Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jan 1.
Published in final edited form as: Harv Rev Psychiatry. 2022 Jan-Feb;30(1):40–53. doi: 10.1097/HRP.0000000000000326

Brick by brick: building a transdiagnostic understanding of inflammation in psychiatry

David S Thylur 1, David R Goldsmith 1
PMCID: PMC8849593  NIHMSID: NIHMS1759932  PMID: 34995034

Abstract

Inflammatory phenomena are found in many psychiatric disorders, notably depression, schizophrenia, and posttraumatic stress disorder. Inflammation has been linked to severity and treatment resistance and may both contribute to as well as result from the pathophysiology of some psychiatric illnesses. Emerging research now suggests that inflammation may contribute to symptom domains of reward, motor processing, and threat reactivity across different psychiatric diagnoses. Reward processing deficits contribute to motivational impairments in depression and schizophrenia, and motor processing deficits contribute to psychomotor slowing in depression as well as in schizophrenia. A number of experimental models and clinical trials suggest that inflammation produces deficits in reward and motor processing through common pathways connecting the cortex and the striatum, which includes the nucleus accumbens, the caudate nucleus, and the putamen.

The observed effects of inflammation on psychiatric disorders may cut across traditional conceptualizations of psychiatric diagnoses, and further study may lead to targeted immunomodulating treatments that address difficult-to-treat symptoms in a number of psychiatric disorders. In this review, we use a Research Domain Criteria framework to discuss proposed mechanisms for inflammation and its effects on the domains of reward processing, psychomotor slowing, and threat reactivity. Furthermore, we discuss data supporting contributing roles of metabolic dysregulation and sex differences on the behavioral outcomes of inflammation. Finally, we discuss ways that future studies can help disentangle this complex topic to yield fruitful results that will help advance the field of psychoneuroimmunology.

Keywords: inflammation, cytokine, C reactive protein, mental disorders/physiopathology, immunomodulation

Introduction

There is a robust body of literature that now allows us to articulate pathways between peripheral inflammation, disruption of neural circuits, and specific symptom domains of psychiatric disorders: reward processing, psychomotor slowing, and to a lesser extent, threat reactivity. Early support for this relationship stemmed from administration of inflammatory stimuli in animal models and was later reinforced when symptoms of depression emerged in individuals receiving cytokines as therapy for cancer or chronic viral infection.14 Continued work in this area has shown that a subset of individuals with depression exhibit persistent inflammation that has both direct and indirect effects on the functioning of brain circuits that mediate core aspects of depression and represent an important source of treatment resistance.59 With depression serving as the blueprint for this relationship, data has emerged showing similar relationships for psychomotor deficits in schizophrenia and reward processing deficits in schizophrenia and PTSD.5,10,11 Furthermore, inflammation has been associated with alterations to threat reactivity in both depression and posttraumatic stress disorder (PTSD).12,13 We are now confronted with mounting evidence that inflammation may produce strikingly similar outcomes psychiatric illness, including depression, schizophrenia, and PTSD, which suggests that the impact that inflammation has on psychiatric symptomology may be relatively conserved across different psychiatric diagnoses and may occur through similar pathways.5,1418 This realization could have profound implications for our understanding of the role between inflammation and human behavior and the ways that we conceptualize and treat psychiatric illnesses.

Meta-analyses measuring circulating cytokines in depression, schizophrenia, and PTSD demonstrate dysregulation in a substantial proportion of individuals when compared with healthy controls.1922 Inflammation has been linked to worse treatment outcomes in both depression as well as in first-episode psychosis.8,23,24 Recent evidence indicates that increased concentrations of C-Reactive Protein (CRP) and inflammatory markers, such as cytokines, are associated with acute illness in patients with schizophrenia, bipolar disorder, and major depressive disorder and improves with treatment. Importantly, a number of markers remain significantly elevated in patients with chronic symptoms of these disorders, which suggests the possibility that some inflammatory markers may represent state and trait markers of psychiatric illness.19,25 Trait markers, which suggest risk for, or presence of illness, can be helpful for understanding pathophysiology or developing preventive treatments. State markers may change dynamically with severity and phase of illness and can be useful for monitoring treatment response. Studying inflammatory dysregulation in psychiatric disorders therefore presents an opportunity to “carve nature at its joints” to understand patterns that manifest in ways are not adequately reflected in current psychiatric nosology.

At the present moment, there is limited evidence implicating specific inflammatory markers with specific psychiatric disorders. Given that there is evidence for increased peripheral inflammation in patients with psychiatric disorders, it is plausible that inflammation and different inflammatory markers may play a specific role in individual psychiatric disorders. Alternatively, it is also plausible to consider that inflammation may impact the brain and behavior in a transdiagnostic fashion. In this narrative review, we consider this latter approach and use the Research Domain Criteria as a unifying framework to suggest that chronic low-grade inflammation may produce similar deficits in reward processing, motor processing, and threat reactivity independent of psychiatric diagnosis (Table 1). We consider these deficits at not only the level of symptoms, but also at the level of the brain through the impact of inflammatory cytokines on brain regions and circuits that have been shown to drive these associated behaviors. Finally, we also discuss the interdependent relationship between metabolic dysregulation and inflammation in psychiatric disorders as well as research hinting at sex differences in the behavioral outcomes of inflammation, and then conclude with suggestions for future work.

Table 1.

Inflammation produces similar behavioral outcomes in different psychiatric illnesses through common pathways

RDoC Domain* RDoC Construct* Behavioral outcome of inflammation Neurocircuitry affected by inflammation
DEP SCZ PTSD
Positive Valence Reward Amotivation Amotivation Anhedonia Mesolimbic
Corticostriatal
Sensorimotor Motor Psychomotor slowing Psychomotor slowing No data Corticostriatal
Negative Valence Threat Threat sensitivity No data Threat sensitivity Limbic§
*

RDoC: The Research Domain Criteria is a framework proposed by the National Institute of Mental Health to help reorganize psychiatric symptomatology into biologically-aligned categories. The Research Domain Criteria are divided into six domains of human functioning, which are further categorized into different components called constructs and subconstructs. As an example, the RDoC domain for positive valence includes a construct for reward responsiveness, which is further divided into subconstructs for reward anticipation, initial response to reward, and reward satiation.

DEP: depression, SCZ: schizophrenia, PTSD: posttraumatic stress disorder

Mesolimbic: This pathway plays a central role in reward processing. It connects the ventral tegmental area with the ventral striatum, which contains the nucleus accumbens.

Corticostratial: This pathway connects the ventromedial prefrontal cortex and striatum. The ventral striatum is implicated in reward processing, while the dorsal striatum, which contains the putamen and caudate, is implicated in motor processing.

§

Limbic: This pathway connects the amygdala, dorsal anterior cingulate cortex, and insula, all of which are involved in threat processing.

Using the Research Domain Criteria framework to understand the role of inflammation in psychiatric disorders

The diagnostic classification system provided by the Diagnostic and Statistical Manual of Mental Disorders, 5th Edition (DSM-5) provides a helpful construct for the clinical approach to mental disorders, but the heterogeneity present in many of these diagnoses creates artificial barriers that impede research into understanding why certain behavioral phenotypes arise and how to best treat them.2628 For example, if a DSM-defined psychiatric illness were to occur due to multiple pathophysiological pathways, a medication that only targets one of these pathways could appear to fail in a clinical trial that enrolls individuals with that diagnosis irrespective of the underlying biology. Furthermore, the diagnostic overlap occurring with some DSM-5 diagnoses may in fact represent distinct entities that occur due to common neurobiological pathways, which could help explain why certain treatments such as antipsychotic medications have indications for schizophrenia, bipolar disorder, and as augmentation for major depression.29,30 In order to address these types of dilemmas, the National Institute of Mental Health created an initiative to establish a more biologically-oriented nosology of mental illness through the Research Domain Criteria (RDoC) project, which aims to characterize domains of human functioning using different units of analysis, such as genes, brain circuits, or neurotransmitters. In alignment with this initiative, we aim to present the hypothesis that inflammation produces several specific patterns of disruptions to neurocircuitry that produce similar behavioral outcomes in a cross-cutting manner. In order to minimize bias in this narrative selective review, a structured search of PubMed was performed to ensure that relevant studies were included using the following search strategy: (INFLAMMATION or CYTOKINE) and (NEUROIMAGING or MRI or NEUROCIRCUITRY or BRAIN CIRCUIT or NEURAL CIRCUIT) and (DEPRESSION or SCHIZOPHRENIA or POSTTRAUMATIC STRESS DISORDER). Results from this search strategy were used to identify articles with data that emphasized multiple units of analysis from the Research Domain Criteria framework (i.e. genes, molecules, cells, circuits, physiology, behavior) for inclusion. Studies that did not assess the effect of inflammation using at least two units of analysis for a specific topic were excluded.

Inflammation: from cytokines to circuits to behavior

Peripheral inflammation can exert effects on the central nervous system (CNS) through a number of mechanisms that challenge conventional notions that the CNS is immunologically privileged: autonomic nervous tone, cytokine transporters located within the blood-brain barrier, volume diffusion of cytokines produced in circumventricular organs and the choroid plexus, and activity of immune cells and cytokine receptors in areas adjacent to the brain.31 Postmortem and neuroimaging studies suggest that blood-brain barrier integrity may frequently be compromised in schizophrenia; postmortem data indicates that this may be the case in affective disorders as well.32 Furthermore, receptors for cytokines are expressed throughout the CNS.33 Cytokines act through these routes to facilitate neuroplastic changes in brain circuits, notably by modulating the synthesis, reuptake, and release of neurotransmitters, including serotonin, dopamine, norepinephrine, and glutamate.34

Studies administering cytokines and other inflammatory stimuli to human subjects have provided robust evidence of the relationship between cytokines, neurocircuitry, and specific behavioral domains of depression.1,31 Perhaps the most well-studied of these models is the antiviral cytokine interferon-α (IFN- α), which provokes a syndrome characterized by symptoms of depressed mood, fatigue, anxiety, amotivation, and motor slowing.1 Other commonly used models include the T-cell-stimulating cytokine interleukin-2 (IL-2) as well as typhoid vaccination and endotoxin administration, which both elicit acute inflammation.31,35,36 These agents have been used in a number of experimental approaches to characterize neurocircuitry changes that occur as a result of inflammation. Reward deficits induced by inflammation have been linked to disruption in the dopamine-mediated mesolimbic pathway, which is composed of the ventral tegmental area and its projections to the ventral striatum, which contains the nucleus accumbens, as well as in corticostriatal pathways between the ventral striatum and ventromedial prefrontal cortex.5,6,34 Motor processing deficits occurring due to inflammation have been principally associated with disruption to dopamine-mediated corticostriatal signaling between the dorsal striatum (which consists of the putamen and caudate), and the ventromedial prefrontal cortex, which is involved in emotional regulation and decision-making.5,34,37 In contrast, alterations to threat reactivity in the setting of inflammation are associated with functional changes to structures involved in fear processing: the amygdala, anterior cingulate gyrus, and insula.3841 As will be discussed in further detail below, disruption to the mesolimbic pathway has been associated with reward processing deficits occurring due to inflammation in depression, schizophrenia, and PTSD, while disruption to corticostriatal signaling has been associated with reward processing and motor slowing due to inflammation in depression and schizophrenia.5,11,12,34 Alterations to brain structures related to fear processing have been associated with changes in threat reactivity occurring in the setting of inflammation in both PTSD and depression.12,42

It is important to note that sustained inflammation also produces considerable metabolic dysregulation. In addition to its neuropsychiatric consequences, inflammation has been strongly linked to other diseases such as obesity, diabetes, and cardiovascular disease.4345 Obesity contributes to systemic inflammation through accumulation and activation of macrophages in adipose tissue, which then release cytokines into the circulation.46,47 The pro-inflammatory cytokines released through this process exert substantial effects on insulin resistance and metabolic regulation.4850 It is increasingly apparent that the immune system, metabolism, and mental health can affect each other in a complex, closely interlinked manner.51,52 Obesity is associated with an increased risk of depression and vice versa, and depressed individuals with metabolic dysregulation may represent a treatment-resistant group.51,53 Surgical intervention for obesity is associated with reductions in both inflammation and depression.54,55 In addition, inflammation has been hypothesized to play a role in the metabolic dysfunction that occurs widely in schizophrenia independent of the effects of antipsychotic medication.5658 Kappelman et al. analyzed genome-wide association study data and found a relationship between CRP, body mass index, and neurovegetative symptoms of depression, supporting the role of triadic relationship between inflammation, metabolic dysregulation, and depression.59 In a similar approach, Perry et al. found that inflammation may drive both insulin resistance and schizophrenia risk.60 The interplay of inflammation and metabolic disruption has important implications for understanding the effects of inflammation on psychiatric illnesses and will be discussed briefly throughout this paper; this topic has been reviewed more extensively elsewhere.52,61

An evolutionary link between inflammation and mental health: sickness behavior

Continued research into the connection between cytokines and behavior suggests that there may be an adaptive component to the influence of inflammation on mental health. Sickness behavior, or behavior that occurs during periods of acute infection, is postulated to have an evolutionary origin: the fatigue, amotivation, social avoidance, and anhedonia experienced during illness may encourage individuals to conserve energy needed for healing and isolation, thereby preventing the spread of pathogens.6 Tumor necrosis factor-α (TNF) and interleukin-1β, which both act as initiators of inflammation, appear to play major roles in sickness behavior and are commonly produced by inflammatory stimuli such as endotoxins.6,31,62 These behavioral symptoms that are subsequent to acute inflammation are thought to be evolutionarily conserved and drive the hypothesis that the behavioral manifestations of chronic, low grade inflammation in psychiatric illness stems from brain regions and circuits that have been shown to be sensitive to the effects of inflammation. In order words, the chronic inflammation associated with a subset of cases of depression, schizophrenia, and PTSD may represent a maladaptive consequence of sickness behavior. Sustained low-grade inflammation in a portion of individuals with these disorders likely contributes to motivational deficits, psychomotor slowing, and altered threat reactivity in a diagnosis-independent manner.

The far-reaching role of inflammation in psychiatric disorders

Acute inflammation serves important roles in responding to infection and injury, but when it fails to resolve, the ensuing imbalance in immune, metabolic, and neuroendocrine systems contributes to a wide array of human diseases, including mental illness.63,64 In the setting of chronic inflammation, cytokine production becomes dysregulated, leading to diverse consequences in a myriad of tissue types, including the central nervous system, adipose tissue, and the cardiovascular system.64 Cytokines are immune signaling peptides produced by both immune and non-immune cells that can have both pro-inflammatory and anti-inflammatory effects. Cytokines can influence the production, release, and reuptake of neurotransmitters, leading to functional changes to brain circuitry that produce specific patterns of psychiatric symptoms.34

The systemic marker of inflammation, C-reactive protein (CRP), may be useful to quantify the degree of inflammatory burden in psychiatric illness. CRP is an acute phase reactant synthesized by the liver in response to cytokines such as interleukin-6 (IL-6). Though not thought to be an inflammatory cytokine, CRP can be considered a marker of systemic inflammation. Though there are other biomarkers that are useful to characterize systemic inflammation, CRP remains widely used in clinical settings because of the ability to be easily assayed in Clinical Laboratory Improvement Amendments (CLIA)-certified laboratories. Moreover, CRP can be used to stratify individuals into groups with low inflammation (<1 mg/L), medium inflammation (1 to 3 mg/L), or high inflammation (>3mg/L), which are thresholds based on guidelines for evaluating cardiovascular risk due to inflammation that were developed by the Centers for Disease Control and Prevention and the American Heart Association.6567 A meta-analysis assessing inflammatory burden in depression showed that 60% of patients with depression exhibit low-grade inflammation as indicated by a C-reactive protein (CRP) concentration above 1 mg/L, while 30% exhibit high inflammation as measured by a CRP greater than 3 mg/L.68 Markers of inflammation in healthy individuals are associated with an increased likelihood of later developing major depression, and for individuals with depression, inflammatory markers correlate with treatment resistance.9,69 In bipolar disorder, CRP concentrations are elevated in all phases of the illness but appear to be worst during mania.70 A number of inflammatory phenomena have been described in schizophrenia, including elevated systemic inflammation (including CRP as well as a number of inflammatory cytokines – most commonly interleukin 6 and tumor necrosis factor), dysregulation of microglial cells, and excess complement activity.19,7174 In addition, persistent low-grade inflammation has been consistently described in PTSD (e.g. CRP, IL-6, TNF, IL-1beta, and interferon-gamma).20 Of note, inflammation has been linked to other psychiatric disorders, including anxiety and eating disorders, but these will not be included in this review due to limited availability of research analyzing the role of inflammation across multiple levels of analysis.12,75,76

Although individual cytokines have been shown to be increased in individual psychiatric illnesses relative to healthy controls, it is premature to suggest that individual markers may be implicated in specific disorders, or in psychiatric illness in general. The findings in the literature, for example, may be skewed by individual markers that are more commonly assayed. It should be noted that various groups have used data-driven approaches to parse the heterogeneity of the various inflammatory markers that have been implicated in the literature. These data-driven approaches that include using principle component analysis and other clustering approaches, may help identify groups of cytokines that may be implicated in psychiatric disorders in addition to behavior and brain function.7779

These findings spur a host of intriguing questions: where does inflammation come from in psychiatric disorders? Does inflammation lead to psychiatric illness, or is the inverse true? Is there is a bidirectional relationship between illness and inflammation, or is the presence of inflammation purely an epiphenomenon? A number of common factors predispose to inflammation and likely play a role in perpetuating immune dysregulation in these disorders, including childhood mistreatment, stress, poor sleep, dietary factors and obesity, the gut-brain axis, and physical inactivity.8082 Early childhood trauma and socioeconomic adversity appear to modulate inflammation that influences the risk of developing subsequent PTSD.81 Less well understood are the underlying cellular and molecular processes that mediate the inflammation that is present in a subset of individuals with psychiatric illness. In depression, substantial attention has been directed at the relationship between stress, the hypothalamic-pituitary axis, and the sympathetic nervous system.83 Stress and adverse childhood experiences can modulate the hypothalamic-pituitary-adrenal (HPA) axis; glucocorticoids produced in response to stress can promote inflammation and alter immune functioning.34,83 Glucocorticoids reduce levels of pro-inflammatory cytokines and increase levels of anti-inflammatory cytokines, which can then act on neurocircuitry to produce behavioral changes.34,83 More recently, production of protein complexes in cells from myeloid lineages have been shown to drive inflammation in depression (so-called inflammasomes).84 In schizophrenia, maternal immune activation and prenatal infection may spur immune dysregulation that mediates the risk of developing schizophrenia.85 Large-scale genome-wide association study findings showing enrichment of enhancer elements for immune cell lineages and epidemiological data showing increasing comorbidity of autoimmune disorders and increased proportions of circulating autoantibodies also point to a possible contribution from B cells in the pathogenesis of schizophrenia.8688 Pathological microglial activation has been hypothesized to play an important role in both bipolar disorder and schizophrenia.72,89 Microglial cells play an important role in maintaining immune homeostasis in the central nervous system, and overactivation could lead to excessive synaptic pruning that may explain the reduced synaptic density noted in postmortem analysis of brain samples from deceased individuals with schizophrenia.9092 In summary, inflammation seen in these psychiatric disorders occur due to different sources of immune dysregulation that vary widely not only between different diagnoses but also within the same DSM-5 diagnosis.

Accumulating evidence suggests that inflammation likely plays a bidirectional role in the development of some psychiatric disorders. Despite substantial heterogeneity in the underlying pathogenesis of depression, bipolar disorder, and schizophrenia, longitudinal data and results from clinical trials with immunomodulating agents support the hypothesis that immune dysregulation likely plays a bidirectional role in at least a subset of cases.6,7,18,73 Early research hints at bidirectionality in PTSD as well. Though further research is needed to dissect this relationship, current research suggests that inflammation both increases risk of developing PTSD and ensues from it.81,93,94 Inflammation in the immediate period after a trauma may also influence the likelihood of developing chronic PTSD.9597

It is unclear whether specific types of inflammation confer risk for certain psychiatric disorders, or whether inflammation generally increases the risk of developing multiple psychiatric disorders. Using a prospective birth cohort of about 4500 individuals, researchers from the Avon Longitudinal Study of Parents and Children found that levels of IL-6 and CRP at age 9 were associated with risk of developing psychosis (unadjusted odds ratios of 1.34 and 1.21 for IL-6 and CRP, respectively) and depression (unadjusted odds ratios of 1.27 and 1.13 for IL-6 and CRP, respectively) in early adulthood.98 Interestingly, the timing of stress during development may differentially effect circuitry that can predispose to either depression or psychosis. Using inescapable footshocks and restraints in a rat model, Gomes et al. found that stress in adolescent rats resulted in circuit deficits and behaviors that resembled those of humans with psychosis, while stress in adult rats instead led to circuit changes and behaviors that mimicked those of depression.99 These are intriguing findings, though future research will need to disentangle the effects of environmental factors and genetic risk on the development of psychiatric illness as it relates to inflammation. This dilemma is readily apparent with schizophrenia in the emerging hypothesis that dysregulated complement activation occurring due to genetic and environmental factors may influence microglial activity that leads to excessive synaptic pruning during critical periods of neurodevelopment.71,91,100102 Peripheral complement proteins, which are important proteins in the innate immune system, have recently been studied as potential biomarkers in patients with psychosis and depression.103105 Moreover, there is a growing literature on the use of positron emission tomography (PET) imaging with ligands to the translocator protein (TSPO), thought to reflect activated microglia in the brain, in individuals with psychiatric illness.106108 It should be noted that there are a number of limitations to this literature that are beyond the scope of this paper, that have been reviewed elsewhere.109111

Positive valence systems in the RDoC framework: the effects of inflammation on reward processing in depression, schizophrenia, and PTSD

The RDoC domain for positive valence systems includes biological pathways that motivate and reinforce the production of actions and behaviors. Reward processing has multiple constructs within the RDoC framework for positive valence systems, including reward responsiveness, reward learning, and reward anticipation. Reward processing determines how motivated an individual will be to perform a given behavior based on how eagerly they desire the reward from that behavior. Reward processing deficits occur commonly in schizophrenia, depression, and PTSD, and often create major life limitations: poor motivation can lead to worsened performance at work that culminates in termination, while reduced reward-seeking could lead to social isolation.112114

In depression, inflammation has been linked to deficits in reward processing via the mesolimbic system115 as well as corticostriatal circuitry that has also been implicated in inflammation-mediated deficits to reward processing.115 Inflammation may play a role in the altered calculation of effort and reward observed in depression, creating a preference for low effort/low reward strategies over those that are more rewarding but more effortful.116118 Early research using neuroimaging in PTSD has also linked inflammation with abnormalities in the mesolimbic system.11 Anhedonia and motivational deficits commonly seen in schizophrenia have been linked to dysregulated inflammation in a number of studies, but data integrating these finding across multiple levels of analysis are not yet available.10

Inflammation affects reward processing through mesolimbic and corticostrial circuits

Immune challenge with inflammatory stimuli has been shown to alter reward processing via the mesolimbic and corticostriatal pathways, which suggests that inflammation may act broadly in psychiatric illness to produce deficits in reward.6 Administration of endotoxin in humans correlated with deficits to reward anticipation and reward responsiveness in several clinical studies by the Eisenberger group.119,120 One study assessing the effects of endotoxin challenge or placebo in 39 participants without a history of mental illness showed a relationship between inflammatory markers, depressive symptoms, and anhedonia.35 Endotoxin provoked depressive symptoms that were associated with increased serum levels of the pro-inflammatory cytokines IL-6 and TNF. Though reward learning was unaffected as measured by performance on the monetary incentive delay task, the authors observed a reduction in ventral striatum activity in anticipation of reward in the endotoxin group. Mediation analysis indicated that left ventral striatal activity mediated the relationship between endotoxin administration and mood.

The ventral striatum and its projections to the cortex serve core functions in modulating effort and reward.121 To study the effects of IFN-α on reward pathways, Capuron et al. compared individuals receiving IFN-α for treatment of chronic hepatitis C with those who were waitlisted for the drug.122 They found that IFN-α caused reduced activation of the bilateral ventral striatum in response to a gambling task and linked this reduced activity to anhedonia. Felger et al. used IFN-α in a nonhuman primate model to ascertain changes in striatal dopamine signaling.123 IFN-α administration led to reduced dopamine release in the striatum, and the timing of this change correlated with anhedonia-like behavior.

Inflammation alters corticostriatal and mesolimbic circuitry in depression and PTSD

Research studying the connection between inflammation and reward processing in psychiatric disorders points to a possible mediating role of corticostriatal and mesolimbic neurocircuitry. In a study of individuals with major depression and bipolar depression, Felger et al. demonstrated that levels of CRP in individuals mediated anhedonia via connectivity between the left inferior ventral striatum and the ventromedial prefrontal cortex, which aids in decision-making and emotional regulation.124 In a study using a cohort of 56 trauma-exposed women, Mehta et al. found that higher peripheral plasma CRP negatively correlated with bilateral ventral striatum-ventromedial prefrontal cortex functional connectivity, and a separate analysis using an inflammatory composite score indicated that mesolimbic connectivity was specifically altered in women with worse PTSD symptoms and prior major trauma.11 Left ventral striatum-ventromedial prefrontal cortex functional connectivity also correlated with higher anhedonia scores in subscales from the Beck Depression Inventory and the PTSD Symptom Scale when comparing groups after stratifying for high inflammation/low inflammation.

A number of studies have also linked immunometabolic dysregulation to specific neurocircuitry changes in depression, primarily in dopaminergic corticostriatal circuits.52 In an fMRI study of 42 individuals with depression, metabolic markers and CRP interacted together to influence functional connectivity in reward and motor circuits.125 Exploratory transcriptional analysis showed that enrichment for inflammatory and metabolic pathways correlated with these functional changes. Since inflammation, mental health, and metabolism are highly interrelated, treatment of individuals with psychiatric illness that have a high inflammatory burden may also need to account for the role of metabolism in the pursuit of more effective therapies. Research on the use of the tumor necrosis factor (TNF)-α antagonist infliximab in depression showed that gene transcription of biomarkers related to lipid and glucose metabolism helped predict its antidepressant response.126,127

Inflammation influences reward processing in schizophrenia

Deficits in reward processing and motivation are core negative symptoms of schizophrenia and represent a difficult-to-treat aspect of the illness.10 Unfortunately, much of the research in this area does not consider reward processing separately from negative symptoms as a whole, though recent evidence supports a role for separate domains of negative symptoms including deficits in motivated behaviors and deficits in expressivity.128131 A number of studies have linked inflammation to these symptoms and though no studies to date have directly linked inflammation to reward processing at the level of the brain or behavior, there is a growing literature on the association between inflammation and negative symptoms of schizophrenia, spurring intriguing hypothesis-driven questions as to whether inflammation may impact similar reward circuits in the brain in patients with schizophrenia. In the largest study on this topic, Liemburg at al. found that CRP correlated with the negative symptoms on the Positive and Negative Syndrome Scale in sample of 2,132 individuals with psychotic disorders.132 Individuals with deficit schizophrenia, marked by prominent and persistent negative symptoms, have been shown to have higher concentrations of TNF and IL-6 compared to both healthy controls and individuals with non-deficit schizophrenia; increased TNF and IL-6 were associated with worse negative symptom severity.133 Inflammation may also be predictive of the development of negative symptoms in individuals at risk for developing a psychotic disorder. Data from the North American Longitudinal Prodromal Study showed that increased concentrations of TNF predicted later development of negative symptoms, even after controlling for depressive symptoms.134 Neuroimaging studies in psychotic disorders have frequently linked abnormalities in the ventral striatal to reward processing deficits in schizophrenia, but research is not yet available examining the possible mediating role of inflammation for this circuit for schizophrenia.135137 Future work in this area could benefit from reorganizing negative symptoms into discrete RDoC constructs such as reward processing and deficits in expressivity in order to help disentangle how inflammation influences each individual component.

As it relates to the interplay of inflammation and metabolic dysfunction, increased inflammatory and cholesterol markers have been shown to predict worse clinical outcomes after one year of treatment in individuals with first episode psychosis, even after controlling for the effects of antipsychotic medication on glucose metabolism.23 Moreover, the interactive effect of inflammation and lipid biomarkers were shown to be associated with worse negative symptom severity in patients with schizophrenia.138 Further work should seek to investigate these interactions as there appears to be evidence for their impact within this RDoC positive valence system and could represent novel and important treatment targets.

Sensorimotor systems in the RDoC framework: the effects of inflammation on psychomotor speed and motor planning in depression and schizophrenia

The RDoC framework for sensorimotor systems encapsulates biological pathways that plan, produce, and refine movement. Constructs and subconstructs from sensorimotor systems that are relevant to inflammation include motor processing and planning, deficits to which are commonly observed in depression and schizophrenia.37,139,140 Conversely, this relationship has not been shown in PTSD, and for unclear reasons, individuals with PTSD that go on to develop depression may actually be less likely to exhibit psychomotor slowing.141 Psychomotor slowing associated with inflammation represents a marker of treatment-resistant depression, which hints at the inability of currently-available treatments to address the inflammation underlying this phenotype.142144 Similarly, psychomotor slowing in schizophrenia does not respond well to usual treatment with antipsychotic medication.140

Inflammation affects motor processing through corticostriatal circuitry

IFN-α helped provide early evidence of the connection between inflammation, depression, and psychomotor slowing. Administration of IFN-α frequently produces a depressive syndrome with prominent neurovegetative symptoms, including fatigue and motor slowing.1 In a clinical trial studying depressive symptoms induced by IFN-α in the treatment of chronic hepatitis C, participants experienced reduced motor speed and worsened reaction times, and these deficits were worst in individuals meeting criteria for a major depressive episode.2 Further experiments to understand the downstream effects of IFN-α have helped elucidate the impact this cytokine has on brain networks and neurotransmitter signaling. Psychomotor slowing induced by IFN-α appears to be mediated by activity in the corticostriatal network, including basal ganglia structures and the prefrontal cortex.145,146 In vivo data using nonhuman primates showed that chronic administration of IFN-α reduced dopamine release in the striatum.123 In another experimental model, stimulated release of cytokines via typhoid vaccination in humans provokes psychomotor slowing that is linked to activity in another basal ganglia structure, the substantia nigra.36

Inflammation affects motor processing via corticostriatal circuitry in depression

The impact that administration of IFN-α has on brain circuitry resembles the phenotype of psychomotor slowing that is seen in depressed individuals with high levels of inflammation. One study assessing inflammation in 93 participants with major depression and bipolar depression showed that elevated inflammatory markers correlated with worsened performance on a battery of psychomotor tasks. In multiple regression analyses, the cytokine IL-6 was identified as a significant predictor of psychomotor performance.77 Accumulating evidence indicates that naturalistically-occurring inflammation may influence motor activity through modulation of dopaminergic corticostriatal circuitry. In a neuroimaging study of 48 participants with major depression and bipolar depression, mediation analysis showed that connectivity between the right dorsal caudal putamen and the ventromedial prefrontal cortex mediated psychomotor slowing in participants with elevated CRP (as measured with Finger Tapping Test in the dominant hand).124 New data further supports a relationship between immunometabolic pathways and psychomotor slowing in depression.147 Using a sample of unmedicated patients with major depression, motor speed and motor processing deficits were associated with transcriptional enrichment of immunometabolic genes in monocytes, plasmacytoid dendritic cells, and natural killer cells.

Inflammation influences motor processing in schizophrenia

Corticostriatal circuit dysfunction has been consistently linked to psychomotor slowing in schizophrenia and may represent an important phenotype of the disorder that occurs independently of antipsychotic medication side effects.148,149 Early evidence now suggests that psychomotor processing in schizophrenia may also be influenced by inflammation. A 2020 study assessed the relationship between peripheral immune markers and psychomotor slowing in 43 participants with schizophrenia and 29 healthy controls. After correction for multiple testing, a number of inflammatory cytokines were associated with psychomotor performance among participants with schizophrenia but not healthy controls, most consistently tumor necrosis factor, IL-6 soluble receptor, and the anti-inflammatory cytokine IL-10.150 It is unclear at this point whether inflammation serves as a mediating factor in the relationship between corticostriatal circuit abnormalities and psychomotor slowing in schizophrenia, and this area merits further study.

Negative valence systems in the RDoC framework: the effect of inflammation on threat reactivity

In the RDoC framework, reactivity to potential threat (anxiety), acute threat (fear), and sustained threat are negative valence systems. Symptoms of PTSD that are mediated by negative valence systems include threat vigilance and avoidance. Inflammation appears to correlate with threat sensitivity, which contributes to impairment in PTSD.12 In a study of 735 individuals with current PTSD, higher levels of CRP correlated with higher threat reactivity.151 Another large study of 2,698 individuals showed that CRP correlated with threat reactivity as measured by a fear-potentiated startle paradigm.152 Inflammatory stimuli can modulate neurocircuitry involved in emotional processing and threat response, particularly the amygdala, dorsal anterior cingulate cortex, and insula.3841 Similarly, the amygdala, dorsal anterior cingulate cortex, and related structures exhibit functional alterations in PTSD in response to threat.12,153 There are few studies that specifically evaluate the mediating role of inflammation with paradigms that measure threat reactivity in psychiatric illness. Preliminary evidence has linked inflammation to disruption in threat-related circuitry in depression. In an fMRI study of 48 participants with major depression or bipolar depression, higher peripheral plasma CRP correlated with reduced (right) amygdala-ventromedial prefrontal cortex connectivity.42

Sex differences in the effects of inflammation on behavior

Despite the important role that sex plays in biological functioning, sex has been historically overlooked as a biological variable in everything from preclinical cell culture data to human clinical trials.154 Researchers studying the effects of inflammation on mental health are placing increased emphasis on integrating sex as a biological variable into study design.155,156 Sex differences in the prevalence of mental illnesses discussed in this paper have been well documented: women appear to be nearly twice as likely to develop depression as men, are twice as likely to develop PTSD after a traumatic event, and though are somewhat less likely to develop schizophrenia, they are often diagnosed at a later age.157159 Sex differences in immune functioning may help explain these epidemiological puzzles.160162 A large body of research indicates that immune functioning differs between men and women (reviewed extensively by Klein and Flanagan163), and emerging data indicate that these differences can influence behavioral outcomes. Women may appear more likely to experience increases in inflammatory cytokines after a psychosocial stressor, and this inflammatory state seems to persist longer for women.164166 Women may be more susceptible to inflammation-mediated depressive symptoms after receiving an inflammatory stimulus.167,168 Inflammatory markers may better correlate with reward processing deficits and psychomotor slowing in depressed women,67,169,170 though this effect is not always replicated.35,171 There also appear to be sex differences in complement gene expression that may help explain the reduced vulnerability to schizophrenia in women.172

Sex hormones help modulate the immune response independent of the effects of gender, and their interactions with the immune and neuroendocrine systems during critical periods of neurodevelopment likely contribute to differences in inflammation and depression susceptibility between males and females.161,163 Sex differences appear particularly prominent in microglia: research in rodent models has demonstrated that estradiol exposure modulates microglial functioning in select regions of the rodent brain, leading to changes in dendritic spine density and ultimately to behavior.160,173 Sex differences in the relationship between inflammation and human behavior are substantive enough that many researchers are now urging the field to explicitly evaluate sex as a biological variable in future research.157,158,160,168,174

Data explicitly examining the role of sex in the relationship between inflammation, neurocircuitry, and the three RDoC components examined in this paper (reward processing, motor processing, and threat reactivity) are currently sparse, but a few preliminary observations can be posited. First, there may be sex differences in the relationship between reward processing and inflammation that are mediated by corticostriatal circuitry. In a clinical trial conducted by the Eisenberger group, an endotoxin stimulus or placebo was given to a group of 115 participants to ascertain sex-based differences in reward sensitivity. Female participants showed reduced ventral striatal activity in response to reward cues after administration of endotoxin, while male participants did not.155 Secondly, sex differences may contribute to the impact of inflammation on threat reactivity. In a study of a sample of 172 undergraduate students, higher levels of CRP correlated with increased amygdala activity in response to threat paradigms in men but not women.175 The possible underlying processes in these sex differences and their potential impact in the setting of mental illness are unclear and merit further study.

Discussion

Inflammation appears to produce similar effects in multiple different psychiatric disorders, particularly in positive valence systems and sensorimotor systems as described by the RDoC framework. Moreover, individuals with high inflammation may exhibit similar phenotypes that are characterized by disruptions to common neurocircuitry pathways, particularly in mesolimbic and corticostriatal circuits. Further research in this area could inform the development of targeted immunomodulating therapy that may improve treatment for subsets of patients with depression, schizophrenia, and PTSD, particularly those for whom traditional therapies have not worked well due to the mediating role of inflammation.

Approaches to mitigating the impact of inflammation

There is a possibility that a marker such as peripheral plasma CRP could someday be used to help identify and treat subsets of psychiatrically ill individuals whose symptoms occur in part due to a high inflammatory burden. In two different studies of depression that stratified using a CRP threshold of 1, individuals below the threshold had a better response to selective serotonin reuptake inhibitor (SSRI) medications, while those above the threshold had a better response with an alternative therapy: bupropion-SSRI combination in one study, and nortriptyline in the other.176,177 The ability of these drugs to modulate norepinephrine and its downstream effects on the immune system has been proposed as a reason for their greater effectiveness in inflammation-mediated depression.177,178 An alternate possibility is through norepinephrine-mediated modulation of dopamine signaling,179,180 which could also explain the greater effectiveness of these agents given the impact of inflammation on dopaminergic neurocircuitry. In a proof-of-concept study, Raison et al. performed a clinical trial to assess the differential effects of the TNF-α antagonist infliximab on patients with treatment-resistant depression across varying levels of CRP.142 Though TNF antagonism did not result in generalized improvement in depression, participants with a CRP greater than 5 experienced a clinically-meaningful improvement in the Hamilton Scale for Depression, with scores specifically improving for symptoms linked to inflammation: anhedonia, psychomotor retardation, and psychic anxiety.

Suggestions for future work in this area

Clinical trials that stratify patients based on the degree of inflammation may be able to lead to the development of novel treatments that address inflammation-mediated symptom burden. A common issue in this area is stratifying results in a post hoc manner, which limits the conclusions that can be drawn.61,181 It is especially important to enrich for inflammation, as positive results in a high-inflammation subgroup could be lost in a heterogeneous sample. Longitudinal research assessing how inflammation and psychiatric symptomatology evolve over time could be especially useful, particularly in the case of bipolar disorder: inflammation-mediated psychomotor slowing and motivational deficits have been noted in bipolar depression, but it remains unclear how manic episodes fit into this narrative, as they are also associated with inflammation but present instead with psychomotor agitation.70 Additionally, it is important to recognize that though there is a strong link between inflammation and some symptoms of psychiatric disorders, this is not the only way that these symptoms occur. Finally, sex is a critical biological variable that should be more consistently and explicitly assessed in future work in this area.

In summary, inflammation appears to exert similar behavioral phenotypes that are mediated by mesolimbic and corticostriatial circuitry across different psychiatric disorders, most notably depression, schizophrenia, and PTSD. Future research should emphasize a dimensional approach to identify common inflammatory pathways between different psychiatric disorders. It remains to be seen whether inflammation-mediated phenotypes across different psychiatric diagnoses respond similarly to immunomodulating treatment. As an example, a study by Nettis et al. using adjunctive minocycline in depression showed a benefit for individuals with an elevated CRP.182 However, a separate study by Deakin et al. did not find any benefit from minocycline for negative symptoms of schizophrenia, even when stratifying for inflammation.183 One possibility for these discrepant findings could be due to the off-target effects of many anti-inflammatory drugs, including minocycline. Similarly, it is important that anti-inflammatory trials demonstrate that they actually engage the system that is being targeted; otherwise the interpretation of negative findings are especially challenging.61,184 For example, demonstrating evidence that an anti-inflammatory medication decreases inflammation is imperative, since only those individuals with increased inflammation would be expected to respond to an anti-inflammatory medication. Moreover, given the evidence that inflammation targets specific neurocircuitry to lead to specific symptoms, anti-inflammatory trials should focus on hypothesized changes at the level of brain and behavior; for example, improvements in connectivity in brain reward circuits should correlate with improvement in symptoms of anhedonia.

Given the broad role that inflammation can play in psychiatric illness, a more granular understanding of the specific pathways involved could enable a new understanding of how certain phenotypes of mental illness arise and lead to targeted immunomodulating treatments that address their underlying inflammation. Careful attention should be paid to the role of immunometabolism and sex differences that contribute to the complex relationship between inflammation and mental health, and accounting for these factors could lead to novel treatments that can address the overlap between inflammation, neuropsychiatric impairment, and metabolic dysregulation in a multipronged fashion. Integrating a dimensional study of inflammation into traditional conceptualizations of psychiatric illness will help explain the considerable overlap in some features of psychiatric disorders while respecting the different components of psychopathology and neurobiology that underlie each.

Acknowledgements:

This study was supported by grants K23MH114037 (Goldsmith) and R25MH101079 (Thylur) from the National Institute of Mental Health, and the Walter Wellborn Endowed Fellowship (Thylur).

Footnotes

Conflicts of Interest Statement: The authors have no conflicts of interest to disclose.

References

  • 1.Capuron L, Miller AH. Cytokines and psychopathology: Lessons from interferon-α. Biol Psychiatry. 2004;56(11):819–824. doi: 10.1016/j.biopsych.2004.02.009 [DOI] [PubMed] [Google Scholar]
  • 2.Majer M, Welberg LAM, Capuron L, Pagnoni G, Raison CL, Miller AH. IFN-alpha-induced motor slowing is associated with increased depression and fatigue in patients with chronic hepatitis C. Brain Behav Immun. 2008;22(6):870–880. doi: 10.1016/j.bbi.2007.12.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Capuron L, Gumnick JF, Musselman DL, et al. Neurobehavioral effects of interferon-alpha in cancer patients: phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology. 2002;26(5):643–652. doi: 10.1016/S0893-133X(01)00407-9 [DOI] [PubMed] [Google Scholar]
  • 4.Shuto H, Kataoka Y, Horikawa T, Fujihara N, Oishi R. Repeated interferon-alpha administration inhibits dopaminergic neural activity in the mouse brain. Brain Res. 1997;747(2):348–351. doi: 10.1016/s0006-8993(96)01371-6 [DOI] [PubMed] [Google Scholar]
  • 5.Felger JC, Treadway MT. Inflammation Effects on Motivation and Motor Activity: Role of Dopamine. Neuropsychopharmacology. 2017;42(1):216–241. doi: 10.1038/npp.2016.143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16(1):22–34. doi: 10.1038/nri.2015.5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Miller AH, Haroon E, Felger JC. Therapeutic Implications of Brain-Immune Interactions: Treatment in Translation. Neuropsychopharmacology. 2017;42(1):334–359. doi: 10.1038/npp.2016.167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Haroon E, Daguanno AW, Woolwine BJ, et al. Antidepressant treatment resistance is associated with increased inflammatory markers in patients with major depressive disorder. Psychoneuroendocrinology. 2018;95:43–49. doi: 10.1016/j.psyneuen.2018.05.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Strawbridge R, Arnone D, Danese A, Papadopoulos A, Herane Vives A, Cleare AJ. Inflammation and clinical response to treatment in depression: A meta-analysis. Eur Neuropsychopharmacol. 2015;25(10):1532–1543. doi: 10.1016/j.euroneuro.2015.06.007 [DOI] [PubMed] [Google Scholar]
  • 10.Goldsmith DR, Rapaport MH. Inflammation and Negative Symptoms of Schizophrenia: Implications for Reward Processing and Motivational Deficits. Front psychiatry. 2020;11:46. doi: 10.3389/fpsyt.2020.00046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Mehta ND, Stevens JS, Li Z, et al. Inflammation, reward circuitry and symptoms of anhedonia and PTSD in trauma-exposed women. Soc Cogn Affect Neurosci. 2020;15(10):1046–1055. doi: 10.1093/scan/nsz100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Michopoulos V, Powers A, Gillespie CF, Ressler KJ, Jovanovic T. Inflammation in Fear-and Anxiety-Based Disorders: PTSD, GAD, and beyond. Neuropsychopharmacology. 2017;42(1):254–270. doi: 10.1038/npp.2016.146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Felger JC. Role of Inflammation in Depression and Treatment Implications. In: Macaluso M, Preskorn SH, eds. Antidepressants: From Biogenic Amines to New Mechanisms of Action. Cham: Springer International Publishing; 2019:255–286. doi: 10.1007/164_2018_166 [DOI] [PubMed] [Google Scholar]
  • 14.Miller AH. Beyond depression: the expanding role of inflammation in psychiatric disorders. World Psychiatry. 2020;19(1):108–109. doi: 10.1002/wps.20723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bauer ME, Teixeira AL. Inflammation in psychiatric disorders: what comes first? Ann N Y Acad Sci. 2019;1437(1):57–67. doi: 10.1111/nyas.13712 [DOI] [PubMed] [Google Scholar]
  • 16.Sumner JA, Nishimi KM, Koenen KC, Roberts AL, Kubzansky LD. Posttraumatic Stress Disorder and Inflammation: Untangling Issues of Bidirectionality. Biol Psychiatry. 2020;87(10):885–897. doi: 10.1016/j.biopsych.2019.11.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Miller BJ, Goldsmith DR. Towards an Immunophenotype of Schizophrenia: Progress, Potential Mechanisms, and Future Directions. Neuropsychopharmacology. 2017;42(1):299–317. doi: 10.1038/npp.2016.211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pereira AC, Oliveira J, Silva S, Madeira N, Pereira CMF, Cruz MT. Inflammation in Bipolar Disorder (BD): Identification of new therapeutic targets. Pharmacol Res. 2021;163:105325. doi: 10.1016/j.phrs.2020.105325 [DOI] [PubMed] [Google Scholar]
  • 19.Goldsmith DR, Rapaport MH, Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: Comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21(12):1696–1709. doi: 10.1038/mp.2016.3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yang JJ, Jiang W. Immune biomarkers alterations in post-traumatic stress disorder: A systematic review and meta-analysis. J Affect Disord. 2020;268(November 2019):39–46. doi: 10.1016/j.jad.2020.02.044 [DOI] [PubMed] [Google Scholar]
  • 21.Munkholm K, Vinberg M, Vedel Kessing L. Cytokines in bipolar disorder: a systematic review and meta-analysis. J Affect Disord. 2013;144(1–2):16–27. doi: 10.1016/j.jad.2012.06.010 [DOI] [PubMed] [Google Scholar]
  • 22.Passos IC, Vasconcelos-Moreno MP, Costa LG, et al. Inflammatory markers in post-traumatic stress disorder: a systematic review, meta-analysis, and meta-regression. The lancet Psychiatry. 2015;2(11):1002–1012. doi: 10.1016/S2215-0366(15)00309-0 [DOI] [PubMed] [Google Scholar]
  • 23.Nettis MA, Pergola G, Kolliakou A, et al. Metabolic-inflammatory status as predictor of clinical outcome at 1-year follow-up in patients with first episode psychosis. Psychoneuroendocrinology. 2019;99:145–153. doi: 10.1016/j.psyneuen.2018.09.005 [DOI] [PubMed] [Google Scholar]
  • 24.Mondelli V, Ciufolini S, Belvederi Murri M, et al. Cortisol and Inflammatory Biomarkers Predict Poor Treatment Response in First Episode Psychosis. Schizophr Bull. 2015;41(5):1162–1170. doi: 10.1093/schbul/sbv028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Jacomb I, Stanton C, Vasudevan R, et al. C-Reactive Protein: Higher During Acute Psychotic Episodes and Related to Cortical Thickness in Schizophrenia and Healthy Controls. Front Immunol. 2018;9:2230. doi: 10.3389/fimmu.2018.02230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cuthbert BN, Insel TR. Toward the future of psychiatric diagnosis: the seven pillars of RDoC. BMC Med. 2013;11:126. doi: 10.1186/1741-7015-11-126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tamminga CA, Clementz BA. Biological fingerprints for psychosis. Neuropsychopharmacology. 2020;45(1):235–237. doi: 10.1038/s41386-019-0505-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sullivan PF, Agrawal A, Bulik CM, et al. Psychiatric Genomics: An Update and an Agenda. Am J Psychiatry. 2018;175(1):15–27. doi: 10.1176/appi.ajp.2017.17030283 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Clementz BA, Sweeney JA, Hamm JP, et al. Identification of Distinct Psychosis Biotypes Using Brain-Based Biomarkers. Am J Psychiatry. 2016;173(4):373–384. doi: 10.1176/appi.ajp.2015.14091200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tamminga CA, Clementz BA, Pearlson G, et al. Biotyping in psychosis: using multiple computational approaches with one data set. Neuropsychopharmacology. 2021;46(1):143–155. doi: 10.1038/s41386-020-00849-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat Rev Neurosci. 2008;9(1):46–56. doi: 10.1038/nrn2297 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kealy J, Greene C, Campbell M. Blood-brain barrier regulation in psychiatric disorders. Neurosci Lett. 2020;726:133664. doi: 10.1016/j.neulet.2018.06.033 [DOI] [PubMed] [Google Scholar]
  • 33.Kennedy RH, Silver R. Neuroimmune Signaling: Cytokines and the CNS. In: Pfaff DW, Volkow ND, eds. Neuroscience in the 21st Century. New York, NY: Springer New York; 2016:1–41. doi: 10.1007/978-1-4614-6434-1_174-1 [DOI] [Google Scholar]
  • 34.Miller AH, Haroon E, Raison CL, Felger JC. Cytokine targets in the brain: Impact on neurotransmitters and neurocircuits. Depress Anxiety. 2013;30(4):297–306. doi: 10.1002/da.22084 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Eisenberger NI, Berkman ET, Inagaki TK, Rameson LT, Mashal NM, Irwin MR. Inflammation-induced anhedonia: Endotoxin reduces ventral striatum responses to reward. Biol Psychiatry. 2010;68(8):748–754. doi: 10.1016/j.biopsych.2010.06.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Brydon L, Harrison NA, Walker C, Steptoe A, Critchley HD. Peripheral inflammation is associated with altered substantia nigra activity and psychomotor slowing in humans. Biol Psychiatry. 2008;63(11):1022–1029. doi: 10.1016/j.biopsych.2007.12.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Northoff G, Hirjak D, Wolf RC, Magioncalda P, Martino M. All roads lead to the motor cortex: psychomotor mechanisms and their biochemical modulation in psychiatric disorders. Mol Psychiatry. 2021;26(1):92–102. doi: 10.1038/s41380-020-0814-5 [DOI] [PubMed] [Google Scholar]
  • 38.Slavich GM, Way BM, Eisenberger NI, Taylor SE. Neural sensitivity to social rejection is associated with inflammatory responses to social stress. Proc Natl Acad Sci U S A. 2010;107(33):14817–14822. doi: 10.1073/pnas.1009164107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Muscatell KA, Dedovic K, Slavich GM, et al. Greater amygdala activity and dorsomedial prefrontal-amygdala coupling are associated with enhanced inflammatory responses to stress. Brain Behav Immun. 2015;43:46–53. doi: 10.1016/j.bbi.2014.06.201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Critchley HD. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol Psychiatry. 2009;66(5):407–414. doi: 10.1016/j.biopsych.2009.03.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Inagaki TK, Muscatell KA, Irwin MR, Cole SW, Eisenberger NI. Inflammation selectively enhances amygdala activity to socially threatening images. Neuroimage. 2012;59(4):3222–3226. doi: 10.1016/j.neuroimage.2011.10.090 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mehta ND, Haroon E, Xu X, Woolwine BJ, Li Z, Felger JC. Inflammation negatively correlates with amygdala-ventromedial prefrontal functional connectivity in association with anxiety in patients with depression: Preliminary results. Brain Behav Immun. 2018;73(August):725–730. doi: 10.1016/j.bbi.2018.07.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ruparelia N, Chai JT, Fisher EA, Choudhury RP. Inflammatory processes in cardiovascular disease: a route to targeted therapies. Nat Rev Cardiol. 2017;14(5):314. doi: 10.1038/nrcardio.2017.33 [DOI] [PubMed] [Google Scholar]
  • 44.Wu H, Ballantyne CM. Metabolic Inflammation and Insulin Resistance in Obesity. Circ Res. 2020;126(11):1549–1564. doi: 10.1161/CIRCRESAHA.119.315896 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107. doi: 10.1038/nri2925 [DOI] [PubMed] [Google Scholar]
  • 46.Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–1808. doi: 10.1172/JCI19246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.de Luca C, Olefsky JM. Inflammation and insulin resistance. FEBS Lett. 2008;582(1):97–105. doi: 10.1016/j.febslet.2007.11.057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Popa C, Netea MG, van Riel PLCM, van der Meer JWM, Stalenhoef AFH. The role of TNF-alpha in chronic inflammatory conditions, intermediary metabolism, and cardiovascular risk. J Lipid Res. 2007;48(4):751–762. doi: 10.1194/jlr.R600021-JLR200 [DOI] [PubMed] [Google Scholar]
  • 49.Pal M, Febbraio MA, Whitham M. From cytokine to myokine: the emerging role of interleukin-6 in metabolic regulation. Immunol Cell Biol. 2014;92(4):331–339. doi: 10.1038/icb.2014.16 [DOI] [PubMed] [Google Scholar]
  • 50.Nieto-Vazquez I, Fernández-Veledo S, Krämer DK, Vila-Bedmar R, Garcia-Guerra L, Lorenzo M. Insulin resistance associated to obesity: the link TNF-alpha. Arch Physiol Biochem. 2008;114(3):183–194. doi: 10.1080/13813450802181047 [DOI] [PubMed] [Google Scholar]
  • 51.Capuron L, Lasselin J, Castanon N. Role of Adiposity-Driven Inflammation in Depressive Morbidity. Neuropsychopharmacology. 2017;42(1):115–128. doi: 10.1038/npp.2016.123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Treadway MT, Cooper JA, Miller AH. Can’t or Won’t? Immunometabolic Constraints on Dopaminergic Drive. Trends Cogn Sci. 2019;23(5):435–448. doi: 10.1016/j.tics.2019.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Vogelzangs N, Beekman ATF, van Reedt Dortland AKB, et al. Inflammatory and metabolic dysregulation and the 2-year course of depressive disorders in antidepressant users. Neuropsychopharmacology. 2014;39(7):1624–1634. doi: 10.1038/npp.2014.9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Emery CF, Fondow MDM, Schneider CM, et al. Gastric bypass surgery is associated with reduced inflammation and less depression: a preliminary investigation. Obes Surg. 2007;17(6):759–763. doi: 10.1007/s11695-007-9140-0 [DOI] [PubMed] [Google Scholar]
  • 55.Musselman D, Shenvi N, Manatunga A, Miller AH, Lin E, Gletsu-Miller N. The effects of roux en y gastric bypass surgery on neurobehavioral symptom domains associated with severe obesity. Physiol Behav. 2019;204:86–92. doi: 10.1016/j.physbeh.2019.02.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Perry BI, McIntosh G, Weich S, Singh S, Rees K. The association between first-episode psychosis and abnormal glycaemic control: systematic review and meta-analysis. The lancet Psychiatry. 2016;3(11):1049–1058. doi: 10.1016/S2215-0366(16)30262-0 [DOI] [PubMed] [Google Scholar]
  • 57.Pillinger T, Beck K, Gobjila C, Donocik JG, Jauhar S, Howes OD. Impaired Glucose Homeostasis in First-Episode Schizophrenia: A Systematic Review and Meta-analysis. JAMA psychiatry. 2017;74(3):261–269. doi: 10.1001/jamapsychiatry.2016.3803 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Leonard BE, Schwarz M, Myint AM. The metabolic syndrome in schizophrenia: is inflammation a contributing cause? J Psychopharmacol. 2012;26(5 Suppl):33–41. doi: 10.1177/0269881111431622 [DOI] [PubMed] [Google Scholar]
  • 59.Kappelmann N, Arloth J, Georgakis MK, et al. Dissecting the Association between Inflammation, Metabolic Dysregulation, and Specific Depressive Symptoms: A Genetic Correlation and 2-Sample Mendelian Randomization Study. JAMA Psychiatry. 2021;78(2):161–170. doi: 10.1001/jamapsychiatry.2020.3436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Perry BI, Burgess S, Jones HJ, et al. The potential shared role of inflammation in insulin resistance and schizophrenia: A bidirectional two-sample mendelian randomization study. PLoS Med. 2021;18(3):1–21. doi: 10.1371/JOURNAL.PMED.1003455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lucido MJ, Bekhbat M, Goldsmith DR, et al. Aiding and Abetting Anhedonia: Impact of Inflammation on the Brain and Pharmacological Implications. Pharmacol Rev. 2021;73(3):1084–1117. doi: 10.1124/pharmrev.120.000043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lasselin J, Lekander M, Benson S, Schedlowski M, Engler H. Sick for science: experimental endotoxemia as a translational tool to develop and test new therapies for inflammation-associated depression. Mol Psychiatry. September 2020. doi: 10.1038/s41380-020-00869-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lawrence T, Gilroy DW. Chronic inflammation: a failure of resolution? Int J Exp Pathol. 2006;88(2):85–94. doi: 10.1111/j.1365-2613.2006.00507.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Furman D, Campisi J, Verdin E, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med. 2019;25(12):1822–1832. doi: 10.1038/s41591-019-0675-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Pearson TA, Mensah GA, Alexander RW, et al. Markers of inflammation and cardiovascular disease: application to clinical and public health practice: A statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation. 2003;107(3):499–511. doi: 10.1161/01.cir.0000052939.59093.45 [DOI] [PubMed] [Google Scholar]
  • 66.Baysak E, Guden DS, Aricioglu F, Halaris A. C-Reactive Protein as a Potential Biomarker in Psychiatric Practice: Are We There Yet? World J Biol Psychiatry. July 2021:1–37. doi: 10.1080/15622975.2021.1961502 [DOI] [PubMed] [Google Scholar]
  • 67.Felger JC, Haroon E, Patel TA, et al. What does plasma CRP tell us about peripheral and central inflammation in depression? Mol Psychiatry. 2020;25(6):1301–1311. doi: 10.1038/s41380-018-0096-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Osimo EF, Baxter LJ, Lewis G, Jones PB, Khandaker GM. Prevalence of low-grade inflammation in depression: A systematic review and meta-Analysis of CRP levels. Psychol Med. 2019;49(12):1958–1970. doi: 10.1017/S0033291719001454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wium-Andersen MK, Ørsted DD, Nielsen SF, Nordestgaard BG. Elevated C-reactive protein levels, psychological distress, and depression in 73, 131 individuals. JAMA psychiatry. 2013;70(2):176–184. doi: 10.1001/2013.jamapsychiatry.102 [DOI] [PubMed] [Google Scholar]
  • 70.Fernandes BS, Steiner J, Molendijk ML, et al. C-reactive protein concentrations across the mood spectrum in bipolar disorder: a systematic review and meta-analysis. The lancet Psychiatry. 2016;3(12):1147–1156. doi: 10.1016/S2215-0366(16)30370-4 [DOI] [PubMed] [Google Scholar]
  • 71.Sekar A, Bialas AR, De Rivera H, et al. Schizophrenia risk from complex variation of complement component 4. Nature. 2016;530(7589):177–183. doi: 10.1038/nature16549 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Corsi-Zuelli F, Deakin B. Impaired regulatory T cell control of astroglial overdrive and microglial pruning in schizophrenia. Neurosci Biobehav Rev. 2021. doi: 10.1016/j.neubiorev.2021.03.004 [DOI] [PubMed] [Google Scholar]
  • 73.Miller BJ, Goldsmith DR. Evaluating the Hypothesis That Schizophrenia Is an Inflammatory Disorder. Focus (Madison). 2020;18(4):391–401. doi: 10.1176/appi.focus.20200015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kindler J, Lim CK, Weickert CS, et al. Dysregulation of kynurenine metabolism is related to proinflammatory cytokines, attention, and prefrontal cortex volume in schizophrenia. Mol Psychiatry. 2020;25(11):2860–2872. doi: 10.1038/s41380-019-0401-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Solmi M, Veronese N, Favaro A, et al. Inflammatory cytokines and anorexia nervosa: A meta-analysis of cross-sectional and longitudinal studies. Psychoneuroendocrinology. 2015;51:237–252. doi: 10.1016/j.psyneuen.2014.09.031 [DOI] [PubMed] [Google Scholar]
  • 76.Gibson D, Mehler PS. Anorexia Nervosa and the Immune System-A Narrative Review. J Clin Med. 2019;8(11). doi: 10.3390/jcm8111915 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Goldsmith DR, Haroon E, Woolwine BJ, et al. Inflammatory markers are associated with decreased psychomotor speed in patients with major depressive disorder. Brain Behav Immun. 2016;56:281–288. doi: 10.1016/j.bbi.2016.03.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fillman SG, Weickert TW, Lenroot RK, et al. Elevated peripheral cytokines characterize a subgroup of people with schizophrenia displaying poor verbal fluency and reduced Broca’s area volume. Mol Psychiatry. 2016;21(8):1090–1098. doi: 10.1038/mp.2015.90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lizano P, Lutz O, Xu Y, et al. Multivariate relationships between peripheral inflammatory marker subtypes and cognitive and brain structural measures in psychosis. Mol Psychiatry. 2021;26(7):3430–3443. doi: 10.1038/s41380-020-00914-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Berk M, Williams LJ, Jacka FN, et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med. 2013;11(1):1–16. doi: 10.1186/1741-7015-11-200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Muniz Carvalho C, Wendt FR, Maihofer AX, et al. Dissecting the genetic association of C-reactive protein with PTSD, traumatic events, and social support. Neuropsychopharmacology. 2020;(October 2019). doi: 10.1038/s41386-020-0655-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lee CH, Sinclair D, O’Donnell M, et al. Transcriptional changes in the stress pathway are related to symptoms in schizophrenia and to mood in schizoaffective disorder. Schizophr Res. 2019;213:87–95. doi: 10.1016/j.schres.2019.06.026 [DOI] [PubMed] [Google Scholar]
  • 83.Irwin MR, Cole SW. Reciprocal regulation of the neural and innate immune systems. Nat Rev Immunol. 2011;11(9):625–632. doi: 10.1038/nri3042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Iwata M, Ota KT, Duman RS. The inflammasome: pathways linking psychological stress, depression, and systemic illnesses. Brain Behav Immun. 2013;31:105–114. doi: 10.1016/j.bbi.2012.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Brown AS, Derkits EJ. Prenatal infection and schizophrenia: a review of epidemiologic and translational studies. Am J Psychiatry. 2010;167(3):261–280. doi: 10.1176/appi.ajp.2009.09030361 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ezeoke A, Mellor A, Buckley P, Miller B. A systematic, quantitative review of blood autoantibodies in schizophrenia. Schizophr Res. 2013;150(1):245–251. doi: 10.1016/j.schres.2013.07.029 [DOI] [PubMed] [Google Scholar]
  • 87.Ripke S, Neale BM, Corvin A, et al. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511(7510):421–427. doi: 10.1038/nature13595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Jeppesen R, Benros ME. Autoimmune diseases and psychotic disorders. Front Psychiatry. 2019;10(MAR):1–11. doi: 10.3389/fpsyt.2019.00131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Naaldijk YM, Bittencourt MC, Sack U, Ulrich H. Kinins and microglial responses in bipolar disorder: a neuroinflammation hypothesis. Biol Chem. 2016;397(4):283–296. doi: 10.1515/hsz-2015-0257 [DOI] [PubMed] [Google Scholar]
  • 90.Saijo K, Glass CK. Microglial cell origin and phenotypes in health and disease. Nat Rev Immunol. 2011;11(11):775–787. doi: 10.1038/nri3086 [DOI] [PubMed] [Google Scholar]
  • 91.Nimgaonkar VL, Prasad KM, Chowdari KV, Severance EG, Yolken RH. The complement system: a gateway to gene-environment interactions in schizophrenia pathogenesis. Mol Psychiatry. 2017;22(11):1554–1561. doi: 10.1038/mp.2017.151 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Keshavan M, Lizano P, Prasad K. The synaptic pruning hypothesis of schizophrenia: promises and challenges. World Psychiatry. 2020;19(1):110–111. doi: 10.1002/wps.20725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Eraly SA, Nievergelt CM, Maihofer AX, et al. Assessment of plasma C-reactive protein as a biomarker of posttraumatic stress disorder risk. JAMA psychiatry. 2014;71(4):423–431. doi: 10.1001/jamapsychiatry.2013.4374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Friend SF, Nachnani R, Powell SB, Risbrough VB. C-Reactive Protein: Marker of risk for post-traumatic stress disorder and its potential for a mechanistic role in trauma response and recovery. Eur J Neurosci. November 2020. doi: 10.1111/ejn.15031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Michopoulos V, Beurel E, Gould F, et al. Association of Prospective Risk for Chronic PTSD Symptoms With Low TNFα and IFNγ Concentrations in the Immediate Aftermath of Trauma Exposure. Am J Psychiatry. 2020;177(1):58–65. doi: 10.1176/appi.ajp.2019.19010039 [DOI] [PubMed] [Google Scholar]
  • 96.Segman RH, Shefi N, Goltser-Dubner T, Friedman N, Kaminski N, Shalev AY. Peripheral blood mononuclear cell gene expression profiles identify emergent post-traumatic stress disorder among trauma survivors. Mol Psychiatry. 2005;10(5):500–513, 425. doi: 10.1038/sj.mp.4001636 [DOI] [PubMed] [Google Scholar]
  • 97.Pervanidou P, Kolaitis G, Charitaki S, et al. Elevated morning serum interleukin (IL)-6 or evening salivary cortisol concentrations predict posttraumatic stress disorder in children and adolescents six months after a motor vehicle accident. Psychoneuroendocrinology. 32(8–10):991–999. doi: 10.1016/j.psyneuen.2007.07.001 [DOI] [PubMed] [Google Scholar]
  • 98.Khandaker GM, Pearson RM, Zammit S, Lewis G, Jones PB. Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: a population-based longitudinal study. JAMA psychiatry. 2014;71(10):1121–1128. doi: 10.1001/jamapsychiatry.2014.1332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gomes FV, Zhu X, Grace AA. The pathophysiological impact of stress on the dopamine system is dependent on the state of the critical period of vulnerability. Mol Psychiatry. 2020;25(12):3278–3291. doi: 10.1038/s41380-019-0514-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Severance EG, Leister F, Lea A, Yang S, Dickerson F, Yolken RH. Complement C4 associations with altered microbial biomarkers exemplify gene-by-environment interactions in schizophrenia. Schizophr Res. February 2021. doi: 10.1016/j.schres.2021.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sellgren CM, Gracias J, Watmuff B, et al. Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat Neurosci. 2019;22(3):374–385. doi: 10.1038/s41593-018-0334-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.van Berckel BN, Bossong MG, Boellaard R, et al. Microglia activation in recent-onset schizophrenia: a quantitative (R)-[11C]PK11195 positron emission tomography study. Biol Psychiatry. 2008;64(9):820–822. doi: 10.1016/j.biopsych.2008.04.025 [DOI] [PubMed] [Google Scholar]
  • 103.Heurich M, Föcking M, Mongan D, Cagney G, Cotter DR. Dysregulation of complement and coagulation pathways: emerging mechanisms in the development of psychosis. Mol Psychiatry. July 2021. doi: 10.1038/s41380-021-01197-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mongan D, Sabherwal S, Susai SR, Föcking M, Cannon M, Cotter DR. Peripheral complement proteins in schizophrenia: A systematic review and meta-analysis of serological studies. Schizophr Res. 2020;222:58–72. doi: 10.1016/j.schres.2020.05.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Feng T, Tripathi A, Pillai A. Inflammatory Pathways in Psychiatric Disorders: The case of Schizophrenia and Depression. Curr Behav Neurosci reports. 2020;7(3):128–138. doi: 10.1007/s40473-020-00207-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Yrondi A, Aouizerate B, El-Hage W, et al. Assessment of Translocator Protein Density, as Marker of Neuroinflammation, in Major Depressive Disorder: A Pilot, Multicenter, Comparative, Controlled, Brain PET Study (INFLADEP Study). Front psychiatry. 2018;9:326. doi: 10.3389/fpsyt.2018.00326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Plavén-Sigray P, Matheson GJ, Collste K, et al. Positron Emission Tomography Studies of the Glial Cell Marker Translocator Protein in Patients With Psychosis: A Meta-analysis Using Individual Participant Data. Biol Psychiatry. 2018;84(6):433–442. doi: 10.1016/j.biopsych.2018.02.1171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Setiawan E, Wilson AA, Mizrahi R, et al. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA psychiatry. 2015;72(3):268–275. doi: 10.1001/jamapsychiatry.2014.2427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Plavén-Sigray P, Matheson GJ, Coughlin JM, et al. Meta-analysis of the Glial Marker TSPO in Psychosis Revisited: Reconciling Inconclusive Findings of Patient–Control Differences. Biol Psychiatry. 2021;89(3):e5–e8. doi: 10.1016/j.biopsych.2020.05.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Notter T, Coughlin JM, Gschwind T, et al. Translational evaluation of translocator protein as a marker of neuroinflammation in schizophrenia. Mol Psychiatry. 2018;23(2):323–334. doi: 10.1038/mp.2016.248 [DOI] [PubMed] [Google Scholar]
  • 111.De Picker L, Morrens M. Perspective: Solving the Heterogeneity Conundrum of TSPO PET Imaging in Psychosis. Front Psychiatry. 2020;11. doi: 10.3389/fpsyt.2020.00362 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Treadway MT, Zald DH. Parsing Anhedonia: Translational Models of Reward-Processing Deficits in Psychopathology. Curr Dir Psychol Sci. 2013;22(3):244–249. doi: 10.1177/0963721412474460 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Whitton AE, Treadway MT, Pizzagalli DA. Reward processing dysfunction in major depression, bipolar disorder and schizophrenia. Curr Opin Psychiatry. 2015;28(1):7–12. doi: 10.1097/YCO.0000000000000122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lokshina Y, Nickelsen T, Liberzon I. Reward Processing and Circuit Dysregulation in Posttraumatic Stress Disorder. Front Psychiatry. 2021;12. doi: 10.3389/fpsyt.2021.559401 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Höflich A, Michenthaler P, Kasper S, Lanzenberger R. Circuit Mechanisms of Reward, Anhedonia, and Depression. Int J Neuropsychopharmacol. 2019;22(2):105–118. doi: 10.1093/ijnp/pyy081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Treadway MT, Bossaller NA, Shelton RC, Zald DH. Effort-based decision-making in major depressive disorder: a translational model of motivational anhedonia. J Abnorm Psychol. 2012;121(3):553–558. doi: 10.1037/a0028813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Treadway MT, Buckholtz JW, Schwartzman AN, Lambert WE, Zald DH. Worth the “EEfRT”? The effort expenditure for rewards task as an objective measure of motivation and anhedonia. PLoS One. 2009;4(8):e6598. doi: 10.1371/journal.pone.0006598 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Vichaya EG, Hunt SC, Dantzer R. Lipopolysaccharide reduces incentive motivation while boosting preference for high reward in mice. Neuropsychopharmacology. 2014;39(12):2884–2890. doi: 10.1038/npp.2014.141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Muscatell KA, Moieni M, Inagaki TK, et al. Exposure to an inflammatory challenge enhances neural sensitivity to negative and positive social feedback. Brain Behav Immun. 2016;57:21–29. doi: 10.1016/j.bbi.2016.03.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Inagaki TK, Muscatell KA, Irwin MR, et al. The role of the ventral striatum in inflammatory-induced approach toward support figures. Brain Behav Immun. 2015;44:247–252. doi: 10.1016/j.bbi.2014.10.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Salamone JD, Correa M, Farrar A, Mingote SM. Effort-related functions of nucleus accumbens dopamine and associated forebrain circuits. Psychopharmacology (Berl). 2007;191(3):461–482. doi: 10.1007/s00213-006-0668-9 [DOI] [PubMed] [Google Scholar]
  • 122.Capuron L, Pagnoni G, Drake DF, et al. Dopaminergic mechanisms of reduced basal ganglia responses to hedonic reward during interferon alfa administration. Arch Gen Psychiatry. 2012;69(10):1044–1053. doi: 10.1001/archgenpsychiatry.2011.2094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Felger JC, Mun J, Kimmel HL, et al. Chronic interferon-α decreases dopamine 2 receptor binding and striatal dopamine release in association with anhedonia-like behavior in nonhuman primates. Neuropsychopharmacology. 2013;38(11):2179–2187. doi: 10.1038/npp.2013.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Felger JC, Li Z, Haroon E, et al. Inflammation is associated with decreased functional connectivity within corticostriatal reward circuitry in depression. Mol Psychiatry. 2016;21(10):1358–1365. doi: 10.1038/mp.2015.168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Goldsmith DR, Bekhbat M, Le NA, et al. Protein and gene markers of metabolic dysfunction and inflammation together associate with functional connectivity in reward and motor circuits in depression. Brain Behav Immun. 2020;88(May):193–202. doi: 10.1016/j.bbi.2020.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Bekhbat M, Treadway MT, Goldsmith DR, et al. Gene signatures in peripheral blood immune cells related to insulin resistance and low tyrosine metabolism define a sub-type of depression with high CRP and anhedonia. Brain Behav Immun. 2020;88:161–165. doi: 10.1016/j.bbi.2020.03.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Mehta D, Raison CL, Woolwine BJ, et al. Transcriptional signatures related to glucose and lipid metabolism predict treatment response to the tumor necrosis factor antagonist infliximab in patients with treatment-resistant depression. Brain Behav Immun. 2013;31:205–215. doi: 10.1016/j.bbi.2013.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Strauss GP, Horan WP, Kirkpatrick B, et al. Deconstructing negative symptoms of schizophrenia: avolition-apathy and diminished expression clusters predict clinical presentation and functional outcome. J Psychiatr Res. 2013;47(6):783–790. doi: 10.1016/j.jpsychires.2013.01.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Kirschner M, Hager OM, Bischof M, et al. Deficits in context-dependent adaptive coding of reward in schizophrenia. NPJ Schizophr. 2016;2:16020. doi: 10.1038/npjschz.2016.20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Hartmann MN, Hager OM, Reimann AV, et al. Apathy but not diminished expression in schizophrenia is associated with discounting of monetary rewards by physical effort. Schizophr Bull. 2015;41(2):503–512. doi: 10.1093/schbul/sbu102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wolf DH, Satterthwaite TD, Kantrowitz JJ, et al. Amotivation in schizophrenia: integrated assessment with behavioral, clinical, and imaging measures. Schizophr Bull. 2014;40(6):1328–1337. doi: 10.1093/schbul/sbu026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Liemburg EJ, Nolte IM, PHAMOUS investigators, Klein HC, Knegtering H. Relation of inflammatory markers with symptoms of psychotic disorders: a large cohort study. Prog Neuropsychopharmacol Biol Psychiatry. 2018;86:89–94. doi: 10.1016/j.pnpbp.2018.04.006 [DOI] [PubMed] [Google Scholar]
  • 133.Goldsmith DR, Haroon E, Miller AH, Strauss GP, Buckley PF, Miller BJ. TNF-α and IL-6 are associated with the deficit syndrome and negative symptoms in patients with chronic schizophrenia. Schizophr Res. 2018;199:281–284. doi: 10.1016/j.schres.2018.02.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Goldsmith DR, Haroon E, Miller AH, et al. Association of baseline inflammatory markers and the development of negative symptoms in individuals at clinical high risk for psychosis. Brain Behav Immun. 2019;76(October 2018):268–274. doi: 10.1016/j.bbi.2018.11.315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Juckel G, Schlagenhauf F, Koslowski M, et al. Dysfunction of ventral striatal reward prediction in schizophrenia. Neuroimage. 2006;29(2):409–416. doi: 10.1016/j.neuroimage.2005.07.051 [DOI] [PubMed] [Google Scholar]
  • 136.Nielsen MØ, Rostrup E, Wulff S, et al. Alterations of the brain reward system in antipsychotic naïve schizophrenia patients. Biol Psychiatry. 2012;71(10):898–905. doi: 10.1016/j.biopsych.2012.02.007 [DOI] [PubMed] [Google Scholar]
  • 137.Grimm O, Heinz A, Walter H, et al. Striatal response to reward anticipation: evidence for a systems-level intermediate phenotype for schizophrenia. JAMA psychiatry. 2014;71(5):531–539. doi: 10.1001/jamapsychiatry.2014.9 [DOI] [PubMed] [Google Scholar]
  • 138.Goldsmith DR, Massa N, Miller BJ, Miller AH, Duncan E. The interaction of lipids and inflammatory markers predict negative symptom severity in patients with schizophrenia. npj Schizophr. October 2021. doi: 10.1038/s41537-021-00179-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Sobin C, Sackeim HA. Psychomotor symptoms of depression. Am J Psychiatry. 1997;154(1):4–17. doi: 10.1176/ajp.154.1.4 [DOI] [PubMed] [Google Scholar]
  • 140.Morrens M, Hulstijn W, Sabbe B. Psychomotor slowing in schizophrenia. Schizophr Bull. 2007;33(4):1038–1053. doi: 10.1093/schbul/sbl051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ballenger JC, Davidson JR, Lecrubier Y, et al. Consensus statement on posttraumatic stress disorder from the International Consensus Group on Depression and Anxiety. J Clin Psychiatry. 2000;61 Suppl 5:60–66. http://www.ncbi.nlm.nih.gov/pubmed/10761680. [PubMed] [Google Scholar]
  • 142.Raison CL, Rutherford RE, Woolwine BJ, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA psychiatry. 2013;70(1):31–41. doi: 10.1001/2013.jamapsychiatry.4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Bruder GE, Alvarenga JE, Alschuler D, et al. Neurocognitive predictors of antidepressant clinical response. J Affect Disord. 2014;166:108–114. doi: 10.1016/j.jad.2014.04.057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Taylor BP, Bruder GE, Stewart JW, et al. Psychomotor slowing as a predictor of fluoxetine nonresponse in depressed outpatients. Am J Psychiatry. 2006;163(1):73–78. doi: 10.1176/appi.ajp.163.1.73 [DOI] [PubMed] [Google Scholar]
  • 145.Martinot M, Bragulat V, Artiges E, et al. Decreased presynaptic dopamine function in the left caudate of depressed patients with affective flattening and psychomotor retardation. Am J Psychiatry. 2001;158(2):314–316. doi: 10.1176/appi.ajp.158.2.314 [DOI] [PubMed] [Google Scholar]
  • 146.Juengling FD, Ebert D, Gut O, et al. Prefrontal cortical hypometabolism during low-dose interferon alpha treatment. Psychopharmacology (Berl). 2000;152(4):383–389. doi: 10.1007/s002130000549 [DOI] [PubMed] [Google Scholar]
  • 147.Bekhbat M, Goldsmith DR, Woolwine BJ, Haroon E, Miller AH, Felger JC. Transcriptomic signatures of psychomotor slowing in peripheral blood of depressed patients: evidence for immunometabolic reprogramming. Mol Psychiatry. September 2021. doi: 10.1038/s41380-021-01258-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Osborne KJ, Walther S, Shankman SA, Mittal VA. Psychomotor Slowing in Schizophrenia: Implications for Endophenotype and Biomarker Development. Biomarkers in neuropsychiatry. 2020;2(3):100016. doi: 10.1016/j.bionps.2020.100016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hirjak D, Thomann PA, Kubera KM, Wolf ND, Sambataro F, Wolf RC. Motor dysfunction within the schizophrenia-spectrum: A dimensional step towards an underappreciated domain. Schizophr Res. 2015;169(1–3):217–233. doi: 10.1016/j.schres.2015.10.022 [DOI] [PubMed] [Google Scholar]
  • 150.Goldsmith DR, Massa N, Pearce BD, et al. Inflammatory markers are associated with psychomotor slowing in patients with schizophrenia compared to healthy controls. npj Schizophr. 2020;6(1):1–8. doi: 10.1038/s41537-020-0098-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.O’Donovan A, Ahmadian AJ, Neylan TC, Pacult MA, Edmondson D, Cohen BE. Current posttraumatic stress disorder and exaggerated threat sensitivity associated with elevated inflammation in the Mind Your Heart Study. Brain Behav Immun. 2017;60:198–205. doi: 10.1016/j.bbi.2016.10.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Michopoulos V, Rothbaum AO, Jovanovic T, et al. Association of CRP genetic variation and CRP level with elevated PTSD symptoms and physiological responses in a civilian population with high levels of trauma. Am J Psychiatry. 2015;172(4):353–362. doi: 10.1176/appi.ajp.2014.14020263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Etkin A, Wager TD. Functional neuroimaging of anxiety: a meta-analysis of emotional processing in PTSD, social anxiety disorder, and specific phobia. Am J Psychiatry. 2007;164(10):1476–1488. doi: 10.1176/appi.ajp.2007.07030504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Arnegard ME, Whitten LA, Hunter C, Clayton JA. Sex as a Biological Variable: A 5-Year Progress Report and Call to Action. J Women’s Heal. 2020;29(6):858–864. doi: 10.1089/jwh.2019.8247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Moieni M, Tan KM, Inagaki TK, et al. Sex Differences in the Relationship Between Inflammation and Reward Sensitivity: A Randomized Controlled Trial of Endotoxin. Biol Psychiatry Cogn Neurosci Neuroimaging. 2019;4(7):619–626. doi: 10.1016/j.bpsc.2019.03.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Kokkosis AG, Tsirka SE. Neuroimmune Mechanisms and Sex/Gender-Dependent Effects in the Pathophysiology of Mental Disorders. J Pharmacol Exp Ther. 2020;375(1):175–192. doi: 10.1124/jpet.120.266163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Derry HM, Padin AC, Kuo JL, Hughes S, Kiecolt-Glaser JK. Sex Differences in Depression: Does Inflammation Play a Role? Curr Psychiatry Rep. 2015;17(10):78. doi: 10.1007/s11920-015-0618-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Fonkoue IT, Michopoulos V, Park J. Sex differences in post-traumatic stress disorder risk: autonomic control and inflammation. Clin Auton Res. 2020;30(5):409–421. doi: 10.1007/s10286-020-00729-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Abel KM, Drake R, Goldstein JM. Sex differences in schizophrenia. Int Rev Psychiatry. 2010;22(5):417–428. doi: 10.3109/09540261.2010.515205 [DOI] [PubMed] [Google Scholar]
  • 160.McCarthy MM. Sex differences in neuroimmunity as an inherent risk factor. Neuropsychopharmacology. 2019;44(1):38–44. doi: 10.1038/s41386-018-0138-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Slavich GM, Sacher J. Stress, sex hormones, inflammation, and major depressive disorder: Extending Social Signal Transduction Theory of Depression to account for sex differences in mood disorders. Psychopharmacology (Berl). 2019;236(10):3063–3079. doi: 10.1007/s00213-019-05326-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Bekhbat M, Neigh GN. Sex differences in the neuro-immune consequences of stress: Focus on depression and anxiety. Brain Behav Immun. 2018;67:1–12. doi: 10.1016/j.bbi.2017.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol. 2016;16(10):626–638. doi: 10.1038/nri.2016.90 [DOI] [PubMed] [Google Scholar]
  • 164.Rohleder N, Schommer NC, Hellhammer DH, Engel R, Kirschbaum C. Sex differences in glucocorticoid sensitivity of proinflammatory cytokine production after psychosocial stress. Psychosom Med. 63(6):966–972. doi: 10.1097/00006842-200111000-00016 [DOI] [PubMed] [Google Scholar]
  • 165.Steptoe A, Owen N, Kunz-Ebrecht S, Mohamed-Ali V. Inflammatory cytokines, socioeconomic status, and acute stress responsivity. Brain Behav Immun. 2002;16(6):774–784. doi: 10.1016/s0889-1591(02)00030-2 [DOI] [PubMed] [Google Scholar]
  • 166.Prather AA, Carroll JE, Fury JM, McDade KK, Ross D, Marsland AL. Gender differences in stimulated cytokine production following acute psychological stress. Brain Behav Immun. 2009;23(5):622–628. doi: 10.1016/j.bbi.2008.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Eisenberger NI, Inagaki TK, Rameson LT, Mashal NM, Irwin MR. An fMRI study of cytokine-induced depressed mood and social pain: the role of sex differences. Neuroimage. 2009;47(3):881–890. doi: 10.1016/j.neuroimage.2009.04.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Moieni M, Irwin MR, Jevtic I, Olmstead R, Breen EC, Eisenberger NI. Sex differences in depressive and socioemotional responses to an inflammatory challenge: implications for sex differences in depression. Neuropsychopharmacology. 2015;40(7):1709–1716. doi: 10.1038/npp.2015.17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Birur B, Amrock EM, Shelton RC, Li L. Sex Differences in the Peripheral Immune System in Patients with Depression. Front psychiatry. 2017;8:108. doi: 10.3389/fpsyt.2017.00108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Vogelzangs N, Comijs HC, Oude Voshaar RC, Stek ML, Penninx BWJH. Late-life depression symptom profiles are differentially associated with immunometabolic functioning. Brain Behav Immun. 2014;41:109–115. doi: 10.1016/j.bbi.2014.05.004 [DOI] [PubMed] [Google Scholar]
  • 171.Jha MK, Miller AH, Minhajuddin A, Trivedi MH. Association of T and non-T cell cytokines with anhedonia: Role of gender differences. Psychoneuroendocrinology. 2018;95:1–7. doi: 10.1016/j.psyneuen.2018.05.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Kamitaki N, Sekar A, Handsaker RE, et al. Complement genes contribute sex-biased vulnerability in diverse disorders. Nature. 2020;582(June). doi: 10.1038/s41586-020-2277-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Lenz KM, Nugent BM, Haliyur R, McCarthy MM. Microglia are essential to masculinization of brain and behavior. J Neurosci. 2013;33(7):2761–2772. doi: 10.1523/JNEUROSCI.1268-12.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Raison CL, Miller AH. Pathogen-Host Defense in the Evolution of Depression: Insights into Epidemiology, Genetics, Bioregional Differences and Female Preponderance. Neuropsychopharmacology. 2017;42(1):5–27. doi: 10.1038/npp.2016.194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Swartz JR, Prather AA, Hariri AR. Threat-related amygdala activity is associated with peripheral CRP concentrations in men but not women. Psychoneuroendocrinology. 2017;78:93–96. doi: 10.1016/j.psyneuen.2017.01.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Jha MK, Minhajuddin A, Gadad BS, et al. Can C-reactive protein inform antidepressant medication selection in depressed outpatients? Findings from the CO-MED trial. Psychoneuroendocrinology. 2017;78:105–113. doi: 10.1016/j.psyneuen.2017.01.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Uher R, Tansey KE, Dew T, et al. An inflammatory biomarker as a differential predictor of outcome of depression treatment with escitalopram and nortriptyline. Am J Psychiatry. 2014;171(12):1278–1286. doi: 10.1176/appi.ajp.2014.14010094 [DOI] [PubMed] [Google Scholar]
  • 178.Martino M, Rocchi G, Escelsior A, Fornaro M. Immunomodulation Mechanism of Antidepressants: Interactions between Serotonin/Norepinephrine Balance and Th1/Th2 Balance. Curr Neuropharmacol. 2012;10(2):97–123. doi: 10.2174/157015912800604542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Morón JA, Brockington A, Wise RA, Rocha BA, Hope BT. Dopamine uptake through the norepinephrine transporter in brain regions with low levels of the dopamine transporter: evidence from knock-out mouse lines. J Neurosci. 2002;22(2):389–395. http://www.ncbi.nlm.nih.gov/pubmed/11784783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Valentini V, Frau R, Di Chiara G. Noradrenaline transporter blockers raise extracellular dopamine in medial prefrontal but not parietal and occipital cortex: differences with mianserin and clozapine. J Neurochem. 2004;88(4):917–927. doi: 10.1046/j.1471-4159.2003.02238.x [DOI] [PubMed] [Google Scholar]
  • 181.Girgis RR, Ciarleglio A, Choo T, et al. A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Tocilizumab, An Interleukin-6 Receptor Antibody, For Residual Symptoms in Schizophrenia. Neuropsychopharmacology. 2018;43(6):1317–1323. doi: 10.1038/npp.2017.258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Nettis MA, Lombardo G, Hastings C, et al. Augmentation therapy with minocycline in treatment-resistant depression patients with low-grade peripheral inflammation: results from a double-blind randomised clinical trial. Neuropsychopharmacology. 2021;46(5):939–948. doi: 10.1038/s41386-020-00948-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Deakin B, Suckling J, Barnes TRE, et al. The benefit of minocycline on negative symptoms of schizophrenia in patients with recent-onset psychosis (BeneMin): a randomised, double-blind, placebo-controlled trial. The lancet Psychiatry. 2018;5(11):885–894. doi: 10.1016/S2215-0366(18)30345-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Husain MI, Chaudhry IB, Khoso AB, et al. Minocycline and celecoxib as adjunctive treatments for bipolar depression: a multicentre, factorial design randomised controlled trial. The lancet Psychiatry. 2020;7(6):515–527. doi: 10.1016/S2215-0366(20)30138-3 [DOI] [PubMed] [Google Scholar]

RESOURCES