Loading

Journal of Neurological Disorders and Stroke

Microglial Abnormalities in the Pathophysiology of Schizophrenia

Review Article | Open Access

  • 1. Department of Psychiatry, Saga University Hospital, Japan
  • 2. Department of Neuropsychiatry, Kyushu University, Japan
+ Show More - Show Less
Corresponding Authors
Akira Monji, Department of Psychiatry, Faculty of Medicine, Saga University Hospital, Nabeshima 5-1-1, Saga 849-8501, Japan
KEYWORDS

•    Schizophrenia
•    Neuroinflammation
•    Microglia
•    Antipsychotics
•    Cytokines
•    Free radicals

INTRODUCTION

Research into neuropsychiatric disorders has been “neuronocentric”. However, several categories of neuropsychiatric disorders show no common alterations of brain structure dictated by neuronal architecture, leaving neuropathologists and neuroanatomists clueless. Schizophrenia, for example, was famously named “the graveyard of neuropathologists” (Iritani, 2007). However, recent neurochemical and brain imaging studies have moved microglia into the center of attention. To evaluate microglia activity in vivo, a few radioligands have been developed. One radiotracer, [11C]- (R)-PK11195 ([11C](R)-(1-[2-chrorophynyl]-N-methyl-N-[1- methylpropyl]-3 isoquinoline carboxamide) which is a specific ligand of peripheral benzodiazepine receptors, specifically binds to activated microglia and is widely used (Benavides et al., 1988, Pike et al., 1993). The availability of well-preserved post-mortem brain samples and several established microglial histological markers in recent years also allows neurochemical evaluations of microglia in diseased brains. Microglia are highly mobile, incessantly surveying the brain environment, and ready to react to even the slightest perturbation. At the experimental biology level, innovative molecular genetic and biophotonic approaches begin to enable investigators to visualize the dynamic microglial behavior in health and disease in vivo, and to introduce microglia-targeted deletions and ablations (Wake et al., 2009 & Li et al., 2012). Therefore, we are at the threshold of making fundamental discoveries regarding microglial contributions to neuropsychiatric disorders.

Here, we will review one of the prominent neuropsychiatric disorders, schizophrenia, to exemplify the pathological roles of microglia, either acting as primary instigators or responding to pre-natal, perinatal, or postnatal insults as well as environmental impacts specific to the diseases of interest. In the pathophysiology of schizophrenia, emphasis has been placed on the gene-environment interplay in the pathophysiology (van Os et al., 2010), as microglia serve as interphase between environmental alterations and regulated brain responses.

SCHIZOPHRENIA

Schizophrenia is a severe neuropsychiatric disorder affecting about 1% of the world population. The onset of full-blown schizophrenia is typically in late adolescence or early adulthood and includes distinct symptom classes which are commonly referred to as positive (hallucinations, delusions, and psychomotor excitement), negative (avolition and amotivation), and cognitive symptoms. In addition to severely disrupting the life of the patient and their family, schizophrenia incurs a great cost to society in terms of lost productivity and treatment-related expenses. Current treatments are ineffective at addressing the full spectrum of symptoms. The etiology of schizophrenia still remains to be elusive while dopaminergic hyperfunction in the limbic system and dopaminergic hypofunction in the frontal cortex as well as glutamatergic hypofunction are known to play important roles in the pathophysiology of schizophrenia (Lieberman, 1999). We herein review the role of neuroinflammation in the pathophysiology of schizophrenia especially focused on microglia. And also, we suggest the therapeutic strategy of schizophrenia through the inhibition of microglial activation.

NEUROINFLAMMATION AND SCHIZOPHRENIA

Jurius Wagner-Jauregg, who was awarded the Nobel Prize in Medicine in 1927, proposed the treatment of mental diseases by inducing fever. His well-known “pyrotherapy” might be the beginning of the study on the immunological concepts of schizophrenia. Many recent neuroimaging studies using magnetic resonance imaging (MRI) have shown progressive brain atrophy in schizophrenia and these results suggest that even schizophrenia has an aspect of neurodegenerative disorder (Davis et al., 2003, Kumra et al., 2005, Salisbury et al., 2007, Hulshoff Pol and Kahn, 2008). Recent genome-wide studies in schizophrenia have shown the association of schizophrenia with markers in the MHC (major histocompatibility complex) region and suggest immune system involvement in schizophrenia (Stefansson et al., 2009, Jia et al., 2010). An accumulating body of evidences point to the significance of neuroinflammation in schizophrenia, characterized by an increased serum concentration of several pro-inflammatory cytokines (Drzyzga et al., 2006, Potvin et al., 2008, Meyer, 2011, Miller et al., 2011). Increased serum and cerebrospinal fluid (CSF) levels of S100B, a suitable marker for the destruction of CNS tissue in the context of different disease including neurodegenerative disorder, were reported in schizophrenia patients with negative symptoms or chronic duration (Schmitt et al., 2005). Increased serum concentrations of interleukin (IL)-2, IL-6 and IL-8 have been observed in schizophrenic patients (Lin et al., 1998, Zhang et al., 2004). Moreover, a recent report has shown the elevated IL-1β levels in the cerebrospinal fluids of the patients with drug-naïve schizophrenia (Soderlund et al., 2009). A recent meta-analysis has demonstrated that the cytokine abnormalities an acute exacerbation of schizophrenia is independent of antipsychotic medication (Miller et al., 2011). Peripheral inflammatory responses in schizophrenia have also been linked to the changes in the numbers of circulating monocytes and T-cells (Drexhage et al., 2010). It has recently been reported that blood lymphocyte abnormalities in drug -naïve first episode psychosis suggest an effect may be independent of antispychtoic medications (Miller et al., 2013). Epidemiologic studies demonstrate significant environmental impact of maternal viral infection and obstetric complications on the risk of schizophrenia. Elevated inflammatory process is known to play an important role under these circumstances. Infection during the perinatal period of life acts as a vulnerability factor for late-life alterations in cytokine production, and marked changes in cognitive and affective behaviors throughout the remainder of lifespan. A series of infection during pregnancy have been associated with risk of schizophrenia in offspring, including influenza, rubella, herpes simplex virus-type2, and toxoplasma gondii. Evidence from recent animal studies suggests that most viral infection do not appear to cross the placenta; therefore, the teratogenic influence might be more related to maternal, fetal, and/or placental response to infection. Two nested case-controlled studies have demonstrated an association between elevated serum levels of maternal tumor necrosis factor α (TNF-α) or interleukin-8 (IL-8) with increased risk for schizophrenia in the offspring (Nawa and Takei, 2006, Ashdown et al., 2006, Deverman and Patterson, 2009, Ellman and Susser, 2009, Brown and Derkits, 2010). A DNA microarray study has shown the increased expression of genes related to immune and chaperone function in the prefrontal cortex (BA46) in schizophrenia (Arion et al., 2007). Another study using prefrontal cortex of schizophrenia has shown that molecular basis for schizophrenia changes from early to chronic stages, providing evidence for a changing nature of schizophrenia with disease progression. Short-term illness was particularly associated with disruption in gene transcription, metal-binding, RNA expression and vesicle-mediated transport. In contrast, while long-term illness was associated with inflammation, stimulus-response and immune functions (Narayan et al., 2008). The latest two studies using postmortem human tissues have also demonstrated increased inflammatory markers identified in BA46 of the patients with schizophrenia (Fillman et al., 2013, Dean et al., 2013). Dean et al. have demonstrated that levels of TNF receptor 1 mRNA are increased in BA46 (82%) in patients with schizophrenia (Dean et al., 2013).

Patients with schizophrenia have a reduced lifespan of > 20 years, with cardiovascular disorders being the main cause of death while diabetes mellitus is a particularly strong risk factor for cardiovascular mortality, being a risk equivalent of myocardial infarction. There has been an exponential increase in the schizophrenia literature discussing the high prevalence of type2 diabetes mellitus and pre-diabetic states such as the metabolic syndrome while diabetes mellitus is well-known to be a pro-inflammatory state. These results are very important because insulin resistance can be observed even in antipsychoticnaïve patients with schizophrenia and second generation antipsychotics, especially olanzapine and clozapine, have been associated with the metabolic syndrome and the development of type2 diabetes mellitus (Meyer and Stahl, 2009, Nielsen et al., 2010, Devaraj et al., 2010, Steiner et al., 2014). Beumer et al have recently demonstrated the increased levels of serum cytokines, chemokines and adipokines in patients with schizophrenia is associated with disease and metabolic syndrome (Beumer et al., 2012). C-reactive protein (CRP) is a pentameric protein which is generated largely in the liver and secreted in the blood. CRP in the blood provides a reliable marker of chronic inflammation. It has recently been reported that high sensitivity CRP (hs CRP) is elevated in schizophrenia while cardiovascular risk factors during second generation antipsychotic (a newer antipsychotic with fewer side effects than first generation antipsychotic such as haloperidol) treatment are associated with increased hs CRP (Dieset et al., 2012, Dickerson et al., 2013).

SCHIZOPHRENIA AND MICROGLIAL ACTIVATION

Bilbo et al have shown the hypothesis that long-term changes in brain glial cell function underlie this vulnerability. They hypothesize that a subsets of microglia are permanently maintained in an activated or primed state into adulthood as a consequence of perinatal infection and that a subsequent immune challenge in adulthood can cause exaggerated levels of cytokines from primed microglia (Bilbo and Schwarz, 2009, Bland et al., 2010). Interestingly, some recent animal studies have shown that even psychological stress, which may be relevant to the pathology of schizophrenia, can induce microglial activation in vivo (Frank et al., 2007, Schiavone et al., 2009, Tynan et al., 2010, Wohleb et al., 2011, Hinwood et al., 2012). A recent study using an analog of viral double-strand RNA (polynsinic-polycytidylic acid sodium salt [poly I:C] has shown that maternal infection during embryogenesis contributes to microglial activation in the offspring (Juckel et al., 2011). Other known risk factors for schizophrenia such as malnutrition and stress involve upregulation of inflammatory cytokines in maternal serum (Deverman and Patterson, 2009). It has recently reported that there is increased prevalence of Chlamydophila DNA in post-mortem brain frontal cortex from patients with schizophrenia. (Fellerhoff and Wank, 2011) Many infections have been shown to induce symptoms of mental illness, but these symptoms generally disappear after recovery from the acute illness. However, some symptoms may not disappear if acute infection becomes chronic. Microbes, which have the ability to permanently reside in the body, could permanently disturb brain functions. The primary targets of Chlamydophila infection in the blood are monocytes while the primary targets of Chlamydophila infection in the brain are probably microglia, which arise from monocyte populations. Persistent Chlamydophila- infected microglia or neuronal cells may impair neuronal circuits and thus be mechanism for causing illness in the patients with schizophrenia (Fellerhoff and Wank, 2011). Prolonged microglial hyperactivity may lead to neuronal apoptosis and brain damage which are commonly seen in neurodegenerative disorder such as Parkinson disease (PD) and Alzheimer’s disease (AD) through the overproduction of inflammatory cytokines and free radicals (Block and Hong, 2005). A neurodegenerative and neurodevelopmental process is indicated in the course of schizophrenia (Lieberman, 1999, Perez-Neri et al., 2006) and may be associated with the microglial activation. Hypoglutamatergic states and impaired N-metyl-D-aspartate (NMDA) signaling underlie the pathophysiology of schizophrenia. NMDA antagonists such as phencyclidine (PCP), ketamine, and MK-801 offer an appropriate animal model of schizophrenia. All three NMDA antagonists are known to induce microglial activation in rodent brains (Nakki et al., 1995, Nakki et al., 1996). Interestingly, microglial activation or increased microglial cellular density has also been suggested by postmortem studies, at least in subpopulations of individuals with schizophrenia (Bayer et al., 1999, Radewicz et al., 2000, Steiner et al., 2006, Schnieder and Dwork, 2011). Highly elevated microglial cell numbers has been demonstrated in the anterior cingulate cortex and mediodorsal thalamus of patients with schizophrenia who had committed suicide during acute psychosis (Steiner et al., 2006). By using the technique of [11C] (R)-PK11195 used to systematically study microglial activation in vivo, researchers recently have reported increased microglial activation in the grey matters or hippocampus of patients with schizophrenia, while it is unclear whether or not the effects of this microglial activation is neuroprotective or detrimental (van Berckel et al., 2008, Doorduin et al., 2009). These positron emission computed tomography (PET) studies have demonstrated that activated microglia are present in schizophrenia patients within the first 5 years of disease onset (van Berckel et al., 2008) or in a psychotic state (Doorduin et al., 2009), respectively. Another recent PET study in chronic schizophrenia has shown that there are no significant differences between microglial activation of the cortical regions of normal controls and the patients with schizophrenia while microglial activation is positively correlated with positive symptoms scores as well as the duration of illness (Takano et al., 2010). A recent PET study has shown microglial activation in young patients with autism spectrum disorders (Suzuki et al., 2013). These results are very interesting because there are many similarities between schizophrenia and autism spectrum disorders (Lugnegard et al., 2013).

SCHIZOPHRENIA AND NEUROGENESIS

The relationship between depression and neurogenesis has been described in general (Warner-Schmidt and Duman, 2006) while one recent human postmortem brain study using Ki-67 immunoreactivity indicated that the phenomenon of neurogenesis is much more related to the pathophysiology of schizophrenia than that of depression (Reif et al., 2006). Repeated administration of PCP as well as MK-801 has recently been reported to inhibit hippocampal neurogenesis in vivo (Juan et al., 2006, Maeda et al., 2007). Mice harboring compound disruption in the neuronal PAS domain protein 3 (NPAS3) and related NPAS1 genes manifest behavioral and neuroanatomical abnormalities reminiscent of schizophrenia (Pickard et al., 2006). Basal neural precursor cell proliferation in the dentate gyrus of NPAS3 gene deficiency mice has been found to be reduced significantly, which indicated the impaired neurogenesis involved in schizophrenia (Pieper et al., 2005). Disrupted-In- Schizophrenia 1 (DISC1) is a well-known schizophrenia susceptibility gene. A recent study has shown that DISC1 regulates integration of newly generated neurons in the adult brain (Duan et al., 2007). The above results indicate the close relationship between schizophrenia and neurogenesis. With regard to neurogenesis, atypical antipsychotics not typical antipsychotics induced neurogenesis in adult brain of rodents (Maeda et al., 2007, Wakade et al., 2002, Kodama et al., 2004). On the other hand, CNS inflammation is detrimental for adult hippocampal neurogenesis (Monje et al., 2003, Ekdahl et al., 2003). The negative effects of inflammation on differentiation and survival of the neuronal cells are due, in vitro, to microglia-derived TNF-α and NO (Monje et al., 2003, Cacci et al., 2005). Pro-inflammatory cytokines such as Il-1β and TNF-α have been reported to inhibit neurogenesis in vivo (Iosif et al., 2006, Kaneko et al., 2006). In addition, in vivo, neurogenesis can be restored by anti-inflammatory drugs such as minocycline and indomethacin that inhibit microglial activation (Monje et al., 2003, Ekdahl et al., 2003).

SCHIZOPHRENIA AND APOPTOSIS

Structural brain abnormalities have been extensively and consistently described in schizophrenic patients. Longitudinal studies using high-resolution MRI to examine brain structure have found that MRI volume changes were progressive over time and related to the course of illness and treatment outcome in schizophrenic patients (Davis et al., 2003, Kumra et al., 2005, Salisbury et al., 2007). A recent review has shown that continuous progressive brain tissue decreases and lateral ventricle volume increases in chronically ill patients with schizophrenia, up to at least 20 years after the first symptoms (Hulshoff Pol and Kahn, 2008). In fact, multiple lines of evidence combine to implicate the increased susceptibility for apoptotic death in the pathophysiology of schizophrenia. Reduced neuronal and glial cell numbers (mainly in astrocyte), decreased neuropil (especially of the synapse elements), lack of gliosis, and in vivo neuroimaging evidence of progressive gray matter loss early in the disorder, as mentioned above, make apoptosis as a plausible mechanism to explain the neurodegenerative course of schizophrenia. The activation of apoptotic process can lead to rapid neuronal death. However, emerging data also indicate that sub-lethal apoptotic activity can lead to a limited form of apoptosis in terminal neuritis and individual synapses to cause elimination without cell death (Glantz et al., 2006, Jarskog et al., 2005). Inappropriate activation of apoptosis occurs not only in the neurons, but also in the oligodendrocytes and synapses (Glantz et al., 2006). Proinflammatory cytokines such as TNF-α has been well characterized as a mediator of oxidative stress, and they induce the apoptosis in the human cortical neuron as well as oligodendrocytes (Medina et al., 2002, Buntinx et al., 2004). In addition, NO has been reported to directly induce neuronal apoptosis, but also to be involved in cytokine-mediated neuronal apoptosis (Palluy and Rigaud, 1996, Hu et al., 1997). The interaction between NO and superoxide anion (O2-), which can be generated from activated microglia, forms peroxynitrite (ONOO- ). Peroxynitrite is highly toxic and triggers apoptotic cell death. Moreover, high levels of NO and TNF-α may also affect synaptogenesis, synaptic plasticity and connectivity, and the composition of synaptic membranes (Sunico et al., 2005, Stellwagen and Malenka, 2006). The alteration in the synaptic organization of the brain is one of the key features of schizophrenia (Roberts et al., 2005). Several postmortem studies have examined dendritic spine density changes in brain regions showing the greatest indice of gray matter loss in schizophrenia and these results support the view that spine density changes directly contribute to gray matter loss in the disease. Reduced spine density with gray matter loss is reported in dorsolateral prefrontal cortex (DLPFC), superior temporal gyrus, and hippocampus. These results reveal a strong association between brain region-specific loss of gray matter, reduced spine density and functional hypoactivity in schizophrenia (Penzes et al., 2011).

SCHIZOPHRENIA AND WHITE MATTER DISORDERS

Neuroimaging studies have shown that first-episode schizophrenia patients had a significant volume reduction in white matter with abnormal brain connectivity (Price et al., 2006, Schlosser et al., 2007, Lee et al., 2013, Wang et al., 2013). The reduced density and compromised morphology of the oligodendroglia cells as well as signs of deviant myelination have been evident in schizophrenia (Uranova et al., 2004, Bernstein et al., 2009, Uranova et al., 2007). Combined with the evidence of dysregulation of the myelination-related genes, a disruption of the oligodendrocyte function in schizophrenia is strongly implicated (McCullumsmith et al., 2007). Microglial activation in the CNS has been implicated in the pathogenesis of white matter disorders and it has recently been reported that microglial cytotoxicity of oligodendrocyte is mediated through free radicalrelated molecules such as NO and peroxynitrite generated by activated microglia (Li et al., 2005, Merrill et al., 1993) and inflammatory cytokines such as TNF-α and IFN-γ (Buntinx et al., 2004). In addition, TNF-α has been shown to compromise the growth of oligodendrocytes and the expression of mRNA for myelin basic protein (MBP) in cultures (Cammer and Zhang, 1999). Furthermore, it inhibited the survival and proliferation of the oligodendrocyte progenitors and their subsequent differentiation into mature myelinating phenotypes (Feldhaus et al., 2004). These results are intriguing because Mittelbronn et al. have demonstrated that local distribution of microglia in the normal adult human braindiffers by up to one order of magnitude and that there are significantly more microglia in white matter than in gray matter (Mittelbronn et al., 2001).

TREATMENT OF SCHIZOPHRENIA THROUGH THE CONTROL OF NEUROINFLAMMATION

Although here we describe two apparently disparate groups of disorders, they do share some clinical characteristics such as social cognition (King and Lord, 2011, Sugranyes et al., 2011, Lugnegard et al., 2013). Epidemiology studies reveal that maternal infections and obstetric complications impose high risk for both schizophrenia and autism. A recent complex network and computational analysis revealed that genetic variations associated with schizophrenia can occur in the same molecular pathways and functional domains (Cristino et al., 2013). From the above discussion, it is clear that abnormal microglial activation or intrinsic microglial abnormalities are also a shared feature of schizophrenia. Taming microglia-associated neuroinflammation as a therapeutic approach has been extensively studied in schizophrenia

ANTI-INFLAMMATORY STRATEGY FOR SCHIZOPHRENIA

Second generation antipsychotics such as olanzapine and risperidone are becoming standard drugs for the treatment of schizophrenia due to their less adverse effects and more effectiveness for the negative symptoms of schizophrenia (Lieberman et al., 2005). Some recent reports have suggested the possibility of specific second generation antipsychotics having pharmacological properties that could produce neurotrophic, neurogenetic, or neuroprotective effects. Namely, specific atypical antipsychotics such as olanzapine and risperidone have been reported to decrease the reduction of MRI volume during the clinical course of schizophrenia (Lieberman et al., 2005, Chakos et al., 2005, Massana et al., 2005, Girgis et al., 2006) while a recent report has shown a brain tissue loss due to the long-time antipsychotic treatment (Ho et al., 2011). There have some reports that studied the effect of antipsychotics on neuroinflammation in vitro or in vivo. Kowalski et al. demonstrated that flupentixol and trifluperidol reduced the secretion of TNF-α and NO by the activated microglia (Kowalski et al., 2003), and flupentixol, trifluperidol, chlorpromazine and loxapine have been reported to reduce IL-1β and IL-2 release by the activated microglia (Kowalski et al., 2004, Labuzek et al., 2005). Until recently, the pharmacological action of second generation antipsychotics on microglial cells has not been well understood. Hou et al. demonstrated that olanzapine inhibited NO release from the activated microglia, while haloperidol and clozapine did not (Hou et al., 2006). We recently demonstrated that risperidone significantly inhibited the IFN-γ-activated microglia-derived production of NO and pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α in comparison to haloperidol a typical first generation antipsychotics (Kato et al., 2007). There have been some reports that suggested the relationship between schizophrenia and IFN-γ, a major immunoactivator in the CNS. The most important immunological studies in schizophrenia have shown a shift fromTh1-like cellular to Th2-like humoral immune reactivity to be the most characteristic common immune findings and these studies have suggested a blunted IFN-γ signal in schizophrenia (Schwarz et al., 2001). However, Rothermundt et al. have argued that the reduced IFN-γ production in vitro may reflect an increased production in vivo, as it is found in several autoimmune disorders (Rothermundt et al., 2001). Furthermore, the serum levels of IL-2 and IFN-γ, and the production of these cytokines from the peripheral blood mononuclear cells (PBMC) stimulated by phytohemagglutinin (PHA) have been reported to be significantly higher in schizophrenic patients than in controls (Cazzullo et al., 2001). A recent systematic quantitative review on the inflammatory cytokine alterations in schizophrenia did not necessarily support the Th2 shift hypothesis of schizophrenia while the levels of IL-6, the Th2 related cytokine, were increased after eliminating medication effects (Potvin et al., 2008). We furthermore demonstrated the same inhibitory effects on IFN-γ-induced microglial activation by other atypical antipsychotics such as perospirone and quetiapine (Bian et al., 2008). Spiperone, a typical antipsychotic, also inbibited the production of NO and pro-inflammatory cytokines such as IL-1β and TNF-α from activated microglia while spiperone was neuroprotective, as the drug reduced microglia-mediated neuroblastoma cell death in the microglia/neuron co-culture (Zheng et al., 2008). Sugino et al. have demonstrated that clozapine, olanzapine, and risperidone, but not haloperidol suppress production of proinflammatory cytokines such as TNF-α and IL-6 and up-regulate anti-inflammatory cytokines such as IL-10 in LPS -treated mice. They have also demonstrated that clozapine alone suppress poly I:C-induced inflammation (Sugino et al., 2009). Risperidone have also recently been reported to normalize increased inflammatory parameters and restores anti-inflammatory pathways in a model of neuroinflammation (MacDowell et al., 2013). Aripiprazole is a novel second generation antipsychotic, which is a high-affinity dopamine D2 receptor partial agonist. We also demonstraretd that aripiprazole significantly inhibited the generation of nitric oxide (NO) and TNF-α from IFN-γ-activated microglia while quinpirole, dopamine D2 full agonist did not in vitro. Our results demonstrated that not only antipsychotics which have dopamine D2 receptor antagonism but also aripiprazole, a dopamine D2 receptor partial agonist, have anti-inflammatory effects via the inhibition of microglial activation (Kato et al., 2008). Social isolation has been reported to cause behavioral and pathological alterations in rats due to the oxidative stress by superoxide derived from the nicotinamide dinucleotide phosphate (NADPH) oxidase (NOX) system in activated microglia (Schiavone et al., 2009). We have recently demonstrated that aripiprazole inhibits superoxide generation through the NOX system in phorbolmyristate-acetate (PMA)-stimulated microglia in vitro (Kato et al., 2007). These results are very intriguing because the loss of fast-spiking, parvalbumin (PV)-positive interneurons found in the prefrontal cortex of ketamine mice model of schizophrenia is reported to be mediated by the oxidative stress through the Nox system. Repetitive adult exposure to the NMDAR antagonist ketamine has been reported to increase the levels of IL-6 in brain which, through the activation of NOX2 system, lead to the loss of the GABAergic phenotype of PV-interneurons and to the decreased inhibitory activity in prefrontal cortex (Behrens et al., 2007, Behrens and Sejnowski, 2009, Powell et al., 2012). Microglia are known to have some neurotransmitter receptors including dopamine D2 receptors (Pocock and Kettenmann, 2007). However, since second generation drugs such as olanzapine have positive effects on neuronal cell growth and survival by unique signaling pathways (Lu and Dwyer, 2005), the pharmacological basis for their neuroprotective effect appears not to be only directly related to the conventional neurotransmitter receptors. All of these studies suggest that some antipsychotics may therefore have a potentially useful therapeutic effect on patients with schizophrenia by reducing the microglial inflammatory reactions, which may cause the apoptotic process, and the white matter abnormalities in the brains of patients with schizophrenia. It is consistent with the evidence showing their influences on slowing the progressive reduction in cortical gray matter in schizophrenia (Lieberman et al., 2005). However, some recent reports have shown some antipsychotics to increase the production of pro-inflammatory cytokines (Kluge et al., 2009, Himmerich et al., 2011). Some recent reports have demonstrated the possible antipsychotic effect of minocycline which is a potent inhibitor of microglial activation. In these studies, adjunctive therapy of minocycline to antipsychotics were reported to be beneficial for the treatment of schizophrenia (Miyaoka et al., 2007, Miyaoka et al., 2008, Levkovitz et al., 2010, Kelly et al., 2011). A recent study by Chaudhry et al has demonstrated that adjunctive therapy of minocycline to antipsychotics predominatly improved negative symptoms, which are usually treatment –resistant to antipsychotics (Chaudhry et al., 2012).

On the other hand, microglia can secrete neurotrophic factors other than proinflammatory cytokines including BDNF and free radicals. BDNF is well known to have a key role in the brain developmental problems associated with schizophrenia (Weickert et al., 2003). In addition, some in vitro data indicate an impairment of intracellular Ca2+ signaling in patients with schizophrenia (Bojarski et al., 2010). Recently, we have shown that BDNF induces sustained elevation of intracellular Ca2+ in rodent microglia, which might be important for the pathopsysiology and/or the treatment of schizophrenia (Mizoguchi et al., 2009). A recent study has shown that α7 nicotinic acetylcholine receptor agonist (α7nAChR) can modify microglial activation toward a neuroprotective role by suppressing the inflammatory state and strengthening the protective function (Suzuki et al., 2006). These results are very interesting because some α7nAChR agonists are known to improve the cognitive dysfunction of schizophrenia (Lieberman et al., 2008). Omega-3 fatty acids, which can inhibit microglial activation, have recently been reported to reduce the rate of progression to first-episode psychotic disorders (Lu et al., 2010, Amminger et al., 2010). Based upon the above results, the appropriate control of microglial activation may thus be a promising target for the prevention and treatment of schizophrenia.

Immunomodulatory drugs such as cyclooxygenase-2 (COX2) inhibitors have recently been reported to show beneficial effects on schizophrenic symptoms. Another recent randomized, double-blind, placebo-controlled trial has shown that adjuvant acetylsalicylic acid (aspirin) therapy reduces symptoms of schizophrenia spectrum disorders (Akhondzadeh et al., 2007, Muller et al., 2005, Muller et al., 2010, Laan et al., 2010). A recent meta-analysis on the use of non-steroidal anti-inflammatory agents (NSAIDs) in schizophrenia have demonstrated that NSAID augmentation could be a potentially useful strategy for the treatment of positive and negative symptoms of schizophrenia and that aspirin may have the additional benefit reducing cardiac and cancer mortality in schizophrenia (Sommer et al., 2012&2014). Successful treatment of psychosis with infliximab, a monoclonal antibody against TNF-α has also been reported in a patients with Crohn’s disease (Reimer et al., 2009). Immunosuppressive or immunomodulatory drugs may thus be beneficial at least for the treatment of acute schizophrenia (Knight et al., 2007). Controlling anti-inflammatory signaling is also important for the treatment of schizophrenia (Meyer, 2011). Reactive oxygen species derived from activated microglia are also relevant to the pathophysiology of schizophrenia. Anti-oxidants such as N-Acetyl-Cysteine (NAC) may thus be useful for the treatment of schizophrenia. Some randomized, double-blind studies shown the effectiveness as the augmentation therapy of schizophrenia of NAC (Sansone and Sansone, 2011).

CONCLUSIONS

In many aspects, the neuropathology of schizophrenia is closely associated with neuroinflammation, especially microglial activation. As described above, their respective anti-neuroinflammation therapies are also strikingly similar. Our understanding about the connection between microglial abnormalities and neuropsychiatric disorders is at its infant stage, but we foresee the tremendous potential of therapeutic approaches targeting microglia. This anticipation is based on two considerations. First, microglia quickly respond to various stimuli with an amazingly dynamic behavioral repertoire, therefore are highly transmutable by therapeutic interventions. Second, our view of the brain being “immune privileged” is fundamentally changed in recent years, with the realization that the systemic immune system has the ability to modulate multiple brain functions (Lynch and Mills, 2012), in part through interactions between peripheral immune cells such as dendritic cells and mast cells with their CNS counterparts, such as microglia. Therefore, approaches to modify the neuroinflammatory environment that is conducive to neuropsychiatric disorders can be launched peripherally. Peripheral immune cells are relatively straightforward to isolate and manipulate ex vivo, and are able to enter the CNS via endogenous mechanisms (Derecki et al., 2013), providing a foundation for novel cell-based therapies or “vaccines”. To achieve this goal, a fundamental question that needs to be investigated in depth is how peripheral immune modulation affects brain function. In our view, schizophrenia provides excellent opportunities for gaining such understanding of the complex interactions between CNS and systemic immune system, a gateway to a true “mind-body” problem (Feigenson et al., 2014).

THE BULLET POINTS

*An accumulating body of evidences point to the significance of neuroinflammation in schizophrenia.

*The neuropathology of schizophrenia is closely associated with microglial abnormalities in many aspects.

*Some typical or atypical antipsychotics have been reported to inhibit the release of inflammatory cytokines and free radicals from activated microglia.

*The treatment through the control of microglial abnormalities may shed new light on the therapeutic strategy of schizophrenia.

REFERENCES

1. Akhondzadeh S, Tabatabaee M, Amini H, Ahmadi Abhari SA, Abbasi SH, Behnam B. Celecoxib as adjunctive therapy in schizophrenia: a double-blind, randomized and placebo-controlled trial. Schizophr Res. 2007; 90: 179-185.

2. Amminger GP, Schafer MR, Papageorgiou K, klier CM, Cotton SM, Harrigan SM, et al. Long-chain omega-3 fatty acids for indicated prevention of psychotic disorders: a randomized, placebo-controlled trial. Archives of General Psychiatry. 2010; 67: 146-154.

3. Arion D, Unger T, Lewis DA, Levitt P, Mirnics K. Molecular evidence for increased expression of genes related to immune and chaperone function in the prefrontal cortex in schizophrenia. Biological Psychiatry. 2007; 62: 711-721.

4. Asadabadi M, Mohammadi MR, Ghanizadeh A, Modabbernia A, Ashrafi M, Hassanzadeh E, Forghani S. Celecoxib as adjunctive treatment to risperidone in children with autistic disorder: a randomized, doubleblind, placebo-controlled trial. Psychopharmacology (Berl). 2013; 225: 51-59.

5. Ashdown H, Dumont Y, Ng M, Poole S, Boksa P, Luheshi GN. The role of cytokines in mediating effects of prenatal infection on the fetus: implications for schizophrenia. Mol Psychiatry. 2006; 11: 47-55.

6. Bayer TA, Buslei R, Havas L, Falkai P. Evidence for activation of microglia in patients with psychiatric illnesses. Neurosci Lett. 1999; 271: 126-128.

7. Behrens MM, Ali SS, Dao DN, Lucero J, Shekhtman G, Quick KL, et al. Ketamine-induced loss of phenotype of fast-spiking interneurons is mediated by NADPH-oxidase. Science. 2007; 318: 1645-1647.

8. Behrens MM, Sejnowski TJ. Does schizophrenia arise from oxidative dysregulation of parvalbumin-interneurons in the developing cortex? Neuropharmacology. 2009; 57: 193-200.

9. Benavides J, Cornu P, Dennis T, Dubois A, Hauw JJ, MacKenzie ET, et al. Imaging of human brain lesions with an omega 3 site radioligand. Ann Neurol. 1988; 24: 708-712.

10. Bernstein HG, Steiner J, Bogerts B. Glial cells in schizophrenia: pathophysiological significance and possible consequences for therapy. Expert Rev Neurother. 2009; 9: 1059-1071.

11. Beumer W, Drexhage RC, Dewit H, Versnel MA, Drexhage HA, Cohen D. Increased level of serum cytokines, chemokines and adipokines in patients with schizophrenia is associated with disease and metabolic syndrome. Psychoneuroendocrinology. 2012; 37: 1901-1911.

12. Bian Q, Kato T, Monji A, Hashioka S, Mizoguchi Y, Horikawa H, et al. The effect of atypical antipsychotics, perospirone, ziprasidone and quetiapine on microglial activation induced by interferon-gamma. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2008; 32: 42-48.

13. Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the immune system. Front Behav Neurosci. 2009; 3: 14.

14. Bland ST, Beckley JT, Young S, Tsang V, Watkins LR, Maier SF, et al. Enduring consequences of early-life infection on glial and neural cell genesis within cognitive regions of the brain. Brain, Behavior, and Immunity. 2010; 24: 329-338.

15. Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol. 2005; 76: 77-98.

16. Bojarski L, Debowska K, Wojda U. In vitro findings of alterations in intracellular calcium homeostasis in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2010; 34: 1367-1374.

17. Brown AS, Derkits EJ. Prenatal infection and schizophrenia: a review of epidemiologic and translational studies. Am J Psychiatry. 2010; 167: 261-280.

18. Buntinx M, Moreels M, Vandenabeele F, Lambrichts I, Raus J, Steels P, et al. Cytokine-induced cell death in human oligodendroglial cell lines: I. Synergistic effects of IFN-gamma and TNF-alpha on apoptosis. J Neurosci Res. 2004; 76: 834-845.

19. Cacci E, Claasen JH, kokaia Z. Microglia-derived tumor necrosis factor-alpha exaggerates death of newborn hippocampal progenitor cells in vitro. Journal of Neuroscience Research. 2005; 80: 789-797.

20. Cammer W, Zhang H. Maturation of oligodendrocytes is more sensitive to TNF alpha than is survival of precursors and immature oligodendrocytes. J Neuroimmunol. 1999; 97: 37-42.

21. Cazzullo CL, Sacchetti E, Galluzzo A, Panariello A, Colombo F, Zagliani A, et al. Cytokine profiles in drug-naive schizophrenic patients. Schizophr Res. 2001; 47: 293-298.

22. Chakos MH, Schobel SA, Gu H, Gerig G, Bradford D, Charles C, et al. Duration of illness and treatment effects on hippocampal volume in male patients with schizophrenia. Br J Psychiatry. 2005; 186: 26-31.

23. Chaudhry IB, Hallak J, Husain N, Minhas F, Stirling J, Richardson P, et al. 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.

24. Chavez B, Chavez-Brown M, Sopko MA Jr, Rey JA. Atypical antipsychotics in children with pervasive developmental disorders. Paediatr Drugs. 2007; 9: 249-266.

25. Cristino AS, Williams SM, Hawi Z, An JY, Bellgrove MA, Schwartz CE, et al. Neurodevelopmental and neuropsychiatric disorders represent an interconnected molecular system. Molecular Psychiatry. 2014; 19: 294-301.

26. Dansie LE, Phommahaxay K, Okusanya AG, Uwadia J, Huang M, Rotschafer SE, et al. Long-lasting effects of minocycline on behavior in young but not adult Fragile X mice. Neuroscience. 2013; 246: 186-198.

27. Davis KL, Stewart DG, Friedman JI, Buchsbaum M, Harvey PD, Hof PR, et al. White matter changes in schizophrenia: evidence for myelin-related dysfunction. Arch Gen Psychiatry. 2003; 60: 443-456.

28. Dean B, Gibbons AS, Tawadros N, Brooks L, Everall IP, Scarr E. Different changes in cortical tumor necrosis factor-α-related pathways in schizophrenia and mood disorders. Mol Psychiatry. 2013; 18: 767- 773.

29. Derecki NC, Cronk JC, Kipnis J. The role of microglia in brain maintenance: implications for Rett syndrome. Trends Immunol. 2013; 34: 144-150.

30. Devaraj S, Dasu MR, Jialal I. Diabetes is a proinflammatory state: a translational perspective. Expert Rev Endocrinol Metab. 2010; 5: 19- 28.

31. Deverman BE, Patterson PH. Cytokines and CNS development. Neuron. 2009; 64: 61-78.

32. Dickerson F, Stallings C, Origoni A, Vaughan C, Khushalani S, Yang S, et al. C-reactive protein is elevated in schizophrenia. Schizophr Res. 2013; 143: 198-202.

33. Dieset I, Hope S, Ueland T, Bjella T, Agartz I, Melle I, et al. Cardiovascular risk factors during second generation antipsychotic treatment are associated with increased C-reactive protein. Schizophrenia Research. 2012; 140: 169-174.

34. Doorduin J, de Vries EF, Willemsen AT, de Groot JC, Dierckx RA, Klein HC. Neuroinflammation in schizophrenia-related psychosis: a PET study. J Nucl Med. 2009; 50: 1801-1807.

35. Drexhage RC, Knijff EM, Padmos RC, Heul-Nieuwenhuijzen Lv, Beumer W, Versnel MA, et al. The mononuclear phagocyte system and its cytokine inflammatory networks in schizophrenia and bipolar disorder. Expert Rev Neurother. 2010; 10: 59-76.

36. Drzyzga L, Obuchowicz E, Marcinowska A, Herman ZS. Cytokines in schizophrenia and the effects of antipsychotic drugs. Brain Behav Immun. 2006; 20: 532-545.

37. Duan X, Chang JH, Ge S, Faulkner RL, Kim JY, Kitabatake Y, et al. Disrupted-In-Schizophrenia 1 regulates integration of newly generated neurons in the adult brain. Cell. 2007; 130: 1146-1158.

38. Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A. 2003; 100: 13632-13637.

39. Ellman LM, Susser ES. The promise of epidemiologic studies: neuroimmune mechanisms in the etiologies of brain disorders. Neuron. 2009; 64: 25-27.

40. Feldhaus B, Dietzel ID, Heumann R, Berger R. Effects of interferon-gamma and tumor necrosis factor-alpha on survival and differentiation of oligodendrocyte progenitors. Journal of the Society for Gynecologic Investigation. 2004; 1: 89-96.

41. Fellerhoff B, Wank R. Increased prevalence of Chlamydophila DNA in post-mortem brain frontal cortex from patients with schizophrenia. Schizophr Res. 2011; 129: 191-195.

42. Feigenson KA, Kusnecov AW, Silverstein SM. Inflammation and the two-hit hypothesis of schizophrenia. Neurosci Biobehav Rev. 2014; 38: 72-93.

43. Fillman SG, Cloonan N, Catts VS, Miller LC, Wong J, Mccrossin T, et al. Increased inflammatory markers identified in the dorsolateral prefrontal cortex of individuals with schizophrenia. Molecular Psychiatry. 2013; 18: 206-214.

44. Frank MG, Baratta MV, Sprunger DB, Watkins LR, Maier SF. Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav Immun. 2007; 21: 47-59.

45. Ghanizadeh A, Moghimi-Sarani E. A randomized double blind placebo controlled clinical trial of N-Acetylcysteine added to risperidone for treating autistic disorders. BMC Psychiatry. 2013; 13: 196.

46. Ghanizadeh A, Sahraeizadeh A, Berk M. A head-to-head comparison of aripiprazole and risperidone for safety and treating autistic disorders, a randomized double blind clinical trial. Child Psychiatry Hum Dev. 2014; 45: 185-192.

47. Girgis RR, Diwadkar VA, Nutche JJ, Sweeney JA, Keshavan MS, Hardan AY. Risperidone in first-episode psychosis: a longitudinal, exploratory voxel-based morphometric study. Schizophr Res. 2006; 82: 89-94.

48. Glantz LA, Gilmore JH, Lieberman JA, Jarskog LF. Apoptotic mechanisms and the synaptic pathology of schizophrenia. Schizophr Res. 2006; 81: 47-63.

49. Himmerich H, Schönherr J, Fulda S, Sheldrick AJ, Bauer K, Sack U. Impact of antipsychotics on cytokine production in-vitro. J Psychiatr Res. 2011; 45: 1358-1365.

50. Hinwood M, Morandini J, Day TA, Walker FR. Evidence that Microglia Mediate the Neurobiological Effects of Chronic Psychological Stress on the Medial Prefrontal Cortex. Cerebral Cortex. 2012; 22: 1442-1454.

51. Ho BC, Andreasen NC, Ziebell S, Pierson R, Magnotta V. Long-term antipsychotic treatment and brain volumes: a longitudinal study of first-episode schizophrenia. Arch Gen Psychiatry. 2011; 68: 128-137.

52. Hou Y, Wu CF, Yang JY, He X, Bi XL, Yu L, et al. Effects of clozapine, olanzapine and haloperidol on nitric oxide production by lipopolysaccharide-activated N9 cells. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2006; 30: 1523-1528.

53. Hu S, Peterson PK, Chao CC. Cytokine-mediated neuronal apoptosis. Neurochem Int. 1997; 30: 427-431.

54. Hulshoff Pol HE, Kahn RS. What happens after the first episode? A review of progressive brain changes in chronically ill patients with schizophrenia. Schizophr Bull. 2008; 34: 354-366.

55. Iosif RE, Ekdahl CT, Ahlenius H, Pronk CJH, Bonde S, Kokaia Z, et al. Tumor necrosis factor receptor 1 is a negative regulator of progenitor proliferation in adult hippocampal neurogenesis. Journal of Neuroscience. 2006; 26: 9703-9712.

56. Iritani S. Neuropathology of schizophrenia: a mini review. Neuropathology. 2007; 27: 604-608.

57. Jarskog LF, Glantz LA, Gilmore JH, Lieberman JA. Apoptotic mechanisms in the pathophysiology of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2005; 29: 846-858.

58. Jia P, Wang L, Meltzer HY, Zhao Z. Common variants conferring risk of schizophrenia: a pathway analysis of GWAS data. Schizophr Res. 2010; 122: 38-42.

59. Liu J, Suzuki T, Seki T, Namba T, Tanimura A, Arai H. Effects of repeated phencyclidine administration on adult hippocampal neurogenesis in the rat. Synapse. 2006; 60: 56-68.

60. Juckel G, Manitz MP, Brüne M, Friebe A, Heneka MT, Wolf RJ. Microglial activation in a neuroinflammational animal model of schizophrenia--a pilot study. Schizophr Res. 2011; 131: 96-100.

61. Kaneko N, Kudo K, Mabuchi T, Takemoto K, Fujimaki K, Wati H, et al. Suppression of cell proliferation by interferon-alpha through interleukin-1 production in adult rat dentate gyrus. Neuropsychopharmacology. 2006; 31: 2619-2626.

62. Kato T, Mizoguchi Y, Monji A, Horikawa H, Suzuki SO, Seki Y, et al. Inhibitory effects of aripiprazole on interferon-gamma-induced microglial activation via intracellular Ca2+ regulation in vitro. J Neurochem. 2008; 106: 815-825.

63. Kato T, Monji A, Hashioka S, Kanba S. Risperidone significantly inhibits interferon-gamma-induced microglial activation in vitro. Schizophr Res. 2007; 92: 108-115.

64. Kelly DL, Vyas G, Richardson CM, Koola M, McMahon RP, Buchanan RW, et al. Adjunct minocycline to clozapine treated patients with persistent schizophrenia symptoms. Schizophr Res. 2011; 133: 257- 258.

65. King BH, Lord C. Is schizophrenia on the autism spectrum? Brain Res. 2011; 1380: 34-41.

66. Kluge M, Schuld A, Schacht A, Himmerich H, Dalal MA, Wehmeier PM, et al. Effects of clozapine and olanzapine on cytokine systems are closely linked to weight gain and drug-induced fever. Psychoneuroendocrinology. 2009; 34: 118-128.

67. Knight JG, Menkes DB, Highton J, Adams DD. Rationale for a trial of immunosuppressive therapy in acute schizophrenia. Mol Psychiatry. 2007; 12: 424-431.

68. Kodama M, Fujioka T, Duman RS. Chronic olanzapine or fluoxetine administration increases cell proliferation in hippocampus and prefrontal cortex of adult rat. Biol Psychiatry. 2004; 56: 570-580.

69. Kowalski J, Labuzek K, Herman ZS. Flupentixol and trifluperidol reduce secretion of tumor necrosis factor-alpha and nitric oxide by rat microglial cells. Neurochem Int. 2003; 43: 173-178.

70. Kowalski J, Labuzek K, Herman ZS. Flupentixol and trifluperidol reduce interleukin-1 beta and interleukin-2 release by rat mixed glial and microglial cell cultures. Pol J Pharmacol. 2004; 56: 563-570.

71. Kumra S, Ashtari M, Cervellione KL, Henderson I, Kester H, Roofeh D, et al. White matter abnormalities in early-onset schizophrenia: a voxel-based diffusion tensor imaging study. J Am Acad Child Adolesc Psychiatry. 2005; 44: 934-941.

72. Laan W, Grobbee DE, Selten JP, Heijnen CJ, Kahn RS, Burger H. Adjuvant aspirin therapy reduces symptoms of schizophrenia spectrum disorders: results from a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry. 2010; 7: 520-527.

73. Labuzek K, Kowalski J, Gabryel B, Herman ZS. Chlorpromazine and loxapine reduce interleukin-1beta and interleukin-2 release by rat mixed glial and microglial cell cultures. Eur Neuropsychopharmacol. 2005; 15: 23-30.

74. Lee SH, Kubicki M, Asami T, Seidman LJ, Goldstein JM, Mesholam-Gately RI, et al. Extensive white matter abnormalities in patients with first-episode schizophrenia: a Diffusion Tensor Imaging (DTI) study. Schizophrenia Research. 2013; 143: 231-238.

75. Leigh MJ, Nguyen DV, Mu Y, Winarni TI, Schneider A, Chechi T, et al. A randomized double-blind, placebo-controlled trial of minocycline in children and adolescents with fragile x syndrome. J Dev Behav Pediatr. 2013; 34: 147-155.

76. Levkovitz Y, Mendlovich S, Riwkes S, Braw Y, Levkovitch-Verbin H, Gal G, et al. A double-blind, randomized study of minocycline for the treatment of negative and cognitive symptoms in early-phase schizophrenia. J Clin Psychiatry. 2010; 71: 138-149.

77. Li J, Baud O, Vartanian T, Volpe JJ, Rosenberg PA. Peroxynitrite generated by inducible nitric oxide synthase and NADPH oxidase mediates microglial toxicity to oligodendrocytes. Proc Natl Acad Sci U S A. 2005; 102: 9936-9941.

78. Li Y, Du XF, Liu CS, Wen ZL, Du JL. Reciprocal regulation between resting microglial dynamics and neuronal activity in vivo. Dev Cell. 2012; 23: 1189-1202.

79. Lieberman JA. Is schizophrenia a neurodegenerative disorder? A clinical and neurobiological perspective. Biol Psychiatry. 1999; 46: 729-739.

80. Lieberman JA, Javitch JA, Moore H. Cholinergic agonists as novel treatments for schizophrenia: the promise of rational drug development for psychiatry. Am J Psychiatry. 2008; 165: 931-936.

81. Lieberman JA, Tollefson GD, Charles C, Zipursky R, Sharma T, Kahn RS, et al. Antipsychotic drug effects on brain morphology in first-episode psychosis. Arch Gen Psychiatry. 2005; 62: 361-370.

82. Lin A, Kenis G, Bignotti S, Tura GJ, De Jong R, Bosmans E, et al. The inflammatory response system in treatment-resistant schizophrenia: increased serum interleukin-6. Schizophr Res. 1998; 32: 9-15.

83. Lu DY, Tsao YY, Leung YM, Su KP. Docosahexaenoic acid suppresses neuroinflammatory responses and induces heme oxygenase-1 expression in BV-2 microglia: implications of antidepressant effects for omega-3 fatty acids. Neuropsychopharmacology. 2010; 35: 2238- 2248.

84. Lu XH, Dwyer DS. Second-generation antipsychotic drugs, olanzapine, quetiapine, and clozapine enhance neurite outgrowth in PC12 cells via PI3K/AKT, ERK, and pertussis toxin-sensitive pathways. J Mol Neurosci. 2005; 27: 43-64.

85. Lugnegård T, Unenge Hallerbäck M, Hjärthag F, Gillberg C. Social cognition impairments in Asperger syndrome and schizophrenia. Schizophr Res. 2013; 143: 277-284.

86. Lynch MA, Mills KH. Immunology meets neuroscience--opportunities for immune intervention in neurodegenerative diseases. Brain Behav Immun. 2012; 26: 1-10.

87. MacDowell KS, García-Bueno B, Madrigal JL, Parellada M, Arango C, Micó JA, et al. Risperidone normalizes increased inflammatory parameters and restores anti-inflammatory pathways in a model of neuroinflammation. Int J Neuropsychopharmacol. 2013; 16: 121-135.

88. Maeda K, Sugino H, Hirose T, Kitagawa H, Nagai T, Mizoguchi H, et al. Clozapine prevents a decrease in neurogenesis in mice repeatedly treated with phencyclidine. J Pharmacol Sci. 2007; 103: 299-308.

89. Massana G, Salgado-Pineda P, Junque C, Perez M, Baeza I, Pons A, et al. Volume changes in gray matter in first-episode neuroleptic-naive schizophrenic patients treated with risperidone. Journal of Clinical Psychopharmacology. 2005; 25: 111-117.

90. McCracken JT, McGough J, Shah B, Cronin P, Hong D, Aman MG, et al. Risperidone in children with autism and serious behavioral problems. N Engl J Med. 2002; 347: 314-321.

91. McCullumsmith RE, Gupta D, Beneyto M, Kreger E, Haroutunian V, Davis KL, et al. Expression of transcripts for myelination-related genes in the anterior cingulate cortex in schizophrenia. Schizophr Res. 2007; 90: 15-27.

92. McDougle CJ, Stigler KA, Erickson CA, Posey DJ. Atypical antipsychotics in children and adolescents with autistic and other pervasive developmental disorders. J Clin Psychiatry. 2008; 69 Suppl 4: 15-20.

93. Medina S, Martinez M, Hernanz A. Antioxidants inhibit the human cortical neuron apoptosis induced by hydrogen peroxide, tumor necrosis factor alpha, dopamine and beta-amyloid peptide 1-42. Free Radical Research. 2002; 36: 1179-1184.

94. Merrill JE, Ignarro LJ, Sherman MP, Melinek J, Lane TE. Microglial cell cytotoxicity of oligodendrocytes is mediated through nitric oxide. J Immunol. 1993; 151: 2132-2141.

95. Meyer JM, Stahl SM. The metabolic syndrome and schizophrenia. Acta Psychiatrica Scandinavica. 2009; 119: 4-14.

96. Meyer U. Anti-inflammatory signaling in schizophrenia. Brain Behav Immun. 2011; 25: 1507-1518.

97. Miller BJ, Buckley P, Seabolt W, Mellor A, Kirkpatrick B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2011; 70: 663-671.

98. Miller BJ, Gassama B, Sebastian D, Buckley P, Mellor A. Meta-analysis of lymphocytes in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2013; 73: 993-999.

99. Mittelbronn M, Dietz K, Schluesener HJ, Meyermann R. Local distribution of microglia in the normal adult human central nervous system differs by up to one order of magnitude. Acta Neuropathol. 2001; 101: 249-255.

100. Miyaoka T, Yasukawa R, Yasuda H, Hayashida M, Inagaki T, Horiguchi J. Possible antipsychotic effects of minocycline in patients with schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2007; 31: 304-307.

101. Miyaoka T, Yasukawa R, Yasuda H, Hayashida M, Inagaki T, Horiguchi J. Minocycline as adjunctive therapy for schizophrenia: an openlabel study. Clin Neuropharmacol. 2008; 31: 287-292.

102. Mizoguchi Y, Monji A, Kato T, Seki Y, Gotoh L, Horikawa H, et al. Brain-derived neurotrophic factor induces sustained elevation of intracellular Ca2+ in rodent microglia. J Immunol. 2009; 183: 7778- 7786.

103. Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science. 2003; 302: 1760-1765.

104. Morgan JT, Chana G, Pardo CA, Achim C, Semendeferi K, Buckwalter J, et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry. 2010; 68: 368-376.

105. Muller N, Krause, D., Dehning, S., Musil, R., Schennach-Wolff, R., Obermeier, M., Moller, H. J., Klauss, V., Schwarz, M. J. & Riedel, M. 2010. Celecoxib treatment in an early stage of schizophrenia: results of a randomized, double-blind, placebo-controlled trial of celecoxib augmentation of amisulpride treatment. Schizophrenia Research, 12, 118-24.

106. Müller N, Riedel M, Schwarz MJ, Engel RR. Clinical effects of COX-2 inhibitors on cognition in schizophrenia. Eur Arch Psychiatry Clin Neurosci. 2005; 255: 149-151.

107. Näkki R, Koistinaho J, Sharp FR, Sagar SM. Cerebellar toxicity of phencyclidine. J Neurosci. 1995; 15: 2097-2108.

108. NAKKI, R., NICKOLENKO, J., CHANG, J., SAGAR, S. M. & SHARP, F. R. 1996. Haloperidol prevents ketamine- and phencyclidine-induced HSP70 protein expression but not microglial activation. Exp Neurol, 137, 234-41.

109. Narayan S, Tang B, Head SR, Gilmartin TJ, Sutcliffe JG, Dean B, Thomas EA. Molecular profiles of schizophrenia in the CNS at different stages of illness. Brain Res. 2008; 1239: 235-248.

110. NAWA, H. & TAKEI, N. 2006. Recent progress in animal modeling of immune inflammatory processes in schizophrenia: Implication of specific cytokines. Neuroscience Research, 56, 2-13.

111. Newschaffer CJ, Croen LA, Daniels J, Giarelli E, Grether JK, Levy SE, Mandell DS. The epidemiology of autism spectrum disorders. Annu Rev Public Health. 2007; 28: 235-258.

112. Nielsen J, Skadhede S, Correll CU. Antipsychotics associated with the development of type 2 diabetes in antipsychotic-naïve schizophrenia patients. Neuropsychopharmacology. 2010; 35: 1997-2004.

113. Owen R, Sikich L, Marcus RN, Corey-Lisle P, Manos G, McQuade RD, Carson WH. Aripiprazole in the treatment of irritability in children and adolescents with autistic disorder. Pediatrics. 2009; 124: 1533- 1540.

114. Palluy O, Rigaud M. Nitric oxide induces cultured cortical neuron apoptosis. Neurosci Lett. 1996; 208: 1-4.

115. Pardo CA, Buckley A2, Thurm A2, Lee LC3, Azhagiri A, Neville DM2,Swedo SE2. A pilot open-label trial of minocycline in patients with autism and regressive features. J Neurodev Disord. 2013; 5: 9.

116. Paribello C, Tao L, Folino A, Berry-Kravis E, Tranfaglia M, Ethell IM, Ethell DW. Open-label add-on treatment trial of minocycline in fragile X syndrome. BMC Neurol. 2010; 10: 91.

117. Penzes P, Cahill ME, Jones KA, VanLeeuwen JE, Woolfrey KM. Dendritic spine pathology in neuropsychiatric disorders. Nat Neurosci. 2011; 14: 285-293.

118. Pérez-Neri I, Ramírez-Bermúdez J, Montes S, Ríos C. Possible mechanisms of neurodegeneration in schizophrenia. Neurochem Res. 2006; 31: 1279-1294.

119. Pickard BS, Pieper AA, Porteous DJ, Blackwood DH, Muir WJ. The NPAS3 gene--emerging evidence for a role in psychiatric illness. Ann Med. 2006; 38: 439-448.

120. Pieper AA, Wu X, Han TW, Estill SJ, Dang Q, Wu LC, Reece-Fincanon S. The neuronal PAS domain protein 3 transcription factor controls FGF-mediated adult hippocampal neurogenesis in mice. Proc Natl Acad Sci U S A. 2005; 102: 14052-14057.

121. PIKE, V. W., HALLDIN, C., CROUZEL, C., BARRE, L., NUTT, D. J., OSMAN, S., SHAH, F., TURTON, D. R. & WATERS, S. L. 1993. Radioligands for PET studies of central benzodiazepine receptors and PK (peripheral benzodiazepine) binding sites--current status. Nucl Med Biol, 20, 503-25.

122. Pocock JM, Kettenmann H. Neurotransmitter receptors on microglia. Trends Neurosci. 2007; 30: 527-535.

123. 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: 801-808.

124. POWELL, S. B., SEJNOWSKI, T. J. & BEHRENS, M. M. 2012. Behavioral and neurochemical consequences of cortical oxidative stress on parvalbumin-interneuron maturation in rodent models of schizophrenia. Neuropharmacology, 62, 1322-31.

125. PRICE, G., CERCIGNANI, M., BAGARY, M. S., BARNES, T. R., BARKER, G. J., JOYCE, E. M. & RON, M. A. 2006. A volumetric MRI and magnetization transfer imaging follow-up study of patients with first-episode schizophrenia. Schizophrenia Research, 87, 100-8.

126. Radewicz K, Garey LJ, Gentleman SM, Reynolds R. Increase in HLA-DR immunoreactive microglia in frontal and temporal cortex of chronic schizophrenics. J Neuropathol Exp Neurol. 2000; 59: 137- 150.

127. Reif A, Fritzen S, Finger M, Strobel A, Lauer M, Schmitt A, Lesch KP. Neural stem cell proliferation is decreased in schizophrenia, but not in depression. Mol Psychiatry. 2006; 11: 514-522.

128. Reimer J, Fink T, Bläker M, Schäfer I, Otte C. Successful treatment of psychosis with infliximab in a patient with Crohn’s disease. Schizophr Res. 2009; 109: 194-195.

129. Roberts RC, Roche JK, Conley RR. Synaptic differences in the postmortem striatum of subjects with schizophrenia: a stereological ultrastructural analysis. Synapse. 2005; 56: 185-197.

130. Rothermundt M, Arolt V, Bayer TA. Review of immunological and immunopathological findings in schizophrenia. Brain Behav Immun. 2001; 15: 319-339.

131. Sahin M. Targeted treatment trials for tuberous sclerosis and autism: no longer a dream. Curr Opin Neurobiol. 2012; 22: 895-901.

132. Salisbury DF, Kuroki N, Kasai K, Shenton ME, McCarley RW. Progressive and interrelated functional and structural evidence of post-onset brain reduction in schizophrenia. Arch Gen Psychiatry. 2007; 64: 521-529.

133. Sansone RA, Sansone LA. Getting a Knack for NAC: N-Acetyl-Cysteine. Innov Clin Neurosci. 2011; 8: 10-14.

134. Schiavone S, Sorce S, Dubois-Dauphin M, Jaquet V, Colaianna M, Zotti M, Cuomo V. Involvement of NOX2 in the development of behavioral and pathologic alterations in isolated rats. Biol Psychiatry. 2009; 66: 384-392.

135. Schlösser RG, Nenadic I, Wagner G, Güllmar D, von Consbruch K, Köhler S, Schultz CC. White matter abnormalities and brain activation in schizophrenia: a combined DTI and fMRI study. Schizophr Res. 2007; 89: 1-11.

136. SCHMITT, A., BERTSCH, T., HENNING, U., TOST, H., KLIMKE, A., HENN, F. A. & FALKAI, P. 2005. Increased serum S100B in elderly, chronic schizophrenic patients: Negative correlation with deficit symptoms. Schizophrenia Research, 80, 305-313.

137. Schnieder TP, Dwork AJ. Searching for neuropathology: gliosis in schizophrenia. Biol Psychiatry. 2011; 69: 134-139.

138. Schwarz MJ, Müller N, Riedel M, Ackenheil M. The Th2-hypothesis of schizophrenia: a strategy to identify a subgroup of schizophrenia caused by immune mechanisms. Med Hypotheses. 2001; 56: 483- 486.

139. SHANG, Y. C., CHONG, Z. Z., WANG, S. H. & MAIESE, K. 2013. Tuberous Sclerosis Protein 2 (TSC2) Modulates CCN4 Cytoprotection During Apoptotic Amyloid Toxicity in Microglia. Current Neurovascular Research, 10, 29-38.

140. Skaper SD, Giusti P, Facci L. Microglia and mast cells: two tracks on the road to neuroinflammation. FASEB J. 2012; 26: 3103-3117.

141. Söderlund J, Schröder J, Nordin C, Samuelsson M, Walther-Jallow L, Karlsson H, Erhardt S. Activation of brain interleukin-1beta in schizophrenia. Mol Psychiatry. 2009; 14: 1069-1071.

142. Sommer IE, de Witte L, Begemann M, Kahn RS. Nonsteroidal anti-inflammatory drugs in schizophrenia: ready for practice or a good start? A meta-analysis. J Clin Psychiatry. 2012; 73: 414-419.

143. Sommer IE, van Westrhenen R, Begemann MJ, de Witte LD, Leucht S, Kahn RS. Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophr Bull. 2014; 40: 181-191.

144. Sommer IE, van Westrhenen R, Begemann MJ, de Witte LD, Leucht S, Kahn RS. Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophr Bull. 2014; 40: 181-191.

145. STEINER J., BERSTEIN H. G., SCHILTZ K., MULLER U. J., WESTPHAL S.,

146. Steiner J, Bernstein HG, Schiltz K, Müller UJ, Westphal S, Drexhage HA, Bogerts B. Immune system and glucose metabolism interaction in schizophrenia: a chicken-egg dilemma. Prog Neuropsychopharmacol Biol Psychiatry. 2014; 48: 287-294.

147. STEFANSSON, H., OPHOFF, R. A., STEINBERG, S., ANDREASSEN, O. A., CICHON, S., RUJESCU, D., WERGE, T., PIETILAINEN, O. P. H., MORS, O., MORTENSEN, P. B., SIGURDSSON, E., GUSTAFSSON, O., NYEGAARD, M., TUULIO-HENRIKSSON, A., INGASON, A., HANSEN, T., SUVISAARI, J., LONNQVIST, J., PAUNIO, T., BORGLUM, A. D., HARTMANN, A., FINK-JENSEN, A., NORDENTOFT, M., HOUGAARD, D., NORGAARDPEDERSEN, B., BOTTCHER, Y., OLESEN, J., BREUER, R., MOLLER, H. J., GIEGLING, I., RASMUSSEN, H. B., TIMM, S., MATTHEISEN, M., BITTER, I., RETHELYI, J. M., MAGNUSDOTTIR, B. B., SIGMUNDSSON, T., OLASON, P., MASON, G., GULCHER, J. R., HARALDSSON, M., FOSSDAL, R., THORGEIRSSON, T. E., THORSTEINSDOTTIR, U., RUGGERI, M., TOSATO, S., FRANKE, B., STRENGMAN, E., KIEMENEY, L. A., MELLE, I., DJUROVIC, S., ABRAMOVA, L., KALEDA, V., SANJUAN, J., DE FRUTOS, R., BRAMON, E., VASSOS, E., FRASER, G., ETTINGER, U., PICCHIONI, M., WALKER, N., TOULOPOULOU, T., NEED, A. C., GE, D., YOON, J. L., SHIANNA, K. V., FREIMER, N. B., CANTOR, R. M., MURRAY, R., KONG, A., GOLIMBET, V., CARRACEDO, A., ARANGO, C., COSTAS, J., JONSSON, E. G., TERENIUS, L., AGARTZ, I., PETURSSON, H., NOTHEN, M. M., RIETSCHEL, M., MATTHEWS, P. M., MUGLIA, P., PELTONEN, L., ST CLAIR, D., GOLDSTEIN, D. B., STEFANSSON, K., COLLIER, D. A. & GROUP 2009. Common variants conferring risk of schizophrenia. Nature, 460, 744-U99.

148. STEINER, J., MAWRIN, C., ZIEGELER, A., BIELAU, H., ULLRICH, O., BERNSTEIN, H. G. & BOGERTS, B. 2006. Distribution of HLA-DRpositive microglia in schizophrenia reflects impaired cerebral lateralization. Acta Neuropathol, 112, 305-16.

149. Stellwagen D, Malenka RC. Synaptic scaling mediated by glial TNF-alpha. Nature. 2006; 440: 1054-1059.

150. SUGINO, H., FUTAMURA, T., MITSUMOTO, Y., MAEDA, K. & MARUNAKA, Y. 2009. Atypical antipsychotics suppress production of proinflammatory cytokines and up-regulate interleukin-10 in lipopolysaccharide-treated mice. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 33, 303-7.

151. Sunico CR, Portillo F, González-Forero D, Moreno-López B. Nitric-oxide-directed synaptic remodeling in the adult mammal CNS. J Neurosci. 2005; 25: 1448-1458.

152. SUZUKI, T., HIDE, I., MATSUBARA, A., HAMA, C., HARADA, K., MIYANO, K., ANDRA, M., MATSUBAYASHI, H., SAKAI, N., KOHSAKA, S., INOUE, K. & NAKATA, Y. 2006. Microglial alpha7 nicotinic acetylcholine receptors drive a phospholipase C/IP3 pathway and modulate the cell activation toward a neuroprotective role. J Neurosci Res, 83, 1461-70.

153. Takano A, Arakawa R, Ito H, Tateno A, Takahashi H, Matsumoto R, Okubo Y. Peripheral benzodiazepine receptors in patients with chronic schizophrenia: a PET study with [11C]DAA1106. Int J Neuropsychopharmacol. 2010; 13: 943-950.

154. SUZUKI, K., SUGIHARA, G., OUCHI, Y., NAKAMURA, K., FUTATSUBASHI, M., TAKEBAYASHI, K., YOSHIHARA, Y., OMATA, K., MATSUMOTO, K., TSUCHIYA, K. J., IWATA, Y., TSUJII, M., SUGIYAMA, T. & MORI, N. 2013. Microglial Activation in Young Adults With Autism Spectrum Disorder. JAMA Psychiatry, 70, 49-58.

155. Tynan RJ, Naicker S, Hinwood M, Nalivaiko E, Buller KM, Pow DV, Day TA. Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions. Brain Behav Immun. 2010; 24: 1058-1068.

156. Uranova NA, Vostrikov VM, Orlovskaya DD, Rachmanova VI. Oligodendroglial density in the prefrontal cortex in schizophrenia and mood disorders: a study from the Stanley Neuropathology Consortium. Schizophr Res. 2004; 67: 269-275.

157. Uranova NA, Vostrikov VM, Vikhreva OV, Zimina IS, Kolomeets NS, Orlovskaya DD. The role of oligodendrocyte pathology in schizophrenia. Int J Neuropsychopharmacol. 2007; 10: 537-545.

158. van Berckel BN, Bossong MG, Boellaard R, Kloet R, Schuitemaker A, Caspers E, Luurtsema G. Microglia activation in recent-onset schizophrenia: a quantitative (R)-[11C]PK11195 positron emission tomography study. Biol Psychiatry. 2008; 64: 820-822.

159. van Os J, Kenis G, Rutten BP. The environment and schizophrenia. Nature. 2010; 468: 203-212.

160. Vismara LA, Rogers SJ. Behavioral treatments in autism spectrum disorder: what do we know? Annu Rev Clin Psychol. 2010; 6: 447- 468.

161. Wakade CG, Mahadik SP, Waller JL, Chiu FC. Atypical neuroleptics stimulate neurogenesis in adult rat brain. J Neurosci Res. 2002; 69: 72-79.

162. Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J Neurosci. 2009; 29: 3974-3980.

163. Wang H, Doering LC. Reversing autism by targeting downstream mTOR signaling. Front Cell Neurosci. 2013; 7: 28.

164. WANG, Q., CHEUNG, C., DENG, W., LI, M., HUANG, C., MA, X., WANG, Y., JIANG, L., SHAM, P. C., COLLIER, D. A., GONG, Q., CHUA, S. E., MCALONAN, G. M. & LI, T. 2013. White-matter microstructure in previously drug-naive patients with schizophrenia after 6 weeks of treatment. Psychological Medicine, 1-9.

165. Warner-Schmidt JL, Duman RS. Hippocampal neurogenesis: opposing effects of stress and antidepressant treatment. Hippocampus. 2006; 16: 239-249.

166. Weickert CS, Hyde TM, Lipska BK, Herman MM, Weinberger DR, Kleinman JE. Reduced brain-derived neurotrophic factor in prefrontal cortex of patients with schizophrenia. Mol Psychiatry. 2003; 8: 592-610.

167. WOHLEB, E. S., HANKE, M. L., CORONA, A. W., POWELL, N. D., STINER, L. M., BAILEY, M. T., NELSON, R. J., GODBOUT, J. P. & SHERIDAN, J. F. 2011. beta-Adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat. J Neurosci, 3, 6277-88.

168. Zhang XY, Zhou DF, Cao LY, Zhang PY, Wu GY, Shen YC. Changes in serum interleukin-2, -6, and -8 levels before and during treatment with risperidone and haloperidol: relationship to outcome in schizophrenia. J Clin Psychiatry. 2004; 65: 940-947.

169. ZHENG, L. T., HWANG, J., OCK, J., LEE, M. G., LEE, W. H. & SUK, K. 2008. The antipsychotic spiperone attenuates inflammatory response in cultured microglia via the reduction of proinflammatory cytokine expression and nitric oxide production. J Neurochem, 107, 1225-35.

ABSTRACT

The etiology of schizophrenia remains unclear while, in many aspects, the neuropathology of schizophrenia has recently been reported to be closely associated with microglia dysfunction. Microglia, which are the major players of innate immunity in the CNS, respond rapidly to even minor pathological changes in the brain and contribute directly to neuroinflammation by producing various pro-inflammatory cytokines and free radicals. Recent human studies have revealed microglial activation in schizophrenia using postmortem brains or in vivo neuroimaging techniques. We and other researchers have recently shown the inhibitory effects of some antipsychotics on the release of inflammatory cytokines and free radicals from activated microglia, both of which have recently been known to cause the synaptic pathology, a decrease in neurogenesis, and white matter abnormalities often found in the brains of patients with schizophrenia. In addition, recent evidence strongly suggests a neurodevelopmental role of microglia in regulating synapse formation/function by their interaction with synapses and phagocytotic activity. It is not known whether microglia dysfunction and microglia-orchestrated neuroinflammation are the primary cause of schizophrenia but they are closely related to the progression and outcomes of schizophrenia. Understanding microglial pathology may shed new light on the therapeutic strategies for schizophrenia.

CITATION

Monji A, Mizoguchi Y, Kato TA (2014) Microglial Abnormalities in the Pathophysiology of Schizophrenia. J Neurol Disord Stroke 2(3): 1059.

Monji A, Mizoguchi Y, Kato TA (2014) Microglial Abnormalities in the Pathophysiology of Schizophrenia. J Neurol Disord Stroke 2(3): 1059.

Received : 21 Jan 2014
Accepted : 28 Feb 2014
Published : 07 Mar 2014
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X