Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-19T23:52:07.778Z Has data issue: false hasContentIssue false

Chapter 1 - Basic Concepts in Immunobiology

Published online by Cambridge University Press:  02 September 2021

Golam Khandaker
Affiliation:
University of Cambridge
Neil Harrison
Affiliation:
Cardiff University Brain Research Imaging Centre (CUBRIC)
Edward Bullmore
Affiliation:
University of Cambridge
Robert Dantzer
Affiliation:
University of Texas, MD Anderson Cancer Center

Summary

Once thought to be an immune-privileged site, we now know that there is a complex and essential bidirectional interplay between the central nervous system (CNS) and the immune system (1). Technological advances in imaging, genomic medicine and immunology have resulted in major revisions to some of the most fundamental and long-held assumptions in neuroscience, and we now understand that the immune system is critically involved not only in brain pathology, but also in the normal processes of brain development and homeostasis.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

1.1 Introduction

Once thought to be an immune-privileged site, we now know that there is a complex and essential bidirectional interplay between the central nervous system (CNS) and the immune system (Reference Savitz and Harrison1). Technological advances in imaging, genomic medicine and immunology have resulted in major revisions to some of the most fundamental and long-held assumptions in neuroscience, and we now understand that the immune system is critically involved not only in brain pathology, but also in the normal processes of brain development and homeostasis.

‘Immunopsychiatry’, namely the study of the interactions between neuroscience, mental health and the immune system, has rapidly become a major priority for psychiatric research. Accumulating evidence indicates roles for the immune system in the pathophysiology of many neurodegenerative and psychiatric disorders including Alzheimer’s disease (AD), schizophrenia, major depressive disorder (MDD), multiple sclerosis (MS) and others. In particular, pro-inflammatory cytokines associated with inflammation, which is common to many of these disorders, can influence the brain to bring about a host of physiological as well as behavioural alterations such as changes in mood and cognition. Raised pro-inflammatory cytokines are consistently reported in MDD (Reference Dowlati, Herrmann and Swardfager2) and schizophrenia (Reference Miller, Buckley, Seabolt, Mellor and Kirkpatrick3). Moreover, systemic inflammation brought about by acute infection, diabetes, obesity and atherosclerosis can accelerate cognitive decline in the elderly and present significant risk for the development or acceleration of AD and delirium (Reference Cunningham and Hennessy4). With this new knowledge come new targets to be exploited as biomarkers for diagnosis or informing treatment. A number of immune-modulating therapeutics are currently being trialled for the treatment of MDD, schizophrenia, AD and others.

In this chapter we aim to provide an overview of the key concepts in immunology and lay the foundations for understanding the role of the immune system in normal CNS homeostasis, and its contribution to neurodegeneration and psychiatric illness.

1.2 Overview of the Immune System

The most fundamental function of the immune system is to prevent and eradicate threats to bodily integrity. Frequently these threats are exogenous, invading microbes for example; but can also be endogenous in the form of damaged or dying tissue and malignancy.

The immune system can be broadly classified into two arms: the innate immune system and the adaptive immune system. Innate or natural immunity is always present in the individual. It consists of barriers that physically prevent entry of invading microbes, and a range of cells and non-cellular products that recognize and respond to threat. The innate immune system is critical for conveying messages to the brain warning of infection, resulting in the fever response. Additionally, components of the innate immune system enhance the adaptive immune response to pathogens. The adaptive immune system consists of T and B cells and their products. Where the innate immune system recognizes structures that are shared by many classes of microbes, and mounts the same response on repeated exposure, the adaptive immune system recognizes a wide variety of molecules with exquisite specificity, generates memory, and mounts a stronger response with each subsequent exposure. The innate and adaptive arms of the immune system work co-operatively to respond to threats and preserve bodily integrity. Failure can result in overwhelming infection, autoimmunity, or tissue damage, such as occurs in neurodegeneration. The immune system is increasingly recognized for its additional non-immune functions: wound healing, organ development and maintenance of homeostasis. The innate and adaptive immune systems and their contributions to immunopsychiatry will be discussed in this chapter.

1.3 Innate Immune System: Barriers

The first line of defence against invading microbes are the skin and mucosal epithelium, including the digestive tract, lungs and urogenital tract, which together provide a continuous physical barrier preventing the entry of microbes into the bloodstream. Within the healthy CNS, the blood-brain-barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) play analogous roles, blocking entry of microbes and selectively restricting the entry of immune cells. However, it is important to recognize that they are highly dynamic interfaces that communicate with the adjacent environments and serve the needs of the CNS (Reference Banks5). The BBB is a complex system comprising highly specialized endothelial cells that are joined by tight junctions, an underlying basement membrane embedded with pericytes, perivascular antigen presenting cells (APCs) and astrocytic endfeet (Reference Engelhardt and Sorokin6). The BCSFB on the other hand consists of the choroid plexus, which provides a physical interface between the blood and the cerebrospinal fluid (CSF). The choroid plexus epithelial cells generate approximately two-thirds of the CSF and help to control its composition by regulating the passage of ions, metabolites and molecules between the blood and the CSF (Reference Liddelow7). Tight junctions between the cells of the BBB and BCSFB inhibit the unrestricted diffusion of water soluble molecules, and both interfaces express active and passive cellular transporters that allow passage of select molecules, such as cytokines, hormones, amino acids and peptides between the blood and the CNS (Reference Banks5) (Figure 1.1).

Figure 1.1 Schematic representation of the Blood Brain Barrier and the blood cerebrospinal fluid barrier (BCSFB). A. The BBB consists of highly specialized endothelial cells joined by tight junctions. They deposit an endothelial basement membrane (yellow) in which pericytes are embedded. The parenchymal basement membrane (orange) is deposited by astrocytes and together with astrocytic endfeet forms the glia limitans perivascularis. The endothelial layer prevents the unrestricted movement of large solutes, antibodies and immune cells while allowing the passage of smaller solutes, cytokines and other proteins through dedicated transport systems.B. The BCSFB is made up by the choroid plexus epithelial cells which similarly prevent the unrestricted passage of molecules between the blood and the CSF. Central memory T cells are the predominant immune population in the CSF, and it is thought that they may cross the BCSFB, although the molecular mechanisms behind this are still unclear.

The BBB should not be thought of as an uninterrupted physical barrier. Certain regions of the brain, such as the circumventricular organs (CVOs) and the nucleus tractus solitarius have a specialized BBB with fenestrated capillary walls that enhance sensing of circulating molecules. Others, like the large vessels of the pial and subarachnoid space, and the sensory ganglia of the spinal and cranial nerves, contain ‘functional leaks’, allowing passage of molecules and substances into the Virchow–Robin spaces and participation in glymphatic flow. While the amounts of material entering the CNS through these pathways is small, it is probably the route through which natural and therapeutic antibodies gain access to the brain, such as anti-amyloid-β antibodies currently in trials for the treatment of AD (Reference Sevigny, Chiao and Bussière8).

1.4 Innate Immune System: Cells

In the periphery, a range of innate immune cells interact to prevent or limit the spread of infection. These cells include phagocytic neutrophils and monocytes, dendritic cells (DCs), mast cells and eosinophils, as well as specific subsets of lymphocytes such as innate lymphoid cells (ILCs), natural killer (NK) cells and NK-T cells. Within the CNS the range of innate immune cells is more limited, the most prevalent being the tissue-resident macrophage population known as microglia, which make up 10–15% of all brain cells (Reference Ransohoff and Cardona9). These cells populate the CNS early in embryonic development, performing typical phagocytic functions such as clearance of apoptotic cell debris and sensing local tissue damage, as well as tissue-specific functions including synapse elimination, regulation of neurogenesis and remodelling of neural circuits (Reference Li and Barres10). While microglia are the resident immune population of the brain parenchyma, they are not the only innate cells to populate the CNS. Macrophages, NK cells, mast cells and DCs also reside within the choroid plexus, perivascular spaces of the CNS and its meningeal coverings, sampling local debris and communicating with surrounding cells (Reference Ransohoff and Engelhardt11). Within the body, histamine released from mast cells plays an important role in dilating postcapillary venules and increasing blood vessel permeability leading to the oedema, warmth and redness associated with classical inflammation. Within the CNS, mast cells also play a critical role in regulating BBB permeability and have been implicated in increases in BBB permeability associated with stress (Reference Esposito, Gheorghe and Kandere12).

While the innate immune system comprises a diverse array of cells with specific functions in host defence, this discussion will focus on selected cell types and their contributions to immunopsychiatry.

1.4.1 Macrophages/Microglia

Macrophages and monocytes are closely related professional phagocytes that engulf and digest microbes and dead and dying host cells. During an inflammatory reaction, circulating blood monocytes can extravasate into tissues where they differentiate into specialized macrophages. The majority of tissues in the body contain phenotypically distinct tissue-resident macrophage populations which act as immune sentinels and perform additional tissue-specific functions. Langerhans cells (skin), Kupffer cells (liver), alveolar macrophages (lungs) and microglia (brain) are examples of tissue-resident macrophage populations. Fate mapping studies indicate that most tissue-resident macrophage populations derive from yolk sac or foetal liver origins, with blood monocytes existing in tissues only transiently (Reference Davies, Jenkins, Allen and Taylor13). The microglia of the brain are established in their niche prenatally, and derive from cells in the yolk sac (Reference Ginhoux, Greter and Leboeuf14). Experimental evidence suggests that microglia undergo self-renewal via local proliferation; however under certain circumstances infiltrating blood monocytes can differentiate into a microglial-like phenotype within the brain parenchyma (Reference Ajami, Bennett, Krieger, Tetzlaff and Rossi15,Reference Mildner, Schmidt and Nitsche16).

Macrophages are a highly dynamic cell population which undergo significant structural and functional changes depending on their local environment. They are critically important not only in the innate immune recognition of foreign and self-antigens, but also in the presentation of antigen to T cells and in guiding the outcome of the adaptive immune response through the secretion of specific cytokines.

Several studies have associated peripheral monocyte/macrophage numbers or activation status with psychiatric disorders. Monocytes/macrophages in both the blood and CSF may be increased in patients with schizophrenia or during acute psychotic episodes (Reference Nikkilä, Müller and Ahokas17,Reference Jackson and Miller18). While the number of monocytes does not appear to be altered in bipolar disorder, gene expression studies suggest that monocytes may be shifted towards a pro-inflammatory phenotype (Reference Padmos, Hillegers and Knijff19).

Traditionally, microglia have been thought of as existing in two states: ‘resting’ or ‘activated’, a misleadingly simplistic classification. Imaging studies have shown that ‘resting’ microglia are in fact highly active, continuously sampling the local environment with extremely motile processes (Reference Nimmerjahn, Kirchhoff and Helmchen20). Microglia express an array of receptors including: purinergic, neurotransmitter, cytokine, complement and Fc receptors, as well as pattern recognition receptors (PRRs), that allow them to detect subtle changes in their microenvironment (Reference Nimmerjahn, Kirchhoff and Helmchen20). In this ‘resting’ state microglia can secrete many neurotrophic factors including brain-derived neurotrophic factor (BDNF), transforming growth factor (TGF)-β, insulin-like growth factor 1 (IGF1), as well as pro- and anti-inflammatory cytokines (Reference Nimmerjahn, Kirchhoff and Helmchen20). In response to homeostatic disruption such as injury, infection or increased neuronal activity, microglia become activated and switch from undirected monitoring to targeted movement of microglial processes towards the site of insult (Reference Nimmerjahn, Kirchhoff and Helmchen20,Reference Davalos, Grutzendler and Yang21). Additional changes to morphology, motility, cell number, phagocytic capacity and cytokine secretion can also occur. Importantly, the shift from ‘resting’ (M1) to ‘activated’ (M2) phenotypes is accompanied by increased expression of the translocator protein (TSPO) which can be detected with positron emission tomography (PET) tracers such PK11195, FEPPA and DTA714 (Reference Setiawan, Wilson and Mizrahi22). Microglial dysregulation is associated with a number of psychiatric and neurodegenerative disorders including amyotrophic lateral sclerosis (ALS), AD, Parkinson’s disease, MDD and schizophrenia (Reference Li and Barres10,Reference Tay, Béchade and D’Andrea23).

A range of cytokines are secreted by microglia in both the healthy and the diseased brain. Microglial-derived IL-1β appears to be critical for normal hippocampus-dependent cognition, learning and memory formation (Reference Williamson, Sholar, Mistry, Smith and Bilbo24), whereas abnormally high levels of IL-1β and tumour necrosis factor (TNF)-α profoundly impair memory and are associated with the development of AD and other neurodegenerative disorders (Reference Marin and Kipnis25,Reference Holmes, Cunningham and Zotova26).

Microglia are critically dependent on colony stimulating factor 1 receptor (CSF1R) signalling for survival (Reference Erblich, Zhu, Etgen, Dobrenis and Pollard27,Reference Elmore, Najafi and Koike28). Congenital absence of CSF1R in mice results in abnormal postnatal brain development, with enlarged ventricles, increased neuronal density in the cortex, elevated numbers of astrocytes and reduced oligodendrocytes, with these animals rarely surviving to adulthood (Reference Erblich, Zhu, Etgen, Dobrenis and Pollard27). Whereas adult mice briefly depleted of microglia using CSF1R inhibitors show no readily apparent behavioural abnormalities or cognitive deficits, suggesting that microglia may not be essential for these functions in the fully developed brain (Reference Elmore, Najafi and Koike28). Pharmacological inhibition of CSF1R in mice with AD-like pathology halts microglial proliferation, prevents synaptic degeneration and improves cognitive performance (Reference Olmos-Alonso, Schetters and Sri29). Interestingly, microglial activation as a result of cranial irradiation for the treatment of brain cancer is associated with progressive and severe cognitive dysfunction. Experimental evidence suggests that elimination of microglia prior to cranial irradiation of mice ameliorates these cognitive defects (Reference Acharya, Green and Allen30). Therapies designed to deplete microglia using CSF1 R inhibitors or similar in the human brain could have efficacy for disorders associated with microglial activation and cognitive decline.

1.4.2 NK Cells

NK cells detect the absence of major histocompatibility complex (MHC) Class I proteins on infected, damaged or malignant cells and respond by inducing apoptosis of the target cell, independent of the adaptive immune system. Reduced NK cell numbers have been repeatedly found in patients with MDD, with some studies suggesting that reduced NK cell number or activity is associated with treatment resistance (Reference Grosse, Hoogenboezem and Ambrée31Reference Grosse, Carvalho and Birkenhager33). Abnormal NK cell activity has also been associated with schizophrenia; however, the direction of association is less consistent, possibly owing to clinical heterogeneity in the patient cohorts and small sample sizes (Reference Yovel, Sirota and Mazeh34). Of general relevance to psychiatry is the concept that psychological stress, a common feature across psychiatric disorders, is often accompanied by reduced NK cell activity (Reference Schedlowski, Jacobs and Stratmann35Reference Duggal, Upton, Phillips, Hampson and Lord37).

1.5 Innate Immune System: Molecules

The innate immune system employs a network of soluble and cell surface associated molecules to detect and combat invading microbial pathogens, remove potentially damaging dead and dying cells, and promote tissue repair. Like much of the immune system, these molecules are understood to also play roles in the maintenance of homeostasis, during development and in various aspects of normal and pathological mental function.

Innate immune cells express PRRs that recognize distinct microbial molecules known as pathogen-associate molecular patterns (PAMPs), or molecules released from damaged or dying host cells known as damage-associated molecular patterns (DAMPs). PAMPs are generally microbial structures that are essential for the survival or infectivity of the microbe, making this feature of innate immune recognition highly effective.

1.5.1 Toll-Like Receptors

TLRs are expressed on the cell surface or endosomal membranes of innate immune cells, including macrophages, NK cells, and DCs, as well as some non-immune cells such as endothelial cells. Importantly, TLRs are expressed by microglia, astrocytes, oligodendrocytes, neurons, neural progenitor cells and the BBB endothelium (Reference Okun, Griffioen and Mattson38). They are recognized for their importance in innate immune defence, and more recently they have been acknowledged for their roles in CNS plasticity and regulation of cognitive function in the absence of pathogen-derived ligands.

Individual TLRs recognize PAMPs that are shared across various classes of pathogen, and host-derived DAMPs. For example, TLR4 recognizes bacterial lipopolysaccharide (LPS) that is common to the outer membrane of all Gram-negative bacteria, as well as endogenous proteins such as heat shock protein-70 (HSP-70). In the CNS, TLR2 and TLR4 are expressed predominantly by glial cells including microglia, and both have been implicated in neuropathic pain (Reference Tanga, Nutile-McMenemy and DeLeo39,Reference Hutchinson, Zhang and Brown40). Interestingly, opioids such as morphine are capable of activating microglia by binding to the TLR4 accessory protein MD2 (Reference Hutchinson, Zhang and Shridhar41), and by stimulating the release of HSP-70 from neurons which in turn potentiates the TLR4 mediated inflammatory response (Reference Qu, Tao and Teng42). Furthermore, activation of TLR4 signalling by opioids is thought to contribute to the development of opioid tolerance and dependence, supporting a role for immune signalling in drug reward and opening up new possibilities for the treatment of addictions (Reference Hutchinson, Northcutt and Hiranita43). TLR2, TLR3 and TLR4 are expressed by human neural progenitor cells and influence cell proliferation and differentiation (Reference Okun, Griffioen and Mattson38). Increased gene expression of TLRs has been found in the brains of depressed suicide victims (Reference Pandey, Rizavi, Ren, Bhaumik and Dwivedi44), and peripheral blood of patients with MDD (Reference Hung, Kang, Huang and Huang45).

1.5.2 NOD-Like Receptors and the Inflammasome

The innate immune system utilizes a number of other PRRs in addition to TLRs. The nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) are a family of cytosolic receptors that sense both PAMPs and DAMPs in the cytoplasm of host cells. Engagement of NLRs by their ligand results in activation of the nuclear factor κ-light-chain-enhancer of activated B cells (NFκB) transcription factor and upregulation of pro-inflammatory cytokine gene expression. The NLRs form an important component of most inflammasomes. These are multimeric protein complexes that consist of an inflammasome sensor (often an NLR), the adaptor protein ASC, and caspase 1. Activation of the inflammasome generally results in cleavage of the inactive cytokine precursors, pro-IL-1β and pro-IL-18 and the subsequent release of their active forms, IL-1β and IL-18, respectively (Reference Latz, Xiao and Stutz46).

Of particular importance for immunopsychiatry is the NLR family pyrin domain containing 3 (NLRP3) inflammasome, a cytosolic inflammasome complex that responds to a diverse range of PAMPs and DAMPs by enhancing production of the pro-inflammatory cytokines IL-1β and IL-18 and has been implicated in the pathophysiology of MDD (Reference Alcocer-Gómez, de Miguel and Casas-Barquero47,Reference Zhang, Liu and Liu48), bipolar disorder (Reference Kim, Andreazza, Elmi, Chen and Young49) and AD (Reference Heneka, Kummer and Stutz50). Psychological stress can activate the NLRP3 inflammasome via adenosine triphosphate (ATP) stimulation of the P2X7 receptor in the mouse hippocampus, ultimately leading to an increase in IL-1β secretion and increased neuroinflammation (Reference Iwata, Ota and Li51). Interestingly, deletion of the NLRP3 gene from mice renders them resistant to the depressive behaviour normally induced by chronic unpredictable stress, possibly implicating the NLRP3 inflammasome in the well-established links between stress and increased risk of MDD (Reference Iwata, Ota and Li51). Together, these studies identify potential therapeutic targets for the treatment of depressive and neuroinflammatory disorders.

The final group of PRRs of importance to immunopsychiatry are the secreted forms, particularly C-reactive protein (CRP). CRP is an acute-phase protein synthesized in the liver with the primary function of activating the complement cascade. CRP is commonly measured in the clinic as an indicator of inflammation, and has consistently been shown to be raised in a proportion of patients with MDD (Reference Smith, Au, Ollis and Schmitz52Reference Wium-Andersen, Ørsted, Nielsen and Nordestgaard54). CRP shows promise as a peripherally accessible biomarker to predict the development of depression (Reference Khandaker, Pearson, Zammit, Lewis and Jones55) and for predicting therapeutic response to serotonergic and noradrenergic antidepressants (Reference Uher, Tansey and Dew56). CRP has additionally been implicated in schizophrenia (Reference Inoshita, Numata and Tajima57,Reference Fernandes, Steiner and Bernstein58); bipolar disorder, where it appears to be particularly increased during periods of mania (Reference Fernandes, Steiner and Molendijk59); and autism (Reference Khakzad, Javanbakht and Shayegan60).

1.5.3 The Complement System

The complement system is a network of more than 30 circulating and membrane-associated proteins responsible for induction of the complement cascade and inflammatory response. While complement components are synthesized predominantly by the liver, they can also be produced selectively by various other cell types including monocytes, fibroblasts, epithelial cells and, notably, all brain cell populations (Reference Orsini, De Blasio, Zangari, Zanier and De Simoni61). Initially recognized for its role in innate immune defence against pathogens, the complement system is now also known for its importance in the maintenance of brain homeostasis, synaptic plasticity and regeneration. Over-activation of the complement system has been implicated in the pathology of stroke, traumatic brain injury, AD, Parkinson’s disease and ALS (Reference Orsini, De Blasio, Zangari, Zanier and De Simoni61). More recently, excessive complement activity in the brain has been functionally linked to genetic risk for schizophrenia (Reference Sekar, Bialas and de Rivera62). Sekar et al. found that gene expression of complement component 4 (C4A) is elevated in the brains of individuals with schizophrenia and that higher levels of C4A expression are associated with a greater risk of schizophrenia. In addition to its critical role in innate immunity, many complement components including C4 are involved in synaptic pruning. Frequently documented pathological finding in the brains of individuals with schizophrenia include cortical grey matter loss and reduced numbers of synaptic structures (Reference Haijma, Van Haren and Cahn63,Reference Osimo, Beck, Reis Marques and Howes64). Given that microglia express the majority of complement receptors in the brain and are critically important in synaptic pruning, Sekar and colleagues offer an intriguing mechanistic explanation: that excessive synaptic pruning by microglia, driven by C4 deposition at the synapses, contributes to the pathology of schizophrenia (Reference Sekar, Bialas and de Rivera62). While C4 and other complement components have potential as diagnostic biomarkers or therapeutic treatment targets, much work still needs to be done before complement therapies reach the clinic.

1.6 Linking Innate and Adaptive Immunity: Cytokines and Chemokines

Cytokines are small proteins secreted by immune and non-immune cells that are responsible for intercellular communication, mediating many of the cellular responses of both innate and adaptive immunity. In innate immunity, most cytokines are produced by mast cells, DCs and macrophages, whereas the main cytokine producers of the adaptive immune system are T helper cells. Chemokines are a class of cytokines that act as chemoattractants for immune cells. Together, the cytokine network includes the interleukins, interferons, tumour necrosis factors, TGFs, colony stimulating factors and chemokines (CCL and CXCL nomenclature). Under steady-state conditions these molecules circulate in very low (picomolar) concentrations, but during infection or trauma blood concentrations can increase up to 1,000-fold. Cytokines can be pro- or anti-inflammatory, and can act in an autocrine, paracrine or endocrine manner. Examples of the latter include the systemic fever response triggered by IL-1β and TNF-α acting on the hypothalamus, a component of sickness behaviours (Reference Dantzer, O’Connor, Freund, Johnson and Kelley65) (covered in detail in Chapter 8), and IL-6 acting on the liver to stimulate production of acute-phase proteins such as CRP. Cytokines typically display a high degree of redundancy and pleiotropy and can enhance or inhibit the action of other cytokines in complex interactions, impeding simple functional classification. Nevertheless, one clinically useful division is between pro-inflammatory cytokines such as IL-1β, TNF-α, IL-6, IFN-γ and IL-12 – which share the common function of promoting inflammation; and anti-inflammatory cytokines – such as IL-4 and IL-10 that reduce inflammation and promote healing and tissue regeneration. Classification of cytokines can also be based on their roles in T helper cell polarization and function. IFN-γ and IL-12 are typically considered Th1 cytokines; IL-4, IL-5 and IL-13 are regarded as Th2 cytokines; and IL-17 and IL-22 as Th17 cytokines.

Some cytokines may cross the BBB through dedicated transporter systems or when barrier function is disrupted (Reference Banks66). Within the CNS, microglial activation results in the production of a number of pro-inflammatory cytokines and chemokines such as IL-1β, TNF-α, IL-6 and CCL2. Like many features of the immune system, cytokines are now known to play additional non-immunological roles, such as in regulation of complex cognitive processes like sleep (Reference Krueger, Rector and Roy67) and hippocampus-dependent learning and memory formation (Reference Rachal Pugh, Fleshner, Watkins, Maier and Rudy68). There is also extensive evidence that some CNS cytokines, IL-1β in particular, are directly involved in the pathogenesis of neurodegenerative disorders (Reference Allan, Tyrrell and Rothwell69).

CCL2, also known as monocyte chemoattractant protein 1 (MCP1) controls monocyte and macrophage migration. CCR2, the receptor that binds CCL2, is constitutively expressed by immune cells and can also be expressed by microglia, astrocytes, and dopaminergic and cholinergic neurons (Reference Conductier, Blondeau, Guyon, Nahon and Rovère70). Increased CCL2 gene expression by monocytes or astrocytes has been linked to a number of neuroinflammatory disorders including MS, stroke, AD, epilepsy, traumatic brain injury and schizophrenia (Reference Conductier, Blondeau, Guyon, Nahon and Rovère70). Elevated peripheral CCL2 has been consistently found in MDD and schizophrenia (Reference Eyre, Air and Pradhan71Reference Stuart and Baune73).

Numerous studies show increased levels of circulating pro-inflammatory cytokines, particularly IL-6 and TNF-α, and the acute-phase protein CRP, in MDD and schizophrenia (Reference Dowlati, Herrmann and Swardfager2,Reference Haapakoski, Mathieu, Ebmeier, Alenius and Kivimäki74,Reference Rodrigues-Amorim, Rivera-Baltanás and Spuch75). Importantly, peripheral levels of IL-6 are thought to precede depressive episodes (Reference Khandaker, Pearson, Zammit, Lewis and Jones55,Reference Miller and Cole76) and are often higher in individuals with a history of childhood adversity (Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli77). There is considerable interest in exploiting IL-6 as a therapeutic target, with a number of humanized monoclonal antibodies targeting IL-6 or the IL-6 receptor currently in clinical trials. Evidence linking pro-inflammatory cytokines and depression will be covered in detail in Chapter 10. Another group of cytokines important to immunopsychiatry are the interferons, key players in the antiviral response that have been used therapeutically for the treatment of certain cancers and the chronic viral infection hepatitis C. Although it has been an effective treatment for hepatitis C, sustained therapeutic use of IFN-α precipitates major depressive episodes in approximately one-third of patients, although the risk can be minimized through pretreatment with the antidepressant paroxetine (Reference Musselman, Lawson and Gumnick78).

1.7 Adaptive Immune System

In contrast to the innate immune system, which functions through the recognition of shared microbial structures and does not possess any form of memory, the adaptive immune system is characterized by the generation of memory responses by lymphocytes that recognize microbes with exquisite specificity. The adaptive immune system can be broadly classified into two functional arms: cellular immunity which is mediated by T cells, and humoral immunity which is mediated by B cells and their secreted antibodies. Key features of the adaptive immune system are the antigen receptors: namely, membrane-bound antibodies in the case of B cells, and the T cell receptor (TCR) complex on T cells. These receptors recognize antigens with a high degree of specificity. The ability of the immune system to discriminate self from non-self, and thereby avoid mounting a pathological response to self antigens, is known as tolerance and is achieved through two processes, termed central and peripheral tolerance. Central tolerance largely occurs in the primary lymphoid organs (thymus and bone marrow) and is a process whereby lymphocytes that strongly recognize self-antigens presented by MHC molecules are removed before they enter the circulation. Additionally, some of the immature CD4+ T cells that do recognize self-antigen develop into T regulatory cells (Treg) that are important for peripheral tolerance (discussed in Section 1.8.1 below). If self-reactive T cells do leave the primary lymphoid organs, they can be further controlled by peripheral tolerance mechanisms through which mature self-reactive T cells are rendered functionally inactive (anergic) or their response is controlled by Tregs. Autoimmunity occurs when immune tolerance breaks down, causing the adaptive immune system to mount a pathogenic response against self-antigens. While the precise aetiology of the inflammatory demyelinating disorder MS is incompletely understood, breakdown of immune tolerance leading to auto-reactive T and B cells plays a major role.

1.7.1 Major Histocompatibility Complex (MHC)

All nucleated cells express MHC Class I, while professional APCs such as DCs and B cells express MHC Class II. CD4+ and CD8+ T cells can only recognize antigen bound to MHC Class I or Class II, respectively. The MHC locus contains more than 200 genes and includes those that encode the MHC Class I and Class II molecules themselves, and other genes collectively known as the Class III genes which include complement components, cytokines, and some genes with no known immune function. The MHC genes are highly polymorphic, with over 10,000 Class I alleles and 3,000 Class II alleles estimated in the human population. The polymorphic gene variants are inherited and determine which peptides can be presented by the MHC molecules. Given their crucial role in immunity it is unsurprising that single nucleotide polymorphisms (SNPs) in the MHC region have been linked to a diverse range of diseases. Over the last decade, several independent genome wide association studies (GWAS) have revealed persuasive evidence for involvement of MHC genes in schizophrenia susceptibility (Reference Corvin and Morris79,80).

1.7.2 Adaptive Immunity: T Cells

Microbial antigens that enter the body are transported to and concentrated within peripheral lymphoid organs where they are most likely to come into contact with lymphocytes bearing antigen receptors that can recognize and respond to them. Lymphocytes continuously circulate between the bloodstream and the peripheral lymphoid organs with perhaps only 2% present in the blood. T cells are functionally classified into three main subsets; CD4+ T helper cells (Th), CD4+ Tregs and CD8+ cytotoxic T cells (Tc).

Th cells do not kill infected cells or microbes directly, but as their name implies, help other cell types to eliminate microbes. They are the primary source of cytokines in the adaptive immune system. Following antigen recognition, naïve Th cells rapidly proliferate (a process known as clonal expansion) and differentiate into effector cells of various subtypes that perform distinct and targeted functions in host defence. The three main subsets of Th cells are Th1, Th2 and Th17 cells, each distinguished by their cytokine profiles, chemokine receptor expression and targeted functions. There is considerable plasticity in the cytokine profile of T cells, allowing one subset to convert to another given the right conditions (See Figure 1.2).

Figure 1.2 Developmental origins of immune cells. Lymphocytes and some dendritic cells derive from the common lymphoid progenitor cell line. CD4+ T cells can differentiate into T helper cells with specific functions, defined by their cytokine profiles. Activated B cells give rise to antibody secreting plasma cells. Granulocytes (mast cells, neutrophils, eosinophils and basophils), monocytes, some dendritic cells and microglia derive from the common myeloid progenitor cell line. However, microglia are established in the brain prenatally during development from yolk sac progenitors and are transcriptionally distinct from other myeloid cells. Monocytes give rise to macrophages, some in peripheral tissues and others in the CNS.

The differentiation of Th effector cells into Th1 cells occurs in the presence of IL-12 and IFN-γ. Th1 cells secrete an abundance of IFN-γ and enhance the efficiency of macrophage killing of ingested microbes. In contrast, Th2 cells are induced during parasitic worm infections and allergy, and are characterized by the secretion of IL-4, IL-5 and IL-13. Th17 cells are important for the control of extracellular bacteria and fungi, but also play a detrimental role in many chronic inflammatory disorders. They are induced by IL-6, IL1-β, IL-23 and TGF-β and characterized by their secretion of IL-17 and IL-22.

Autoimmune Th cells of both the Th1 and Th17 subsets have been well described in MS, where they cross the BBB and accumulate in inflammatory lesions (Reference Kebir, Kreymborg and Ifergan81,Reference Reboldi, Coisne and Baumjohann82). Notably, the ratio of Th1:Th17 cells appears to determine where in the CNS the inflammation will occur. Myelin-specific Th cells infiltrate the meninges regardless of lineage; however, inflammation of the brain parenchyma only occurs when the balance is shifted towards Th17 cells, resulting in an increase in IL-17 in the brain (Reference Stromnes, Cerretti, Liggitt, Harris and Goverman83). While each of the Th subsets has been variously implicated in psychiatric disorders (Reference Grosse, Hoogenboezem and Ambrée31,Reference Avgustin, Wraber and Tavcar84,Reference Brambilla, Bellani and Isola85), their roles are still unclear and many studies regarding Th17 cells in schizophrenia report contradictory findings (Reference Debnath and Berk86Reference Fernandez-Egea, Vértes and Flint88). The discrepancies in these studies could be attributed to a number of factors such as inadequate sample size, ill-defined disease status (e.g., chronic versus recent onset psychosis), age, sex, body mass index (BMI) and treatment history. Moreover, antipsychotics may well have an effect on the inflammatory profile of immune cells (Reference Røge, Møller, Andersen, Correll and Nielsen89Reference Chen, Tsai and Wang91). Risperidone and clozapine, two antipsychotics commonly used for the treatment of schizophrenia, have shown therapeutic potential in ameliorating experimental autoimmune encephalitis (EAE), an experimental mouse model of MS (Reference O’Sullivan, Green and Stone92). The immunomodulatory activity of these antipsychotics occurs primarily in the CNS, significantly reducing microglial activation in EAE mice. Although the precise molecular mode of action of most antipsychotics is incompletely understood, they are generally thought to exert their therapeutic effects through the blockade of dopamine and serotonin receptors, both of which have immunomodulatory properties themselves. It is thus conceivable that antipsychotics may have therapeutic potential in non-psychotic inflammatory disorders.

In contrast to Th cells, Tc cells recognize antigen bound to MHC Class I, present on all nucleated cells including neurons (Reference Shatz93). Following activation, naïve Tc cells differentiate into cytotoxic T lymphocytes (CTLs) that are adept at killing infected cells, thereby eliminating the reservoir of infection. Tc cells have been less well-studied in immunopsychiatry, although there is some evidence that increased proportions of Tc cells may predict non-responsiveness to antidepressant treatment (Reference Grosse, Carvalho and Birkenhager33).

Following resolution of infection, a fraction of Th and Tc cells persist as long-lived memory cells. These cells circulate throughout the lymphoid organs, mucosa, peripheral tissues and the blood stream, and upon exposure to their antigen they rapidly and vigorously respond. Memory cells are defined by the lack of CD45RA and by the expression of CD45RO. While the numbers of lymphocytes and leukocytes in the CSF of normal healthy humans are low, there is a predominance of central memory CD45RO+ Th cells (Reference de Graaf, Smitt and Luitwieler94). Few studies have investigated the phenotype of immune cells in the CSF of patients with psychiatric disorders. However, there is some evidence that these populations differ from those found in healthy CSF (Reference Nikkilä, Müller and Ahokas17,Reference Nikkilä, Müller and Ahokas95). The question of whether or not these cells are detrimental in the CNS is an interesting one. Early studies showed that in response to CNS injury, effector T cells and Treg cells become activated, with autoimmune brain-reactive T cells playing a protective role and preventing further neurodegeneration, and Tregs playing a detrimental role by dampening the effector response (Reference Moalem, Leibowitz-Amit and Yoles96Reference Kipnis, Cardon and Avidan98). However, subsequent studies have shown that both depletion and administration of Tregs is associated with impaired neuronal survival (Reference Walsh, Zheng, Smirnov, Lorenz, Tung and Kipnis99). While this seems paradoxical, alterations in Treg numbers correlate with macrophage phenotype at the injury site. The broader relevance of this is yet to be defined. In addition to their preventative or pathologic roles in neurodegeneration, T cells also appear to be important in cognition and learning behaviour, as mice lacking T cells exhibit abnormal cognitive function. In the absence of T cells, meningeal myeloid cells acquire a pro-inflammatory phenotype which is associated with impaired learning behaviour (Reference Kipnis, Gadani and Derecki100). Taken together these studies suggest an important role for CNS T cells in maintaining homeostasis, that may have pathological implications when disrupted.

1.7.3 Adaptive Immunity: B Cells

B cells are able to recognize macromolecules of many types including proteins, lipids, carbohydrates and nucleic acids, without the requirement for MHC presentation. Activation of naïve B cells upon antigen exposure results in their clonal expansion and differentiation into antibody secreting plasma cells. The antibodies that are secreted have the same antigen specificity as the surface antibody receptor of the naïve B cell that initiated the response. Th cells are critical for the activation of B cells and the maturation of the humoral immune response. B cells can also have specificity for self-antigen, secreting antibodies that recognize self-antigens (autoantibodies) and mounting an unwanted immune response. In 2007, the first cases of antibody-mediated encephalitis and associated psychosis were reported by Josep Dalmau and colleagues (Reference Dalmau, Tüzün and Wu101) and since this seminal publication, autoantibodies to multiple neuronal antigens or synaptic proteins have been described (Reference Lennox, Palmer-Cooper and Pollak102). Psychiatric or behavioural manifestations are often seen in patients with these autoantibodies. However, they have also frequently been detected in healthy control patients without psychiatric symptomology. A recent study estimates approximately 9% of patients presenting with first episode psychosis have serum anti-neuronal antibodies, which may be treatable with immunotherapy (Reference Lennox, Palmer-Cooper and Pollak102). This will be discussed in further detail in Chapter 5. Some preliminary evidence suggests that maternal brain-reactive antibodies may contribute to the development of autism in the child (Reference Brimberg, Mader and Jeganathan103). Autoantibodies are also associated with Parkinson’s disease, where the presence of anti-α-synuclein antibodies is reduced compared with healthy controls (Reference Brudek, Winge and Folke104). While the function of these autoantibodies in the healthy CNS is currently unknown, it is possible that they help to clear pathological α-synuclein, a key characteristic of Parkinson’s disease. Thus, like CNS T cells, CNS autoantibodies may serve both protective and pathologic functions which could be exploited for therapeutic or diagnostic benefit.

1.8 Glymphatics

Until recently the CNS was thought to be devoid of lymphatic vasculature, despite scattered reports over the years indicating its presence. With the 2012 demonstration of the ‘glymphatic system’ which functions as a fluid and solute clearance pathway (Reference Iliff, Wang and Liao105), and the subsequent 2015 discovery of meningeal lymphatic vasculature lining the dural sinuses and meningeal arteries to drain lymphatic fluid, immune cells and other solutes (Reference Louveau, Smirnov and Keyes106,Reference Aspelund, Antila and Proulx107), our mechanistic understanding of CNS drainage has undergone major revision. These two systems are now believed to work in concert to maintain homeostasis and contribute to CNS function and health (Reference Da Mesquita, Fu and Kipnis108).

In the periphery, toxic metabolites, cell debris, infiltrating microbes, DCs carrying microbial antigens, lymphocytes and interstitial fluid (IF) are drained from tissues via the lymphatic system. Located strategically along the lymphatic vessels are the lymph nodes through which the draining fluid passes before its return to the blood circulation. As lymphatic fluid passes through the lymph nodes its contents are sampled by B cells which can respond by producing antibodies, and APCs which internalize, process and present antigen to T cells, initiating the cell-mediated immune response. However, the CNS parenchyma lacks a classical lymphatic drainage system. Nedergard and colleagues demonstrated that CSF enters the brain within the periarterial spaces and then exchanges with IF, facilitated by aquaporin-4 (AQP4) water channels positioned within perivascular astrocyte endfoot processes. This fluid collects within perivenous spaces and drains to the subarachnoid CSF, which subsequently drains either to the venous circulation, extracranial lymphatic vessels or lymph nodes. Named the glial-associated lymphatic system, or ‘glymphatic system’, due to the dependence on AQP4 on astrocytes and the lymphatic function it serves. The glymphatic system allows nutrients such as glucose to diffuse through the brain parenchyma and functions to clear extracellular metabolites, waste products such as lactate, and protein aggregates such as amyloid-β from the brain parenchyma and into the CSF. Interestingly, glymphatic clearance of amyloid-β declines with age, and amyloid-β in turn suppresses glymphatic influx in a detrimental feedback loop. The drainage of amyloid-β is further impeded by APOE4, the most important genetic risk factor for late-onset AD, demonstrating the importance of the glymphatic system in the pathology of AD and highlighting its potential as a therapeutic target (Reference Iliff, Wang and Liao105).

The meninges that surround the brain and spinal cord are composed of three layers: the pia, arachnoid and dura maters. Despite scattered reports of the presence of lymphatic vessels within the meninges, it was not until 2015 that their presence gained wider attention thanks to work from Kipnis and Alitalo (Reference Louveau, Smirnov and Keyes106,Reference Aspelund, Antila and Proulx107). These two studies, published simultaneously, used advanced imaging techniques and fluorescent tracer experiments to demonstrate draining lymphatic vessels adjacent to arteries, major venous sinuses and cranial nerves (Figure 1.3).

Figure 1.3 CNS lymphatic and glymphatic systems. A. Schematic of the glymphatic system. CSF enters the brain parenchyma along paraarterial routes, exchanges with interstitial fluid and is cleared along paravenous routes. Convective flow is facilitated by AQP4 pores which are abundantly expressed on astrocytic endfeet. Clearance of solutes, proteins and waste products such as amyloid-β occur along this pathway. Draining fluid may be dispersed into the subarachnoid CSF, venous circulation or lymphatics.B. Schematic representation of lymphatic drainage of the brain. Meningeal lymphatic vessels (green) adjacent to the venous sinuses and arteries drain molecules and meningeal immune cells in the CSF into the deep cervical lymph nodes. Adapted with permission from Louveau et al. Neuron. 2016; 91:957–73 (Reference Louveau, Da Mesquita and Kipnis109).

Though still in the early stages, intensive research is underway to exploit these systems therapeutically for the treatment of disorders with CNS involvement.

1.9 Central and Peripheral Communication

Bidirectional crosstalk between the CNS and the peripheral immune system is now well established. An important example of immune-brain communication is the inflammatory reflex which consists of two arcs. The afferent or sensory arc involves visceral sensory neurons which detect and respond to inflammatory stimuli, such as changes in pro-inflammatory cytokines, by relaying information to the brain stem via the vagus nerve. Inflammation detected in the periphery can have profound effects on the brain, resulting in impaired learning and memory, cognitive abnormalities and altered mood (Reference Harrison, Brydon and Walker110,Reference Harrison, Doeller, Voon, Burgess and Critchley111) and this pathway is important in the generation of sickness behaviours (see Section 1.10 below). The efferent arc in which immunomodulatory signals are delivered from the brain stem to immune cells via the vagus nerve, is also known as the anti-inflammatory cholinergic pathway and signals through acetylcholine receptors expressed by immune cells, leading to NFκB activation and modulation of immune response (Reference Tracey112). Additionally, all primary and secondary lymphoid organs, including the thymus, bone marrow and spleen, receive direct sympathetic innervation (Reference Bellinger, Millar and Perez113). These sympathetic nerve fibres release the catecholamine norepinephrine (NE) in response to stress and other stimuli, which binds to β-adrenergic receptors on immune cells and modulates diverse immunological processes including lymphocyte activation, cytokine production and cell trafficking (Reference Sloan, Capitanio and Tarara114).

It is clear that some classical neurotransmitters, like acetylcholine in the inflammatory reflex, are not only expressed by immune cells but also functionally important for immune system signalling. Dopamine signalling, in particular, has been linked to modulation of the immune synapse that forms between an APC, typically a DC and a naïve T cell (Reference Pacheco, Riquelme and Kalergis115). This interaction determines the fate of the T cell and is conditioned by dopamine released presynaptically from DCs activating dopamine receptors (DR) postsynaptically expressed by T cells. For example, activation of D3DR inhibits the suppressive capability of Tregs (Reference Kipnis, Cardon and Avidan98) which may be important for dysregulated cognition. Dysregulated dopaminergic neurotransmission has been implicated in the pathogenesis of Parkinson’s disease, schizophrenia, dementia with Lewy bodies and attention deficit hyperactivity disorder. In addition to these central defects in dopamine function, Parkinson’s disease has been replicably associated with increased (Reference Debnath and Berk86,Reference Ilani, Ben-Shachar and Strous116,Reference Brito-Melo, Nicolato and de Oliveira117) and schizophrenia with decreased (Reference Nagai, Ueno and Saeki118) peripheral expression of the gene coding the D3DR gene. Collectively these studies indicate that the peripheral immune system is capable of interacting with the nervous system through the secretion and recognition of neurotransmitters. With further research, the peripheral expression of neurotransmitter-related genes may prove useful as biomarkers for dopaminergic disorders such as schizophrenia and Parkinson’s disease.

1.10 Sickness Behaviour

In addition to immune activation, the physiological response to infection is accompanied by behavioural changes such as lethargy, malaise, irritability, impaired concentration, behavioural depression, decreased social activity, anhedonia and somnolence. Collectively these symptoms are known as ‘sickness behaviours’, notable for their remarkable similarity to common symptoms of depression. The main pro-inflammatory cytokines responsible for sickness behaviour are IL-1β and TNF-α, both of which are increased in the brain following peripheral infection or experimental administration of LPS in the absence of infection (Reference Dantzer, O’Connor, Freund, Johnson and Kelley65,Reference Thomson, McColl, Cavanagh and Graham119). Conversely, anti-inflammatory cytokines such as IL-10 limit sickness behaviour and attenuate the behavioural effects induced by LPS stimulation (Reference Dantzer, O’Connor, Freund, Johnson and Kelley65). Pro-inflammatory cytokines act on the brain to reorient motivation towards prioritizing wound healing and fighting infection. The behavioural phenotypes of social avoidance and anhedonia help to protect from further insult and pathogen exposure (Reference Miller and Raison120). When inflammation is severe or prolonged, as in chronic inflammatory conditions, major depressive episodes frequently arise. Roughly one-third of patients receiving recombinant IL-2 or IFN-α develop major depression, even previously euthymic individuals (Reference Musselman, Lawson and Gumnick78), and similar patterns of brain dysfunction are seen in both IFN-α induced depression and idiopathic MDD (Reference Harrison121). See Chapter 8 for an in-depth review of sickness behaviour.

1.10.1 Kynurenine Pathway

Cytokine immunotherapy in cancer patients is accompanied by a marked reduction in plasma tryptophan concentration that is positively correlated with depression scores (Reference Miller and Raison120,Reference Capuron, Gumnick and Musselman123). Actively transported into the brain, tryptophan is an essential precursor for the synthesis of serotonin, a neurotransmitter well known for its involvement in mood regulation. Decreased plasma tryptophan in patients undergoing immunotherapy could be due to the action of the tryptophan metabolizing enzymes tryptophan dioxygenase (TDO) or indoleamine 2,3 dioxygenase (IDO), both of which divert tryptophan metabolism away from serotonin synthesis and towards kynurenine synthesis. Under normal circumstances TDO is the dominant enzyme, however the enzymatic activity of IDO is enhanced during inflammation and is potently induced by several pro-inflammatory cytokines including IFN-γ and TNF-α (Figure 1.4). Kynurenine is readily transported across the BBB into the CNS where its metabolites can be either neuroprotective or neurotoxic (Reference Dantzer, O’Connor, Freund, Johnson and Kelley65). Kynurenine metabolites may prove useful as biomarkers; for example, a high kynurenine/tryptophan ratio predicted remission in a recent trial of the anti-inflammatory COX-2 inhibitor celecoxib (Reference Krause, Myint and Schuett124). Interestingly, previous clinical trials suggest that the antidepressant effects of celecoxib are linked to its capacity to reduce serum IL-6 (Reference Abbasi, Hosseini, Modabbernia, Ashrafi and Akhondzadeh125). While the causative role of IDO activation in MDD still needs to be thoroughly investigated in humans, there is convincing evidence from rodent studies that therapeutic targeting of IDO may help to alleviate inflammation-associated depression (Reference O’Connor, Lawson and André126).

Figure 1.4 The enzymatic activity of IDO is enhanced during inflammation and diverts tryptophan metabolism away from serotonin synthesis and towards the kynurenine pathway. Kynurenine is readily transported across the BBB where it can be metabolized to 3-hydroxykynurenine (3-HK), quinolinic acid (QA) or kynurenic acid (KA). 3-HK can increase oxidative stress and may be neurotoxic. QA is preferentially produced by microglia and is an NMDA-receptor agonist which may be neurotoxic, whereas KA is produced by astrocytes and is an NMDA-receptor antagonist which may be neuroprotective. In the periphery KA is thought to modulate immune cell phenotypes. Impaired glutamatergic and serotonergic neurotransmission are both associated with the development of depressive behaviour.

1.11 Conclusion

This chapter serves as a broad introduction to some of the immunological concepts relevant to the study of neurodegenerative and psychiatric disorders. A greater understanding of the causative role of the immune system in these disorders could generate novel immunotherapeutic targets for diagnosis and treatment. Convincing evidence from preclinical studies indicate that inflammation is a key contributor to MDD; several clinical trials using anti-inflammatory immunotherapies to treat MDD are in various stages of completion and are showing promising results. Recent advances in our understanding of how the immune system interacts with the brain to cause pathology offer a range of new therapeutic targets for researchers and clinicians to utilize. Although the field of immunopsychiatry is still relatively young, close collaborations between researchers, clinicians and industry will deliver high quality research and therapeutic targets for tackling what are currently some of the most common and debilitating human illnesses.

References

Savitz, J, Harrison, NA. Interoception and Inflammation in Psychiatric Disorders. Biol Psychiatry Cogn Neurosci Neuroimaging. 2018 Jun;3(6):514–24.Google Scholar
Dowlati, Y, Herrmann, N, Swardfager, W, et al. A meta-analysis of cytokines in major depression. Biological Psychiatry. 2010 Mar 1;67(5):446–57.CrossRefGoogle ScholarPubMed
Miller, BJ, Buckley, P, Seabolt, W, Mellor, A, Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biological Psychiatry. 2011 Oct 1;70(7):663–71.Google Scholar
Cunningham, C, Hennessy, E. Co-morbidity and systemic inflammation as drivers of cognitive decline: new experimental models adopting a broader paradigm in dementia research. Alzheimers Res Ther. BioMed Central Ltd. 2015;7(1):33.Google Scholar
Banks, WA. From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery. Nature Publishing Group. 2016 Apr;15(4):275–92.Google Scholar
Engelhardt, B, Sorokin, L. The blood-brain and the blood-cerebrospinal fluid barriers: function and dysfunction. Semin Immunopathol. 2009 Nov;31(4):497511.Google Scholar
Liddelow, SA. Development of the choroid plexus and blood-CSF barrier. Front Neurosci. Frontiers. 2015 Mar 3;9(154):319–13.Google Scholar
Sevigny, J, Chiao, P, Bussière, T, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016 Sep 1;537(7618):50–6.Google Scholar
Ransohoff, RM, Cardona, AE. The myeloid cells of the central nervous system parenchyma. Nature. 2010 Nov 11;468(7321):253–62.Google Scholar
Li, Q, Barres, BA. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol. 2017 Nov 20.Google Scholar
Ransohoff, RM, Engelhardt, B. The anatomical and cellular basis of immune surveillance in the central nervous system. Nat Rev Immunol. 2012 Sep;12(9):623–35.Google Scholar
Esposito, P, Gheorghe, D, Kandere, K, et al. Acute stress increases permeability of the blood-brain-barrier through activation of brain mast cells. Brain Res. 2001 Jan 5;888(1):117–27.Google Scholar
Davies, LC, Jenkins, SJ, Allen, JE, Taylor, PR. Tissue-resident macrophages. Nat Immunol. 2013 Oct;14(10):986–95.CrossRefGoogle ScholarPubMed
Ginhoux, F, Greter, M, Leboeuf, M, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010 Nov 5;330(6005):841–5.Google Scholar
Ajami, B, Bennett, JL, Krieger, C, Tetzlaff, W, Rossi, FMV. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 2007 Dec;10(12):1538–43.Google Scholar
Mildner, A, Schmidt, H, Nitsche, M, et al. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci. 2007 Dec;10(12):1544–53.Google Scholar
Nikkilä, HV, Müller, K, Ahokas, A, et al. Accumulation of macrophages in the CSF of schizophrenic patients during acute psychotic episodes. Am J Psychiatry. 1999 Nov;156(11):1725–9.CrossRefGoogle ScholarPubMed
Jackson, AJ, Miller, BJ. Meta-analysis of total and differential white blood cell counts in schizophrenia. Acta Psychiatr Scand. 2020 Jul;142(1):1826.Google Scholar
Padmos, RC, Hillegers, MHJ, Knijff, EM, et al. A discriminating messenger RNA signature for bipolar disorder formed by an aberrant expression of inflammatory genes in monocytes. Arch Gen Psychiatry. American Medical Association; 2008 Apr;65(4):395407.Google Scholar
Nimmerjahn, A, Kirchhoff, F, Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005 May 27;308(5726):1314–8.CrossRefGoogle ScholarPubMed
Davalos, D, Grutzendler, J, Yang, G, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005 Jun;8(6):752–8.Google Scholar
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 Mar;72(3):268–75.Google Scholar
Tay, TL, Béchade, C, D’Andrea, I, et al. Microglia gone rogue: impacts on psychiatric disorders across the lifespan. Front Mol Neurosci. 2017;10:421.Google Scholar
Williamson, LL, Sholar, PW, Mistry, RS, Smith, SH, Bilbo, SD. Microglia and memory: modulation by early-life infection. J Neurosci. 2011 Oct 26;31(43):15511–21.Google Scholar
Marin, I, Kipnis, J. Learning and memory … and the immune system. Learn Mem. 2013 Sep 19;20(10):601–6.Google Scholar
Holmes, C, Cunningham, C, Zotova, E, et al. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009 Sep 8;73(10):768–74.Google Scholar
Erblich, B, Zhu, L, Etgen, AM, Dobrenis, K, Pollard, JW. Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS ONE. 2011;6(10):e26317.CrossRefGoogle ScholarPubMed
Elmore, MRP, Najafi, AR, Koike, MA, et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron. 2014 Apr 16;82(2):380–97.Google Scholar
Olmos-Alonso, A, Schetters, STT, Sri, S, et al. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain. 2016 Mar;139(Pt 3):891907.Google Scholar
Acharya, MM, Green, KN, Allen, BD, et al. Elimination of microglia improves cognitive function following cranial irradiation. Sci Rep. 2016 Aug 12;6(1):31545.Google Scholar
Grosse, L, Hoogenboezem, T, Ambrée, O, et al. Deficiencies of the T and natural killer cell system in major depressive disorder: T regulatory cell defects are associated with inflammatory monocyte activation. Brain Behavior and Immunity. 2016 May;54:3844.Google Scholar
Suzuki, H, Savitz, J, Teague, TK, et al. Altered populations of natural killer cells, cytotoxic T lymphocytes, and regulatory T cells in major depressive disorder: Association with sleep disturbance. Brain Behavior and Immunity. 2017 Nov 1;66:193200.Google Scholar
Grosse, L, Carvalho, LA, Birkenhager, TK, et al. Circulating cytotoxic T cells and natural killer cells as potential predictors for antidepressant response in melancholic depression. Restoration of T regulatory cell populations after antidepressant therapy.Psychopharmacology (Berl). 2016 May;233(9):1679–88.Google Scholar
Yovel, G, Sirota, P, Mazeh, D, et al. Higher natural killer cell activity in schizophrenic patients: the impact of serum factors, medication, and smoking. Brain Behavior and Immunity. 2000 Sep;14(3):153–69.Google Scholar
Schedlowski, M, Jacobs, R, Stratmann, G, et al. Changes of natural killer cells during acute psychological stress. J Clin Immunol. 1993 Mar;13(2):119–26.Google Scholar
Segerstrom, SC, Miller, GE. Psychological stress and the human immune system: a meta-analytic study of 30 years of inquiry. Psychol Bull. 2004 Jul;130(4):601–30.Google Scholar
Duggal, NA, Upton, J, Phillips, AC, Hampson, P, Lord, JM. NK cell immunesenescence is increased by psychological but not physical stress in older adults associated with raised cortisol and reduced perforin expression. Age (Dordr). 2015 Feb;37(1):9748.Google Scholar
Okun, E, Griffioen, KJ, Mattson, MP. Toll-like receptor signaling in neural plasticity and disease. Trends Neurosci. 2011 May;34(5):269–81.Google Scholar
Tanga, FY, Nutile-McMenemy, N, DeLeo, JA. The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proceedings of the National Academy of Sciences. National Academy of Sciences. 2005 Apr 19;102(16):5856–61.Google Scholar
Hutchinson, MR, Zhang, Y, Brown, K, et al. Non-stereoselective reversal of neuropathic pain by naloxone and naltrexone: involvement of toll-like receptor 4 (TLR4). Eur J Neurosci. 2008 Jul;28(1):20–9.CrossRefGoogle ScholarPubMed
Hutchinson, MR, Zhang, Y, Shridhar, M, et al. Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behavior and Immunity. 2010 Jan;24(1):8395.Google Scholar
Qu, J, Tao, X-Y, Teng, P, et al. Blocking ATP-sensitive potassium channel alleviates morphine tolerance by inhibiting HSP70-TLR4-NLRP3-mediated neuroinflammation. J Neuroinflammation. BioMed Central. 2017 Nov 25;14(1):228.Google Scholar
Hutchinson, MR, Northcutt, AL, Hiranita, T, et al. Opioid Activation of Toll-Like Receptor 4 Contributes to Drug Reinforcement. Journal of Neuroscience. 2012 Aug 15;32(33):11187–200.Google Scholar
Pandey, GN, Rizavi, HS, Ren, X, Bhaumik, R, Dwivedi, Y. Toll-like receptors in the depressed and suicide brain. Journal of Psychiatric Research. 2014 Jun 1;53(C):62–8.Google Scholar
Hung, Y-Y, Kang, H-Y, Huang, K-W, Huang, T-L. Association between toll-like receptors expression and major depressive disorder. Psychiatry Research. 2014 Dec 15;220(1–2):283–6.Google Scholar
Latz, E, Xiao, TS, Stutz, A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013 Jun;13(6):397411.Google Scholar
Alcocer-Gómez, E, de Miguel, M, Casas-Barquero, N, et al. NLRP3 inflammasome is activated in mononuclear blood cells from patients with major depressive disorder. Brain Behavior and Immunity. 2014 Feb;36:111–7.Google Scholar
Zhang, Y, Liu, L, Liu, YZ, et al. NLRP3 Inflammasome Mediates Chronic Mild Stress-Induced Depression in Mice via Neuroinflammation. Int J Neuropsychopharm. 2015 May 29;18(8):pyv0066.Google Scholar
Kim, HK, Andreazza, AC, Elmi, N, Chen, W, Young, LT. Nod-like receptor pyrin containing 3 (NLRP3) in the post-mortem frontal cortex from patients with bipolar disorder: A potential mediator between mitochondria and immune-activation. Journal of Psychiatric Research. 2016 Jan 1;72(C):4350.Google Scholar
Heneka, MT, Kummer, MP, Stutz, A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013 Jan 23;493(7434):674–8.Google Scholar
Iwata, M, Ota, KT, Li, X-Y, et al. Psychological Stress Activates the Inflammasome via Release of Adenosine Triphosphate and Stimulation of the Purinergic Type 2X7 Receptor. Biological Psychiatry. 2016 Jul 1;80(1):1222.Google Scholar
Smith, KJ, Au, B, Ollis, L, Schmitz, N. The association between C-reactive protein, Interleukin-6 and depression among older adults in the community: A systematic review and meta-analysis. Exp Gerontol. 2018 Feb 1;102:109–32.Google Scholar
Valkanova, V, Ebmeier, KP, Allan, CL. CRP, IL-6 and depression: a systematic review and meta-analysis of longitudinal studies. J Affect Disord. 2013 Sep 25;150(3):736–44.Google Scholar
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 Feb;70(2):176–84.Google Scholar
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 Oct;71(10):1121–8.CrossRefGoogle ScholarPubMed
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 Dec 1;171(12):1278–86.Google Scholar
Inoshita, M, Numata, S, Tajima, A, et al. A significant causal association between C-reactive protein levels and schizophrenia. Sci Rep. 2016 May 19;6(1):26105.Google Scholar
Fernandes, BS, Steiner, J, Bernstein, H-G, et al. C-reactive protein is increased in schizophrenia but is not altered by antipsychotics: meta-analysis and implications. Mol Psychiatry. 2016 Apr;21(4):554–64.Google Scholar
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. 2016 Dec;3(12):1147–56.Google Scholar
Khakzad, MR, Javanbakht, M, Shayegan, MR, et al. The complementary role of high sensitivity C-reactive protein in the diagnosis and severity assessment of autism. Research in Autism Spectrum Disorders. 2012 May 5;6(3):1032–7.Google Scholar
Orsini, F, De Blasio, D, Zangari, R, Zanier, ER, De Simoni, M-G. Versatility of the complement system in neuroinflammation, neurodegeneration and brain homeostasis. Front Cell Neurosci. 2014;8:380.Google Scholar
Sekar, A, Bialas, AR, de Rivera, H, et al. Schizophrenia risk from complex variation of complement component 4. Nature. 2016 Feb 11;530(7589):177–83.Google Scholar
Haijma, SV, Van Haren, N, Cahn, W, et al. Brain volumes in schizophrenia: a meta-analysis in over 18 000 subjects. Schizophr Bull. 2013 Sep;39(5):1129–38.Google Scholar
Osimo, EF, Beck, K, Reis Marques, T, Howes, OD. Synaptic loss in schizophrenia: a meta-analysis and systematic review of synaptic protein and mRNA measures. Mol Psychiatry. 2019 Apr;24(4):549–61.Google Scholar
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 Jan;9(1):4656.Google Scholar
Banks, WA. Blood-brain barrier transport of cytokines: a mechanism for neuropathology. Curr Pharm Des. 2005;11(8):973–84.Google Scholar
Krueger, JM, Rector, DM, Roy, S, et al. Sleep as a fundamental property of neuronal assemblies. 2008 Nov 5;9(12):910–9.CrossRefGoogle Scholar
Rachal Pugh, C, Fleshner, M, Watkins, LR, Maier, SF, Rudy, JW. The immune system and memory consolidation: a role for the cytokine IL-1beta. Neurosci Biobehav Rev. 2001 Jan;25(1):2941.Google Scholar
Allan, SM, Tyrrell, PJ, Rothwell, NJ. Interleukin-1 and neuronal injury. Nat Rev Immunol. 2005 Aug;5(8):629–40.Google Scholar
Conductier, G, Blondeau, N, Guyon, A, Nahon, J-L, Rovère, C. The role of monocyte chemoattractant protein MCP1/CCL2 in neuroinflammatory diseases. Journal of Neuroimmunology. 2010 Jul 27;224(1–2):93100.Google Scholar
Eyre, HA, Air, T, Pradhan, A, et al. A meta-analysis of chemokines in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2016 Jul 4;68:18.Google Scholar
Leighton, SP, Nerurkar, L, Krishnadas, R, et al. Chemokines in depression in health and in inflammatory illness: a systematic review and meta-analysis. Mol Psychiatry. 2017 Nov 14;7(1):72–58.Google Scholar
Stuart, MJ, Baune, BT. Chemokines and chemokine receptors in mood disorders, schizophrenia, and cognitive impairment: a systematic review of biomarker studies. Neurosci Biobehav Rev. 2014 May;42:93115.Google Scholar
Haapakoski, R, Mathieu, J, Ebmeier, KP, Alenius, H, Kivimäki, M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behavior and Immunity. 2015 Oct;49:206–15.Google Scholar
Rodrigues-Amorim, D, Rivera-Baltanás, T, Spuch, C, et al. Cytokines dysregulation in schizophrenia: A systematic review of psychoneuroimmune relationship. Schizophrenia Research. 2018 Jul;197:1933.Google Scholar
Miller, GE, Cole, SW. Clustering of depression and inflammation in adolescents previously exposed to childhood adversity. Biological Psychiatry. 2012 Jul 1;72(1):3440.Google Scholar
Baumeister, D, Akhtar, R, Ciufolini, S, Pariante, CM, Mondelli, V. Childhood trauma and adulthood inflammation: a meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-α. Mol Psychiatry. Nature Publishing Group; 2016 May;21(5):642–9.Google Scholar
Musselman, DL, Lawson, DH, Gumnick, JF, et al. Paroxetine for the prevention of depression induced by high-dose interferon alfa. N Engl J Med. 2001 Mar 29;344(13):961–6.Google Scholar
Corvin, A, Morris, DW. Genome-wide Association Studies: Findings at the Major Histocompatibility Complex Locus in Psychosis. Biological Psychiatry. 2014 Feb 15;75(4):276–83.Google Scholar
Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014 Jul 24;511(7510):421–7.Google Scholar
Kebir, H, Kreymborg, K, Ifergan, I, et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med. 2007 Oct;13(10):1173–5.Google Scholar
Reboldi, A, Coisne, C, Baumjohann, D, et al. C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat Immunol. 2009 May;10(5):514–23.Google Scholar
Stromnes, IM, Cerretti, LM, Liggitt, D, Harris, RA, Goverman, JM. Differential regulation of central nervous system autoimmunity by T(H)1 and T(H)17 cells. Nat Med. 2008 Mar;14(3):337–42.Google Scholar
Avgustin, B, Wraber, B, Tavcar, R. Increased Th1 and Th2 immune reactivity with relative Th2 dominance in patients with acute exacerbation of schizophrenia. Croatian Medical Journal. 2005 Apr;46(2):268–74.Google Scholar
Brambilla, P, Bellani, M, Isola, M, et al. Increased M1/decreased M2 signature and signs of Th1/Th2 shift in chronic patients with bipolar disorder, but not in those with schizophrenia. Transl Psychiatry. 2014 Jul 1;4(7):e406–6.Google Scholar
Debnath, M, Berk, M. Th17 pathway-mediated immunopathogenesis of schizophrenia: mechanisms and implications. Schizophrenia Bulletin. Oxford University Press; 2014 Nov;40(6):1412–21.Google Scholar
Ding, M, Song, X, Zhao, J, et al. Activation of Th17 cells in drug naïve, first episode schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2014 Jun 3;51:7882.Google Scholar
Fernandez-Egea, E, Vértes, PE, Flint, SM, et al. Peripheral Immune Cell Populations Associated with Cognitive Deficits and Negative Symptoms of Treatment-Resistant Schizophrenia. PLoS ONE. 2016;11(5):e0155631.Google Scholar
Røge, R, Møller, BK, Andersen, CR, Correll, CU, Nielsen, J. Immunomodulatory effects of clozapine and their clinical implications: what have we learned so far? Schizophrenia Research. Elsevier B.V. 2012 Sep 1;140(1–3):204–13.Google Scholar
Chen, M-L, Tsai, T-C, Lin, Y-Y, et al. Antipsychotic drugs suppress the AKT/NF-kB pathway and regulate the differentiation of T-cell subsets. Immunology Letters. Elsevier B.V. 2011 Oct 30;140(1–2):8191.Google Scholar
Chen, M-L, Tsai, T-C, Wang, L-K, et al. Clozapine inhibits Th1 cell differentiation and causes the suppression of IFN-γ production in peripheral blood mononuclear cells. Immunopharmacol Immunotoxicol. 2012 Aug;34(4):686–94.Google Scholar
O’Sullivan, D, Green, L, Stone, S, et al. Treatment with the antipsychotic agent, risperidone, reduces disease severity in experimental autoimmune encephalomyelitis. PLoS ONE. 2014;9(8):e104430.Google Scholar
Shatz, CJ. MHC class I: an unexpected role in neuronal plasticity. Neuron. 2009 Oct 15;64(1):40–5.Google Scholar
de Graaf, MT, Smitt, PAES, Luitwieler, RL, et al. Central memory CD4+ T cells dominate the normal cerebrospinal fluid. Cytometry B Clin Cytom. 2011 Jan;80(1):4350.Google Scholar
Nikkilä, H, Müller, K, Ahokas, A, et al. Abnormal distributions of T-lymphocyte subsets in the cerebrospinal fluid of patients with acute schizophrenia. Schizophrenia Research. 1995 Feb;14(3):215–21.Google Scholar
Moalem, G, Leibowitz-Amit, R, Yoles, E, et al. Autoimmune T cells protect neurons from secondary degeneration after central nervous system axotomy. Nat Med. 1999 Jan;5(1):4955.Google Scholar
Kipnis, J, Mizrahi, T, Hauben, E, et al. Neuroprotective autoimmunity: naturally occurring CD4+CD25+ regulatory T cells suppress the ability to withstand injury to the central nervous system. Proc Natl Acad Sci USA. National Acad Sciences. 2002 Nov 26;99(24):15620–5.Google Scholar
Kipnis, J, Cardon, M, Avidan, H, et al. Dopamine, through the extracellular signal-regulated kinase pathway, downregulates CD4+CD25+ regulatory T-cell activity: implications for neurodegeneration. J Neurosci. Society for Neuroscience. 2004 Jul 7;24(27):6133–43.Google ScholarPubMed
Walsh, JT, Zheng, J, Smirnov, I, Lorenz, U, Tung, K, Kipnis, J. Regulatory T cells in central nervous system injury: a double-edged sword. J Immunol. 2014 Nov 15;193(10):5013–22.Google Scholar
Kipnis, J, Gadani, S, Derecki, NC. Pro-cognitive properties of T cells. Nat Rev Immunol. 2012 Sep;12(9):663–9.Google Scholar
Dalmau, J, Tüzün, E, Wu, H-Y, et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol. 2007 Jan;61(1):2536.Google Scholar
Lennox, BR, Palmer-Cooper, EC, Pollak, T, et al. Prevalence and clinical characteristics of serum neuronal cell surface antibodies in first-episode psychosis: a case-control study. Lancet Psychiatry. 2017 Jan;4(1):42–8.Google Scholar
Brimberg, L, Mader, S, Jeganathan, V, et al. Caspr2-reactive antibody cloned from a mother of an ASD child mediates an ASD-like phenotype in mice. Mol Psychiatry. 2016 Dec;21(12):1663–71.Google Scholar
Brudek, T, Winge, K, Folke, J, et al. Autoimmune antibody decline in Parkinson’s disease and Multiple System Atrophy; a step towards immunotherapeutic strategies. Mol Neurodegener. 2017 Jun 7;12(1):44.Google Scholar
Iliff, JJ, Wang, M, Liao, Y, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Science Translational Medicine. American Association for the Advancement of Science. 2012 Aug 15;4(147):147ra1111.Google Scholar
Louveau, A, Smirnov, I, Keyes, TJ, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015 Jun 1;523(7560):337–41.Google Scholar
Aspelund, A, Antila, S, Proulx, ST, et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med. 2015 Jun 29;212(7):991–9.Google Scholar
Da Mesquita, S, Fu, Z, Kipnis, J. The Meningeal Lymphatic System: A New Player in Neurophysiology. Neuron. 2018 Oct 24;100(2):375–88.Google Scholar
Louveau, A, Da Mesquita, S, Kipnis, J. Lymphatics in neurological disorders: a neuro-lympho-vascular component of multiple sclerosis and Alzheimer’s disease? Neuron. Elsevier Inc. 2016 Sep 7;91(5):957–73.Google Scholar
Harrison, NA, Brydon, L, Walker, C, et al. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biological Psychiatry. 2009 Sep 1;66(5):407–14.Google Scholar
Harrison, NA, Doeller, CF, Voon, V, Burgess, N, Critchley, HD. Peripheral Inflammation Acutely Impairs Human Spatial Memory via Actions on Medial Temporal Lobe Glucose Metabolism. Biological Psychiatry. 2014 Oct 1;76(7):585–93.Google Scholar
Tracey, KJ. Reflex control of immunity. Nat Rev Immunol. 2009 Jun;9(6):418–28.Google Scholar
Bellinger, DL, Millar, BA, Perez, S, et al. Sympathetic modulation of immunity: Relevance to disease. Cell Immunol. 2008;252(1–2):2756.Google Scholar
Sloan, EK, Capitanio, JP, Tarara, RP, et al. Social Stress Enhances Sympathetic Innervation of Primate Lymph Nodes: Mechanisms and Implications for Viral Pathogenesis. Journal of Neuroscience. 2007 Aug 15;27(33):8857–65.Google Scholar
Pacheco, R, Riquelme, E, Kalergis, AM. Emerging evidence for the role of neurotransmitters in the modulation of T cell responses to cognate ligands. Cent Nerv Syst Agents Med Chem. 2010 Mar;10(1):6583.Google Scholar
Ilani, T, Ben-Shachar, D, Strous, RD, et al. A peripheral marker for schizophrenia: Increased levels of D3 dopamine receptor mRNA in blood lymphocytes. Proc Natl Acad Sci USA. National Acad Sciences. 2001 Jan 16;98(2):625–8.Google Scholar
Brito-Melo, GEA, Nicolato, R, de Oliveira, ACP, et al. Increase in dopaminergic, but not serotoninergic, receptors in T-cells as a marker for schizophrenia severity. Journal of Psychiatric Research. 2012 Jun 1;46(6):738–42.Google Scholar
Nagai, Y, Ueno, S, Saeki, Y, et al. Decrease of the D3 dopamine receptor mRNA expression in lymphocytes from patients with Parkinson’s disease. Neurology. 1996 Mar 1;46(3):791–5.Google Scholar
Thomson, CA, McColl, A, Cavanagh, J, Graham, GJ. Peripheral inflammation is associated with remote global gene expression changes in the brain. J Neuroinflammation. 2014 Apr 8;11(1):73.Google Scholar
Miller, AH, Raison, CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nature. 2016 Jan;16(1):2234.Google Scholar
Harrison, NA. Brain Structures Implicated in Inflammation-Associated Depression. Curr Top Behav Neurosci. 2017;31:221–48.Google Scholar
Capuron, L, Ravaud, A, Neveu, PJ, et al. Association between decreased serum tryptophan concentrations and depressive symptoms in cancer patients undergoing cytokine therapy. Mol Psychiatry. 2002 Jun 18;7(5):468–73.CrossRefGoogle ScholarPubMed
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 May;26(5):643–52.Google Scholar
Krause, D, Myint, A-M, Schuett, C, et al. High Kynurenine (a Tryptophan Metabolite) Predicts Remission in Patients with Major Depression to Add-on Treatment with Celecoxib. Front Psychiatry. 2017;8:16.Google Scholar
Abbasi, S-H, Hosseini, F, Modabbernia, A, Ashrafi, M, Akhondzadeh, S. Effect of celecoxib add-on treatment on symptoms and serum IL-6 concentrations in patients with major depressive disorder: randomized double-blind placebo-controlled study. J Affect Disord. 2012 Dec 10;141(2–3):308–14.Google Scholar
O’Connor, JC, Lawson, MA, André, C, et al. Induction of IDO by bacille Calmette-Guérin is responsible for development of murine depressive-like behaviour. J Immunol. 2009 Mar 1;182(5):3202–12.Google Scholar
Figure 0

Figure 1.1 Schematic representation of the Blood Brain Barrier and the blood cerebrospinal fluid barrier (BCSFB). A. The BBB consists of highly specialized endothelial cells joined by tight junctions. They deposit an endothelial basement membrane (yellow) in which pericytes are embedded. The parenchymal basement membrane (orange) is deposited by astrocytes and together with astrocytic endfeet forms the glia limitans perivascularis. The endothelial layer prevents the unrestricted movement of large solutes, antibodies and immune cells while allowing the passage of smaller solutes, cytokines and other proteins through dedicated transport systems.B. The BCSFB is made up by the choroid plexus epithelial cells which similarly prevent the unrestricted passage of molecules between the blood and the CSF. Central memory T cells are the predominant immune population in the CSF, and it is thought that they may cross the BCSFB, although the molecular mechanisms behind this are still unclear.

Figure 1

Figure 1.2 Developmental origins of immune cells.Lymphocytes and some dendritic cells derive from the common lymphoid progenitor cell line. CD4+ T cells can differentiate into T helper cells with specific functions, defined by their cytokine profiles. Activated B cells give rise to antibody secreting plasma cells. Granulocytes (mast cells, neutrophils, eosinophils and basophils), monocytes, some dendritic cells and microglia derive from the common myeloid progenitor cell line. However, microglia are established in the brain prenatally during development from yolk sac progenitors and are transcriptionally distinct from other myeloid cells. Monocytes give rise to macrophages, some in peripheral tissues and others in the CNS.

Figure 2

Figure 1.3 CNS lymphatic and glymphatic systems.A. Schematic of the glymphatic system. CSF enters the brain parenchyma along paraarterial routes, exchanges with interstitial fluid and is cleared along paravenous routes. Convective flow is facilitated by AQP4 pores which are abundantly expressed on astrocytic endfeet. Clearance of solutes, proteins and waste products such as amyloid-β occur along this pathway. Draining fluid may be dispersed into the subarachnoid CSF, venous circulation or lymphatics.B. Schematic representation of lymphatic drainage of the brain. Meningeal lymphatic vessels (green) adjacent to the venous sinuses and arteries drain molecules and meningeal immune cells in the CSF into the deep cervical lymph nodes. Adapted with permission from Louveau et al. Neuron. 2016; 91:957–73 (109).

Figure 3

Figure 1.4 The enzymatic activity of IDO is enhanced during inflammation and diverts tryptophan metabolism away from serotonin synthesis and towards the kynurenine pathway. Kynurenine is readily transported across the BBB where it can be metabolized to 3-hydroxykynurenine (3-HK), quinolinic acid (QA) or kynurenic acid (KA). 3-HK can increase oxidative stress and may be neurotoxic. QA is preferentially produced by microglia and is an NMDA-receptor agonist which may be neurotoxic, whereas KA is produced by astrocytes and is an NMDA-receptor antagonist which may be neuroprotective. In the periphery KA is thought to modulate immune cell phenotypes. Impaired glutamatergic and serotonergic neurotransmission are both associated with the development of depressive behaviour.

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×