Hostname: page-component-78c5997874-xbtfd Total loading time: 0 Render date: 2024-11-05T05:55:20.139Z Has data issue: false hasContentIssue false

Inflammatory neuropsychiatric disorders and COVID-19 neuroinflammation

Published online by Cambridge University Press:  30 April 2021

Siu Wa Tang*
Affiliation:
University of California, Irvine, CA, USA
Daiga Helmeste
Affiliation:
University of California, Irvine, CA, USA
Brian Leonard
Affiliation:
National University of Ireland, Galway, Ireland
*
Author for correspondence: Siu Wa Tang, Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Neuropsychiatric sequalae to coronavirus disease 2019 (COVID-19) infection are beginning to emerge, like previous Spanish influenza and severe acute respiratory syndrome episodes. Streptococcal infection in paediatric patients causing obsessive compulsive disorder (PANDAS) is another recent example of an infection-based psychiatric disorder. Inflammation associated with neuropsychiatric disorders has been previously reported but there is no standard clinical management approach established. Part of the reason is that it is unclear what factors determine the specific neuronal vulnerability and the efficacy of anti-inflammatory treatment in neuroinflammation. The emerging COVID-19 data suggested that in the acute stage, widespread neuronal damage appears to be the result of abnormal and overactive immune responses and cytokine storm is associated with poor prognosis. It is still too early to know if there are long-term-specific neuronal or brain regional damages associated with COVID-19, resulting in distinct neuropsychiatric disorders. In several major psychiatric disorders where neuroinflammation is present, patients with abnormal inflammatory markers may also experience less than favourable response or treatment resistance when standard treatment is used alone. Evidence regarding the benefits of co-administered anti-inflammatory agents such as COX-2 inhibitor is encouraging in selected patients though may not benefit others. Disease-modifying therapies are increasingly being applied to neuropsychiatric diseases characterised by abnormal or hyperreactive immune responses. Adjunct anti-inflammatory treatment may benefit selected patients and is definitely an important component of clinical management in the presence of neuroinflammation.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2021. Published by Cambridge University Press on behalf of Scandinavian College of Neuropsychopharmacology

Summations

  • This review summarises the evidence that both acute and chronic inflammation may have significant neuropsychiatric sequalae.

  • Although inflammation is present in patients suffering from a number of major neuropsychiatric disorders, factors determining the specific neuronal vulnerability are still unknown.

  • Patients suffering from some major neuropsychiatric disorders with abnormal inflammatory markers may experience less than favourable response or treatment resistance when standard treatment is used alone.

  • Managing the inflammation with anti-inflammatory agents, disease-modifying therapies, and modulation of the hyperactive immune response are becoming an important part of clinical management of neuropsychiatric disorders where inflammation is present.

Considerations

  • Inflammation could be beneficial or harmful. Suppressing overactive immune response has been found to be important to prevent inflammatory-associated damages in COVID-19 but could be harmful in other inflammation, such as viral influenza.

  • Inflammation in different neuropsychiatric disorders may require different anti-inflammatory management protocols.

  • Results from well-designed clinical trials to test the efficacy of anti-inflammatory agents or immune modulatory therapies, alone or in combination with standard treatment, in each major neuropsychiatric disorder, are not available yet.

Introduction

Recent reports on serious neuropsychiatric sequalae in coronavirus disease 2019 (COVID-19), SARS, and streptococcal infection in paediatric patients show the important role of the immune system in maintaining health and in disorders of the central nervous system (CNS). Presence of neuroinflammation in neurodegenerative disorders and affective disorders is already well documented and has been extended to include schizophrenia, obsessive compulsive disorder (OCD), post-traumatic stress disorder (PTSD), other anxiety disorders, and neuropsychiatric diseases in recent years. If systemic or regional brain inflammation may indeed inflict damage to neurocircuits or neurons, then the factors determining the susceptibility, selectivity, and vulnerability of the neuro-targets should be determined. Clinically, it is important to know if anti-inflammatory medications should be considered in all patients as neuroprotective, such as in COVID-19, or as adjunct treatment, such as in major depression, or only in specific cases. In addition to common anti-inflammatory agents, disease-modifying therapies (DMTs) are becoming an important and novel approach in modulating neuropsychiatric disorders with an inflammatory component. We review the literature on neuroinflammation and management issues associated with major neuropsychiatric disorders.

Method

We searched the English language literature, including foreign language publications with informative abstracts in English, up to November 30, 2020, using PubMed (www.ncbi.nlm.nih.gov), crossing the keyword inflammation and neuroinflammation, respectively, in turn with the following words: COVID-19, psychiatry, psychosis, psychiatric disorders, bipolar disorders, OCD, anxiety disorders, dementia, neuron death, neurodegeneration, brain circuits, neurotransmitters, brain areas, serotonin (5HT), dopamine (DA), glutamine (Glu), cholinergic (ACh), adenosine, herbs, plants, gastrointestinal disorders, gut microbes, probiotics, fecal transplant, and treatment. Manuscripts were identified and then included in this review after evaluation of quality of research and data, and relevancy to our search.

Inflammation in neuropsychiatric disorders

Acute inflammation

Coronavirus disease 2019 (COVID-19)

COVID-19 is an excellent example of neuropsychiatric complications in acute neuroinflammation. The previous Spanish flu epidemic of 1918–1919, the severe acute stress respiratory syndrome (SARS) and the Middle East respiratory syndrome showed that the damage from these virus infections of the respiratory tract is not limited to the lung. Immunopathological studies on a cohort of COVID-19 patients in the Netherlands in the first 3 months of 2020 reported that an extensive inflammatory response was present not only in the lungs, but also in the heart, liver, kidney, and the brain (Schrink et al., 2020). In the brain itself, extensive inflammation was seen in the olfactory bulbs and medulla oblongata, reflecting the loss of smell and many of the CNS symptoms (Banerjee and Vikswanath, Reference Banerjee and Viswamath2020). The COVID-19 virus may enter through the angiotensin-converting enzyme two receptors (ACE-2) which are present on endothelial cells of cerebral vessels (Garg Reference Garg2020) and also likely by crossing the damaged blood–brain barrier (BBB), which is highly susceptible to peripheral immune changes (Schwartz et al., Reference Schwartz, Kipris, Rivets and Prat2013) and certainly under the characteristic cytokine storm of COVID-19.

Many cases of neuropsychiatric disorders have now been reported in COVID-19 patients, less than 1 year since the pandemic started (Cothran et al., Reference Cothran, Kellman, Singh, Beck, Powell, Bolton and Tam2020; Huang and Zhao Reference Huang and Zhao2020; Rogers et al., Reference Rogers, Chesney, Oliver, Pollak, McGuire, Fusar-Poli, Zandi, Lewis and David2020; Troyer et al., Reference Troyer, Kohn and Hong2020). Neurological manifestations occur early in the illness (Orsucci et al., Reference Orsucci, Ienco, Nocita, Napolitano and Vista2020). Delirium, from hypoxia and metabolic abnormalities (Garg Reference Garg2020), particularly occurred in the more vulnerable aged population and those with dementia (Butler et al.,Reference Butler, Pollak, Rooney, Michael and Nicholson2020; Mcloughlan et al.,2020). Acute neuropsychiatric symptoms which include altered mental status, psychosis, and suicidal ideation (Chacko et al., Reference Chacko, Job, Caston, George, Yacoub and Cáceda2020; Correa-Palacio et al., Reference Correa-Palacio, Hernandez-Huerta, Gómez-Arnau, Loeck and Caballero2020; Ferrando et al., Reference Ferrando, Klepacz, Lynch, Shahar, Dornbush, Smiley, Miller, Tavakkoli, Regan and Bartell2020; Finatti et al., Reference Finatti, Pigato, Pavan, Toffanin and Favaro2020; Ng et al., Reference Ng, Yeo, Lim and Chee2020; Sher Reference Sher2020; Valdés-Florido et al., Reference Valdés-Florido, López-Díaz, Palermo-Zeballos, Martínez-Molina, Martín-Gil, Crespo-Facorro and Ruiz-Veguilla2020) were the second most common presentation. Encephalopathy or encephalitis occurred in younger patients as well (Varatharaj et al., Reference Varatharaj, Thomas, Ellul, Davies, Pollak, Tenorio, Sultan, Easton, Breen, Zandi, Coles, Manji, Al-Shahi Salman, Menon, Nicholson, Benjamin, Carson, Smith, Turner, Solomon, Kneen, Pett, Galea, Thomas and Michael2020). An increase of stress response is reflected in the low mood, anxiety, and severe fatigue, which may persist in about 20% of patients following their apparent full recovery (Rogers et al.,Reference Rogers, Chesney, Oliver, Pollak, McGuire, Fusar-Poli, Zandi, Lewis and David2020; Sterdolt and Verkhratsky 2020). PTSD also occurred (Mazza et al., Reference Mazza, De Lorenzo, Conte, Poletti, Vai, Bollettini, Melloni, Furlan, Ciceri, Rovere-Querini and Benedetti2020), as well as Guillain-Barré syndrome (Garg Reference Garg2020; Webb et al., Reference Webb, Wallace, Martin-Lopez and Yogarajah2020) and other forms of neuropathy and myopathy (Ottaviani et al., Reference Ottaviani, Boso, Tranquillini, Gapeni, Pedrotti, Cozzio, Guarrera and Giometto2020). Severe and debilitating fatigue and myalgia could be present, and elevated creatine kinase levels indicate serious myopathy (Garg Reference Garg2020; Orsucci et al., Reference Orsucci, Ienco, Nocita, Napolitano and Vista2020). Cognitive defects (Troyer et al., Reference Troyer, Kohn and Hong2020; Zhou et al, Reference Zhou, Lu, Chen, Wei, Wang, Lyu, Shi and Hu2020) may persist for many months after apparent recovery. The damage to neuronal networks initiated by the COVID-19 virus and sustained by the chronic inflammation and disruption of metabolic homoeostasis is likely to result in the long-term CNS disabilities, and the term “long COVID” has been applied in the UK and Ireland.

Both acute and long-term neuropsychiatric consequences of viral infections have been documented historically (Davydow et al., Reference Davydow, Desai, Needham and Bienvenu2008). These include schizophrenia cases in the 1918 Spanish flu (Kępińska et al., Reference Kępińska, Iyegbe, Vernon, Yolken, Murray and Pollak2020), depression, anxiety, and PTSD cases in SARS (Mak et al., Reference Mak, Chu, Pan, Yiu and Chan2009), and Parkinson’s symptoms with H5N1 influenza (Henry et al., Reference Henry, Smeyne, Jang, Miller and Okun2010).

The immediate impact of the COVID-19 virus is attributed to its binding to the widely distributed ACE-2, which is distributed along the respiratory and gastrointestinal epithelium as well as endothelial cell surfaces. The spread of the virus into the brain occurs following a high viral load coupled with susceptibility due to the age of the patient, abnormal or over-reactive immune function, chronic medical illness, and frequently a history of neurotropic virus infections (Razanamahery et al., Reference Razanamahery, Malinowski, Humbert, Brunel, Lepiller, Chirouze and Bouiller2020; Singhou Reference Singhou2020). The spread of the virus through the brain is heterogeneous but in experimental studies it has been shown to rapidly infect the olfactory bulbs (approximately after 4 days) and later the piriform cortex (Perlman et al, Reference Perlman, Evans and Afift2020). The remainder of the cortex, hypothalamus, basal ganglia, and brain stem are affected later. Microglia are important regulators of COVID-19 expression in the brain since their ablation results in increased viral loads, 7–8 days post-infection. Neurons appear to be the main targets of infection in vivo (Mangale et al., Reference Mangale, Syage, Ekiz, Skinner, Cheng, Stone, Brown, O’Connell, Green and Lane2020). As ACE-2 receptors are expressed in the olfactory lining, a main target for COVID-19, this likely accounts for the anosmia and hypo-osmia experienced early in the infection, which occurs with a frequency of 12–32% (Lechien et al, Reference Le chien, Chiesa-Estomba, De Siati, Horoi, Le Bon, Rodriguez, Dequanter, Blecic, El Afia, Distinguin, Chekkoury-Idrissi, Hans, Delgado, Calvo-Henriquez, Lavigne, Falanga, Barillari, Cammaroto, Khalife, Leich, Souchay, Rossi, Journe, Hsieh, Edjlali, Carlier, Ris, Lovato, De Filippis, Coppee, Fakhry, Ayad and Saussez2020), and loss of smell results from the neurodegenerative effect of the virus on the olfactory bulbs. Dinein and kinesin have been identified as the proteins responsible for the transmission of the virus and the nucleus solitarius of the brain stem is preferentially affected (Wu et al.,Reference Wu, Xu, Chen, Duan, Hashimoto, Yang, Liu and Yang2020), accounting for the central effects of the virus on breathing.

C-reactive protein (CRP) is a commonly used early marker to grade the severity of systemic inflammation from infection (Nehring et al., Reference Nehring, Goyal, Bansal and Patel2020). Relatively mild to moderate elevations are seen in obesity, diabetes, depression, periodontitis, sedentary lifestyle and cigarette smoking rheumatoid arthritis, myocardial infarction, pancreatitis, and bronchitis, but marked and severe elevations in CRP (more than 10.0 mg/dL and >50.0 mg/dL, respectively) require acute bacterial or viral infections, systemic vasculitis or major trauma. In early-stage COVID-19, CRP levels show a positive correlation with lung lesions and disease severity (Wang, Reference Wang2020). Importantly, only a subset of COVID-19 patients shows severe elevations in CRP, for which there appears to be emerging evidence of genetic susceptibility (Zeberg and Pääbo, Reference Zeberg and Pääbo2020).

The vulnerability of the elderly to severe COVID-19 infection is well known. Ageing is usually associated with overall neurodegenerative changes and diminished homoeostatic brain mechanisms. In ageing, synaptic plasticity decreases, brain metabolism is reduced, and the vulnerability to exogenous toxins is increased. The increases in specific pro-inflammatory cytokines have been correlated with specific symptoms of the virus infection. For example, IL-1 beta has been linked to depression in later life, IL-6 with anhedonia and suicidal behaviour, while tumour necrosis factor (TNF)-alpha and IL-2 have been linked to apathy and motor inhibition (Thomas Reference Thomas2005; Schmidt 2016).

Apart from psychological support and treatment of accompanying neuropsychiatric disorders such as anxiety, depression, psychosis, and neuropathic and myopathic pain (Drożdżal et al., Reference Drożdżal, Rosik, Lechowicz, Machaj, Szostak, Majewski, Rotter and Kotfis2020), management of the neuroinflammation is important. Intravenous immunoglobulins (Novak Reference Novak2020) and careful steroidal immunosuppressant therapy appear to be useful in some cases to avoid complications (Rajabally et al., Reference Rajabally, Goedee, Attarian and Hartung2020). A recent report summarised the recommendations from the Italian Societies of Neurology, Cinical Neurophysiology, and Peripheral Nervous System Association in the management of COVID-19 immune-mediated neuropathies (Dubbioso et al., Reference Dubbioso, Nobile-Orazio, Manganelli, Santoro, Briani, Cocito, Tedeschi, Di Lazzaro and Fabrizi2020).

Because of the very recent occurrence of the pandemic, much still has to be learned about the chronic effects of the virus on the brain and behaviour. However, it is already apparent that its impact, both direct and indirect, could result in neuroprogressive changes (as seen in Parkinson’s disease (PD)) and long-term neuropsychiatric consequences. Increased vigilance for all neuropsychiatric symptoms in patients with COVID-19 is certainly warranted (Brown et al., Reference Brown, Gray, Lo Monaco, O’Donoghue, Nelson, Thompson, Francey and McGorry2020). Minimising the relevance of the psychiatric aspects of COVID-19 and mistakenly explaining that “sometimes an abnormal behavior in an abnormal situation is a normal behavior” could be an unforgivable mistake (Steardo and Verkhratsky, 2020).

Autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS)

Compared to the multiple neuropsychiatric consequences of COVID-19 infection, acute neuroinflammation causing a specific psychiatric disorder is highlighted in PANDAS (Frick and Pittenger Reference Frick and Pittenger2016). PANDAS is a medical emergency, in which sudden and severe OCD, with cognitive and behavioural symptoms, developed in children after streptococcal infection. The exact neuro-mechanism is still unclear. It is not known whether serotonin (5HT) or other types of neurons were involved or damaged (Chiarello et al., 2017; Jasper-Fayer et al., 2017), but there is evidence suggesting that human anti-brain autoantibodies induced by Streptococcus pyogenes infections target DA receptors (Cox et al., Reference Cox, Sharma, Leckman, Zuccolo, Zuccolo, Kovoor, Swedo and Cunningham2013; Cunningham and Cox Reference Cunningham and Cox2016; Orefici et al., Reference Orefici, Cardona, Cox, Cunningham, Ferretti, Stevens and Fischetti2016). Acute infections in adults have not been reported to cause OCD, but depression is a fairly common consequence, as reported in influenza (Bornand et al. 2016). Acute systematic inflammation induced by endotoxin also may precipitate sad mood (Benson et al., Reference Benson, Brinkhoff, Lueg, Roderigo, Kribben, Wilde, Witzke, Engler, Schedlowski and Elsenbruch2017) and exacerbation of schizophrenic symptoms has been reported with oseltamivir administration (Lan et al., Reference Lan, Liu and Chen2015). Penicillin, azithromycin, intravenous immunoglobulin, plasma exchange, tonsillectomy, cognitive behaviour therapy, non-steroidal anti-inflammatory drug (NSAID), and corticosteroids (CSs) have been used in the treatment of PANDAS. The rationale and efficacy of antibiotics and immunomodulatory therapies have been reviewed and discussed (Burchi and Pallanti Reference Burchi and Pallanti2018; Sigra et al., Reference Sigra, Hesselmark and Bejerot2018).

Traumatic brain injury (TBI)

Acute physical trauma to the brain, as in boxing and after head injury, triggers a reactive inflammatory process. Though inflammation serves defence and neuroprotective purposes in the early stages when the brain sustains physical injuries, it is a double-edged sword (Loane and Kumar Reference Loane and Kumar2016; Konoshi and Kiyama Reference Konishi and Kiyama2018). The consequences of inflammation are dictated by multiple local and systematic cues of the host. It is hard to predict which direction the multiple processes will end up, but the results could be quite different. In physical head injuries, hyperactivation of glutaminergic (Glu) neurotransmission after injury may result in neuronal death. Amantadine is neuroprotective, supposedly through its N-methyl-D-aspartate receptor (NMDAR) antagonist action. The antioxidants glutathione and N-acetylcysteine reduce brain damage and improve recovery, glial limitans breakdown, and parenchymal cell death by up to approximately 70%, suggesting that reactive oxidative stress may be a primary inducer of cell death and inflammation after focal brain injury (Corps et al. Reference Corps, Roth and McGavern2015). The benefit of NSAIDs is controversial as they block both the tissue damaging and repair-promoting aspects of inflammation. Likewise, the benefit of systemic glucocorticoid treatment in COVID-19 patients is not straightforward (Keller et al., Reference Keller, Kitsis, Arora, Chen, Agarwal, Ross, Tomer and Southern2020). Glucocorticoid administration appears beneficial when CRP levels are high (greater than 20.0 mg/dL) but harmful when CRP levels are low (less than 10 mg/dL), perhaps due to the association between glucocorticoid use and delayed viral clearance (Keller et al., Reference Keller, Kitsis, Arora, Chen, Agarwal, Ross, Tomer and Southern2020).

In traumatic brain injury (TBI), salsalate, an unacetylated salicylate, has been found to be effective, through blockade of NF-κB, pro-inflammatory gene expression and nitrite secretion by microglia, increasing expression of genes associated with neuroprotection and neurogenesis, including neuropeptides, oxytocin, and thyrotropin-releasing hormone (Lagraoui et al. Reference Lagraoui, Sukumar, Latoche, Maynard, Dalgard and Schaefer2017). Apart from these small molecules, cell-based therapeutics are being pursued as well, to manipulate the immune response into a neuroprotective direction (Xiong et al. Reference Xiong, Mahmood and Chopp2018). Apart from neurodegenerative disorders triggered by chronic neuroinflammation post-TBI, it is important to mention that about 1/3 of ischemic stroke patients with neuroinflammation also end up with post-stroke depression. Low mental health literacy in many elderly subjects may result in patients not being able to voice the mental symptoms properly, making a diagnosis difficult (Lee et al., Reference Lee, Tang, Leung, Yu and Cheung2009). Though there were some studies on the relationship between brain areas affected by inflammation and the subsequent development of depression, the specific neurocircuits affected are unclear, and anti-inflammatory measures again seemed to be beneficial in these patients (Villa et al. Reference Villa, Ferrari and Moretti2018).

The cause for the widespread CNS damage following acute neuroinflammation, whether it is from viral or bacterial infections, or physical trauma, appears to be an acute activation and subsequent dysregulation of the immune system (Becher et al., Reference Becher, Bechmann and Greter2006; Rostami and Ciric 2013; Kostic et al., Reference Kostic, Stojanovic, Marjanovic, Zivkovic and Cvetanovic2015), especially the eicosanoids and cytokine systems (cytokine and eicosanoid storms). Cytokine activations in inflammation has been well studied. Eicosanoids and related bioactive lipid mediators derived from polyunsaturated fatty acids were viewed as pro-inflammatory until recently when unique eicosanoids and related docosanoids with anti-inflammatory and pro-resolution functions were discovered (Dennis and Norris Reference Dennis and Norris2015).

Autoimmune encephalitis includes those with systematic lupus erythematosus (Bendorius et al., Reference Bendorius, Po, Muller and Jeltsch-David2018), cases of autoimmune anti-NMDAR antibodies (Kayser and Dalmau Reference Kayser and Dalmau2016), or anti-GABA (γ-aminobutyric acid) receptor antibodies. Regarding anti-GABA antibody encephalitis (Dalmau Reference Dalmau2017), there may be differences in vulnerability in terms of age, as the antibodies in childhood cases appeared to be more viral-related while adult cases are more tumour-related (Spatola et al., Reference Spatola, Petit-Pedrol, Simabukuro, Armangue, Castro, Barcelo Artigues, Julià Benique, Benson, Gorman, Felipe, Caparó Oblitas, Rosenfeld, Graus and Dalmau2017), highlighting the age effect on vulnerability of inflammation again. Kawasaki disease is a good example of age-dependent vulnerability. The excessive inflammatory response is seen in a minority of preschool infants, with Asian ancestry being a suspected vulnerability factor, while adults in the same household rarely experience anything more serious than transient conjunctivitis (Chen et al, 2018; personal communication).

In summary, there are neuropsychiatric consequences following acute neuroinflammation. Apart from the potency of the causative agent such as COVID-19, or SARS, or streptococci bacteria, and co-existing medical disorders such as diabetes and cardiovascular disorders, age appears to be an important factor for susceptibility. Other factors, including genotype, determine the pro- or anti-inflammatory outcome of initial immune responses, selectivity of neuroinflammatory insult, neuronal vulnerability, and the neuropsychiatric consequences.

Chronic neuroinflammation

Chronic inflammation is defined here as inflammation which lasts for long durations, usually years. Acute inflammations, such as viral infections and TBI, may trigger reactions that turn chronic. For example, dementia may result from chronic neuroinflammation after physical traumatic injuries in boxing. Chronic microglial activation and sustained immune response are well known in neurological disorders such as Alzheimer’s disease (AD), PD, and multiple sclerosis (MS) (Pulli and Chen Reference Pulli and Chen2014; Hoehn Reference Hoehn2015; Albrecht et al., Reference Albrecht, Granziera, Hooker and Loggia2016; Hagens et al., Reference Hagens, van Berckel and Barkhof2016; Bevan Jones et al., Reference Bevan-Jones, Surendranathan, Passamonti, Vázquez Rodríguez, Arnold, Mak, Su, Coles, Fryer, Hong, Williams, Aigbirhio, Rowe and O’Brien2017; Lagarde et al., Reference Lagarde, Sarazin and Bottlaender2018; Tommasin et al., 2019). In MS, myelin-specific CD4(+) T cells, activated in the periphery, infiltrate the CNS and start an inflammatory cascade by secreting cytokines and chemokines (Rostami and Ciric 2013), followed by BBB disruption, demyelination, and neurodegeneration (Kostic et al., Reference Kostic, Stojanovic, Marjanovic, Zivkovic and Cvetanovic2015). Semi-acute factors which may create chronic inflammatory reactions include oxidative loads, toxic metals such as aluminium, copper, and iron, nutritional factors, dietary or environmental toxins, and contaminants such as insecticides (Yegambaram et al., Reference Yegambaram, Manivannan, Beach and Halden2015).

Dementia and ACh neurons

ACh neurons appear to be particularly vulnerable to neuroinflammation. ACh neuronal deaths in the basal forebrain occur early in AD (Whitehouse et al., Reference Whitehouse, Price, Clark, Coyle and DeLong1981) but not in normal ageing (McQuail et al., Reference McQuail, Riddle and Nicolle2011). On the other hand, the ACh system has been discovered to exert significant modulatory action on the immune system (Gatta et al., Reference Gatta, Mengod, Reale and Tata2020; Hoover Reference Hoover2017). Muscarinic and nicotinic receptors stimulate pro- and anti-inflammatory cytokines, respectively, to modulate the immune/inflammatory responses (Di Bari et al., Reference Di Bari, Di Pinto, Reale, Mengod and Tata2017). Acetylcholine esterase inhibitors (AChI) or vagal nerve stimulation modulate neuroinflammation via the α7 nicotinic acetylcholine receptor (Treinin et al., Reference Treinin, Papke, Nizri, Ben-David, Mizrachi and Brenner2017).

PD and DA neurons

The susceptibility or vulnerability of DA neurons to neuro- and peripheral inflammation is demonstrated in PD (Matheoud et al., Reference Matheoud, Cannon, Voisin, Penttinen, Ramet, Fahmy, Ducrot, Laplante, Bourque, Zhu, Cayrol, Le Campion, McBride, Gruenheid, Trudeau and Desjardins2019). DA-dependent motor function being easily testable, urinary tract, and chest infections have been reported to cause transient worsening of PD symptoms. Neuromelanin is a catecholamine-derived pigment in DA neurons of the substantia nigra and also in norepinephrine neurons of the locus coeruleus. They are both known to be damaged by inflammation in PD. A close relationship has been described between iron, DA, and neuromelanin. Excess iron or DA is toxic. Excess DA is converted into neuromelanin, and excess iron is chelated by neuromelanin, which also removes pesticides and some other oxidants. Microglia become activated when this DA–iron neuromelanin balance is lost (Fedorow et al., Reference Fedorow, Tribl, Halliday, Gerlach, Riederer and Double2005; Haining and Achat-Mendes Reference Haining and Achat-Mendes2017), resulting in neuronal death. In PD, it is the neurons containing neuromelanin that degenerate (Zecca et al., Reference Zecca, Zucca, Wilms and Sulzer2003, Reference Zecca, Casella, Albertini, Bellei, Zucca, Engelen, Zadlo, Szewczyk, Zareba and Sarna2008; Zucca et al., Reference Zucca, Segura-Aguilar, Ferrari, Muñoz, Paris, Sulzer, Sarna, Casella and Zecca2017). Similar to the relationship between ACh neurons and the immune system, dysregulation of DA transmission may also induce dysfunction in the immune system (Vidal and Pacheco Reference Vidal and Pacheco2019). Astrocyte DA D2 receptor (DRD2) was also shown to modulate immunity through αB-crystallin (CRYAB), which is part of the small heat shock protein family and is anti-neuroinflammatory (Shao et al., Reference Shao, Zhang, Tang, Zhang, Zhou, Yin, Zhou, Huang, Liu, Wawrousek, Chen, Li, Xu, Zhou, Hu and Zhou2013; Zhang et al., Reference Zhang, Chen, Wu, Manaenko, Yang, Tang, Fu and Zhang2015)

Affective disorders and NMDARs (glutamate)

Chronic neuroinflammation appeared to be present in some patients with affective disorders. Transition into depression in these patients has been attributed to the activation of the toxic kynurenine metabolic pathway, resulting in the formation of toxic quinolinic acid, an NMDAR agonist (Leonard Reference Leonard2010, Reference Leonard2015, Reference Leonard2017, 2018; Müller Reference Müller2010; Müller and Schwarz Reference Müller and Schwarz2006, Reference Müller and Schwarz2007; Dantzer and Walker Reference Dantzer and Walker2014; Won and Kim Reference Won and Kim2016; Dantzer Reference Dantzer2017). Bringing in this inflammatory toxicity factor, an integrated toxic brain hypothesis of depression (Tang et al., 2017a) would supplement the other two hypotheses of depression, namely the amine hypothesis (Bunney Reference Bunney1975) and the stress hypercortisolemia hypothesis (Duman and Monteggia Reference Duman and Monteggia2006). In addition to the neurotoxic effects on neurons and astroglia cells, quinolinic acid is also an important substrate for nicotinamide adenine dinucleotide (NAD+), a key component of the electron transport system. As discussed by Leonard (2018), in severe depression, the synthesis of NAD+ is reduced. Unlike non-nervous tissues, brain energy metabolism is largely dependent on glucose and the transport of glucose across the BBB is an insulin-dependent process. As the resistance of the insulin receptors is increased in the inflammatory state associated with severe depression, the transport of glucose into the brain is compromised. The concurrent increase in the activity of superoxide dismutase and the rise in reactive oxygen species further compromise the integrity of neurons and, in addition, damage the mitochondria (Burkunina et al., Reference Burkunina, Pariante and Zunszain2015). As a result of the damage to the mitochondria, the synthesis of ATP and other high-energy intermediates is decreased. These metabolic changes caused by the neurotoxins and metabolic stress help to understand the causes of the neurodegenerative changes associated with chronic depression, particularly in elderly patients.

OCD and anxiety disorders

Recently, heightened inflammatory activities have been described in patients suffering from PTSD, fear, and anxiety disorders (Frick and Pittenger Reference Frick and Pittenger2016; Attwells et al., Reference Attwells, Setiawan, Wilson, Rusjan, Mizrahi, Miler, Xu, Richter, Kahn, Kish, Houle, Ravindran and Meyer2017; Michopoulos et al., Reference Michopoulos, Powers, Gillespie, Ressler and Jovanovic2017; Zaas et al., 2017).

This is also the case in OCD. Altered gut flora has been discovered in OCD (Turna et al., 2016; Reference Turna, Grosman Kaplan, Patterson, Bercik, Anglin, Soreni and Van Ameringen2019) and in animal models (Scheepers et al., 2019). Pregnancy may induce OCD with gut flora changes in pregnancy suggested as possible causes (Rees Reference Rees2014). Reintroduction of beneficial microbes and probiotics into the gut has been proposed as a possible treatment for OCD.

Psychosis

Early clinical evidence arose from epidemiological studies of the 1957 influenza pandemic in which maternal infection during pregnancy was found to correlate with the development of schizophrenia in the offspring in later life (Brown and Patterson,Reference Brown and Patterson2011; Mednick et al,Reference Mednick, Machon, Huttunen and Bonett1988). A growing body of experimental and clinical evidence supports the role of neuroinflammation in the pathophysiology of psychosis and schizophrenia (Doorduin et al., Reference Doorduin, deVries, Willemsen, de Groot, Dierckx and Klein2009), with theories pinpointing parvalbumin interneuron development impacted by inflammation and NMDAR hypofunction (Barron et al., Reference Barron, Hafizi and Mizrahi2017; Najjar et al., Reference Najjar, Steiner, Najjar and Bechter2018). These parvalbumin interneurons are fast spiking GABAergic neurons that synchronise the pyramidal neurons in the cortex and contribute to the generation of cognitive processes and working memory (Zandi et al., Reference Zandi, Irani, Lang, Waters, Jones, McKenna, Coles, Vincent and Lennox2011). Support for the importance of neuroinflammation in schizophrenia and psychosis also comes from genetic studies. Multiple genome-wide association studies implicate the major histocompatibility site on chromosome 6, in particular compliment C4 within the human leucocyte antigen. C4 is involved in pathogen opsonisation and synaptic pruning which could be involved in the initiation of the early disruption of the neuronal network in schizophrenia (Sekar et al., Reference Sekar, Bialas, de Rivera, Davis, Hammond, Kamitaki, Tooley, Presumey, Baum, Van Doren, Genovese, Rose, Handsaker, Daly, Carroll, Stevens and McCarroll2016).

Post-mortem studies of schizophrenic patients have also demonstrated the presence of activated microglia in brain tissue, though results of several studies are inconsistent or confined to a limited number of brain regions and without a specific selection of the patients studied. An early positive emission study using 14C-PK-11195, a peripheral benzodiazepine receptor ligand, showed that neuroinflammation occurred in the hippocampus of schizophrenic patients in the psychotic phase of the illness (Doorduin et al., Reference Doorduin, deVries, Willemsen, de Groot, Dierckx and Klein2009). Later positron emission tomography (PET) studies using a more specific radioligand showed no difference between medicated and drug-naïve first episode psychosis or schizophrenia compared to healthy controls (Hafizi et al., Reference Hafizi, Tseng, Rao, Selvanathan, Kenk, Bazinet, Suridjan, Wilson, Meyer, Remington, Houle, Rusjan and Mizrahi2017; Coughlin et al., Reference Coughlin, Wang, Ambinder, Ward, Minn, Vranesic, Kim, Ford, Higgs, Hayes, Schretlen, Dannals, Kassiou, Sawa and Pomper2016). This suggests that neuroinflammation may be confined to a subgroup of patients and present at an early stage of the illness.

A meta-analysis by Miller et al., (Reference Miller, Buckley, Seabolt, Mellor and Kirkpatrick2011) of 40 studies involving over 2500 schizophrenic patients found that the pro-inflammatory cytokines, TNF-alpha, interferon (IFN)-gamma, IL-12, and IL2 receptor were consistently increased independent of the stage of the illness, suggesting that these cytokines might be trait markers. Conversely, IL-1 beta, IL-6, and TGF-beta were positively associated with the active phase of the illness and were therefore possible state markers.

Finally, celecoxib, a cyclo-oxygenase 2 inhibitor, can enhance the efficacy of risperidone and amisulpride in the treatment of chronic schizophrenia (Muller 2010; Muller et al., 2010), while the anti-inflammatory tetracycline, minocycline, improves the negative symptoms and cognitive dysfunction of schizophrenic patients in the early and acute phases of the disorder (Levkovitz et al., 2009).

MS and 5-HT

Although disruption of the BBB and demyelination are known consequences of neuroinflammation in MS, an interesting relationship between 5-HT and MS exists. 5-HT disturbances of gut origin have been suggested to play a pivotal role in demyelinating disorder, including MS (Malinova et al., Reference Malinova, Dijkstra and de Vries2018). PET showed differential expression of 5-HT transporters in several brain regions in relapsing MS. Lower levels of serotonin reuptake transporters (SERTs) have been reported in the cingulate cortex, the thalamus, insula, and hippocampus, with higher SERTs in the prefrontal cortex. Lower SERT in the insula was correlated with depression scores (Hesse et al., Reference Hesse, Moeller, Petroff, Lobsien, Luthardt, Regenthal, Becker, Patt, Thomae, Seese, Meyer, Bergh and Sabri2014).

Chronic stress causes widespread health problems, including malignancies, ageing, gastrointestinal disorders, and skin problems. Psychological stress and mental disorders have been shown to activate the peripheral immune system (Bendorius et al., Reference Bendorius, Po, Muller and Jeltsch-David2018). The composition of gut microbes, which are far away from the brain, is known to be affected by psychological factors, including pregnancy-related stress, and vice versa (Rees Reference Rees2014). Toxic metabolites generated from pathological microbes in the gut could travel to the CNS and trigger neuroinflammation. Thus, a dynamic triple relationship exists between the CNS, immune system, and gut axis and this intricate balance could be lost in the presence of external insults or stress. Dysregulation of the eicosanoids and cytokine systems is suspected to be the mediator of the psychological stress-damaging effects (Umamaheswaran et al., Reference Umamaheswaran, Dasari, Yang, Lutgendorf and Sood2018).

A neuroprotective role of the endocannabinoid system and a modulating role of the cannabinoid receptor 1 (CB1) have been debated, using data from animal models (Zoppi et al., Reference Zoppi, Pérez Nievas, Madrigal, Manzanares, Leza and García-Bueno2011; Rabasa et al., Reference Rabasa, Pastor-Ciurana, Delgado-Morales, Gómez-Román, Carrasco, Gagliano, García-Gutiérrez, Manzanares and Armario2015). The potential neuroprotective usage of anti-inflammatory CB1 and CB2 agonists would need further research (Mastinu et al., Reference Mastinu, Premoli, Ferrari-Toninelli, Tambaro, Maccarinelli, Memo and Bonini2018).

In these different neuropsychiatric disorders, finding the clues to address the specificity and vulnerability issue of chronic neuroinflammation is important. It is difficult to accept general inflammation as the aetiology of any specific neuropsychiatric disorder, as one would have to explain why the same inflammation would end up with neurodegeneration in some patients and depression, OCD, or schizophrenia in others. In fact, the same difficulty happened in the early hypercortisolemia hypothesis of depression; a question of whether it is the inflammation or hypercortisolemia that created the neurocircuit damage or suppressed neurogenesis (Lau et al. Reference Lau, Qiu, Helmeste, Lee, Tang, So and Tang2007; Qiu et al. Reference Qiu, Helmeste, Samaranayake, Lau, Lee, Tang and So2007). The damages are not specific to depression (Tang et al., Reference Tang, Helmeste and Leonard2012; Tang et al., 2017b).

Profiles of neuroinflammation, anti-inflammatory agents, and disease-modifying agents

In neuroinflammation, chronic activation of astrocytes and microglia, infiltration of peripheral leucocytes, and secretion of inflammatory cytokines are well known. Astrocytes and microglia are key regulators of innate and adaptive immune responses. Their coordinated activities can be pro- or anti-inflammatory, neuroprotective, or neurotoxic (Colombo and Farina Reference Colombo and Farina2016; Jha et al., Reference Jha, Jo, Kim and Suk2019). A good example is the observation that Th1 cells produce IFN-γ and mediate neuroinflammation in MS. In contrast, Th2 cells produce IL-4 which would antagonise Th1 cells and therefore would be beneficial (Rostami and Ciric 2013). These kinds of checks and balances are typical of the immune system but when off-balance, these result in many run-away inflammatory responses and autoimmune disorders. More precise manipulation of the immune system may be useful. One approach is the use of a designer monoclonal antibody to bind directly to the targeted chemo/cytokine or the use of soluble receptors to bind the chemo/cytokine molecules. Small molecule antagonists and neutralising molecules to precisely target one or more cytokine are also possible (Pranzatelli Reference Pranzatelli2018).

The neuroinflammation-triggered immune response may be heterogenous and cytokine/chemokine profiling might provide new insights into disease pathogenesis and improve our ability to monitor inflammation and respond to treatment (Kothur et al., Reference Kothur, Wienholt, Brilot and Dale2016). Different inflammatory markers or profiles of immune response have been described for different neuropsychiatric disorders. The best examples are in neurodegenerative disorders where the profiles of immune responses have been rigorously investigated (Oeckl et al., 2018; Abu Rumeileh et al., Reference Abu-Rumeileh, Steinacker, Polischi, Mammana, Bartoletti-Stella, Oeckl, Baiardi, Zenesini, Huss, Cortelli, Capellari, Otto and Parchi2019). Profiling immune response may eventually provide directions for the type of anti-inflammatory measures to be used. For example, patients with high YKL-40 (glycoprotein marker of inflammation) might benefit from compounds targeting specific neuroinflammatory mechanisms, independently of the initial clinical diagnosis (Baldacci et al., Reference Baldacci, Lista, Palermo, Giorgi, Vergallo and Hampel2019).

Alcoholism is an example of the heterogeneity and complexity of the immune response to inflammation (Orio et al., Reference Orio, Alen, Pavón, Serrano and García-Bueno2019). Alcoholism induces both peripheral and CNS inflammation and activates toll-like receptors 4 (TLR4). The innate lipid transmitter oleoylethanolamide (OEA) is potently anti-inflammatory and neuroprotective in alcohol abuse. In animal models, it has been demonstrated that OEA blocks the alcohol-induced TLR4-mediated pro-inflammatory cascade. The release of pro-inflammatory cytokines and chemokines is blocked, resulting in the blockade of inflammatory neuro damage in the frontal cortex (Sayd et al., Reference Sayd, Antón, Alén, Caso, Pavón, Leza, Rodríguez de Fonseca, García-Bueno and Orio2014).

Pro-inflammatory cytokines, such as IL-6, TNF- α, IL-1β, as well as TNF-alpha and IFN-gamma, have been reported to be elevated in patients suffering from major depression (O’Brien et al., Reference O’Brien, Scott and Dinan2004; Schiepers et al., Reference Schiepers, Wichers and Maes2005; Brites and Fernandes Reference Brites and Fernandes2015; Benatti et al., Reference Benatti, Blom, Rigillo, Alboni, Zizzi, Torta, Brunello and Tascedda2016). Activated pro-inflammatory cytokines may cause HPA axis hyperactivity through interference with the negative feedback inhibition of circulating CSs on the HPA axis. These patients may benefit from anti-inflammatory agents (Kopschina Feltes et al., Reference Kopschina Feltes, Doorduin, Klein, Juárez-Orozco, Dierckx, Moriguchi-Jeckel and de Vries2017). There are other signature cytokines such as interleukin-17A (IL-17A) produced by T helper 17 cells (Th17) that have been suspected to mediate the damaging effects of neuroinflammation (Beurel and Lowell Reference Beurel and Lowell2018). Intestinal Th17 cells are regulated by gut microbes. Immune profiling is therefore the step to target therapy.

Considering the profiles of neuroinflammation, the dynamic interaction between the cytokines and the glucocorticoid system (Kim et al., Reference Kim, Na, Myint and Leonard2016) is an important consideration. The profiles may not be static when the glucocorticoid system is activated, or in response to steroid therapy, when treating hyperinflammatory states, as in cytokine storms in COVID-19.

Neuronal damage in peripheral inflammation and innate neuroinflammation may differ. It has been pointed out that in degenerative diseases, brain resident cells, not blood-borne leucocytes, are the predominant producers of pro-inflammatory cytokines. In neuroinflammatory diseases, such as in MS, invading leucocytes are the main producers of pro-inflammatory cytokines (Becher et al., Reference Becher, Spath and Goverman2017). This may translate into targeted therapeutics with further research.

Use of anti-inflammatory agents such as NSAIDs has been found to be associated with reduced risks in AD studies (Wang et al., Reference Wang, Tan, Wang, Tan, Meng, Wang, Tang and Yu2015), especially if used early (McGeer et al., Reference McGeer, Rogers and McGeer2016). This contrasts with the ineffectiveness of NSAID in treating acute inflammation in TBI as mentioned above. COX-2 inhibitors have been reported to be effective in some but not all studies (Sethi et al., Reference Sethi, Gómez-Coronado, Walker, Robertson, Agustini, Berk and Dodd2019; Westwell-Roper C, Stewart 2020).

Apart from anti-inflammatory agents such as NSAID, an important advancement in targeting the dysregulated immune system in neuroinflammation is in the area of disease-modifying agents or therapies (DMT). There is a paucity of such agents at present for neuropsychiatric disorders other than MS. The US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have already approved a number of DMTs for MS, such as β-IFN-1α, teriflunomide, and natalizumab (Doshi and Chataway, Reference Doshi and Chataway2016). The use of IFNs to modify inflammatory damage in viral infection is already a common practice. In COVID-19 infections, the virus has been discovered to impair IFN λ induction, resulting in severe hyperinflammation. IFN-lambda (IFN λ) is thus a possible DMT for COVID-19-associated hyperinflammation (Andreakos and Tsiodras Reference Andreakos and Tsiodras2020). Cell-based therapies have also been considered. Examples include mesenchymal, neuronal, human embryonic, and induced pluripotent stem cell and hematopoietic stem cell therapy for suppressing hyperinflammation in relapsing MS, thereby improving neurological disability (Cuascut and Hutton Reference Cuascut and Hutton2019).

Neuroprotection and therapeutics

The diagnosis of the cause of acute neuroinflammation is usually straightforward. Treatment would involve targeting and removal of the causative factors and applying neuroprotective measures as fast as possible. In COVID-19 infection, application of steroids and a full plethora of disease-modifying agents illustrate the contemporary approach to mitigating the potential damage of a hyperinflammatory condition, which may proceed into a chronic neuroinflammatory state, such as MS. In PANDAS, for example, prophylactic NSAIDs given within 30 days of onset may shorten neuropsychiatric symptom duration (Brown et al. Reference Brown, Farmer, Freeman, Spartz, Farhadian, Thienemann and Frankovich2017). Small doses of DA antagonists may help to relieve the hallucinations, which are different from the intrusive images of OCD (Thienemann et al. Reference Thienemann, Murphy, Leckman, Shaw, Williams, Kapphahn, Frankovich, Geller, Bernstein, Chang, Elia and Swedo2017). Repinotan, a highly selective 5-HT1A receptor agonist, was found to have pronounced neuroprotective effects in ischemic stroke (Berends et al. Reference Berends, Luiten and Nyakas2005). Other treatment and activities such as enhancing BDNF, Bcl-2, and other neurotrophic hormones, and physical exercise (Tang et al. Reference Tang, Chu, Hui, Helmeste and Law2008), are important in facilitating or enhancing neurogenesis and synaptogenesis.

One of the most exciting developments is repurposing of existing medications for neuroprotection. Fluvoxamine, an SSRI antidepressant with high (agonist) affinity for sigma1 receptors, is beneficial in pre-clinical models of inflammation and sepsis (Rosen et al., Reference Rosen, Seki, Fernandez-Castaneda, Beiter, Eccles, Woodfolk and Gaultier2019). It also lowers clinical deterioration in COVID-19 outpatients (Lenze et al., Reference Lenze, Mattar, Zorumski, Stevens, Schweiger, Nicol, Miller, Yang, Yingling, Avidan and Reiersen2020). The effect appears related to the fact that sigma1 receptors control production of inflammatory cytokines via the endoplasmic reticulum stress sensor IRE1. For reviews on sigma-1 receptor’s role in neurodegeneration and neuroprotection, please see (Nguyen et al., Reference Nguyen, Lucke-Wold, Mookerjee, Cavendish, Robson, Scandinaro and Matsumoto2015, Reference Nguyen, Lucke-Wolds, Mookerjee, Kaushal and Matsumoto2017).

Interestingly, a number of important psychiatric drugs have been found to label sigma receptors in human brain (Helmeste et al., Reference Helmeste, Tang, Bunney, Potkin and Jones1996, Reference Helmeste, Shioiri, Mitsuhashi and Tang1999; Tang et al., Reference Tang, Helmeste, Fang, Li, Vu, Bunney, Potkin and Jones1997). Considering the emerging role of sigma1 receptors in neuroprotection, it is significant that post-mortem brain studies in schizophrenic subjects have shown marked reductions (50%) in sigma receptor numbers compared to normal controls (Helmeste et al., Reference Helmeste, Tang, Fang and Li1996). How this affects long-term neurodegeneration is an important area for further study.

Different neurons exhibit different vulnerabilities to insults. GABA neurons appear to be particularly vulnerable to psychological stress resulting in depression in early life (Gabbay et al., Reference Gabbay, Bradley, Mao, Ostrover, Kang and Shungu2017) or schizophrenia later (Modinos et al., 2018a, b; Romeo et al., Reference Romeo, Choucha, Fossati and Rotge2018).

ACh neurons are the vulnerable neurons in AD and degenerate early. Their number is a good correlate of disease progression while drugs that protect the ACh system are still one of the most promising treatments for AD (Ferreira-Vieira et al., Reference Ferreira-Vieira, Guimaraes, Silva and Ribeiro2016; Hampel et al., Reference Hampel, Mesulam, Cuello, Farlow, Giacobini, Grossberg, Khachaturian, Vergallo, Cavedo, Snyder and Khachaturian2018).

Serotonin (5HT) neurons are vulnerable to certain hormone deficiencies, 5HT depletion medications, and certain psychedelics, which bind strongly to the 5HT2A receptors. Normal function of the 5HT system depends on ovarian steroidal hormones, especially estradiol and deficiency of these hormones in early development adversely influenced the development of 5HT neurons and resulted in fewer 5HT neurons in animal models (Bethea et al., Reference Bethea, Smith, Centeno and Reddy2011, Reference Bethea, Reddy and Christian2017). Decrease in female steroid hormones in middle age or in those with ovariectomy correlates with an increase in the onset of AD and depression. Oestrogen enhances the effect of antidepressant treatment (Hernández-Hernández et al., Reference Hernández-Hernández, Martínez-Mota, Herrera-Pérez and Jiménez-Rubio2019). All these illustrate the value of adjunct or supportive therapeutics.

Drugs of abuse and many psychedelics tend to damage 5HT neurons, by binding strongly to 5HTA2 receptors and via perturbation of 5HT uptake and release. Well-known examples include amphetamine and its analogues. 3,4-methylenedioxymethamphetamine and its analogues damage serotonin (5-HT) neurons through mitochondrially mediated oxidative stress and activate autophagy, with dieback of 5-HT arbour. Rilmenidine antagonises this neurotoxic effect (Mercer et al. 2017). Whether damaged 5HT axons in the adult mammalian brain have the capacity to regrow is controversial (Jin et al. Reference Jin, Dougherty, Wood, Sun, Cudmore, Abdalla, Kannan, Pletnikov, Hashemi and Linden2016).

Glutaminergic pyramidal neurons in hippocampus are sensitive to ischemia and may degenerate after recurrent seizures and stroke. Interestingly, susceptibility differs between CA1 pyramidal neurons, which tend to degenerate after global ischemia and CA3 neurons after limbic seizures. The basis for these differential vulnerabilities is unclear and might be related to the differential entry of Zn2+ into CA1 (delayed and long-lasting) and CA3 (rapid) (Medvedeva et al. Reference Medvedeva, Ji, Yin and Weiss2017).

The above discussion highlights the differential vulnerabilities of neurons to inflammatory insults. The remaining difficulty is to differentiate neuroinflammation from other aetiological factors in causing functional impairment or neuronal degeneration/death in the particular disorder.

Regarding neuroprotection, toll-like receptors (TLRs), such as TLR2 and TLR4, are known inducers of tissue inflammation in trauma and infections. Binding to TLRs, hyaluronan (HA) oligomer and HA tetrasaccharide (HA4) could suppress the expression of pro-inflammatory cytokine IL-1β and was found to significantly prevent hippocampal pyramidal neuronal death even 7 days after ischemic injury (Sunabori et al., Reference Sunabori, Koike, Asari, Oonuki and Uchiyama2016). The polyphenol resveratrol, present in red wine, potently protects against ischemia neuronal damage through its oxygen-free radical scavenging and antioxidant properties (Zhang et al., Reference Zhang, Schools, Lei, Wang, Kimelberg and Zhou2008). Delayed pyramidal neuronal death in ischemia might be due to apoptosis; the NSAID indomethacin, a prostaglandin inhibitor, has been shown to be neuroprotective in an animal model (Kondo et al., Reference Kondo, Kondo, Makino and Ogawa2000). Extensive but selective pyramidal neuronal death occurred in the neocortex and hippocampus in AD. In normal ageing, there is no neuronal death, but synaptic NMDARs, no longer protected by oestrogen, are decreased in certain hippocampal circuits (Morrison and Hof, Reference Morrison and Hof2002). NMDAR antagonists are known to be neuroprotective against hippocampal neuronal death in cell culture models (Pozzo Miller et al., Reference Pozzo Miller, Mahanty, Connor and Landis1994). Pyramidal neuron damage may lead to deafferentation and degeneration of GABAergic neurons (Shih et al., Reference Shih, Lee, Huang, Ko and Fu2004).

Another type of glutaminergic neuron in the anterior cingulate cortex and frontal-insular cortex, the Von Economo neurons, are especially vulnerable to AD pathology, particularly in later stages of pathogenesis. Their densities do not change throughout normal ageing but are more numerous in super-agers with high memory functioning. They are selectively destroyed in frontotemporal dementia (Kaufman et al., Reference Kaufman, Paul, Manaye, Granstedt, Hof, Hakeem and Allman2008), in which activated microglia are prominent (Lall and Baloh Reference Lall and Baloh2017). The intrinsic vulnerabilities, whether high metabolism or high oxidative load of these glutaminergic neurons, which push them into degeneration, will require further investigation (Gefen et al., Reference Gefen, Papastefan, Rezvanian, Bigio, Weintraub, Rogalski, Mesulam and Geula2018). Attempts to ameliorate glutamate-induced cytotoxicity were illustrated in the anti-allergic and anti-inflammatory actions of N-Palmitoyl-5- hydroxytryptamines (Pal-5HT), a cannabinoid, which demonstrated a dose-dependent inhibition of oxidation-induced cell death and suppressed glutamate-induced apoptosis and enhanced Bcl-2 and BDNF (Yoo et al., Reference Yoo, Lee, Sok, Ma and Kim2017).

Many other compounds and medications, as well as physical exercise, have been examined for their neuroprotective properties, but their neuroprotective actions appear to be general and not specific nor selective for specific neurons.

There are other less known but emerging novel neuroprotective compounds and measures to antagonise neuroinflammation. Psychological stress has been shown to change the composition of the gut flora, resulting in the passage of neurotoxic metabolites through the BBB to create damages or changes in the brain. The brain-gut-microbe-inflammation hypothesis of mental disorders and fecal transplantation as treatment is a rapidly emerging area of new research and new concept of treatment (Choi and Cho Reference Choi and Cho2016; Evrensel and Ceylan Reference Evrensel and Ceylan2016; Fung et al. Reference Fung, Olson and Hsiao2017; Inserra et al. Reference Inserra, Rogers, Licinio and Wong2018; Lin et al. Reference Lin, Zheng and Zhang2018; Sun and Shen Reference Sun and Shen2018; Cerovic et al. Reference Cerovic, Forloni and Balducci2019; Kim et al. Reference Kim, Kim, Choi, Kim, Park, Lee, Kim, Kim, Choi, Hyun, Lee, Choi, Lee, Bae and Mook-Jung2019; Sochocka et al. Reference Sochocka, Donskow-Łysoniewska, Diniz, Kurpas, Brzozowska and Leszek2019). Interestingly, processed fecal preparation has been used in traditional Chinese medicine for the treatment of all mental disorders from depression to psychosis (Tang and Tang Reference Tang and Tang2019). The recently announced seaweed-based drug oligomannate (Wang et al. Reference Wang, Sun, Feng, Zhang, Huang, Wang, Xie, Chu, Yang, Wang, Chang, Gong, Ruan, Zhang, Yan, Lian, Du, Yang, Zhang, Lin, Liu, Zhang, Ge, Xiao, Ding and Geng2019), which also works through restoration of normal gut flora, shows that small molecules are not necessarily the only compounds for neuroinflammation treatment.

Imaging inflammation

Investigation of neuroinflammation in psychiatric disorders benefited from recent advances in imaging techniques to visualise and quantify neuroinflammation in vivo (Wu et al., Reference Wu, Li, Niu and Chen2013; Felger Reference Felger2018). Cellular, immunoproteins, and other elements involved in inflammation and infection can now be imaged. Examples include tracking inflammation by PET, tagging targets such as the translocator protein in microglia, with 18F or 11C ligands (Wu et al., Reference Wu, Li, Niu and Chen2013). Other techniques included the use of 2-[(18F]-fluoro-2-deoxy-d-glucose or gallium −68 ligands to label leucocytes and other elements of immune responses (Vaidyanathan et al., 2015). Diagnosis and management of patients suffering from a broad spectrum of infections, such as human immunodeficiency virus (HIV) infections, disorders such as sarcoidosis, autoimmune disorders, and IgG4-related systemic diseases, can benefit from these imaging techniques now. It is foreseeable that more inflammation markers could be imaged soon with new techniques, such as the hybrid PER/magnetic resonance imaging systems (Sollini et al., Reference Sollini, Berchiolli, Kirienko, Rossi, Glaudemans, Slart and Erba2018). These advanced imaging techniques have yielded much data in patients with neurodegenerative disorders. Applications to other neuropsychiatric disorders such as OCD (Attwells et al., Reference Attwells, Setiawan, Wilson, Rusjan, Mizrahi, Miler, Xu, Richter, Kahn, Kish, Houle, Ravindran and Meyer2017) are in the early phases. These new techniques may enable us to quantify or differentiate the types of neuroinflammation in different psychiatric disorders and design targeted therapy to replace a general anti-inflammatory strategy such as the use of COX-2 inhibitors.

Conclusion

Neuroinflammation, triggered by a variety of causes, including viral infections such as COVID-19, plays an important role in the initiation, progression, or enhancement of many neuropsychiatric disorders, including depression and anxiety, OCD, AD, PD, and MS. In patients showing clear and marked elevations of inflammatory activities, or abnormal anti-inflammatory response, management or immune modulation may be crucial and useful as adjunct therapy to the standard medication.

References

Abu-Rumeileh, S, Steinacker, P, Polischi, B, Mammana, A, Bartoletti-Stella, A, Oeckl, P, Baiardi, S, Zenesini, C, Huss, A, Cortelli, P, Capellari, S, Otto, M, Parchi, P (2019) CSF biomarkers of neuroinflammation in distinct forms and subtypes of neurodegenerative dementia. Alzheimer's Research & Therapy 12, 2. doi: 10.1186/s13195-019-0562-4.CrossRefGoogle Scholar
Albrecht, DS, Granziera, C, Hooker, JM, Loggia, ML (2016) In vivo imaging of human neuroinflammation. ACS Chemical Neuroscience 7, 470483.CrossRefGoogle ScholarPubMed
Andreakos, E, Tsiodras, S (2020) COVID-19: lambda interferon against viral load and hyperinflammation. EMBO Molecular Medicine 12, e12465.Google Scholar
Attwells, S, Setiawan, E, Wilson, AA, Rusjan, PM, Mizrahi, R, Miler, L, Xu, C, Richter, MA, Kahn, A, Kish, SJ, Houle, S, Ravindran, L, Meyer, JH (2017) Inflammation in the neurocircuitry of obsessive-compulsive disorder. JAMA Psychiatry 74, 833840.CrossRefGoogle ScholarPubMed
Baldacci, F, Lista, S, Palermo, G, Giorgi, FS, Vergallo, A, Hampel, H (2019) The neuroinflammatory biomarker YKL-40 for neurodegenerative diseases: advances in development. Expert Rev Proteomics 16, 593600.CrossRefGoogle ScholarPubMed
Banerjee, D Viswamath, B (2020) Neuropsychiatric manifestations of covid-19 and possible pathogenic mechanisms: insights from other corona viruses. American Journal of Psychiatry 54, 17.Google Scholar
Barron, H, Hafizi, S, Mizrahi, R (2017) Towards an integrated view of early molecular changes underlying vulnerability to social stress in psychosis. Modern Trends in Pharmacopsychiatry 31, 96106.CrossRefGoogle ScholarPubMed
Becher, B, Bechmann, I, Greter, M (2006) Antigen presentation in autoimmunity and CNS inflammation: how T lymphocytes recognize the brain. Journal of Molecular Medicine (Berlin) 84, 532543.CrossRefGoogle Scholar
Becher, B, Spath, S, Goverman, J (2017) Cytokine networks in neuroinflammation. Nature Reviews Immunology 17, 4959.CrossRefGoogle ScholarPubMed
Benatti, C, Blom, JM, Rigillo, G, Alboni, S, Zizzi, F, Torta, R, Brunello, N, Tascedda, F (2016) Disease-induced neuroinflammation and depression. CNS & Neurological Disorders: Drug Targets 15, 414433.CrossRefGoogle ScholarPubMed
Bendorius, M, Po, C, Muller, S, Jeltsch-David, H (2018) From systemic inflammation to neuroinflammation: the case of neurolupus. International Journal of Molecular Sciences 19, 3588.CrossRefGoogle ScholarPubMed
Benson, S, Brinkhoff, A, Lueg, L, Roderigo, T, Kribben, A, Wilde, B, Witzke, O, Engler, H, Schedlowski, M, Elsenbruch, S (2017) Effects of acute systemic inflammation on the interplaybetween sad mood and affective cognition. Translational Psychiatry 7, 1281.CrossRefGoogle ScholarPubMed
Berends, AC, Luiten, PG, Nyakas, C (2005) A review of the neuroprotective properties of the 5- HT1A receptor agonist repinotan HCl (BAYx3702) in ischemic stroke. CNS Drug Reviews 11, 379402.CrossRefGoogle Scholar
Bethea, CL, Reddy, AP, Christian, FL (2017) How studies of the serotonin system in Macaque models of menopause relate to Alzheimer’s disease. Journal of Alzheimer’s Disease 57, 10011015.CrossRefGoogle Scholar
Bethea, CL, Smith, AW, Centeno, ML, Reddy, AP (2011) Long-term ovariectomy decreases serotonin neuron number and gene expression in free ranging macaques. Neuroscience 192, 675688.CrossRefGoogle ScholarPubMed
Beurel, E, Lowell, JA (2018) Th17 cells in depression. Brain, Behavior, and Immunity 69:2834.CrossRefGoogle ScholarPubMed
Bevan-Jones, WR, Surendranathan, A, Passamonti, L, Vázquez Rodríguez, P, Arnold, R, Mak, E, Su, L, Coles, JP, Fryer, TD, Hong, YT, Williams, G, Aigbirhio, F, Rowe, JB, O’Brien, JT (2017) Neuroimaging of inflammation in memory and related other disorders (NIMROD) study protocol: a deep phenotyping cohort study of the role of brain inflammation in dementia, depression and other neurological illnesses. BMJ Open 7, e013187.CrossRefGoogle Scholar
Brites, D, Fernandes, A (2015) Neuroinflammation and depression: microglia activation, extracellular microvesicles and microRNA dysregulation. Frontiers in Cellular Neuroscience 9, 476.CrossRefGoogle Scholar
Brown, AS Patterson, PH (2011) Maternal infection and schizophrenia: implications for prevention. Schizophrenia Bulletin 37, 284290.CrossRefGoogle ScholarPubMed
Brown, KD, Farmer, C, Freeman, GM Jr, Spartz, EJ, Farhadian, B, Thienemann, M, Frankovich, J (2017) Effect of early and prophylactic nonsteroidal anti-inflammatory drugs on flare duration in pediatric acute-onset neuropsychiatric syndrome: an observational study of patients followed by an academic community-based pediatric acute-onset neuropsychiatric syndrome clinic. Journal of Child and Adolescent Psychopharmacology 27, 619628.CrossRefGoogle ScholarPubMed
Brown, E, Gray, R, Lo Monaco, S, O’Donoghue, B, Nelson, B, Thompson, A, Francey, S, McGorry, P (2020) The potential impact of COVID-19 on psychosis: a rapid review of contemporary epidemic and pandemic research. Schizophrenia Research 222, 7987.CrossRefGoogle ScholarPubMed
Bunney, WE Jr (1975) The current status of research in the catecholamine theories of affective disorders. Psychopharmacology Communications 1, 599609.Google ScholarPubMed
Burchi, E, Pallanti, S (2018) Antibiotics for PANDAS? Limited evidence: review and putative mechanisms of action. Primary Care Companion to CNS Disorders 20, 17r02232.Google ScholarPubMed
Burkunina, N, Pariante, CM, Zunszain, PA (2015) Immune mechanisms linked to depression via oxidative stress and neuroprogression. Immunology 144, 365373.CrossRefGoogle Scholar
Butler, M, Pollak, TA, Rooney, AG, Michael, BD, Nicholson, TR (2020) Neuropsychiatric complications of covid-19. British Medical Journal 371, 12.Google Scholar
Cerovic, M, Forloni, G, Balducci, C (2019) Neuroinflammation and the gut microbiota: possible alternative therapeutic targets to counteract Alzheimer’s disease? Frontiers in Aging Neuroscience 11, 284.CrossRefGoogle ScholarPubMed
Chacko, M, Job, A, Caston, F 3rd, George, P, Yacoub, A, Cáceda, R (2020) COVID-19-induced psychosis and suicidal behavior: case report. SN Comprehensive Clinical Medicine 26, 15.Google Scholar
Choi, HH, Cho, YS (2016) Fecal microbiota transplantation: current applications, effectiveness, and future perspectives. Clinical Endoscopy 49, 257265.CrossRefGoogle ScholarPubMed
Colombo, E, Farina, C (2016) Astrocytes: key regulators of neuroinflammation. Trends in Immunology 37, 608620.CrossRefGoogle ScholarPubMed
Correa-Palacio, AF, Hernandez-Huerta, D, Gómez-Arnau, J, Loeck, C, Caballero, I (2020) Affective psychosis after COVID-19 infection in a previously healthy patient: a case report. Psychiatry Research 290, 113115.CrossRefGoogle Scholar
Corps, KN, Roth, TL, McGavern, DB (2015) Inflammation and neuroprotection in traumatic brain injury. JAMA Neurology 72, 355362.CrossRefGoogle ScholarPubMed
Cothran, TP, Kellman, S, Singh, S, Beck, JS, Powell, KJ, Bolton, CJ, Tam, JW (2020) A brewing storm: the neuropsychological sequelae of hyperinflammation due to COVID-19. Brain, Behavior, and Immunity 88, 957958.CrossRefGoogle ScholarPubMed
Coughlin, JM, Wang, Y, Ambinder, EB., Ward, RE, Minn, I, Vranesic, M, Kim, PK, Ford, CN, Higgs, C, Hayes, LN, Schretlen, DJ, Dannals, RF, Kassiou, M, Sawa, A, Pomper, MG (2016) In vivo markers of inflammatory response in recent-onset schizophrenia: a combined study using [(11)C] DPA-713 PET and analysis of CSF and plasma. Translational Psychiatry 6, e777. https://doi.org/10.1038/ CrossRefGoogle Scholar
Cox, CJ, Sharma, M, Leckman, JF, Zuccolo, J, Zuccolo, A, Kovoor, A, Swedo, SE, Cunningham, MW (2013) Brain human monoclonal autoantibody from sydenham chorea targets dopaminergic neurons in transgenic mice and signals dopamine D2 receptor: implications in human disease. Journal of Immunology 191, 55245541.CrossRefGoogle ScholarPubMed
Cuascut, FX, Hutton, GJ (2019) Stem cell-based therapies for multiple sclerosis: current perspectives. Biomedicines 7, 26.CrossRefGoogle ScholarPubMed
Cunningham, MW, Cox, CJ (2016) Autoimmunity against dopamine receptors in neuropsychiatric and movement disorders: a review of Sydenham chorea and beyond. Acta Physiologica (Oxford) 216, 90100.CrossRefGoogle ScholarPubMed
Dalmau, J (2017) Investigations in GABAA receptor antibody-associated encephalitis. Neurology 88, 10121020.Google Scholar
Dantzer, R (2017) Role of the Kynurenine metabolism pathway in inflammation-induced depression: preclinical approaches. Current Topics in Behavioral Neurosciences 31, 117138.CrossRefGoogle ScholarPubMed
Dantzer, R, Walker, AK (2014) Is there a role for glutamate-mediated excitotoxicity in inflammation-induced depression? Journal of Neural Transmission 121, 925932.CrossRefGoogle Scholar
Davydow, DS, Desai, SV, Needham, DM, Bienvenu, OJ (2008) Psychiatric morbidity in survivors of the acute respiratory distress syndrome: a systematic review. Psychosomatic Medicine 70, 512519.CrossRefGoogle ScholarPubMed
Dennis, EA, Norris, PC (2015) Eicosanoid storm in infection and inflammation. Nature Reviews Immunology 15, 511523.CrossRefGoogle ScholarPubMed
Di Bari, M, Di Pinto, G, Reale, M, Mengod, G, Tata, AM (2017) Cholinergic system and neuroinflammation: implication in multiple sclerosis. Central Nervous System Agents in Medicinal Chemistry 17, 109115.CrossRefGoogle ScholarPubMed
Doorduin, J, deVries, EFJ, Willemsen, ATM, de Groot, JC, Dierckx, RA, Klein, HC (2009) Neuroinflammation in schizophrenia-related psychosis: a PET study. Journal of Nuclear Medicine 50, 18011807.CrossRefGoogle ScholarPubMed
Doshi, A, Chataway, J (2016) Multiple sclerosis, a treatable disease. Clinical Medicine (London) 16 (Suppl 6), s53s59.CrossRefGoogle ScholarPubMed
Drożdżal, S, Rosik, J, Lechowicz, K, Machaj, F, Szostak, B, Majewski, P, Rotter, I, Kotfis, K (2020) COVID 19: pain management in patients with SARS-CoV-2 infection-molecular mechanisms, challenges, and perspectives. Brain Science 10, 465.CrossRefGoogle Scholar
Dubbioso, R, Nobile-Orazio, E, Manganelli, F, Santoro, L, Briani, C, Cocito, D, Tedeschi, G, Di Lazzaro, V, Fabrizi, GM; SIN, SINC and ASNP (2020) Dealing with immune-mediated neuropathies during COVID-19 outbreak: practical recommendations from the task force of the Italian Society of Neurology (SIN), the Italian Society of Clinical Neurophysiology (SINC) and the Italian Peripheral Nervous System Association (ASNP). Neurological Sciences 41, 13451348.CrossRefGoogle Scholar
Duman, RS, Monteggia, LM (2006) A neurotrophic model for stress-related mood disorders. Biological Psychiatry 59,11161127.CrossRefGoogle ScholarPubMed
Evrensel, A, Ceylan, ME (2016) Fecal microbiota transplantation and its usage in neuropsychiatric disorders. Clinical Psychopharmacology and Neuroscience 14, 231237.CrossRefGoogle ScholarPubMed
Fedorow, H, Tribl, F, Halliday, G, Gerlach, M, Riederer, P, Double, KL (2005) Neuromelanin in human dopamine neurons: comparison with peripheral melanins and relevance to Parkinson’s disease. Progress in Neurobiology 75, 109124.CrossRefGoogle ScholarPubMed
Felger, JC (2018) Imaging the role of inflammation in mood and anxiety-related disorders. Current Neuropharmacology 16, 533558.CrossRefGoogle Scholar
Ferrando, SJ, Klepacz, L, Lynch, S, Shahar, S, Dornbush, R, Smiley, A, Miller, I, Tavakkoli, M, Regan, J, Bartell, A (2020) Psychiatric emergencies during the height of the COVID-19 pandemic in the suburban New York City area. Journal of Psychiatric Research 3956, 3103231033.Google Scholar
Ferreira-Vieira, TH, Guimaraes, IM, Silva, FR, Ribeiro, FM (2016) Alzheimer’s disease: targeting the Cholinergic system. Current Neuropharmacology 14, 101115.CrossRefGoogle ScholarPubMed
Finatti, F, Pigato, G, Pavan, C, Toffanin, T, Favaro, A (2020) Psychosis in Patients in COVID-19- related quarantine: a case series. The Primary Care Companion for CNS Disorders 22, 20l02640.CrossRefGoogle ScholarPubMed
Frick, L, Pittenger, C (2016) Microglial dysregulation in OCD, tourette syndrome, PANDAS. Journal of Immunology Research 8606057. doi: 10.1155/2016/8606057 CrossRefGoogle ScholarPubMed
Fung, TC, Olson, CA, Hsiao, EY (2017) Interactions between the microbiota, immune and nervous systems in health and disease. Nature Neuroscience 20, 145155.CrossRefGoogle ScholarPubMed
Gabbay, V, Bradley, KA, Mao, X, Ostrover, R, Kang, G, Shungu, DC (2017) Anterior cingulate cortex γ-aminobutyric acid deficits in youth with depression. Translational Psychiatry 7, e1216.CrossRefGoogle ScholarPubMed
Garg, RK (2020) Spectrum of neurological manifestations in Covid-19: a review. Neurology India 68, 560572.CrossRefGoogle ScholarPubMed
Gatta, V, Mengod, G, Reale, M, Tata, AM (2020) Possible correlation between cholinergic system alterations and neuro/inflammation in multiple sclerosis. Biomedicines 8: 153.CrossRefGoogle ScholarPubMed
Gefen, T, Papastefan, ST, Rezvanian, A, Bigio, EH, Weintraub, S, Rogalski, E, Mesulam, MM, Geula, C (2018) Von economo neurons of the anterior cingulate across the lifespan and in Alzheimer’s disease. Cortex 99, 6977.CrossRefGoogle ScholarPubMed
Hafizi, S, Tseng, HH, Rao, N, Selvanathan, T, Kenk, M, Bazinet, RP, Suridjan, I, Wilson, AA, Meyer, JH, Remington, G, Houle, S, Rusjan, PM, Mizrahi, R (2017) Imaging microglial activation in untreated first-episode psychosis: a PET study with [18F] FEPPA. The American Journal of Psychiatry 174, 118124.CrossRefGoogle ScholarPubMed
Hagens, M, van Berckel, B, Barkhof, F (2016) Novel MRI and PET markers of neuroinflammation in multiple sclerosis. Current Opinion in Neurology 29, 229236.CrossRefGoogle ScholarPubMed
Haining, RL, Achat-Mendes, C (2017) Neuromelanin, one of the most overlooked molecules in modern medicine, is not a spectator. Neural Regeneration Research 12, 372375.CrossRefGoogle Scholar
Hampel, H, Mesulam, MM, Cuello, AC, Farlow, MR, Giacobini, E, Grossberg, GT, Khachaturian, AS, Vergallo, A, Cavedo, E, Snyder, PJ, Khachaturian, ZS (2018) The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 141, 19171933.CrossRefGoogle ScholarPubMed
Helmeste, DM, Shioiri, T, Mitsuhashi, M, Tang, SW (1999) Binding of [3H] U-101958 to σ1 receptor-like sites in human cerebellum and neuroblastoma cells. European Journal of Pharmacology 370, 205209.CrossRefGoogle Scholar
Helmeste, DM, Tang, SW, Bunney, WE, Potkin, SG, Jones, EG (1996) Decrease in σ but no increase in striatal dopamine D4 sites in schizophrenic brains. European Journal of Pharmacology 314, R3R5.CrossRefGoogle ScholarPubMed
Helmeste, DM, Tang, SW, Fang, H, Li, M (1996) Brain σ receptors labelled by [3H] nemonapride. European Journal of Pharmacology 301, R1R3.CrossRefGoogle Scholar
Henry, J, Smeyne, RJ, Jang, H, Miller, B, Okun, MS (2010) Parkinsonism and neurological manifestations of influenza throughout the 20th and 21st centuries. Parkinsonism & Related Disorders 16, 566571.CrossRefGoogle ScholarPubMed
Hernández-Hernández, OT, Martínez-Mota, L, Herrera-Pérez, JJ, Jiménez-Rubio, G (2019) Role of estradiol in the expression of genes involved in serotonin neurotransmission: implications for female depression. Current Neuropharmacology 17, 459471.CrossRefGoogle ScholarPubMed
Hesse, S, Moeller, F, Petroff, D, Lobsien, D, Luthardt, J, Regenthal, R, Becker, GA, Patt, M, Thomae, E, Seese, A, Meyer, PM, Bergh, FT, Sabri, O (2014) Altered serotonin transporter availability in patients with multiple sclerosis. European Journal of Nuclear Medicine and Molecular Imaging 41, 827835.CrossRefGoogle ScholarPubMed
Hoehn, M (2015) Imaging of neuro-inflammation: past, present and future. 2015. Georgian Medical News 243, 8284.Google Scholar
Hoover, DB (2017) Cholinergic modulation of the immune system presents new approaches for treating inflammation. Pharmacology & Therapeutics 179, 116.CrossRefGoogle Scholar
Huang, Y, Zhao, N (2020) Generalized anxiety disorder, depressive symptoms and sleep quality during COVID-19 outbreak in China: a web-based cross-sectional survey. Psychiatry Research 288, 112954.CrossRefGoogle ScholarPubMed
Inserra, A, Rogers, GB, Licinio, J, Wong, ML (2018) The microbiota-inflammasome hypothesis of major depression. Bioessays 40, e1800027.CrossRefGoogle ScholarPubMed
Jha, MK, Jo, M, Kim, JH, Suk, K (2019) Microglia-astrocyte crosstalk: an intimate molecular conversation. Neuroscientist 25, 227240.CrossRefGoogle ScholarPubMed
Jin, Y, Dougherty, SE, Wood, K, Sun, L, Cudmore, RH, Abdalla, A, Kannan, G, Pletnikov, M, Hashemi, P, Linden, DJ (2016) Regrowth of Serotonin Axons in the adult mouse brain following injury. Neuron 91, 748762.CrossRefGoogle ScholarPubMed
Kaufman, JA, Paul, LK, Manaye, KF, Granstedt, AE, Hof, PR, Hakeem, AY, Allman, JM (2008) Selective reduction of Von Economo neuron number in agenesis of the corpus callosum. Acta Neuropathologica 116, 479489.CrossRefGoogle ScholarPubMed
Kayser, MS, Dalmau, J (2016) Anti-NMDA receptor encephalitis, autoimmunity, and psychosis. Schizophrenia Research 176, 3640.CrossRefGoogle ScholarPubMed
Keller, MJ, Kitsis, EA, Arora, S, Chen, J-T, Agarwal, S, Ross, MJ, Tomer, Y, Southern, W (2020) Effect of systemic glucorticoids on mortality or mechanical ventilation in patients with COVID-19. Journal of Hospital Medicine 15, 489493.CrossRefGoogle ScholarPubMed
Kępińska, AP, Iyegbe, CO, Vernon, AC, Yolken, R, Murray, RM, Pollak, TA (2020) Schizophrenia and influenza at the centenary of the 1918–1919 Spanish influenza pandemic: mechanisms of psychosis risk. Front Psychiatry 11, 72.CrossRefGoogle Scholar
Kim, MS, Kim, Y, Choi, H, Kim, W, Park, S, Lee, D, Kim, DK, Kim, HJ, Choi, H, Hyun, DW, Lee, JY, Choi, EY, Lee, DS, Bae, JW, Mook-Jung, I (2019) Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. Epub 2019 Aug 30.Google Scholar
Kim, YK, Na, KS, Myint, AM, Leonard, BE (2016) The role of pro-inflammatory cytokines in neuroinflammation, neurogenesis and the neuroendocrine system in major depression. Progress in Neuro-Psychopharmacology & Biological Psychiatry 64, 277284.CrossRefGoogle ScholarPubMed
Kondo, F, Kondo, Y, Makino, H, Ogawa, N (2000) Delayed neuronal death in hippocampal CA1 pyramidal neurons after forebrain ischemia in hyperglycemic gerbils: amelioration by indomethacin. Brain Research 853, 9398.CrossRefGoogle ScholarPubMed
Konishi, H, Kiyama, H (2018) Microglial TREM2/DAP12 signaling: a double-edged Sword in neural diseases. Frontiers in Cellular Neuroscience 12, 206.CrossRefGoogle ScholarPubMed
Kopschina Feltes, P, Doorduin, J, Klein, HC, Juárez-Orozco, LE, Dierckx, RA, Moriguchi-Jeckel, CM, de Vries, EF (2017) Anti-inflammatory treatment for major depressive disorder: implications for patients with an elevated immune profile and non-responders to standard antidepressant therapy. Journal of Psychopharmacology 31, 11491165.CrossRefGoogle ScholarPubMed
Kostic, M, Stojanovic, I, Marjanovic, G, Zivkovic, N, Cvetanovic, A (2015) Deleterious versus protective autoimmunity in multiple sclerosis. Cellular Immunology 296, 122132.CrossRefGoogle ScholarPubMed
Kothur, K, Wienholt, L, Brilot, F, Dale, RC (2016) CSF cytokines/chemokines as biomarkers in neuroinflammatory CNS disorders: a systematic review. Cytokine 77, 227237.CrossRefGoogle ScholarPubMed
Lagarde, J, Sarazin, M, Bottlaender, M (2018) In vivo PET imaging of neuroinflammation in Alzheimer’s disease. Journal of Neural Transmission 125, 847867.CrossRefGoogle ScholarPubMed
Lagraoui, M, Sukumar, G, Latoche, JR, Maynard, SK, Dalgard, CL, Schaefer, BC (2017) Salsalate treatment following traumatic brain injury reduces inflammation and promotes a neuroprotective and neurogenic transcriptional response with concomitant functional recovery. Brain, Behavior, and Immunity 61, 96109.CrossRefGoogle ScholarPubMed
Lall, D, Baloh, RH (2017) Microglia and C9orf72 in neuroinflammation and ALS and frontotemporal dementia. The Journal of Clinical Investigation 127, 32503258.CrossRefGoogle ScholarPubMed
Lan, CC, Liu, CC, Chen, YS (2015) Acute exacerbation of psychiatric symptoms during influenza treatment with oseltamivir in chronic schizophrenia. Journal of the Chinese Medical Association 78, 374376.CrossRefGoogle ScholarPubMed
Lau, WM, Qiu, G, Helmeste, DM, Lee, TM, Tang, SW, So, KF, Tang, SW (2007) Corticosteroid decreases subventricular zone cell proliferation, which could be reversed by paroxetine. Restorative Neurology and Neuroscience 25, 1723.Google ScholarPubMed
Le chien, JR, Chiesa-Estomba, CM, De Siati, DR, Horoi, M, Le Bon, SD, Rodriguez, A, Dequanter, D, Blecic, S, El Afia, F, Distinguin, L, Chekkoury-Idrissi, Y, Hans, S, Delgado, IL, Calvo-Henriquez, C, Lavigne, P, Falanga, C, Barillari, MR, Cammaroto, G, Khalife, M, Leich, P, Souchay, C, Rossi, C, Journe, F, Hsieh, J, Edjlali, M, Carlier, R, Ris, L, Lovato, A, De Filippis, C, Coppee, F, Fakhry, N, Ayad, T, Saussez, S (2020) Olfactory gustatory dysfunctions as a clinical presentation of mild and moderate forms of covid-19: multi-centre study. European Archives of Otorhinolaryngology 277, 22512261.CrossRefGoogle Scholar
Lee, AC, Tang, SW, Leung, SS, Yu, GK, Cheung, RT (2009) Depression literacy among Chinese stroke survivors. Aging and Mental Health 13, 349356.CrossRefGoogle ScholarPubMed
Lenze, EJ, Mattar, C, Zorumski, CF, Stevens, A, Schweiger, J, Nicol, GE, Miller, JP, Yang, L, Yingling, M, Avidan, MS, Reiersen, AM (2020) Fluovoxamine versus placebo and clinical deterioration in outpatients with symptomatic COVID-19. JAMA. doi: 10:1001/jama.2020.22760 CrossRefGoogle Scholar
Leonard, BE (2010) The concept of depression as a dysfunction of the immune system. Current Immunology Reviews 6, 205212.CrossRefGoogle ScholarPubMed
Leonard, BE (2015) Pain, depression and inflammation: are interconnected causative factors involved? Mod Trends Pharmacopsychiatry 30, 2235.CrossRefGoogle ScholarPubMed
Leonard, BE (2017) Major depression as a neuroprogressive prelude to dementia: what is the evidence? Mod Trends Pharmacopsychiatry 31, 5666.CrossRefGoogle ScholarPubMed
Levkovitz, Y, Mendlovich, S, Riwkes, S, Braw, Y, Levkovitch-Verbin, H, Gal, G, Fennig, S, Treves, I, Kron, S (2010) A double-blind, randomized study of minocycline for the treatment of negative and cognitive symptoms in early-phase schizophrenia. 71, 138149.CrossRefGoogle Scholar
Lin, L, Zheng, LJ, Zhang, LJ (2018) Neuroinflammation, gut microbiome, and Alzheimer’s disease. Molecular Neurobiology 55, 82438250.CrossRefGoogle ScholarPubMed
Loane, DJ, Kumar, A (2016) Microglia in the TBI brain: the good, the bad, and the dysregulated. Experimental Neurology 275 (Pt 3), 316327.CrossRefGoogle ScholarPubMed
Mak, IW, Chu, CM, Pan, PC, Yiu, MG, Chan, VL (2009) Long-term psychiatric morbidities among SARS survivors. General Hospital Psychiatry 31, 318326.CrossRefGoogle ScholarPubMed
Malinova, TS, Dijkstra, CD, de Vries, HE (2018) Serotonin: a mediator of the gut-brain axis in multiple sclerosis. Multiple Sclerosis 24, 11441150.CrossRefGoogle ScholarPubMed
Mangale, V, Syage, AR, Ekiz, HA, Skinner, DD, Cheng, Y, Stone, CL, Brown, RM, O’Connell, RM, Green, KN, Lane, TE (2020) Microglia influence host defense, disease, and repair following murine coronavirus infection of the central nervous system. Glia. doi: 10.1002/glia.23844. doi:10.1002/glia.23844 CrossRefGoogle ScholarPubMed
Mastinu, A, Premoli, M, Ferrari-Toninelli, G, Tambaro, S, Maccarinelli, G, Memo, M, Bonini, SA (2018) Cannabinoids in health and disease: pharmacological potential in metabolic syndrome and neuroinflammation. Hormone Molecular Biology and Clinical Investigation 36.:/j/hmbci.2018.36.issue-2/hmbci 2018-0013/hmbci-2018-0013.xml. doi: 10.1515/hmbci-2018-0013 CrossRefGoogle ScholarPubMed
Matheoud, D, Cannon, T, Voisin, A, Penttinen, AM, Ramet, L, Fahmy, AM, Ducrot, C, Laplante, A, Bourque, MJ, Zhu, L, Cayrol, R, Le Campion, A, McBride, HM, Gruenheid, S, Trudeau, LE, Desjardins, M (2019) Intestinal infection triggers Parkinson’s disease-like symptoms in Pink1-/- mice. Nature 571, 565569.CrossRefGoogle ScholarPubMed
Mazza, MG, De Lorenzo, R, Conte, C, Poletti, S, Vai, B, Bollettini, I, Melloni, EMT, Furlan, R, Ciceri, F, Rovere-Querini, P, COVID-19 BioB Outpatient Clinic Study group, Benedetti, F (2020) Covid-19 Anxiety and depression in covid-19 survivors. Role of inflammatory and clinical predictors. Brain Behav. Immun. doi:10.1016/bbi.2020.07.o37pmid.32738287 CrossRefGoogle Scholar
Mcloughlin, BC, Miles, A, Webb, TE, Knopp, P, Eyres, C, Fabbri, A, Humphries, F, Davis, D (2020) Functional and cognitive outcomes after covid 19 delirium. European Geriatric Medicine. doi: 10.1007/s41999-020-00353-8pmid:3266303 CrossRefGoogle Scholar
McGeer, PL, Rogers, J, McGeer, EG. (2016) Inflammation, antiinflammatory agents, and Alzheimer’s disease: the last 22 years. Journal of Alzheimer's Disease 54, 853857.CrossRefGoogle ScholarPubMed
McQuail, JA, Riddle, DR, Nicolle, MM (2011) Neuroinflammation not associated with cholinergic degeneration in aged-impaired brain. Neurobiol Aging 32, 2322.e12322.e4.CrossRefGoogle Scholar
Mednick, SA, Machon, RA, Huttunen, MO, Bonett, D (1988) Adult schizophrenia following prenatal exposure to an influenza epidemic. Archives of General Psychiatry 45, 189192.CrossRefGoogle Scholar
Medvedeva, YV, Ji, SG, Yin, HZ, Weiss, JH (2017) Differential vulnerability of CA1 versus CA3 pyramidal neurons after ischemia: possible relationship to sources of Zn2+ accumulation and its entry into and prolonged effects on mitochondria. Journal of Neuroscience 37, 726737.CrossRefGoogle ScholarPubMed
Ménard, C, Pfau, ML, Hodes, GE, Russo, SJ (2017) Immune and neuroendocrine mechanisms of stress vulnerability and resilience. Neuropsychopharmacology 42, 6280.CrossRefGoogle ScholarPubMed
Michopoulos, V, Powers, A, Gillespie, CF, Ressler, KJ, Jovanovic, T (2017) Inflammation in fear and anxiety-based disorders: PTSD, GAD, and beyond. Neuropsychopharmacology 42, 254270.CrossRefGoogle ScholarPubMed
Miller, BJ, Buckley, P, Seabolt, W, Mellor, A, Kirkpatrick, B (2011) Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biological Psychiatry 70, 663671.CrossRefGoogle ScholarPubMed
Modinos, G, Şimşek, F, Azis, M, Bossong, M, Bonoldi, I, Samson, C, Quinn, B, Perez, J, Broome, MR, Zelaya, F, Lythgoe, DJ, Howes, OD, Stone, JM, Grace, AA, Allen, P, McGuire, P (2018) Prefrontal GABA levels, hippocampal resting perfusion and the risk of psychosis. Neuropsychopharmacology 43, 26522659.CrossRefGoogle ScholarPubMed
Modinos, G, Simsek, F, Horder, J, Bossong, M, Bonoldi, I, Azis, M, Perez, J, Broome, M, Lythgoe, DJ, Stone, JM, Howes, OD, Murphy, DG, Grace, AA, Allen, P, McGuire, P (2018) Cortical GABA in subjects at ultra-high risk of psychosis: relationship to negative prodromal symptoms. International Journal of Neuropsychopharmacology 21, 114119.CrossRefGoogle ScholarPubMed
Morrison, JH, Hof, PR (2002) Selective vulnerability of corticocortical and hippocampal circuits in aging and Alzheimer’s disease. Progress in Brain Research 136, 467486.CrossRefGoogle ScholarPubMed
Müller, N (2010) COX-2 inhibitors as antidepressants and antipsychotics: clinical evidence. Current Opinion in Investigational Drugs 11, 3142.Google ScholarPubMed
Müller, N, Krause, D, Dehning, S, Musil, R, Schennach-Wolff, R, Obermeier, M, Möller, HJ, Klauss, V, Schwarz, MJ, Riedel, M (2010) Celecoxib treatment in an early stage of schizophrenia: results of a randomized, double-blind, placebo-controlled trial of celecoxib augmentation of amisulpride treatment. Schizophrenia Research 121, 118124.CrossRefGoogle Scholar
Müller, N, Schwarz, MJ (2006) Neuroimmune-endocrine crosstalk in schizophrenia and mood disorders. Expert Review of Neurotherapeutics 6, 10171038.CrossRefGoogle ScholarPubMed
Müller, N, Schwarz, M (2007) The immune-mediated alteration of serotonin and glutamate: towards an integrated view of depression. Journal of Molecular Psychiatry 12, 9881000.CrossRefGoogle ScholarPubMed
Najjar, S, Steiner, J, Najjar, A, Bechter, K (2018) A clinical approach to new-onset psychosis associated with dysregulation: the concept of autoimmune psychosis. Journal of Neuroinflammation 15, 40.CrossRefGoogle ScholarPubMed
Nehring, SM, Goyal, A, Bansal, P, Patel, BC (2020) C reactive protein (CRP). https://www.ncbi.nlm.nih.gov/books/NBK441843/?report=printable.Google Scholar
Ng, QX, Yeo, WS, Lim, DY, Chee, KT (2020) Re-examining the association between COVID-19 and psychosis. Psychosomatics 61, 853855.CrossRefGoogle ScholarPubMed
Nguyen, L, Lucke-Wold, BP, Mookerjee, SA, Cavendish, JZ, Robson, MJ, Scandinaro, AL, Matsumoto, RR (2015) Role of sigma-1 receptors in neurodegenerative diseases. Journal of Pharmaceutical Sciences 127, 1729.CrossRefGoogle ScholarPubMed
Nguyen, L, Lucke-Wolds, BP, Mookerjee, S, Kaushal, N, Matsumoto, RR (2017) Sigma-1 receptors and neurodegenerative diseases: towards a hypothesis of sigma-1 receptors as amplifiers of neurodegeneration and neuroprotection. Advances in Experimental Medicine and Biology 964, 133152.CrossRefGoogle ScholarPubMed
Novak, P (2020) Post COVID-19 syndrome associated with orthostatic cerebral hypoperfusion syndrome, small fiber neuropathy and benefit of immunotherapy: a case report. eNeurologicalSci 21, 100276.CrossRefGoogle Scholar
O’Brien, SM, Scott, LV, Dinan, TG (2004) Cytokines: abnormalities in major depression and implications for pharmacological treatment. Human Psychopharmacology 19, 397403.CrossRefGoogle ScholarPubMed
Oeckl, P, Weydt, P, Steinacker, P, Anderl-Straub, S, Nordin, F, Volk, AE, Diehl-Schmid, J, Andersen, PM, Kornhuber, J, Danek, A, Fassbender, K, Fliessbach, K; German Consortium for Frontotemporal Lobar Degeneration, Jahn, H, Lauer, M, Müller, K, Knehr, A, Prudlo, J, Schneider, A, Thal, DR, Yilmazer-Hanke, D, Weishaupt, JH, Ludolph, AC, Otto, M (2019) Different neuroinflammatory profile in amyotrophic lateral sclerosis and frontotemporal dementia is linked to the clinical phase. Journal of Neurology, Neurosurgery and Psychiatry 90, 410.CrossRefGoogle ScholarPubMed
Orefici, G, Cardona, F, Cox, CJ, Cunningham, MW (2016) Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS). In: Ferretti, JJ, Stevens, DL, Fischetti, VA (eds), Streptococcus Pyogenes: Basic Biology to Clinical Manifestations [Internet]. Oklahoma City, OK: University of Oklahoma Health Sciences Center.Google Scholar
Orio, L, Alen, F, Pavón, FJ, Serrano, A, García-Bueno, B (2019) Oleoylethanolamide, neuroinflammation, and alcohol abuse. Frontiers in Molecular Neuroscience 11, 490.CrossRefGoogle ScholarPubMed
Orsucci, D, Ienco, EC, Nocita, G, Napolitano, A, Vista, M (2020) Neurological features of COVID-19 and their treatment: a review. Drugs Context 9, 2020-5-1.CrossRefGoogle ScholarPubMed
Ottaviani, D, Boso, F, Tranquillini, E, Gapeni, I, Pedrotti, G, Cozzio, S, Guarrera, GM, Giometto, B (2020) Early Guillain-Barré syndrome in coronavirus disease 2019 (COVID-19): a case report from an Italian COVID-hospital. Neurological Sciences 41, 13511354.CrossRefGoogle ScholarPubMed
Perlman, S, Evans, G, Afift, AD (2020) Effect of olfactory bulb ablation on the spread of a neurovirus into the mouse brain. Journal of Experimental Medicine 172, 11271132.CrossRefGoogle Scholar
Pozzo Miller, LD, Mahanty, NK, Connor, JA, Landis, DM (1994) Spontaneous pyramidal cell death in organotypic slice cultures from rat hippocampus is prevented by glutamate receptor antagonists. Neuroscience 63, 471487.CrossRefGoogle ScholarPubMed
Pranzatelli, MR (2018) Advances in biomarker-guided therapy for pediatric- and adult-onset neuroinflammatory disorders: targeting chemokines/cytokines. Frontiers in Immunology 9, 557.CrossRefGoogle ScholarPubMed
Pulli, B, Chen, JW (2014) Imaging neuroinflammation – from bench to bedside. Journal of Clinical & Cellular Immunology 5, 226.Google ScholarPubMed
Qiu, G, Helmeste, DM, Samaranayake, AN, Lau, WM, Lee, TM, Tang, SW, So, KF (2007) Modulation of the suppressive effect of corticosterone on adult rat hippocampal cell proliferation by paroxetine. Neuroscience Bulletin 23, 131136.CrossRefGoogle ScholarPubMed
Rabasa, C, Pastor-Ciurana, J, Delgado-Morales, R, Gómez-Román, A, Carrasco, J, Gagliano, H, García-Gutiérrez, MS, Manzanares, J, Armario, A (2015) Evidence against a critical role of CB1 receptors in adaptation of the hypothalamic-pituitary-adrenal axis and other consequences of daily repeated stress. European Neuropsychopharmacology 25, 12481259.CrossRefGoogle ScholarPubMed
Rajabally, YA, Goedee, HS, Attarian, S, Hartung, HP (2020) Management challenges for chronic dysimmune neuropathies during the COVID-19 pandemic. Muscle Nerve 62, 3440.CrossRefGoogle ScholarPubMed
Razanamahery, J, Malinowski, L, Humbert, S, Brunel, AS, Lepiller, Q, Chirouze, C, Bouiller, K (2020) Predictive factors of poor outcomes in the COVID-19 epidemic: consider the inflammatory response. Médecine et maladies infectieuses 50, 620631.CrossRefGoogle ScholarPubMed
Rees, JC (2014) Obsessive-compulsive disorder and gut microbiota dysregulation. Medical Hypotheses 82, 163166.CrossRefGoogle ScholarPubMed
Rogers, JP, Chesney, E, Oliver, D, Pollak, TA, McGuire, P, Fusar-Poli, P, Zandi, MS, Lewis, G, David, AS (2020) Psychiatric and neuropsychiatric presentations associated with severe coronavirus infections: a systematic review and meta-analysis with comparison to the COVID-19 pandemic. Lancet Psychiatry 7, 611627.CrossRefGoogle ScholarPubMed
Romeo, B, Choucha, W, Fossati, P, Rotge, JY (2018) Meta-analysis of central and peripheral γ aminobutyric acid levels in patients with unipolar and bipolar depression. Journal of Psychiatry and Neuroscience 43, 5866.CrossRefGoogle ScholarPubMed
Rosen, DA, Seki, SM, Fernandez-Castaneda, A, Beiter, RM, Eccles, JD, Woodfolk, JA, Gaultier, A (2019) Modulation of the sigma-1 receptor-IRE1 pathway is beneficial in preclinical models of inflammation and sepsis. Science Translational Medicine 11, eaau5266.CrossRefGoogle Scholar
Sayd, A, Antón, M, Alén, F, Caso, JR, Pavón, J, Leza, JC, Rodríguez de Fonseca, F, García-Bueno, B, Orio, L (2014) Systemic administration of oleoylethanolamide protects from neuroinflammation and anhedonia induced by LPS in rats. International Journal of Neuropsychopharmacology 18, pyu111.CrossRefGoogle ScholarPubMed
Schiepers, OJ, Wichers, MC, Maes, M (2005) Cytokines and major depression. Progress in Neuro-Psychopharmacology & Biological Psychiatry 29, 201217.CrossRefGoogle ScholarPubMed
Schwartz, M, Kipris, J, Rivets, S, Prat, A (2013) How do immune cells support shape the brain in health, disease and aging? The Journal of Neuroscience 33, 1758717596.CrossRefGoogle ScholarPubMed
Sekar, A, Bialas, AR, de Rivera, H, Davis, A, Hammond, TR, Kamitaki, N, Tooley, K, Presumey, J, Baum, M, Van Doren, V, Genovese, G, Rose, SA, Handsaker, RE, Schizophrenia Working Group of the Psychiatric Genomics Consortium, Daly, MJ, Carroll, MC, Stevens, B, McCarroll, SA (2016) Schizophrenia risk from complex variation of complement component 4. Nature 530, 177183.CrossRefGoogle ScholarPubMed
Sethi, R, Gómez-Coronado, N, Walker, AJ, Robertson, OD, Agustini, B, Berk, M, Dodd, S (2019) Neurobiology and therapeutic potential of cyclooxygenase-2 (COX-2) inhibitors for inflammation in neuropsychiatric disorders. Frontiers in Psychiatry, 10, 605.CrossRefGoogle ScholarPubMed
Shao, W, Zhang, SZ, Tang, M, Zhang, XH, Zhou, Z, Yin, YQ, Zhou, QB, Huang, YY, Liu, YJ, Wawrousek, E, Chen, T, Li, SB, Xu, M, Zhou, JN, Hu, G, Zhou, JW (2013) Suppression of neuroinflammation by astrocytic dopamine D2 receptors via αB-crystallin. Nature 494, 9094.CrossRefGoogle ScholarPubMed
Sher, L (2020) The impact of the COVID-19 pandemic on suicide rates. QJM 113, 707712.CrossRefGoogle ScholarPubMed
Shih, YH, Lee, AW, Huang, YH, Ko, MH, Fu, YS (2004) GABAergic neuron death in the striatum following kainate-induced damage of hippocampal neurons: evidence for the role of NO in locomotion. International Journal of Neuroscience 114, 11191132.CrossRefGoogle ScholarPubMed
Sigra, S, Hesselmark, E, Bejerot, S (2018) Treatment of PANDAS and PANS: a systematic review. Neuroscience and Biobehavioral Reviews 86, 5165.CrossRefGoogle ScholarPubMed
Singhou, T (2020) A review of coronavirus disease 2019 (covid-19). Indian Journal of Pediatrics 22, 16.Google Scholar
Sochocka, M, Donskow-Łysoniewska, K, Diniz, BS, Kurpas, D, Brzozowska, E, Leszek, J (2019) The gut microbiome alterations and inflammation-driven pathogenesis of Alzheimer’s disease a critical review. Molecular Neurobiology 56, 18411851.CrossRefGoogle ScholarPubMed
Sollini, M, Berchiolli, R, Kirienko, M, Rossi, A, Glaudemans, AWJM, Slart, R, Erba, PA (2018) PET/MRI in infection and inflammation. Seminars in Nuclear Medicine 48, 225241.CrossRefGoogle ScholarPubMed
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, Armangue, T, Castro, FJ, Barcelo Artigues, MI, Julià Benique, MR, Benson, L, Gorman, M, Felipe, A, Caparó Oblitas, RL, Rosenfeld, MR, Graus, F, Dalmau, J (2017) Investigation in GABA receptor antibody-associated encephalitis. Neurology 88, 10121020.CrossRefGoogle Scholar
Sun, MF, Shen, YQ (2018) Dysbiosis of gut microbiota and microbial metabolites in Parkinson’s Disease. Ageing Research Reviews 45, 5361.CrossRefGoogle ScholarPubMed
Sunabori, T, Koike, M, Asari, A, Oonuki, Y, Uchiyama, Y (2016) Suppression of ischemia-induced hippocampal pyramidal neuron death by Hyaluronan Tetrasaccharide through inhibition of toll-like receptor 2 signaling pathway. American Journal of Pathology 186, 21432151.CrossRefGoogle ScholarPubMed
Tang, SW, Chu, E, Hui, T, Helmeste, D, Law, C (2008) Influence of exercise on serum brain derived neurotrophic factor concentrations in healthy human subjects. Neuroscience Letters 431, 6265.CrossRefGoogle ScholarPubMed
Tang, SW, Helmeste, DM, Fang, H, Li, M, Vu, R, Bunney, W, Potkin, S, Jones, EG (1997) Differential labeling of dopamine and sigma sites by [3H] nemonapride and [3H] raclopride in postmortem human brains. Brain Research 765, 712.CrossRefGoogle Scholar
Tang, SW, Helmeste, D, Leonard, B (2012) Is neurogenesis relevant in depression and in the mechanism of antidepressant drug action? A critical review. The World Journal of Biological Psychiatry 13, 402412.CrossRefGoogle Scholar
Tang, SW, Helmeste, DM, Leonard, BE (2017) Neurodegeneration, neuroregeneration, and neuroprotection in psychiatric disorders. Modern Trends in Pharmacopsychiatry 31, 107123.CrossRefGoogle ScholarPubMed
Tang, SW, Tang, WH (2019) Opportunities in novel psychotropic drug design from natural compounds. International Journal of Neuropsychopharmacology 22, 601607.CrossRefGoogle ScholarPubMed
Tang, SW, Tang, WH, Leonard, BE (2017) Multitarget botanical pharmacotherapy in major depression: a toxic brain hypothesis. International Clinical Psychopharmacology 32, 299308.CrossRefGoogle ScholarPubMed
Thienemann, M, Murphy, T, Leckman, J, Shaw, R, Williams, K, Kapphahn, C, Frankovich, J, Geller, D, Bernstein, G, Chang, K, Elia, J, Swedo, S (2017) Clinical management of pediatric acute onset neuropsychiatric syndrome: part i-psychiatric and behavioral interventions. Journal of Child and Adolescent Psychopharmacology 27, 566573.CrossRefGoogle ScholarPubMed
Thomas, AJ (2005) Increase in IL-1 beta in late-life depression. The American Journal of Psychiatry 162, 175177.CrossRefGoogle ScholarPubMed
Tommasin, S, Giannì, C, De Giglio, L, Pantano, P (2017) Neuroimaging techniques to assess inflammation in multiple sclerosis. Neuroscience 403, 416.CrossRefGoogle ScholarPubMed
Treinin, M, Papke, RL, Nizri, E, Ben-David, Y, Mizrachi, T, Brenner, T (2017) Role of the α7 nicotinic acetylcholine receptor and RIC-3 in the cholinergic anti-inflammatory pathway. Central Nervous System Agents in Medicinal Chemistry 17, 9099.Google ScholarPubMed
Troyer, EA, Kohn, JN, Hong, S (2020) Are we facing a crashing wave of neuropsychiatric sequelae of COVID-19? Neuropsychiatric symptoms and potential immunologic mechanisms. Brain, Behavior, and Immunity 87, 3439.CrossRefGoogle ScholarPubMed
Turna, J, Grosman Kaplan, K, Anglin, R, Van Ameringen, M (2016) What’s bugging the gut in OCD?” A review of the gut microbiome in obsessive-compulsive disorder. Depress Anxiety 33, 171178.CrossRefGoogle ScholarPubMed
Turna, J, Grosman Kaplan, K, Patterson, B, Bercik, P, Anglin, R, Soreni, N, Van Ameringen, M (2019) Higher prevalence of irritable bowel syndrome and greater gastrointestinal symptoms in obsessive-compulsive disorder. Journal of Psychiatric Research 118, 16.CrossRefGoogle ScholarPubMed
Umamaheswaran, S, Dasari, SK, Yang, P, Lutgendorf, SK, Sood, AK (2018) Stress, inflammation, and eicosanoids: an emerging perspective. Cancer and Metastasis Reviews 37, 203211.CrossRefGoogle Scholar
Valdés-Florido, MJ, López-Díaz, Á, Palermo-Zeballos, FJ, Martínez-Molina, I, Martín-Gil, VE, Crespo-Facorro, B, Ruiz-Veguilla, M (2020) Reactive psychoses in the context of the COVID-19 pandemic: clinical perspectives from a case series. Revista de Psiquiatría y Salud Mental 13, 9094.CrossRefGoogle ScholarPubMed
Varatharaj, A, Thomas, N, Ellul, MA, Davies, N, Pollak, TA, Tenorio, EL, Sultan, M, Easton, A, Breen, G, Zandi, M, Coles, JP, Manji, H, Al-Shahi Salman, R, Menon, DK, Nicholson, TR, Benjamin, LA, Carson, A, Smith, C, Turner, MR, Solomon, T, Kneen, R, Pett, SL, Galea, I, Thomas, RH, Michael, BD, Coro Nerve Study Group (2020) Neurological and neuropsychiatric complications of COVID-19 in 153 patients: a UK-wide surveillance study. The Lancet Psychiatry 7, 875882.CrossRefGoogle ScholarPubMed
Vidal, PM, Pacheco, R (2019) Targeting the dopaminergic system in autoimmunity. Journal of Neuroimmune Pharmacology. doi: 10.1007/s11481-019-09834-5.Google ScholarPubMed
Villa, RF, Ferrari, F, Moretti, A (2018) Post-stroke depression: mechanisms and pharmacological treatment. Pharmacology & Therapeutics 184, 131144.CrossRefGoogle ScholarPubMed
Wang, L (2020) C-reactive protein levels in the early stage of COVID-19. Médecine et Maladies Infectieuses, 50, 332334.CrossRefGoogle ScholarPubMed
Wang, J, Tan, L, Wang, HF, Tan, CC, Meng, XF, Wang, C, Tang, SW, Yu, JT (2015) Anti-inflammatory drugs and risk of Alzheimer’s disease: an updated systematic review and metaanalysis. Journal of Alzheimer’s Disease 44, 385396.CrossRefGoogle Scholar
Wang, X, Sun, G, Feng, T, Zhang, J, Huang, X, Wang, T, Xie, Z, Chu, X, Yang, J, Wang, H, Chang, S, Gong, Y, Ruan, L, Zhang, G, Yan, S, Lian, W, Du, C, Yang, D, Zhang, Q, Lin, F, Liu, J, Zhang, H, Ge, C, Xiao, S, Ding, J, Geng, M (2019) Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Research 29, 787803.CrossRefGoogle ScholarPubMed
Webb, S, Wallace, VC, Martin-Lopez, D, Yogarajah, M (2020) Guillain-Barré syndrome following COVID-19: a newly emerging post-infectious complication. BMJ Case Reports 13, e236182.CrossRefGoogle ScholarPubMed
Westwell-Roper, C, Stewart, SE (2020) Commentary: neurobiology and therapeutic potential of cyclooxygenase-2 (COX-2) inhibitors for inflammation in neuropsychiatric disorders. Frontiers Psychiatry 11, 264.CrossRefGoogle ScholarPubMed
Whitehouse, PJ, Price, DL, Clark, AW, Coyle, JT, DeLong, MR (1981) Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus Basalis. Annals of Neurology 10, 122126.CrossRefGoogle ScholarPubMed
Won, E, Kim, YK (2016) Stress, the autonomic nervous system, and the immune kynurenine pathway in the etiology of depression. Current Neuropharmacology 14, 665673.CrossRefGoogle ScholarPubMed
Wu, C, Li, F, Niu, G, Chen, X (2013) PET imaging of inflammation biomarkers. Theranostics 3, 448466.CrossRefGoogle ScholarPubMed
Wu, Y, Xu, K, Chen, Z, Duan, J, Hashimoto, K, Yang, L, Liu, C, Yang, C (2020) Nervous system involvement after infection with covid-19 and other coronavirus diseases. Brain, Behavior, and Immunity 87, 1822.CrossRefGoogle Scholar
Xiong, Y, Mahmood, A, Chopp, M (2018) Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chinese Journal of Traumatology (Zhonghua chuang shang za zhi) 21, 137151.CrossRefGoogle ScholarPubMed
Yegambaram, M, Manivannan, B, Beach, TG, Halden, RU (2015) Role of environmental contaminants in the etiology of Alzheimer’s disease: a review. Current Alzheimer Research 12, 116146.CrossRefGoogle ScholarPubMed
Yoo, JM, Lee, BD, Sok, DE, Ma, JY, Kim, MR (2017) Neuroprotective action of N-acetyl serotonin in oxidative stress-induced apoptosis through the activation of both TrkB/CREB/BDNF pathway and Akt/Nrf2/Antioxidant enzyme in neuronal cells. Redox Biology 11, 592599.CrossRefGoogle ScholarPubMed
Zandi, MS, Irani, SR, Lang, B, Waters, P, Jones, PB, McKenna, P, Coles, AJ, Vincent, A, Lennox, BR (2011) Disease-relevant autoantibodies in first episode schizophrenia. Journal of Neurology 258, 686688.CrossRefGoogle ScholarPubMed
Zass, LJ, Hart, SA, Seedat, S, Hemmings, SM, Malan-Müller, S (2017) Neuroinflammatory genes associated with post-traumatic stress disorder: implications for comorbidity. Psychiatric Genetics 27, 116.CrossRefGoogle ScholarPubMed
Zeberg, H, Pääbo, S (2020) The major genetic risk factor for severe COVID-19 is inherited from Neanderthals. Nature. Epub ahead of print. doi: 10.1038/s41586-020-2818-3 CrossRefGoogle Scholar
Zecca, L, Casella, L, Albertini, A, Bellei, C, Zucca, FA, Engelen, M, Zadlo, A, Szewczyk, G, Zareba, M, Sarna, T (2008) Neuromelanin can protect against iron-mediated oxidative damage in system modeling iron overload of brain aging and Parkinson’s disease. Journal of Neurochemistry 106, 18661875.Google ScholarPubMed
Zecca, L, Zucca, FA, Wilms, H, Sulzer, D (2003) Neuromelanin of the substantia nigra: a neuronal black hole with protective and toxic characteristics. Trends in Neurosciences 26, 578580.CrossRefGoogle ScholarPubMed
Zhang, Y, Chen, Y, Wu, J, Manaenko, A, Yang, P, Tang, J, Fu, W, Zhang, JH (2015) Activation of dopamine D2 receptor suppresses neuroinflammation through αB-crystalline by inhibition of NF-κB nuclear translocation in experimental ICH mice model. Stroke 46, 26372646.CrossRefGoogle ScholarPubMed
Zhang, H, Schools, GP, Lei, T, Wang, W, Kimelberg, HK, Zhou, M (2008) Resveratrol attenuates early pyramidal neuron excitability impairment and death in acute rat hippocampal slices caused by oxygen-glucose deprivation. Experimental Neurology 212, 4452.CrossRefGoogle ScholarPubMed
Zhou, H, Lu, S, Chen, J, Wei, N, Wang, D, Lyu, H, Shi, C, Hu, S (2020) The landscape of cognitive function in recovered COVID-19 patients. Journal of Psychiatric Research 129, 98102.CrossRefGoogle ScholarPubMed
Zoppi, S, Pérez Nievas, BG, Madrigal, JL, Manzanares, J, Leza, JC, García-Bueno, B (2011) Regulatory role of cannabinoid receptor 1 in stress-induced excitotoxicity and neuroinflammation. Neuropsychopharmacology 36, 805818.CrossRefGoogle ScholarPubMed
Zucca, FA, Segura-Aguilar, J, Ferrari, E, Muñoz, P, Paris, I, Sulzer, D, Sarna, T, Casella, L, Zecca, L (2017) Interactions of iron, dopamine and neuromelanin pathways in brain aging and Parkinson’s disease. Progress in Neurobiology 155, 96119.CrossRefGoogle ScholarPubMed