, and accumulative evidence suggests that glutamatergic, serotonergic, and GABA alterations can lead to aberrant functioning of interneurons, which is manifested as cognitive, behavioral, and social dysfunction through altered functioning of a broad range of macro- and microcircuits. Genetic and early-life environmental risk factors and their interactions can contribute to these abnormalities. Unfortunately, none of the single neurotransmitter systems could explain the full picture of the heterogeneity of schizophrenia symptoms. Intriguingly, the dopaminergic system may interact with other neurotransmitter pathways. For example, postsynaptic density has been suggested to be involved in schizophrenia, and is related to dopaminergic and glutamatergic systems [45]. The interactions between neurotransmitter systems lead to complex and diverse patterns of mechanisms potentially involved in the pathophysiology of schizophrenia [46]. Thus, the treatment strategy that exclusively targets a single neurotransmitter system is less likely to be successful.
Therefore, identifying the role of novel treatment agents in such complex neurotransmitter networks could be important for understanding their mechanisms of action. Such interactions can be possibly modeled as nodes and edges using graph theory, a novel tool in computational biology for studying complex network interactions. In this approach, dopaminergic, glutamatergic or serotonergic systems may represent multiple nodes connected at a central hub; imbalance within any of these nodes might affect the entire system [47]. Reversal of anomalous activity in any of these nodes, or any combinations thereof, might have beneficial effects on the entire system.

3. Novel Treatment Strategies

The relationship between neurobiological findings and clinical symptoms of schizophrenia could lead to the development of novel drug treatments targeting different phases of the illness [48,49,50]. In addition, improvement in the understanding of the pathophysiology of schizophrenia enables us to define potential therapeutic targets more systematically [36,37,51,52] and utilize more actionable biomarkers [53]. The development of antipsychotics was supposed to be disease-specific and it targeted the core pathology of schizophrenia encompassing both positive and negative symptom domains. However, current antipsychotics usually show higher efficacy in treating positive symptoms, such as hallucinations or delusions, than negative symptoms or cognitive deficits [54,55]. Therefore, medication that can specifically target negative or other nonpsychotic symptoms may have substantial clinical applications, either being used as an add-on adjuvant or a stand-alone treatment. Additionally, recent discoveries in the genetic susceptibility of schizophrenia may implicate in the treatment of the illness. In the context of the complex heterogeneity of schizophrenia, we discuss these potential therapeutic approaches in this review.

3.1. Specific Treatments for Negative Symptoms

A major factor for severe disability in patients with schizophrenia is its persistent and deteriorating negative symptoms. Nevertheless, few current treatments have shown efficacy in this domain [56]. In recent years, several novel strategies have been evaluated [57], including glutamatergic receptors with glycine transporter inhibitors (e.g., bitopertin) [58] and metabotropic M2/M3 agonists (e.g., pomeglumetad methionil) [59]. These studies have yielded positive results, which have not yet been confirmed in subsequent studies. In addition to agents targeting the glutamatergic system, treatments targeting nicotinic and muscarinic cholinergic agents [24,60], antibiotics (e.g., minocycline) [61], and hormones (e.g., oxytocin [62], estrogen receptor modulator raloxifene [63], and pregnenolone [64]) have also emerged in recent years to treat negative symptoms of schizophrenia. However, additional studies are warranted to evaluate their efficacy and consistency of the treatment response.

3.2. Specific Treatments for Cognitive Deficits

Cognitive impairment associated with schizophrenia is a significant predictor of social and vocational disability [65]. Currently, the exact mechanisms underlying cognitive impairments in schizophrenia are not fully understood, and reduced brain function in the prefrontal cortex (known as hypofrontality) has often been observed in patients with schizophrenia [66]. Some pharmacological targets, such as those targeting the nicotine system, are currently being investigated [54,67,68]. However, no consistent benefits from any of these strategies have been demonstrated.

3.3. Specific Treatment for Different Phases of the Illness

Pathophysiological processes may vary across different stages of schizophrenia, but current pharmacological treatment does not typically differ during the course of the illness [69]. Several risk factors contribute to the clinical manifestations of the illness. Approaches toward prevention are focused on the early identification of those at risk and the development of safe and effective interventions that can eliminate specific risks [70,71]. Treatments that can potentially prevent deteriorative brain processes and approaches to reverse them are currently being evaluated [72].

4. Novel Treatment Targets

4.1. Dopaminergic Antagonists and Stabilizers

All current antipsychotic therapies have been developed based on the antagonism of dopaminergic receptors or dopamine “stabilizers” including D2/D3 partial agonists, such as the currently available aripiprazole. An exception is the recent introduction of pimavanserin as an antipsychotic in Parkinson’s disease [73]. The development of antipsychotics targeting other neurotransmitters may raise the hope of addressing the frequent problem of nonadherence, which is a significant roadblock in the recovery of many patients with schizophrenia.

4.2. Glutamatergic Agents

Increasing attention has been focused on glutamatergic dysfunction as a pathophysiological mechanism in schizophrenia, and various therapeutic approaches for schizophrenia are being evaluated. Among potential targets related to glutamatergic dysfunction, agents stimulating NMDARs have received the most attention. Because direct NMDAR agonists are neurotoxic, efforts to stimulate NMDARs indirectly by using glycinergic agents (e.g., serine and cycloserine) and glycine transport (GlyT1) inhibitors (e.g., bitopertin) have been extensively studied. Glycine and d-serine bind to the NMDAR site as coagonists; thus, an increase in glycine availability by GlyT1 inhibitors is shown to be effective in preclinical studies [74]. However, only moderate benefits on negative symptoms of schizophrenia were demonstrated in the clinical trial [58].
Metabotropic receptors 1 receptor antagonists have been found to be potential targets for positive symptoms of schizophrenia in animal models [75,76], but the development of related treatment agents have not yet reached clinical trials. Additionally, metabotropic 2 and 3 receptor agonists (e.g., pomaglumetad methionil) that reduce excessive glutamate release have also received considerable attention [77]. Similar to the development of bitopertin, initial studies with pomaglumetad methionil were positive [78], whereas other studies have not confirmed its efficacy [59,79]. These agents might be effective only in the earlier stages of schizophrenia [80], or in individuals having specific genetic polymorphisms, such as in neuregulin [81]. These preliminary findings are promising, but require further evaluation through clinical trials.

4.3. Serotonin Agents

Second-generation antipsychotics having serotonin–dopamine antagonism provides certain protection against extrapyramidal symptoms, although these agents, except for clozapine, do not provide additional efficacy relative to the first-generation antipsychotics [54]. Alternative 5-HT approaches in schizophrenia under investigation include the use of 5-HT1A agonists, 5-HT reuptake inhibitors, 5-HT2C antagonists and agonists, 5-HT3 antagonists, 5-HT6 antagonists, and 5HT7 antagonists either individually or in combination with D2 antagonism or 5-HT2A antagonism or both [82]. Although agents with D2/5-HT2A antagonism and 5-HT1A partial agonism are currently available, efforts to develop agents with more potent 5-HT1A activity (targeting negative and cognitive symptoms) with better tolerability are ongoing. Recently, there is a renewed interest in the development of 5-HT3 antagonists (ondansetron, tropisetron and granisetron) as adjunctive agents for negative and cognitive symptoms [83,84,85,86]. For example, Ondansetron is a 5-HT3 receptor antagonist widely used to prevent nausea and vomiting in patients receiving chemotherapy for cancer and is also potentially related to anti-inflammatory treatment strategy for schizophrenia. Adjunctive ondansetron has now entered a phase III trial for improving negative symptoms in schizophrenia [82,86]. Lurasidone, the most recently introduced antipsychotic, has potent 5-HT7 antagonism and may have clinical implications for the mood components of psychosis.

4.4. Gamma-Aminobutyric Acid (GABA) Allosteric Modulators

GABA is implicated in the pathophysiological mechanisms of schizophrenia. Benzodiazepines (that work on GABA-A receptor allosteric sites) have often been used with antipsychotic medications in patients with schizophrenia. Selective GABA-A agonists and GABA-B antagonists, and more recently, allosteric modulators at GABA-A receptor subtypes are being evaluated in schizophrenia treatment [87].

4.5. Cholinergic Agonists

The cholinergic system has long been a focus of studies on the pathophysiology of schizophrenia, given the clinical observation that nicotine smoking, which most patients with schizophrenia are exposed to, has procognitive effects. Both muscarinic and nicotinic receptors have been targeted in the therapy of schizophrenia [24,88,89]. Although several agents targeting these receptors are currently in various stages of study, α-7 nicotinic and M1 muscarinic agonists and positive allosteric modulators are of greatest interest [90,91]. Nicotinic agonists principally target cognitive symptoms [68], whereas muscarinic agonists appear to address positive symptoms [92,93,94].

4.6. Neuropeptides

Drugs targeting the neuropeptides associated with dopaminergic and glutamatergic systems have been proposed as a potential treatment strategy for schizophrenia. Cholecystokinin agonists were among the earliest to be evaluated; however, results have been equivocal [95]. Cannabis has been implicated in schizophrenia in high doses, of which the principal psychoactive constituent is tetrahydrocannabinol (THC), and may be psychotogenic through involvement in dopaminergic, GABA, and glutamatergic neurotransmission [96,97,98]. Interestingly, another major constituent of cannabis, cannabidiol, is able to prevent psychotic-like symptoms induced by high doses of THC by acting as an indirect antagonist of cannabinoid (CB) receptors [99]. As a result, the CB receptor subfamily has received considerable attention as a potential antipsychotic target, with CB antagonists being of the highest interest.
Neurokinin-3 (NK3) receptors have broad modulatory effects on several neurotransmitter systems (including dopamine and GABA); preclinical data with NK3 receptor antagonists have suggested potential efficacy across several symptom dimensions in schizophrenia. Similarly, despite studies indicating the role of neurotensin (NT) in the pathophysiology of schizophrenia [100], the questions of which NT receptor to target and how best to do so remain controversial.

4.7. Anti-Inflammatory Approaches

Many epidemiological, experimental, and clinical studies have demonstrated significant associations between schizophrenia and inflammatory conditions [36]. A broad range of anti-inflammatory strategies have been evaluated in schizophrenia, but results have been inconsistent [37,101]. Anti-inflammatory approaches might be the most effective in the early period of the illness [102,103].

4.8. Genetic-Based Approaches

Genome-wide association studies have identified over a hundred single nucleotide polymorphisms (SNPs) associated with schizophrenia risk. Despite the fact that these SNPs account for a modest fraction of genetic predisposition and almost all of them are in the non-coding region, the identified genetic risks of schizophrenia still implicate novel treatment targets via the understanding of neurotransmitter signaling. Certain genes have been already the drug targets for schizophrenia, such as the gene coding for serine racemase (SRR), which synthesizes d-serine, and one clinical trial has shown positive results with augmenting antipsychotics [104]. Other genes, such as DISC1, NOS1, NOS1AP, GRM, Pdxdc1, or ZNF804A, have been implicated in the treatment target for the schizophrenia in preclinical studies. For example, the dopamine D2 receptor has been shown to interact with the DISC1 protein, and disruption of this interaction with a peptide has antipsychotic-like effects in animal models [105]. More recently, the Pdxdc1 gene is a new antipsychotic treatment target due to its modulatory effect of pre-pulse inhibition [106], a behavioral endophenotype often used to screen for antipsychotic effects [105]. Additionally, genetic risks such as copy number variants (CNV) have also been a potential treatment target of schizophrenia [107]. It is noteworthy that neurobiology involved in these genetic risks is complex and requires extensive research to elucidate their therapeutic implications.

4.9. Other Approaches

In addition to the aforementioned approaches, epigenetic abnormalities may be a potential target for the treatment of patients with schizophrenia [108]. Recent data have indicated that epigenetic abnormalities in the disorder are more complex and include a combination of restrictive chromatin, open chromatin, and dysfunctional communication between various epigenetic mediators, leading to faulty regulation [109,110]. Epigenetic factors can account for both genetic and environmental risks and their interactions in the pathogenesis of schizophrenia. Furthermore, epigenetic factors are modifiable, and a multitude of epigenetic pharmacological treatments are already available or in development for nonpsychiatric disorders [111]. It is noteworthy that none of epigenetic treatments have been investigated in schizophrenia, and problems such as systemic toxicity may prevent it from the clinical use [112]. Finding a suitable delivery method might be necessary to reduce side effects of epigenetic drugs in potential clinical trials [113].

5. Conclusions

Schizophrenia treatment based on the dopamine hypothesis has been successful. However, despite many decades of effort by both scientists and drug companies, all currently available clinical treatments still primarily target the dopamine D2 receptor. The reason behind this inconvenient result may be due to the heterogeneity of psychosis. Patients with schizophrenia exhibit marked variations in symptoms, even the biological characterization of symptom domains of schizophrenia remains unclear, and the responses to different therapeutic interventions also vary significantly. The efforts of finding homogenous groups of psychosis using biological markers such as neuroimaging or genetic data may be of help for future pharmacological studies to evaluate novel treatment strategies. Additionally, the lack of reliable animal models of psychosis also contributes to the difficulty in identifying and validating novel treatment agents of schizophrenia. Alternatively, perhaps the dopamine dysfunction is indeed the core psychopathology of schizophrenia, but because of complex interactions between dopamine and other neurotransmitter systems, the development of novel treatment targets cannot be successful without considering these network-like interactions.
The difficulty to prospectively predict individual responses to specific treatments leads to a trial-and-error therapeutic strategy. Advances in pharmacogenomics (the study of the genetic determinants of drug response) generate optimism about applying these strategies in treating schizophrenia [114,115,116]. Over a hundred compounds, which encompass a broad range of targets in schizophrenia, are currently in various stages of drug development. Although recent results with some of the most novel drug candidates have been disappointing [117], the increasing robustness of genetic findings in schizophrenia have generated optimism about developing more effective and specific treatments for this disorder [52]. Furthermore, incorporation of brain imaging markers, such as those derived from PET or functional magnetic resonance imaging, into the treatment strategy can potentially provide new opportunities for precisely treating the illness and tracking the outcome of schizophrenia [118].

Acknowledgments

This study was supported by the Ministry of Science and Technology (MOST) of Taiwan (MOST 104-2314-B-075-078-MY2 and MOST 103-2314-B-075-067-MY3), and grants V105E17-002-MY2-1, V105C-008, V104C-018 from the Taipei Veterans General Hospital. The author has no other potential conflict of interest to disclose. We acknowledge Wallace Academic Editing for editing this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Howes, O.D.; Kambeitz, J.; Kim, E.; Stahl, D.; Slifstein, M.; Abi-Dargham, A.; Kapur, S. The nature of dopamine dysfunction in schizophrenia and what this means for treatment. Arch. Gen. Psychiatry 2012, 69, 776–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Kapur, S. Psychosis as a state of aberrant salience: A framework linking biology, phenomenology, and pharmacology in schizophrenia. Am. J. Psychiatry 2003, 160, 13–23. [Google Scholar] [CrossRef] [PubMed]
  3. Berridge, K.C. From prediction error to incentive salience: Mesolimbic computation of reward motivation. Eur. J. Neurosci. 2012, 35, 1124–1143. [Google Scholar] [CrossRef] [PubMed]
  4. Howes, O.D.; Kapur, S. The dopamine hypothesis of schizophrenia: Version III—The final common pathway. Schizophr. Bull. 2009, 35, 549–562. [Google Scholar] [CrossRef] [PubMed]
  5. Laruelle, M.; Abi-Dargham, A.; van Dyck, C.H.; Gil, R.; D’Souza, C.D.; Erdos, J.; McCance, E.; Rosenblatt, W.; Fingado, C.; Zoghbi, S.S.; et al. Single photon emission computerized tomography imaging of amphetamine-induced dopamine release in drug-free schizophrenic subjects. Proc. Natl. Acad. Sci. USA 1996, 93, 9235–9240. [Google Scholar] [CrossRef] [PubMed]
  6. Howes, O.D.; Williams, M.; Ibrahim, K.; Leung, G.; Egerton, A.; McGuire, P.K.; Turkheimer, F. Midbrain dopamine function in schizophrenia and depression: A post-mortem and positron emission tomographic imaging study. Brain 2013, 136 Pt 11, 3242–3251. [Google Scholar] [CrossRef] [PubMed]
  7. Egerton, A.; Chaddock, C.A.; Winton-Brown, T.T.; Bloomfield, M.A.; Bhattacharyya, S.; Allen, P.; McGuire, P.K.; Howes, O.D. Presynaptic striatal dopamine dysfunction in people at ultra-high risk for psychosis: Findings in a second cohort. Biol. Psychiatry 2013, 74, 106–112. [Google Scholar] [CrossRef] [PubMed]
  8. Edwards, A.C.; Bacanu, S.A.; Bigdeli, T.B.; Moscati, A.; Kendler, K.S. Evaluating the dopamine hypothesis of schizophrenia in a large-scale genome-wide association study. Schizophr. Res. 2016, 176, 136–140. [Google Scholar] [CrossRef] [PubMed]
  9. Fletcher, P.C.; Frith, C.D. Perceiving is believing: A Bayesian approach to explaining the positive symptoms of schizophrenia. Nat. Rev. 2009, 10, 48–58. [Google Scholar] [CrossRef] [PubMed]
  10. Urs, N.M.; Peterson, S.M.; Caron, M.G. New Concepts in Dopamine D2 Receptor Biased Signaling and Implications for Schizophrenia Therapy. Biol. Psychiatry 2017, 81, 78–85. [Google Scholar] [CrossRef] [PubMed]
  11. Moran, R.J.; Jones, M.W.; Blockeel, A.J.; Adams, R.A.; Stephan, K.E.; Friston, K.J. Losing control under ketamine: Suppressed cortico-hippocampal drive following acute ketamine in rats. Neuropsychopharmacology 2015, 40, 268–277. [Google Scholar] [CrossRef] [PubMed]
  12. Glantz, L.A.; Lewis, D.A. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch. Gen. Psychiatry 2000, 57, 65–73. [Google Scholar] [CrossRef] [PubMed]
  13. Grunder, G.; Cumming, P. The dopamine hypothesis of schizophrenia: Current status. In The Neurobiology of Schizophrenia; Abel, T., Nickl-Jockschat, T., Eds.; Academic Press: Cambridge, MA, USA, 2016; pp. 109–124. [Google Scholar]
  14. Moghaddam, B. Bringing order to the glutamate chaos in schizophrenia. Neuron 2003, 40, 881–884. [Google Scholar] [CrossRef]
  15. Olney, J.W.; Newcomer, J.W.; Farber, N.B. NMDA receptor hypofunction model of schizophrenia. J. Psychiatry Res. 1999, 33, 523–533. [Google Scholar] [CrossRef]
  16. Goff, D.C.; Coyle, J.T. The emerging role of glutamate in the pathophysiology and treatment of schizophrenia. Am. J. Psychiatry 2001, 158, 1367–1377. [Google Scholar] [CrossRef] [PubMed]
  17. Kim, J.S.; Kornhuber, H.H.; Schmid-Burgk, W.; Holzmuller, B. Low cerebrospinal fluid glutamate in schizophrenic patients and a new hypothesis on schizophrenia. Neurosci. Lett. 1980, 20, 379–382. [Google Scholar] [CrossRef]
  18. Farber, N.B. The NMDA receptor hypofunction model of psychosis. Ann. N. Y. Acad. Sci. 2003, 1003, 119–130. [Google Scholar] [CrossRef] [PubMed]
  19. Aghajanian, G.K.; Marek, G.J. Serotonin model of schizophrenia: Emerging role of glutamate mechanisms. Brain Res. 2000, 31, 302–312. [Google Scholar] [CrossRef]
  20. Kapur, S.; Remington, G. Serotonin-dopamine interaction and its relevance to schizophrenia. Am. J. Psychiatry 1996, 153, 466–476. [Google Scholar] [PubMed]
  21. Abi-Dargham, A. Alterations of serotonin transmission in schizophrenia. Int. Rev. Neurobiol. 2007, 78, 133–164. [Google Scholar] [PubMed]
  22. Dalack, G.W.; Becks, L.; Hill, E.; Pomerleau, O.F.; Meador-Woodruff, J.H. Nicotine withdrawal and psychiatric symptoms in cigarette smokers with schizophrenia. Neuropsychopharmacology 1999, 21, 195–202. [Google Scholar] [CrossRef]
  23. Tandon, R. Cholinergic aspects of schizophrenia. Br. J. Psychiatry 1999, 174 (Suppl. S37), 7–11. [Google Scholar]
  24. Raedler, T.J.; Bymaster, F.P.; Tandon, R.; Copolov, D.; Dean, B. Towards a muscarinic hypothesis of schizophrenia. Mol. Psychiatry 2007, 12, 232–246. [Google Scholar] [CrossRef] [PubMed]
  25. Brunzell, D.H.; McIntosh, J.M. α7 nicotinic acetylcholine receptors modulate motivation to self-administer nicotine: Implications for smoking and schizophrenia. Neuropsychopharmacology 2012, 37, 1134–1143. [Google Scholar] [CrossRef] [PubMed]
  26. Forchuk, C.; Norman, R.; Malla, A.; Martin, M.L.; McLean, T.; Cheng, S.; Diaz, K.; McIntosh, E.; Rickwood, A.; Vos, S.; et al. Schizophrenia and the motivation for smoking. Perspect. Psychiatr. Care 2002, 38, 41–49. [Google Scholar] [CrossRef] [PubMed]
  27. Freedman, R.; Olincy, A.; Ross, R.G.; Waldo, M.C.; Stevens, K.E.; Adler, L.E.; Leonard, S. The genetics of sensory gating deficits in schizophrenia. Curr. Psychiatry Rep. 2003, 5, 155–161. [Google Scholar] [CrossRef] [PubMed]
  28. Olincy, A.; Stevens, K.E. Treating schizophrenia symptoms with an α7 nicotinic agonist, from mice to men. Biochem. Pharmacol. 2007, 74, 1192–1201. [Google Scholar] [CrossRef] [PubMed]
  29. Benes, F.M. The GABA system in schizophrenia: Cells, molecules and microcircuitry. Schizophr. Res. 2015, 167, 1–3. [Google Scholar] [CrossRef] [PubMed]
  30. Garbutt, J.C.; van Kammen, D.P. The interaction between GABA and dopamine: Implications for schizophrenia. Schizophr. Bull. 1983, 9, 336–353. [Google Scholar] [CrossRef] [PubMed]
  31. Benes, F.M.; Lim, B.; Matzilevich, D.; Walsh, J.P.; Subburaju, S.; Minns, M. Regulation of the GABA cell phenotype in hippocampus of schizophrenics and bipolars. Proc. Natl. Acad. Sci. USA 2007, 104, 10164–10169. [Google Scholar] [CrossRef] [PubMed]
  32. Uhlhaas, P.J.; Singer, W. Abnormal neural oscillations and synchrony in schizophrenia. Nat. Rev. 2010, 11, 100–113. [Google Scholar] [CrossRef] [PubMed]
  33. Cardin, J.A.; Carlen, M.; Meletis, K.; Knoblich, U.; Zhang, F.; Deisseroth, K.; Tsai, L.H.; Moore, C.I. Driving fast-spiking cells induces gamma rhythm and controls sensory responses. Nature 2009, 459, 663–667. [Google Scholar] [CrossRef] [PubMed]
  34. Lewis, D.A.; Gonzalez-Burgos, G. Pathophysiologically based treatment interventions in schizophrenia. Nat. Med. 2006, 12, 1016–1022. [Google Scholar] [CrossRef] [PubMed]
  35. Wassef, A.; Baker, J.; Kochan, L.D. GABA and schizophrenia: A review of basic science and clinical studies. J. Clin. Psychopharmacol. 2003, 23, 601–640. [Google Scholar] [CrossRef] [PubMed]
  36. Feigenson, K.A.; Kusnecov, A.W.; Silverstein, S.M. Inflammation and the two-hit hypothesis of schizophrenia. Neurosci. Biobehav. Rev. 2014, 38, 72–93. [Google Scholar] [CrossRef] [PubMed]
  37. Na, K.S.; Jung, H.Y.; Kim, Y.K. The role of pro-inflammatory cytokines in the neuroinflammation and neurogenesis of schizophrenia. Prog. Neuro Psychopharmacol. Biol. Psychiatry 2014, 48, 277–286. [Google Scholar] [CrossRef] [PubMed]
  38. Mayilyan, K.R.; Weinberger, D.R.; Sim, R.B. The complement system in schizophrenia. Drug News Perspect. 2008, 21, 200–210. [Google Scholar] [CrossRef] [PubMed]
  39. Sekar, A.; Bialas, A.R.; de Rivera, H.; Davis, A.; Hammond, T.R.; Kamitaki, N.; Tooley, K.; Presumey, J.; Baum, M.; Van Doren, V.; et al. Schizophrenia risk from complex variation of complement component 4. Nature 2016, 530, 177–183. [Google Scholar] [CrossRef] [PubMed]
  40. Sasaki, T.; Oga, T.; Nakagaki, K.; Sakai, K.; Sumida, K.; Hoshino, K.; Miyawaki, I.; Saito, K.; Suto, F.; Ichinohe, N. Developmental expression profiles of axon guidance signaling and the immune system in the marmoset cortex: Potential molecular mechanisms of pruning of dendritic spines during primate synapse formation in late infancy and prepuberty (I). Biochem. Biophys. Res. Commun. 2014, 444, 302–306. [Google Scholar] [CrossRef] [PubMed]
  41. Stephan, A.H.; Barres, B.A.; Stevens, B. The complement system: An unexpected role in synaptic pruning during development and disease. Ann. Rev. Neurosci. 2012, 35, 369–389. [Google Scholar] [CrossRef] [PubMed]
  42. Kayser, M.S.; Dalmau, J. Anti-NMDA receptor encephalitis, autoimmunity, and psychosis. Schizophr. Res. 2016, 176, 36–40. [Google Scholar] [CrossRef] [PubMed]
  43. Dalmau, J.; Gleichman, A.J.; Hughes, E.G.; Rossi, J.E.; Peng, X.; Lai, M.; Dessain, S.K.; Rosenfeld, M.R.; Balice-Gordon, R.; Lynch, D.R. Anti-NMDA-receptor encephalitis: Case series and analysis of the effects of antibodies. Lancet Neurol. 2008, 7, 1091–1098. [Google Scholar] [CrossRef]
  44. Jackson, J.; Eaton, W.; Cascella, N.; Fasano, A.; Warfel, D.; Feldman, S.; Richardson, C.; Vyas, G.; Linthicum, J.; Santora, D.; et al. A gluten-free diet in people with schizophrenia and anti-tissue transglutaminase or anti-gliadin antibodies. Schizophr. Res. 2012, 140, 262–263. [Google Scholar] [CrossRef] [PubMed]
  45. Iasevoli, F.; Tomasetti, C.; Buonaguro, E.F.; de Bartolomeis, A. The glutamatergic aspects of schizophrenia molecular pathophysiology: Role of the postsynaptic density, and implications for treatment. Curr. Neuropharmacol. 2014, 12, 219–238. [Google Scholar] [CrossRef] [PubMed]
  46. De Bartolomeis, A.; Buonaguro, E.F.; Iasevoli, F. Serotonin-glutamate and serotonin-dopamine reciprocal interactions as putative molecular targets for novel antipsychotic treatments: From receptor heterodimers to postsynaptic scaffolding and effector proteins. Psychopharmacology 2013, 225, 1–19. [Google Scholar] [CrossRef] [PubMed]
  47. Steullet, P.; Cabungcal, J.H.; Monin, A.; Dwir, D.; O’Donnell, P.; Cuenod, M.; Do, K.Q. Redox dysregulation, neuroinflammation, and NMDA receptor hypofunction: A “central hub” in schizophrenia pathophysiology? Schizophr. Res. 2016, 176, 41–51. [Google Scholar] [CrossRef] [PubMed]
  48. Dawson, N.; Morris, B.J.; Pratt, J.A. Functional brain connectivity phenotypes for schizophrenia drug discovery. J. Psychopharmacol. 2015, 29, 169–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Horvath, S.; Mirnics, K. Schizophrenia as a disorder of molecular pathways. Biol. Psychiatry 2015, 77, 22–28. [Google Scholar] [CrossRef] [PubMed]
  50. Padmanabhan, J.L.; Tandon, N.; Haller, C.S.; Mathew, I.T.; Eack, S.M.; Clementz, B.A.; Pearlson, G.D.; Sweeney, J.A.; Tamminga, C.A.; Keshavan, M.S. Correlations between brain structure and symptom dimensions of psychosis in schizophrenia, schizoaffective, and psychotic bipolar I disorders. Schizophr. Bull. 2015, 41, 154–162. [Google Scholar] [CrossRef] [PubMed]
  51. Bernstein, H.G.; Steiner, J.; Guest, P.C.; Dobrowolny, H.; Bogerts, B. Glial cells as key players in schizophrenia pathology: Recent insights and concepts of therapy. Schizophr. Res. 2015, 161, 4–18. [Google Scholar] [CrossRef] [PubMed]
  52. Hall, J.; Trent, S.; Thomas, K.L.; O’Donovan, M.C.; Owen, M.J. Genetic risk for schizophrenia: Convergence on synaptic pathways involved in plasticity. Biol. Psychiatry 2015, 77, 52–58. [Google Scholar] [CrossRef] [PubMed]
  53. Pickard, B.S. Schizophrenia biomarkers: Translating the descriptive into the diagnostic. J. Psychopharmacol. 2015, 29, 138–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tandon, R.; Nasrallah, H.A.; Keshavan, M.S. Schizophrenia, “just the facts” 5. Treatment and prevention. Past, present, and future. Schizophr. Res. 2010, 122, 1–23. [Google Scholar] [CrossRef] [PubMed]
  55. Tandon, R.; Gaebel, W.; Barch, D.M.; Bustillo, J.; Gur, R.E.; Heckers, S.; Malaspina, D.; Owen, M.J.; Schultz, S.; Tsuang, M.; et al. Definition and description of schizophrenia in the DSM-5. Schizophr. Res. 2013, 150, 3–10. [Google Scholar] [CrossRef] [PubMed]
  56. Kirkpatrick, B.; Fenton, W.S.; Carpenter, W.T., Jr.; Marder, S.R. The NIMH-MATRICS consensus statement on negative symptoms. Schizophr. Bull. 2006, 32, 214–219. [Google Scholar] [CrossRef] [PubMed]
  57. Davis, M.C.; Horan, W.P.; Marder, S.R. Psychopharmacology of the negative symptoms: Current status and prospects for progress. Eur. Neuropsychopharmacol. 2014, 24, 788–799. [Google Scholar] [CrossRef] [PubMed]
  58. Umbricht, D.; Alberati, D.; Martin-Facklam, M.; Borroni, E.; Youssef, E.A.; Ostland, M.; Wallace, T.L.; Knoflach, F.; Dorflinger, E.; Wettstein, J.G.; et al. Effect of bitopertin, a glycine reuptake inhibitor, on negative symptoms of schizophrenia: A randomized, double-blind, proof-of-concept study. JAMA Psychiatry 2014, 71, 637–646. [Google Scholar] [CrossRef] [PubMed]
  59. Stauffer, V.L.; Millen, B.A.; Andersen, S.; Kinon, B.J.; Lagrandeur, L.; Lindenmayer, J.P.; Gomez, J.C. Pomaglumetad methionil: No significant difference as an adjunctive treatment for patients with prominent negative symptoms of schizophrenia compared to placebo. Schizophr. Res. 2013, 150, 434–441. [Google Scholar] [CrossRef] [PubMed]
  60. Jones, C.K.; Byun, N.; Bubser, M. Muscarinic and nicotinic acetylcholine receptor agonists and allosteric modulators for the treatment of schizophrenia. Neuropsychopharmacology 2012, 37, 16–42. [Google Scholar] [CrossRef] [PubMed]
  61. Chaudhry, I.B.; Hallak, J.; Husain, N.; Minhas, F.; Stirling, J.; Richardson, P.; Dursun, S.; Dunn, G.; Deakin, B. Minocycline benefits negative symptoms in early schizophrenia: A randomised double-blind placebo-controlled clinical trial in patients on standard treatment. J. Psychopharmacol. 2012, 26, 1185–1193. [Google Scholar] [CrossRef] [PubMed]
  62. Modabbernia, A.; Rezaei, F.; Salehi, B.; Jafarinia, M.; Ashrafi, M.; Tabrizi, M.; Hosseini, S.M.; Tajdini, M.; Ghaleiha, A.; Akhondzadeh, S. Intranasal oxytocin as an adjunct to risperidone in patients with schizophrenia: An 8-week, randomized, double-blind, placebo-controlled study. CNS Drugs 2013, 27, 57–65. [Google Scholar] [CrossRef] [PubMed]
  63. Usall, J.; Huerta-Ramos, E.; Iniesta, R.; Cobo, J.; Araya, S.; Roca, M.; Serrano-Blanco, A.; Teba, F.; Ochoa, S. Raloxifene as an adjunctive treatment for postmenopausal women with schizophrenia: A double-blind, randomized, placebo-controlled trial. J. Clin. Psychiatry 2011, 72, 1552–1557. [Google Scholar] [CrossRef] [PubMed]
  64. Marx, C.E.; Bradford, D.W.; Hamer, R.M.; Naylor, J.C.; Allen, T.B.; Lieberman, J.A.; Strauss, J.L.; Kilts, J.D. Pregnenolone as a novel therapeutic candidate in schizophrenia: Emerging preclinical and clinical evidence. Neuroscience 2011, 191, 78–90. [Google Scholar] [CrossRef] [PubMed]
  65. Green, M.F. Cognitive impairment and functional outcome in schizophrenia and bipolar disorder. J. Clin. Psychiatry 2006, 67 (Suppl. S9), 3–8. [Google Scholar] [CrossRef] [PubMed]
  66. Paulman, R.G.; Devous, M.D., Sr.; Gregory, R.R.; Herman, J.H.; Jennings, L.; Bonte, F.J.; Nasrallah, H.A.; Raese, J.D. Hypofrontality and cognitive impairment in schizophrenia: Dynamic single-photon tomography and neuropsychological assessment of schizophrenic brain function. Biol. Psychiatry 1990, 27, 377–399. [Google Scholar] [CrossRef]
  67. Ibrahim, H.M.; Tamminga, C.A. Treating impaired cognition in schizophrenia. Curr. Pharm. Biotechnol. 2012, 13, 1587–1594. [Google Scholar] [CrossRef] [PubMed]
  68. Rowe, A.R.; Mercer, L.; Casetti, V.; Sendt, K.V.; Giaroli, G.; Shergill, S.S.; Tracy, D.K. Dementia praecox redux: A systematic review of the nicotinic receptor as a target for cognitive symptoms of schizophrenia. J. Psychopharmacol. 2015, 29, 197–211. [Google Scholar] [CrossRef] [PubMed]
  69. Keshavan, M.S.; Tandon, R.; Boutros, N.N.; Nasrallah, H.A. Schizophrenia, “just the facts”: What we know in 2008 Part 3: Neurobiology. Schizophr. Res. 2008, 106, 89–107. [Google Scholar] [CrossRef] [PubMed]
  70. Amminger, G.P.; Schafer, M.R.; Papageorgiou, K.; Klier, C.M.; Cotton, S.M.; Harrigan, S.M.; Mackinnon, A.; McGorry, P.D.; Berger, G.E. Long-chain omega-3 fatty acids for indicated prevention of psychotic disorders: A randomized, placebo-controlled trial. Arch. Gen. Psychiatry 2010, 67, 146–154. [Google Scholar] [CrossRef] [PubMed]
  71. Tsuang, M.T.; Van Os, J.; Tandon, R.; Barch, D.M.; Bustillo, J.; Gaebel, W.; Gur, R.E.; Heckers, S.; Malaspina, D.; Owen, M.J.; et al. Attenuated psychosis syndrome in DSM-5. Schizophr. Res. 2013, 150, 31–35. [Google Scholar] [CrossRef] [PubMed]
  72. Dodd, S.; Maes, M.; Anderson, G.; Dean, O.M.; Moylan, S.; Berk, M. Putative neuroprotective agents in neuropsychiatric disorders. Prog. Neuro Psychopharmacol. Biol. Psychiatry 2013, 42, 135–145. [Google Scholar] [CrossRef] [PubMed]
  73. Cummings, J.; Isaacson, S.; Mills, R.; Williams, H.; Chi-Burris, K.; Corbett, A.; Dhall, R.; Ballard, C. Pimavanserin for patients with Parkinson’s disease psychosis: A randomised, placebo-controlled phase 3 trial. Lancet 2014, 383, 533–540. [Google Scholar] [CrossRef]
  74. Alberati, D.; Moreau, J.L.; Lengyel, J.; Hauser, N.; Mory, R.; Borroni, E.; Pinard, E.; Knoflach, F.; Schlotterbeck, G.; Hainzl, D.; et al. Glycine reuptake inhibitor RG1678: A pharmacologic characterization of an investigational agent for the treatment of schizophrenia. Neuropharmacology 2012, 62, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
  75. Chaki, S.; Hikichi, H. Targeting of metabotropic glutamate receptors for the treatment of schizophrenia. Curr. Pharm. Des. 2011, 17, 94–102. [Google Scholar] [CrossRef] [PubMed]
  76. Satow, A.; Maehara, S.; Ise, S.; Hikichi, H.; Fukushima, M.; Suzuki, G.; Kimura, T.; Tanak, T.; Ito, S.; Kawamoto, H.; et al. Pharmacological effects of the metabotropic glutamate receptor 1 antagonist compared with those of the metabotropic glutamate receptor 5 antagonist and metabotropic glutamate receptor 2/3 agonist in rodents: Detailed investigations with a selective allosteric metabotropic glutamate receptor 1 antagonist, FTIDC [4-[1-(2-fluoropyridine-3-yl)-5-methyl-1H-1,2,3-triazol-4-yl]-N-isopropyl-N-methy l-3,6-dihydropyridine-1(2H)-carboxamide]. J. Pharmacol. Exp. Ther. 2008, 326, 577–586. [Google Scholar] [PubMed]
  77. Moghaddam, B. Targeting metabotropic glutamate receptors for treatment of the cognitive symptoms of schizophrenia. Psychopharmacology 2004, 174, 39–44. [Google Scholar] [CrossRef] [PubMed]
  78. Patil, S.T.; Zhang, L.; Martenyi, F.; Lowe, S.L.; Jackson, K.A.; Andreev, B.V.; Avedisova, A.S.; Bardenstein, L.M.; Gurovich, I.Y.; Morozova, M.A.; et al. Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: A randomized Phase 2 clinical trial. Nat. Med. 2007, 13, 1102–1107. [Google Scholar] [CrossRef] [PubMed]
  79. Kinon, B.J.; Zhang, L.; Millen, B.A.; Osuntokun, O.O.; Williams, J.E.; Kollack-Walker, S.; Jackson, K.; Kryzhanovskaya, L.; Jarkova, N.; Group, H.S. A multicenter, inpatient, phase 2, double-blind, placebo-controlled dose-ranging study of LY2140023 monohydrate in patients with DSM-IV schizophrenia. J. Clin. Psychopharmacol. 2011, 31, 349–355. [Google Scholar] [CrossRef] [PubMed]
  80. Goff, D.C. Drug development in schizophrenia: Are glutamatergic targets still worth aiming at? Curr. Opin. Psychiatry 2015, 28, 207–215. [Google Scholar] [CrossRef] [PubMed]
  81. Liu, W.; Downing, A.C.; Munsie, L.M.; Chen, P.; Reed, M.R.; Ruble, C.L.; Landschulz, K.T.; Kinon, B.J.; Nisenbaum, L.K. Pharmacogenetic analysis of the mGlu2/3 agonist LY2140023 monohydrate in the treatment of schizophrenia. Pharm. J. 2012, 12, 246–254. [Google Scholar] [CrossRef] [PubMed]
  82. Garay, R.P.; Bourin, M.; de Paillette, E.; Samalin, L.; Hameg, A.; Llorca, P.M. Potential serotonergic agents for the treatment of schizophrenia. Expert Opin. Investig. Drugs 2016, 25, 159–170. [Google Scholar] [CrossRef] [PubMed]
  83. Ellenbroek, B.A.; Prinssen, E.P. Can 5-HT3 antagonists contribute toward the treatment of schizophrenia? Behav. Pharmacol. 2015, 26, 33–44. [Google Scholar] [CrossRef] [PubMed]
  84. Chaudhry, I.B.; Husain, N.; Drake, R.; Dunn, G.; Husain, M.O.; Kazmi, A.; Hamirani, M.M.; Rahman, R.; Stirling, J.; Deakin, W. Add-on clinical effects of simvastatin and ondansetron in patients with schizophrenia stabilized on antipsychotic treatment: Pilot study. Ther. Adv. Psychopharmacol. 2014, 4, 110–116. [Google Scholar] [CrossRef] [PubMed]
  85. Kishi, T.; Mukai, T.; Matsuda, Y.; Iwata, N. Selective serotonin 3 receptor antagonist treatment for schizophrenia: Meta-analysis and systematic review. Neuromol. Med. 2014, 16, 61–69. [Google Scholar] [CrossRef] [PubMed]
  86. Samadi, R.; Soluti, S.; Daneshmand, R.; Assari, S.; Manteghi, A.A. Efficacy of Risperidone Augmentation with Ondansetron in the Treatment of Negative and Depressive Symptoms in Schizophrenia: A Randomized Clinical Trial. Iran. J. Med. Sci. 2017, 42, 14–23. [Google Scholar] [PubMed]
  87. Gill, K.M.; Grace, A.A. The role of α5 GABAA receptor agonists in the treatment of cognitive deficits in schizophrenia. Curr. Pharm. Des. 2014, 20, 5069–5076. [Google Scholar] [CrossRef] [PubMed]
  88. Tandon, R.; DeQuardo, J.R.; Goodson, J.; Mann, N.A.; Greden, J.F. Effect of anticholinergics on positive and negative symptoms in schizophrenia. Psychopharmacol. Bull. 1992, 28, 297–302. [Google Scholar] [PubMed]
  89. Freedman, R. α7-nicotinic acetylcholine receptor agonists for cognitive enhancement in schizophrenia. Ann. Rev. Med. 2014, 65, 245–261. [Google Scholar] [CrossRef] [PubMed]
  90. Dineley, K.T.; Pandya, A.A.; Yakel, J.L. Nicotinic ACh receptors as therapeutic targets in CNS disorders. Trends Pharmacol. Sci. 2015, 36, 96–108. [Google Scholar] [CrossRef] [PubMed]
  91. Dencker, D.; Thomsen, M.; Wortwein, G.; Weikop, P.; Cui, Y.; Jeon, J.; Wess, J.; Fink-Jensen, A. Muscarinic Acetylcholine Receptor Subtypes as Potential Drug Targets for the Treatment of Schizophrenia, Drug Abuse and Parkinson’s Disease. ACS Chem. Neurosci. 2012, 3, 80–89. [Google Scholar] [CrossRef] [PubMed]
  92. Gibbons, A.; Dean, B. The Cholinergic System: An Emerging Drug Target for Schizophrenia. Curr. Pharm. Des. 2016, 22, 2124–2133. [Google Scholar] [CrossRef] [PubMed]
  93. Money, T.T.; Scarr, E.; Udawela, M.; Gibbons, A.S.; Jeon, W.J.; Seo, M.S.; Dean, B. Treating schizophrenia: Novel targets for the cholinergic system. CNS Neurol. Disord. Drug Targets 2010, 9, 241–256. [Google Scholar] [CrossRef] [PubMed]
  94. Scarr, E.; Dean, B. Role of the cholinergic system in the pathology and treatment of schizophrenia. Expert Rev. Neurother. 2009, 9, 73–86. [Google Scholar] [CrossRef] [PubMed]
  95. Montgomery, S.A.; Green, M.C. The use of cholecystokinin in schizophrenia: A review. Psychol. Med. 1988, 18, 593–603. [Google Scholar] [CrossRef] [PubMed]
  96. McLoughlin, B.C.; Pushpa-Rajah, J.A.; Gillies, D.; Rathbone, J.; Variend, H.; Kalakouti, E.; Kyprianou, K. Cannabis and schizophrenia. Cochrane Database Syst. Rev. 2014, CD004837. [Google Scholar] [CrossRef]
  97. Pushpa-Rajah, J.A.; McLoughlin, B.C.; Gillies, D.; Rathbone, J.; Variend, H.; Kalakouti, E.; Kyprianou, K. Cannabis and schizophrenia. Schizophr. Bull. 2015, 41, 336–337. [Google Scholar] [CrossRef] [PubMed]
  98. D’Souza, D.C.; Sewell, R.A.; Ranganathan, M. Cannabis and psychosis/schizophrenia: Human studies. Eur. Arch. Psychiatry Clin. Neurosci. 2009, 259, 413–431. [Google Scholar] [CrossRef] [PubMed]
  99. Zuardi, A.W.; Crippa, J.A.; Hallak, J.E.; Bhattacharyya, S.; Atakan, Z.; Martin-Santos, R.; McGuire, P.K.; Guimaraes, F.S. A critical review of the antipsychotic effects of cannabidiol: 30 years of a translational investigation. Curr. Pharm. Des. 2012, 18, 5131–5140. [Google Scholar] [CrossRef] [PubMed]
  100. Caceda, R.; Kinkead, B.; Nemeroff, C.B. Neurotensin: Role in psychiatric and neurological diseases. Peptides 2006, 27, 2385–2404. [Google Scholar] [CrossRef] [PubMed]
  101. Kim, Y.K.; Na, K.S.; Myint, A.M.; Leonard, B.E. The role of pro-inflammatory cytokines in neuroinflammation, neurogenesis and the neuroendocrine system in major depression. Prog. Neuro Psychopharmacol. Biol. Psychiatry 2016, 64, 277–284. [Google Scholar] [CrossRef] [PubMed]
  102. Kenk, M.; Selvanathan, T.; Rao, N.; Suridjan, I.; Rusjan, P.; Remington, G.; Meyer, J.H.; Wilson, A.A.; Houle, S.; Mizrahi, R. Imaging neuroinflammation in gray and white matter in schizophrenia: An in vivo PET study with [18F]-FEPPA. Schizophr. Bull. 2015, 41, 85–93. [Google Scholar] [CrossRef] [PubMed]
  103. Muller, N.; Krause, D.; Dehning, S.; Musil, R.; Schennach-Wolff, R.; Obermeier, M.; Moller, H.J.; Klauss, V.; Schwarz, M.J.; Riedel, M. Celecoxib treatment in an early stage of schizophrenia: Results of a randomized, double-blind, placebo-controlled trial of celecoxib augmentation of amisulpride treatment. Schizophr. Res. 2010, 121, 118–124. [Google Scholar] [CrossRef] [PubMed]
  104. Moghaddam, B.; Javitt, D. From revolution to evolution: The glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology 2012, 37, 4–15. [Google Scholar] [CrossRef] [PubMed]
  105. Su, P.; Li, S.; Chen, S.; Lipina, T.V.; Wang, M.; Lai, T.K.; Lee, F.H.; Zhang, H.; Zhai, D.; Ferguson, S.S.; et al. A dopamine D2 receptor-DISC1 protein complex may contribute to antipsychotic-like effects. Neuron 2014, 84, 1302–1316. [Google Scholar] [CrossRef] [PubMed]
  106. Feldcamp, L.A.; Boutros, P.C.; Raymond, R.; Fletcher, P.J.; Nobrega, J.N.; Wong, A.H.C. Pdxdc1 modulates prepulse inhibition of acoustic startle in the mouse. Transl. Psychiatry 2017, 7, e1125. [Google Scholar] [CrossRef] [PubMed]
  107. Bray, N.J.; Leweke, F.M.; Kapur, S.; Meyer-Lindenberg, A. The neurobiology of schizophrenia: New leads and avenues for treatment. Curr. Opin. Neurobiol. 2010, 20, 810–815. [Google Scholar] [CrossRef] [PubMed]
  108. Wockner, L.F.; Morris, C.P.; Noble, E.P.; Lawford, B.R.; Whitehall, V.L.; Young, R.M.; Voisey, J. Brain-specific epigenetic markers of schizophrenia. Transl. Psychiatry 2015, 5, e680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Gavin, D.P.; Floreani, C. Epigenetics of schizophrenia: An open and shut case. Int. Rev. Neurobiol. 2014, 115, 155–201. [Google Scholar] [PubMed]
  110. Guidotti, A.; Auta, J.; Chen, Y.; Davis, J.M.; Dong, E.; Gavin, D.P.; Grayson, D.R.; Matrisciano, F.; Pinna, G.; Satta, R.; et al. Epigenetic GABAergic targets in schizophrenia and bipolar disorder. Neuropharmacology 2011, 60, 1007–1016. [Google Scholar] [CrossRef] [PubMed]
  111. Ivanov, M.; Barragan, I.; Ingelman-Sundberg, M. Epigenetic mechanisms of importance for drug treatment. Trends Pharmacol. Sci. 2014, 35, 384–396. [Google Scholar] [CrossRef] [PubMed]
  112. Heerboth, S.; Lapinska, K.; Snyder, N.; Leary, M.; Rollinson, S.; Sarkar, S. Use of epigenetic drugs in disease: An overview. Genet. Epigenet. 2014, 6, 9–19. [Google Scholar] [PubMed]
  113. Millan, M.J. An epigenetic framework for neurodevelopmental disorders: From pathogenesis to potential therapy. Neuropharmacology 2013, 68, 2–82. [Google Scholar] [CrossRef] [PubMed]
  114. Arranz, M.J.; Kapur, S. Pharmacogenetics in psychiatry: Are we ready for widespread clinical use? Schizophr. Bull. 2008, 34, 1130–1144. [Google Scholar] [CrossRef] [PubMed]
  115. Lotrich, F.E. The emerging potential of pharmacogenetics in psychiatry. Am. J. Psychiatry 2012, 169, 681–683. [Google Scholar] [CrossRef] [PubMed]
  116. Malhotra, A.K.; Zhang, J.P.; Lencz, T. Pharmacogenetics in psychiatry: Translating research into clinical practice. Mol. Psychiatry 2012, 17, 760–769. [Google Scholar] [CrossRef] [PubMed]
  117. Dunlop, J.; Brandon, N.J. Schizophrenia drug discovery and development in an evolving era: Are new drug targets fulfilling expectations? J. Psychopharmacol. 2015, 29, 230–238. [Google Scholar] [CrossRef] [PubMed]
  118. Poels, E.M.; Kegeles, L.S.; Kantrowitz, J.T.; Slifstein, M.; Javitt, D.C.; Lieberman, J.A.; Abi-Dargham, A.; Girgis, R.R. Imaging glutamate in schizophrenia: Review of findings and implications for drug discovery. Mol. Psychiatry 2014, 19, 20–29. [Google Scholar] [CrossRef] [PubMed]
Table 1. Novel treatment targets for schizophrenia.
Table 1. Novel treatment targets for schizophrenia.
HypothesisTargetStrategy
DopamineDopaminergic stabilizersImprove medication adherence
GlutamateNMDAR, AMPA receptor, or metabotropic receptorsImprove negative symptoms and cognitive impairments
Serotonin5-HT1A agonists, 5-HT reuptake inhibitors, 5-HT2C antagonists and agonists, 5-HT3 antagonists, 5-HT6 antagonists, and 5HT7 antagonistsReduce the extrapyramidal effects
Improve negative symptoms and cognitive impairments
Potential treatment for different phases of the illness
Acetylcholineα-7 nicotinic and M1 muscarinic agonists and positive allosteric modulatorsNicotinic agonists for cognitive symptoms
Muscarinic agonists for positive symptoms
Gamma-aminobutyric acidSelective GABA-A agonists, GABA-B antagonists, and allosteric modulators at GABA-A receptor subtypesAugmentation of psychosis treatment
InflammationCytokinesPossibly the early period of the psychosis
NMDAR: N-methyl-d-aspartate receptors; AMPA: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; 5-HT: 5-hydroxytryptamine; GABA: gamma-aminobutyric acid.

Share and Cite

MDPI and ACS Style

Yang, A.C.; Tsai, S.-J. New Targets for Schizophrenia Treatment beyond the Dopamine Hypothesis. Int. J. Mol. Sci. 2017, 18, 1689. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081689

AMA Style

Yang AC, Tsai S-J. New Targets for Schizophrenia Treatment beyond the Dopamine Hypothesis. International Journal of Molecular Sciences. 2017; 18(8):1689. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081689

Chicago/Turabian Style

Yang, Albert C., and Shih-Jen Tsai. 2017. "New Targets for Schizophrenia Treatment beyond the Dopamine Hypothesis" International Journal of Molecular Sciences 18, no. 8: 1689. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081689

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop