Hostname: page-component-8448b6f56d-mp689 Total loading time: 0 Render date: 2024-04-23T06:27:59.090Z Has data issue: false hasContentIssue false

Long COVID, neuropsychiatric disorders, psychotropics, present and future

Published online by Cambridge University Press:  03 March 2022

Siu Wa Tang*
Affiliation:
Department of Psychiatry, University of California, Irvine, Irvine, CA, USA Institute of Brain Medicine, Hong Kong, China
Brian E. Leonard
Affiliation:
Institute of Brain Medicine, Hong Kong, China National University of Ireland, Galway, Ireland
Daiga Maret Helmeste
Affiliation:
Department of Psychiatry, University of California, Irvine, Irvine, CA, USA Institute of Brain Medicine, Hong Kong, China
*
Author for correspondence: Siu Wa Tang, Email: swtang@uci.edu
Rights & Permissions [Opens in a new window]

Abstract

Long COVID refers to the lingering symptoms which persist or appear after the acute illness. The dominant long COVID symptoms in the two years since the pandemic began (2020–2021) have been depression, anxiety, fatigue, concentration and cognitive impairments with few reports of psychosis. Whether other symptoms will appear later on is not yet known. For example, dopamine-dependent movement disorders generally take many years before first symptoms are seen. Post-stroke depression and anxiety may explain many of the early long COVID cases. Hemorrhagic, hypoxic and inflammatory damages of the central nervous system, unresolved systematic inflammation, metabolic impairment, cerebral vascular accidents such as stroke, hypoxia from pulmonary damages and fibrotic changes are among the major causes of long COVID. Glucose metabolic and hypoxic brain issues likely predispose subjects with pre-existing diabetes, cardiovascular or lung problems to long COVID as well. Preliminary data suggest that psychotropic medications may not be a danger but could instead be beneficial in combating COVID-19 infection. The same is true for diabetes medications such as metformin. Thus, a focus on sigma-1 receptor ligands and glucose metabolism is expected to be useful for new drug development as well as the repurposing of current drugs. The reported protective effects of psychotropics and antihistamines against COVID-19, the earlier reports of reduced number of sigma-1 receptors in post-mortem schizophrenic brains, with many antidepressant and antipsychotic drugs being antihistamines with significant affinity for the sigma-1 receptor, support the role of sigma and histamine receptors in neuroinflammation and viral infections. Literature and data in all these areas are accumulating at a fast rate. We reviewed and discussed the relevant and important literature.

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 (https://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), 2022. Published by Cambridge University Press on behalf of Scandinavian College of Neuropsychopharmacology

Summation

  • The neuropsychiatric symptoms in long COVID may represent the consequences of acute viral damage of the central nervous system (CNS), stroke and other cerebrovascular incidents.

  • Stress from prolonged social isolation and reduced physical activities may also contribute to some of the reported symptoms.

  • Safety of psychotropic usage in patients with COVID-19 is always a consideration. Initial reports showed that psychotropics may actually be protective in COVID-19.

  • The role of sigma-1 and histamine receptors and glucose metabolism in COVID-19 infection are important areas for further research and possibly raise novel directions for anti-COVID-19 drug development.

Consideration

  • Stress from quarantine, social isolation, job loss and fear of infection are confounding factors in the study of long COVID neuropsychiatric disorders.

  • The intensity of the prominent symptoms in long COVID, such as depression, anxiety, fatigue, somatic pain, mild memory and cognitive impairments, has to be carefully quantified in future studies of long COVID.

  • Differential efficacy of psychotropics in the treatment of long COVID depression and anxiety should be explored in future studies.

  • As there are already many reports on long COVID neuropsychiatric symptoms, international collaborative survey of long COVID neuropsychiatric symptoms under stringent and careful design should be the next step.

Introduction

The COVID-19 (SARS-COV-2) pandemic has so far resulted in 276,436,619 confirmed cases and 5,374,744 deaths worldwide as of December 23, 2021, according to the WHO COVID-19 Dashboard (https://covid19.who.int). The appearance of 822,278 new cases just in the 24 hours prior to December 23, 2021 highlights the ominous situation the world is facing and that we are far from passing the peak of devastation in this pandemic. Comparatively, there was a total of 8098 people worldwide who became sick and 774 deaths with the 2003 SARS outbreak (https://www.cdc.gov).

Unlike the 2003 SARS and many common viral infectious diseases, whose impact is largely absent or soon after when the infection ends, there is now increasing clinical and post-mortem pathological evidence that COVID-19 continues to have significant adverse health effects long after the acute phase of the infection.

The scope of COVID-19 impacts on recovering patients was reflected in an online survey (Davis et al., Reference Davis, Assaf, McCorkell, Wei, Low, Re’em, Redfield, Austin and Akrami2021). It covered 3762 participants with confirmed or suspected COVID-19 from 56 countries and estimated the presence of 203 symptoms in 10 organ systems. After month 6, fatigue, post-exertional malaise and cognitive dysfunction were the most frequent symptoms. Almost half of the respondents (45.2%) required a reduced work schedule compared to pre-illness, and about a quarter (22.3%) were still not working. Cognitive dysfunction or memory issues were common across all age groups (∼88%).

One of the largest cohort studies with the longest follow-up duration reported very well the health consequences of discharged adult patients recovering from COVID-19 (Huang et al., Reference Huang, Huang, Wang, Li, Ren, Gu, Kang, Guo, Liu, Zhou, Luo, Huang, Tu, Zhao, Chen, Xu, Li, Li, Peng, Li, Xie, Cui, Shang, Fan, Xu, Wang, Wang, Zhong, Wang, Wang, Zhang and Cao2021). Fatigue (Rudroff et al., Reference Rudroff, Fietsam, Deters, Bryant and Kamholz2020; Ortelli et al., Reference Ortelli, Ferrazzoli, Sebastianelli, Engl, Romanello, Nardone, Bonini, Koch, Saltuari, Quartarone, Oliviero, Kofler and Versace2021) or muscle weakness, sleep difficulties and anxiety or depression were common, even at 6 months after symptom onset. It appears that long COVID survivors may follow the same convalescent path of previous Canadian SARS survivors, in that though they had good physical recovery from their illness, a significant number (33%) of them reported a significant decrement in mental health 1 year later (Tansey et al., Reference Tansey, Louie and Loeb2007). Whether this is related to the lingering pulmonary diffusion abnormalities and therefore impaired oxygenation and nutritional supply to the brain (up to 3 months after hospital discharge) (Huang et al., Reference Huang, Tan and Wu2019; Zhao et al., Reference Zhao, Shang and Song2020) is awaiting further investigation. Evidence from the previous SARS pandemic suggests that reduced lung diffusion capacity could persist in 38% of survived SARS patients 15 years after infection (Zhang et al., Reference Zhang, Li and Liu2020).

Similar enduring and debilitating symptoms in COVID-19 patients after recovery also were reported from other countries (Chippa et al., Reference Chippa, Aleem and Anjum2021; Dewanjee et al., Reference Dewanjee, Vallamkondu, Kalra, Puvvada, Kandimalla and Reddy2021; Garg et al., Reference Garg, Maralakunte, Garg, Dhooria, Sehgal, Bhalla, Vijayvergiya, Grover, Bhatia, Jagia, Bhalla, Suri, Goyal, Agarwal, Puri and Sandhu2021; Huang et al., Reference Huang, Huang, Wang, Li, Ren, Gu, Kang, Guo, Liu, Zhou, Luo, Huang, Tu, Zhao, Chen, Xu, Li, Li, Peng, Li, Xie, Cui, Shang, Fan, Xu, Wang, Wang, Zhong, Wang, Wang, Zhang and Cao2021; Sisó-Almirall et al., Reference Sisó-Almirall, Brito-Zerón, Conangla Ferrín, Kostov, Moragas Moreno, Mestres, Sellarès, Galindo, Morera, Basora, Trilla and Ramos-Casals2021; Wang et al., Reference Wang, Kream and Stefano2020). Again, neuropsychiatric symptoms were prominent. Anosmia, the ‘Brain fog’ syndrome, consisting of confusion and cognitive and attention deficits (Asadi-Pooya et al., Reference Asadi-Pooya, Akbari, Emami, Lotfi, Rostamihosseinkhani, Nemati, Barzegar, Kabiri, Zeraatpisheh, Farjoud-Kouhanjani, Jafari, Sasannia, Ashrafi, Nazeri, Nasiri and Shahisavandi2021; Stefano et al., Reference Stefano, Ptacek, Ptackova, Martin and Kream2021b), fatigue, headaches, pain syndrome, anxiety and depression were all common. Data from Germany and the United Kingdom showed post-COVID-19 neuropsychiatric symptoms in 20% to 70% of patients, lasting months after respiratory symptoms resolved, suggesting that CNS symptoms persist long after the acute infection (Boldrini et al., Reference Boldrini, Canoll and Klein2021). If neurodegeneration and new neuropsychiatric disorders are proven to happen in long COVID, it could become a major public health burden (Serrano-Castro et al., Reference Serrano-Castro, Estivill-Torrús, Cabezudo-García, Reyes-Bueno, Ciano Petersen, Aguilar-Castillo, Suárez-Pérez, Jiménez-Hernández, Moya-Molina, Oliver-Martos, Arrabal-Gómez and Rodríguez de Fonseca2020).

The term ‘long COVID’ is used to refer to the lingering or protracted illness, from 4 (Sisó-Almirall et al., Reference Sisó-Almirall, Brito-Zerón, Conangla Ferrín, Kostov, Moragas Moreno, Mestres, Sellarès, Galindo, Morera, Basora, Trilla and Ramos-Casals2021) to 12 weeks (Mohamed-Hussein et al., Reference Mohamed-Hussein, Amin, Makhlouf, Makhlouf, Galal, Abd-Elaal, Abdeltawab, Kholief and Hashem2021) after the acute illness and during recovery. Other terms were also used, including ‘post-acute COVID-19’, ‘ongoing symptomatic COVID-19’, ‘chronic COVID-19’, ‘post COVID-19 syndrome’ and ‘long-haul COVID-19’. Long COVID could be the consequence of COVID-19 viral injury, stroke, hypoxia, maladaptive or abnormal inflammation, or the persistent presence of SARS-CoV-2, hypoxia-induced mitochondria dysfunction (Stefano et al., Reference Stefano, Ptacek, Ptackova, Martin and Kream2021b) and other unknown causes (Viszlayová et al., Reference Viszlayová, Sojka, Dobrodenková, Szabó, Bilec, Turzová, Ďurina, Baloghová, Borbély and Kršák2021). There is no consensus so far.

Long-term psychological or adverse mental health consequences of COVID-19 has just begun to be recognised. The episodic, ever-changing, quarantine policies around the world in the last two years have resulted in difficulties in travel, shutdowns of country borders in many countries, decrease in social contacts and also making essential or urgent visits of dependent relatives and families impossible. It is difficult to know how much some of the vague neuropsychiatric symptoms are quarantine/isolation related and psychosomatic or post-traumatic in nature.

We reviewed and discussed the fast-accumulating literature on long COVID.

Method

We searched the English language literature, including foreign-language publications with informative abstracts in English, up to December 28th, 2021, using PubMed (https://pubmed.ncbi.nlm.nih.gov), crossing the keywords “COVID-19”, “long COVID”, “stress”, “quarantine” and “isolation” respectively and in turn with the following words: psychiatry, psychosis, psychiatric disorders, depression, bipolar disorders, anxiety disorders, post-traumatic stress disorder, dementia, neurodegeneration, brain circuits, neurotransmitters, psychotropics, brain areas, serotonin (5HT), dopamine (DA), norepinephrine (NE), histamine, sigma receptor, cortisol, glucose metabolism, brain metabolism, psychological, treatment. Manuscripts identified were included in this review after evaluating the quality of the research and relevancy to the various sections of this review. Health statistics were obtained from the World Health Organization (WHO) and Centers for Disease Control and Prevention, USA (CDC) websites, accessed on December 28 2021.

Results

The renin–angiotensin aldosterone system (RAAS)

The dipeptidyl carboxypeptidase angiotensin-converting enzyme (ACE) and the mono-carboxypeptidase angiotensin-converting enzyme 2 (ACE2)

Regulatory peptides control various physiological processes in the human body. Active peptides are degraded by peptidases after performing their functions. Two amino peptidases enzymes of common ancestry but with opposing actions on the renin–angiotensin aldosterone system (RAAS), the dipeptidyl carboxypeptidase ACE and the mono-carboxypeptidase ACE2, work together to regulate many important physiological processes.

ACE cleaves the decapeptide angiotensin-1 (AngI) into the potent vasoconstrictor angiotensin-2 (AngII). AngII binds to two major receptors, AT1R (angiotensin type 1receptor) and AT2R (angiotensin type 2 receptor) (Carey, Reference Carey2005). Activation of AT1R produces vasoconstriction, pro-fibrotic and pro-inflammatory effects, all of which are important in SARS-CoV-2 infection lung injury (Simões e Silva et al., Reference Simões e Silva, Silveira, Ferreira and Teixeira2013). The importance of AT1 in the regulation of the cerebrovascular system is also being recognised. The development of biased AT1R agonists has led to new therapeutic strategies to target detrimental effects of AT1R activation (Delaitre et al., Reference Delaitre, Boisbrun, Lecat and Dupuis2021).

Activation of the less investigated AT2 receptor may produce opposite actions, antagonising the AT1R effects (Porrello et al., Reference Porrello, Delbridge and Thomas2009), especially concerning the growth- and differentiation-modulating actions of ANG II. Its effects on the regulation of cell growth, differentiation and apoptosis, may have a role in neurodevelopment and regeneration (Stoll & Unger, Reference Stoll and Unger2001).

The other peptidase ACE2 converts AngII into Ang-(1-7). Ang-(1-7) acts on the G protein-coupled Mas receptors (MasR) and produces vasodilatation. It is anti-apoptotic and anti-proliferative and anti-fibrotic. Thus, ACE2 counteracts actions of ACE, being antihypertensive and cardioprotective, and it reduces lung inflammation (Saponaro et al., Reference Saponaro, Rutigliano, Sestito, Bandini, Storti, Bizzarri and Zucchi2020).

Thus, there are two axes working together: one is the vasoconstrictive axis, renin/ACE/angiotensin II/angiotensin II receptor type 1 (AT1R), and the other is the opposing vasorelaxant axis, ACE2/angiotensin-(1-7)/Mas receptor (MasR) (Povlsen et al., Reference Povlsen, Grimm, Wehland, Infanger and Krüger2020). The two enzymes, ACE and ACE2, work together in concert and maintain homeostasis, not only in maintaining blood pressure, fluid and salt balance, but in many other important physiological processes such as the brain, kidney and heart, many of which are beginning to be discovered. For example, ACE2 may have both positive and negative roles in cancer therapies.

There are two forms of ACE2 (Batlle et al., Reference Batlle, Wysocki and Satchell2020). The full-length mACE2s are located on human cell membranes and are the binding sites for the spike (S) proteins on the envelope of the virus. These S proteins are cleaved into S1 and S2 subunits. The S1 protein/receptor binding interaction will initiate the infection (Samavati & Uhal, Reference Samavati and Uhal2020). The other soluble form, sACE2, goes into the circulation (Batlle et al., Reference Batlle, Wysocki and Satchell2020) in low concentrations. The ratio between the anchored and the soluble forms has been suggested to be related to the severity of symptoms (Scialo et al., Reference Scialo, Daniele, Amato, Pastore, Matera, Cazzola, Castaldo and Bianco2020). Both membrane-bound and soluble ACE2 degrade angiotensin II to angiotensin-(1-7). Consequently, ACE2 receptors limit several detrimental effects resulting from binding of angiotensin II to AT1 receptors, which include vasoconstriction, enhanced inflammation and thrombosis. The increased generation of angiotensin-(1-7) also triggers counter-regulatory protective effects through binding to G protein-coupled Mas receptors.

As stated, the SARS-CoV-2 virus gains entry to the human body through ACE2 (Beyerstedt et al., Reference Beyerstedt, Casaro and Rangel2021; Tai et al., Reference Tai, He, Zhang, Pu, Voronin, Jiang, Zhou and Du2020). SARS-CoV-2 has a spike protein (S protein) binding domain that is nearly identical to the earlier SARS virus, but the binding affinity for ACE2 is much higher, explaining its high virulence in comparison (Wrapp et al., Reference Wrapp, Wang, Corbett, Goldsmith, Hsieh, Abiona, Graham and McLellan2020). Preclinical data suggest ACE2 might be downregulated after SARS-CoV-2 binding; this ACE2 down-regulation induced by the virus may be especially detrimental in people of old age, suffering from hypertension, diabetes and cardiovascular disease, all of whom already shared baseline ACE2 deficiency. As enhanced and unopposed angiotensin II via the ACE→Angiotensin II→AT1 receptor axis may increase pulmonary inflammation and coagulation (Verdecchia et al., Reference Verdecchia, Cavallini, Spanevello and Angeli2020), treatments that increase ACE2 may prevent cardiopulmonary injury. Metformin, by enhancing ACE2 expression, may offer cardiopulmonary protection (Malhotra et al., Reference Malhotra, Hepokoski, McCowen and Shyy2020) and possibly neuroprotection in COVID-19 (Samuel et al., Reference Samuel, Varghese and Büsselberg2021).

Expression of ACE2 also occurred in other organs, highest in the small intestines, high in the salivary glands, testicular, kidney, thyroid and adipose tissues, but lower in the spleen, muscle, pituitary, skin and the brain (Han et al., Reference Han, Kang, Li, Ge and Gu2020; Li et al., Reference Li, Li, Zhang and Wang2020; Song et al., Reference Song, Li, Huang, Chen, Li, Liu, Chen and Duan2020). High expression of ACE2 was found in the olfactory bulb areas in the mouse (Chen et al., Reference Chen, Wang, Yu, Howard, French, Chen, Wen and Xu2021). Though ACE2 is lower in the brain compared to other organs and tissues as described above, it is highly expressed in the choroid plexus and paraventricular nuclei of the thalamus. Nuclear expression of ACE2 was found in neuronal as well as non-neuron cells, including astrocytes, oligodendrocytes and endothelial cells in the human middle temporal gyrus and posterior cingulate cortex.

The high ACE2 expression in the salivary glands and the intestine implies the common infection entry site and the corresponding salivary test for the infection. The high rate of mutations in corona viruses may change their specificity and/or binding affinity to different receptor sites (Scialo et al., Reference Scialo, Daniele, Amato, Pastore, Matera, Cazzola, Castaldo and Bianco2020). Middle East respiratory syndrome-related coronavirus (MERS-CoV), for example, binds to dipeptidyl peptidase-4 (DPP4) (Letko et al., Reference Letko, Marzi and Munster2020) which plays an important role in glucose metabolism. Glucose metabolism is known to play an important role in brain functions (see section on brain glucose metabolism below).

The SARS-CoV-2 causes a disruption in ACE/ACE2 balance (Lubbe et al., Reference Lubbe, Cozier, Oosthuizen, Acharya and Sturrock2020). A high ACE/ACE2 ratio may be detrimental in the COVID-19 infection (Pagliaro & Penna, Reference Pagliaro and Penna2020), and dysregulation of ACE/ACE2 equilibrium by SARS-CoV-2 is now being investigated for its relationship to long-term neurological complications (Haghighi et al., Reference Haghighi, Kakhki, Sato, Ghani and Rogaeva2020). RAAS activation may lead to COVID-19 progression, especially in patients with comorbidities, such as hypertension, diabetes mellitus and cardiovascular disease (Beyerstedt et al., Reference Beyerstedt, Casaro and Rangel2021).

The COVID-19 virus (SARS-CoV-2) entrance into the brain

High prevalence of neurological symptoms in patients at both the acute stage of COVID-19 and in long COVID suggests the extensive damages of SARS-CoV-2 on CNS tissues (Pezzini and Padovani Reference Pezzini and Padovani2020; Rogers et al., Reference Rogers, Watson, Badenoch, Cross, Butler, Song, Hafeez, Morrin, Rengasamy, Thomas, Ralovska, Smakowski, Sundaram, Hunt, Lim, Aniwattanapong, Singh, Hussain, Chakraborty, Burchill, Jansen, Holling, Walton, Pollak, Ellul, Koychev, Solomon, Michael, Nicholson and Rooney2021). 18F-FDG PET study of brain metabolism in COVID-19 (Rodríguez-Alfonso et al., Reference Rodríguez-Alfonso, Ruiz Solís, Silva-Hernández, Pintos Pascual, Aguado Ibáñez and Salas Antón2021) also revealed hypometabolism in areas such as the olfactory/rectus gyrus, amygdala, hippocampus, parahippocampus, cingulate cortex, pre-/post-central gyrus, thalamus/hypothalamus, cerebellum, pons and medulla. Findings reinforce the hypotheses of SARS-CoV-2 neurotropism involving multiple brain structures (Guedj et al., Reference Guedj, Campion, Dudouet, Kaphan, Bregeon, Tissot-Dupont, Guis, Barthelemy, Habert, Ceccaldi, Million, Raoult, Cammilleri and Eldin2021a, b; Sollini et al., Reference Sollini, Morbelli, Ciccarelli, Cecconi, Aghemo, Morelli, Chiola, Gelardi and Chiti2021).

Loss of smell is prominent in SARS-CoV-2 infection. Using real-time quantitative PCR (rt-qPCR), and in-situ hybridisation to detect the SARS-CoV-2 RNA plus immunohistochemistry and electron microscopy, viral RNA was found in the olfactory mucosa, and then the uvula and the medulla oblongata (Meinhardt et al., Reference Meinhardt, Radke, Dittmayer, Franz, Thomas, Mothes, Laue, Schneider, Brünink, Greuel, Lehmann, Hassan, Aschman, Schumann, Chua, Conrad, Eils, Stenzel, Windgassen, Rößler, Goebel, Gelderblom, Martin, Nitsche, Schulz-Schaeffer, Hakroush, Winkler, Tampe, Scheibe, Körtvélyessy, Reinhold, Siegmund, Kühl, Elezkurtaj, Horst, Oesterhelweg, Tsokos, Ingold-Heppner, Stadelmann, Drosten, Corman, Radbruch and Heppner2021). Using this same approach in a mouse model, Bilinska et al. (Reference Bilinska, Jakubowska, Von Bartheld and Butowt2020) showed that ACE2 is expressed in sustentacular cells of the olfactory epithelium and increased with old age. Thus, SARS-CoV-2 may cross the neural-mucosal interface in the olfactory mucosa, enter the olfactory neurons and then migrate up to the medulla oblongata (Banerjee & Viswanath, Reference Banerjee and Viswanath2020; Mahalaxmi et al., Reference Mahalaxmi, Kaavya, Mohana Devi and Balachandar2021). The respiratory and cardiovascular control centres are in the medulla oblongata, and SARS-CoV-2 attack on these neurons may contribute to the respiratory and cardiovascular symptoms. Nasal swab thus is a convenient SARS-CoV-2 detection test (Butowt & Bilinska, Reference Butowt and Bilinska2020). Other possible viral entry points less discussed include the vagal and trigeminal nerve and the compromised blood–brain barrier (BBB) with neuroinflammation (Boldrini et al., Reference Boldrini, Canoll and Klein2021).

Long COVID psychiatric disorders

Long COVID symptoms of a psychiatric nature have been reported globally. While COVID-19 was regarded as a pulmonary respiratory viral disease in the early stage like SARS, its involvement of other organs like the heart, liver, kidney and the CNS was soon recognised. In Germany and the United Kingdom, post-COVID neuropsychiatric symptoms were reported from 20% to a high of 70% (Woo et al., Reference Woo, Malsy, Pöttgen, Seddiq Zai, Ufer, Hadjilaou, Schmiedel, Addo, Gerloff, Heesen, Schulze Zur Wiesch and Friese2020; Meinhardt et al., Reference Meinhardt, Radke, Dittmayer, Franz, Thomas, Mothes, Laue, Schneider, Brünink, Greuel, Lehmann, Hassan, Aschman, Schumann, Chua, Conrad, Eils, Stenzel, Windgassen, Rößler, Goebel, Gelderblom, Martin, Nitsche, Schulz-Schaeffer, Hakroush, Winkler, Tampe, Scheibe, Körtvélyessy, Reinhold, Siegmund, Kühl, Elezkurtaj, Horst, Oesterhelweg, Tsokos, Ingold-Heppner, Stadelmann, Drosten, Corman, Radbruch and Heppner2021).

One of the largest and longest studies was from China (Huang et al., Reference Huang, Huang, Wang, Li, Ren, Gu, Kang, Guo, Liu, Zhou, Luo, Huang, Tu, Zhao, Chen, Xu, Li, Li, Peng, Li, Xie, Cui, Shang, Fan, Xu, Wang, Wang, Zhong, Wang, Wang, Zhang and Cao2021). The study involved 1733 of 2469 discharged patients with COVID-19. Patients had a median age of 57 and 52% were men. The median follow-up time after symptom onset was 186 days. Fatigue or muscle weakness (63%) and sleep difficulties (26%) were common, followed by anxiety or depression (23%). Median 6-min walking distance less than the lower limit of the patient’s normal range was 24%. Fatigue or muscle weakness, sleep difficulties and anxiety or depression continued to even 6 months after symptom onset. Being a woman and severity of illness were risk factors for persistent psychological symptoms.

The Chinese findings were echoed by Schou et al. (Reference Schou, Joca, Wegener and Bay-Richter2021). They reviewed 66 studies from Asia, Europe and North America, covering discharged patients up to 7 months. It showed that depression, post-traumatic stress disorder (PTSD), fatigue and sleep disturbances were common. In 47 studies, the incidence of depression and anxiety ranged from no indication of depression or anxiety to >30% at follow-up (up to 199 days after discharge). Risk factors, similar to the Chinese study, were found to be disease severity, duration of symptoms, and the female sex.

The high incidence of insomnia reported by Schou et al. (Reference Schou, Joca, Wegener and Bay-Richter2021) and Huang et al. (Reference Huang, Huang, Wang, Li, Ren, Gu, Kang, Guo, Liu, Zhou, Luo, Huang, Tu, Zhao, Chen, Xu, Li, Li, Peng, Li, Xie, Cui, Shang, Fan, Xu, Wang, Wang, Zhong, Wang, Wang, Zhang and Cao2021) was confirmed by Li et al. (Reference Li, Chen, Hong, Sun, Dai, Basta, Tang and Qin2021b). Their report documented about 37% of patients with COVID-19 had insomnia in the early stage which rose to 41.8% in the later stage.

Taquet et al. (Reference Taquet, Luciano, Geddes and Harrison2021) analysed US electronic data, covering 62,354 patients from 54 health-care organisations diagnosed with COVID-19. They found that the most common psychiatric diagnosis after COVID-19 diagnosis was anxiety disorder (12.8%), followed by mood disorders (9.9%). Psychotic disorder was rare in the 14–90 days after COVID-19 diagnosis (0.1%). Having a diagnosis of psychiatric disorder in the year before the COVID-19 outbreak was associated with a 65% increased risk of COVID-19. In patients with no previous psychiatric history, a diagnosis of COVID-19 was associated with increased incidence of a first psychiatric diagnosis in the following 14–90 days. The incidence of any psychiatric diagnosis in the 14–90 days after COVID-19 diagnosis was 18.1%, in which 5.8% were a first diagnosis.

A Spanish report (Méndez et al., Reference Méndez, Balanzá-Martínez, Luperdi, Estrada, Latorre, González-Jiménez, Bouzas, Yépez, Ferrando, Reyes and Menéndez2021) covered a total of 179 patients at 2 months and 171 (95.5% retention rate) at 12 months. Screening was by telephone, using questionnaires, self-reporting and screening instruments. At 12 months, the prominent symptoms included fatigue (48.5%), memory complaints (32.2%), arthromyalgia (26.9%), dyspnoea (25.7%), headache (15.8%). Neurocognitive dysfunction and psychiatric morbidity were found in 46.8% and 45% of patients, respectively. Psychiatric morbidity was at a total of 45%, including anxiety (35.1%), depression (32.2%) and PTSD (24.6%).

In summary, psychiatric symptoms are prominent in long COVID and are similar across different countries and cultures. Depression, anxiety, fatigue, insomnia, headache, somatic pain and PTSD were the common neuropsychiatric disorders, while psychosis was rare.

Depression and anxiety are known to be associated with cerebral vascular accidents, but not psychosis. Post-stroke depression (PSD) could occur in up to about 1/3 of the patients (Ahmed et al., Reference Ahmed, Khalil, Kohail, Eldesouky, Elkady and Shuaib2020; Das & Rajanikant, Reference Das and Rajanikant2018; Lee et al., Reference Lee, Tang, Yu and Cheung2007, Reference Lee, Tang, Yu and Cheung2008; Medeiros et al., Reference Medeiros, Roy, Kontos and Beach2020; Robinson & Jorge, Reference Robinson and Jorge2016; Sharma et al., Reference Sharma, Mallick, Llinas and Marsh2020). Other reports also recorded PSD prevalence at 31.1% (Schöttke & Giabbiconi, Reference Schöttke and Giabbiconi2015) and 36% (Ahmed et al., Reference Ahmed, Khalil, Kohail, Eldesouky, Elkady and Shuaib2020), with post-stroke anxiety (PSA) at 20.4% and 32%. Lifetime depression could not predict the emergence of PSD but lifetime anxiety was a good predictor of PSA (Schöttke & Giabbiconi, Reference Schöttke and Giabbiconi2015). Thus, a significant percentage of neuropsychiatric symptoms in long COVID could be the result of stroke and other cerebral vascular damages caused by the SARS-CoV-2 in the acute stage.

Controversial infection and mortality rate in COVID-19 patients with pre-existing mental disorders

Data on the mortality of patients with pre-existing mental disorders in COVID-19 have been contradictory. There are reports and data supporting both lower and higher morbidity and mortality rates in this population in COVID-19 and long COVID.

A lower incidence of symptomatic forms of COVID-19 among patients (4%) than among the clinical staff (14%) was observed in Sainte-Anne hospital, a Paris psychiatric hospital (Plaze et al., Reference Plaze, Attali, Petit, Blatzer, Simon-Loriere, Vinckier, Cachia, Chrétien and Gaillard2020, Reference Plaze, Attali, Prot, Petit, Blatzer, Vinckier, Levillayer, Chiaravalli, Perin-Dureau, Cachia, Friedlander, Chrétien, Simon-Loriere and Gaillard2021). This contradicts another report by Fond et al. (Reference Fond, Pauly, Orleans, Antonini, Fabre, Sanz, Klay, Jimeno, Leone, Lancon, Auquier and Boyer2021), who reported that schizophrenic patients admitted to acute care hospitals in Marseille, France, had much higher mortality compared to the non-SCZ patients (26.7% vs. 8.7%). In examining the data in detail, it became obvious that the report by Fond et al. (Reference Fond, Pauly, Orleans, Antonini, Fabre, Sanz, Klay, Jimeno, Leone, Lancon, Auquier and Boyer2021) had a highly skewed/sample. There were 15 schizophrenic patients only, out of a total of 1092 patients studied. 100% of the schizophrenic patients who died were institutionalised. Health conditions of the schizophrenic patients versus the other patients were also not comparable, with smokers (33.3% vs. 11.1%), suffering from cancers (20.0% vs. 5.5%) and respiratory comorbidities (26.7% vs. 4.9%).

Higher infection and mortality rate in schizophrenic patients and patients with severe mental disorders in other countries were also reported, including Israel (Tzur Bitan et al., Reference Tzur Bitan, Krieger, Kridin, Komantscher, Scheinman, Weinstein, Cohen, Cicurel and Feingold2021), Spain (Garcia-Ribera et al., Reference Garcia-Ribera, Carrasco and Ruiz-Ripoll2021), Italy (Barlati et al., Reference Barlati, Nibbio and Vita2021), Canada (Zhand & Joober, Reference Zhand and Joober2021), the United Kingdom (Hassan et al., Reference Hassan, Peek, Lovell, Carvalho, Solmi, Stubbs and Firth2021) and USA (Teixeira et al., Reference Teixeira, Krause, Ghosh, Shahani, Machado-Vieira, Lane, Boerwinkle and Soares2021; Wang et al., Reference Wang, Xu and Volkow2021b).

Karaoulanis and Christodoulou (Reference Karaoulanis and Christodoulou2021) reviewed the literature, with seven studies meeting their criteria. They found a statistically significant effect for higher infection rates and a strong statistically significant effect for higher mortality rates in patients with schizophrenia.

On the other hand, there are reports contradicting the high infection and mortality rate observations. Apart from the report by Plaze et al. (Reference Plaze, Attali, Petit, Blatzer, Simon-Loriere, Vinckier, Cachia, Chrétien and Gaillard2020, Reference Plaze, Attali, Prot, Petit, Blatzer, Vinckier, Levillayer, Chiaravalli, Perin-Dureau, Cachia, Friedlander, Chrétien, Simon-Loriere and Gaillard2021), Rivas-Ramírez et al. (Reference Rivas-Ramírez, Tendilla-Beltrán, Gómez-Mendoza, Loaiza and Flores2021) from Mexico reported their observation on 198 patients with psychiatric and neurological disorders and hospitalised in Puebla. They found the mortality rate (5.75%) was lower than that reported in Mexico (11.28–13.55%), which was higher than the worldwide average of 2.95–4.98%.

Moga et al. (Reference Moga, Teodorescu, Ifteni, Gavris and Petric2021) studied 101 schizophrenic patients tested positive for COVID-19 and treated with oral antipsychotics in a long-term facility in Brasov, Romania, between April 2020 and April 2021. They found that schizophrenics on antipsychotic treatment, when compared to 101 individuals without schizophrenia in the same hospital, showed a lower risk of SARS-CoV-2 severe infection and a likely better COVID-19 prognosis.

It may be difficult to exclude the confounding factors such as pre-morbid health conditions, comorbidities and availability/accessibility/quality of care for patients with mental disorders, institutionalised versus acute care differences, etc., in different cultures and different countries. This likely will remain controversial until data becomes available from better design studies with ample case numbers. For now, whether patients with severe mental disorders are more vulnerable to COVID-19 or not, further studies will be useful to reveal the neurobiology and interaction between the virus and the CNS.

Neurological complications

The neurological complications/consequences in long COVID can be largely grouped into three major categories:

  1. 1. Direct viral invasion of the brain neuronal and vascular structures and its consequences

  2. 2. Abnormal immune and inflammatory reaction such as ‘cytokine storm’ and its long-term consequences

  3. 3. Neurological consequences secondary to viral pulmonary and associated systemic disease including systemic inflammation, sepsis and multi-organ failure, resulting in hypoxic brain damage, encephalopathy and stroke, Guillain–Barré syndrome (GBS), acute haemorrhagic necrotising encephalopathy (ANE) and acute disseminated encephalomyelitis (ADEM) (Pezzini & Padovani, Reference Pezzini and Padovani2020; Ryoo et al., Reference Ryoo, Pyun, Baek, Suh, Kang, Wang, Youn, Yang, Kim, Park and Kim2020).

Pilotto et al. (Reference Pilotto, Cristillo, Cotti Piccinelli, Zoppi, Bonzi, Sattin, Schiavolin, Raggi, Canale, Gipponi, Libri, Frigerio, Bezzi, Leonardi and Padovani2021) in Italy studied 208 non-neurological patients hospitalised for COVID-19 disease and evaluated 165 survivors at 6 months follow-up, using a structured standardised clinical protocol. They found that these patients displayed a wide array of symptoms, including fatigue (34%), memory/attention (31%) and sleep disorders (30%). Neurological abnormalities were found in 40 % of patients, with hyposmia at 18.0%, cognitive deficits at 17.5%, postural tremor at 13.8% and subtle motor/sensory deficits at 7.6%. Older age, pre-morbid comorbidities and severity of COVID-19 were independent predictors of neurological manifestations. Some motor symptoms however may take longer to develop (Otero-Losada et al., Reference Otero-Losada, Kobiec, Udovin, Chevalier, Quarracino, Maissonave, Bordet, Capani and Perez-Lloret2020).

A report from Saudi Arabia covered 79 patients infected with SARS-CoV-2 and found a high incidence of stroke. The commonest neurological signs and symptoms were altered level of consciousness (45.9%), dizziness (11.5%) and focal neurological deficit (10.4%). Acute ischaemic stroke was seen in 18 of the 79 patients. Diabetic patients were 4 times more at risk to develop stroke while patients with respiratory failure were 21 times more likely to have a stroke (Tawakul et al., Reference Tawakul, Alharbi, Basahal, Almalki, Alharbi, Almaghrabi and Imam2021).

A systematic review by Collantes et al. (Reference Collantes, Espiritu, Sy, Anlacan and Jamora2021) covering 403 articles and 49 studies, with a total of 6,335 confirmed COVID-19 cases, reported headache, dizziness, nausea and vomiting, confusion and myalgia vascular disorders, encephalopathy, encephalitis, oculomotor nerve palsy, isolated sudden-onset anosmia, Guillain–Barré syndrome and Miller-Fisher syndrome. Similar wide spectrum of COVID-19 neurological was also summarised by others (Delorme et al., Reference Delorme, Paccoud, Kas, Hesters, Bombois, Shambrook, Boullet, Doukhi, Le Guennec, Godefroy, Maatoug, Fossati, Millet, Navarro, Bruneteau and Demeret2020; Paterson et al., Reference Paterson, Brown, Benjamin, Nortley, Wiethoff, Bharucha, Jayaseelan, Kumar, Raftopoulos, Zambreanu, Vivekanandam, Khoo, Geraldes, Chinthapalli, Boyd, Tuzlali, Price, Christofi, Morrow, McNamara, McLoughlin, Lim, Mehta, Levee, Keddie, Yong, Trip, Foulkes, Hotton, Miller, Everitt, Carswell, Davies, Yoong, Attwell, Sreedharan, Silber, Schott, Chandratheva, Perry, Simister, Checkley, Longley, Farmer, Carletti, Houlihan, Thom, Lunn, Spillane, Howard, Vincent, Werring, Hoskote, Jäger, Manji and Zandi2020).

Guillain–Barré syndrome (GBS) and its variants, dysfunction of taste and smell, and muscle injury are examples of peripheral nervous system (PNS) involvement. Hemorrhagic and ischaemic stroke, encephalitis, meningitis, encephalopathy (Kas et al., Reference Kas, Soret, Pyatigoskaya, Habert, Hesters, Le Guennec, Paccoud, Bombois and Delorme2021) ADEM, endothelialitis and venous sinus thrombosis are examples of COVID-19 CNS involvement. Thus, COVID-19 poses a large-scale threat to the whole nervous system, acutely and in the long term as well (Jha et al., Reference Jha, Ojha, Jha, Dureja, Singh, Shukla, Chellappan, Gupta, Bhardwaj, Kumar, Jeyaraman, Jain, Muthu, Kar, Kumar, Goswami, Ruokolainen, Kesari, Singh and Dua2021).

There were other less known long COVID symptoms of neurological nature. For example, strange disturbing internal vibration and tremor sensation was reported in newspaper (Wall Street Journal Dec 21, 2021). It is unknown if they were caused by autonomic nervous system damages as suggested by some.

In summary, about 30% of hospitalised COVID-19 patients developed neurological symptoms, including ataxia, agitation, delirium, headache, cerebrovascular disease, epilepsy, loss of taste and smell and diffuse corticospinal tract signs (Mao et al., Reference Mao, Jin, Wang, Hu, Chen, He, Chang, Hong, Zhou, Wang, Miao, Li and Hu2020; Helms et al., Reference Helms, Kremer, Merdji, Clere-Jehl, Schenck, Kummerlen, Collange, Boulay, Fafi-Kremer, Ohana, Anheim and Meziani2020). While the area of the brain first affected by the virus may depend on the distribution of the ACE2 receptors, the neuropsychiatric symptoms in long COVID (Jozuka et al., Reference Jozuka, Kimura, Uematsu, Fujigaki, Yamamoto, Kobayashi, Kawabata, Koike, Inada, Saito, Katsuno and Ozaki2021) are likely secondary to neurological damages from neuroinflammation, stroke, hypoxia and other causes yet to be discovered.

Age effect

Although all patients infected with COVID-19 showed neuroinflammatory changes, those with severe infections and particularly elderly patients seemed more likely to suffer from ‘cytokine storm’, referring to the excessive release of the pro-inflammatory cytokines (IL) interleukin-1,-6,-10 and tumour necrosis factor-alpha (TNF) (Garber et al., Reference Garber, Vasek, Vollmer, Sun, Jiang and Klein2018). In the brain, these cytokines activate the microglia and astrocytes to release more inflammatory cytokines in addition to neurotoxins and complement proteins (Vasek et al., Reference Vasek, Garber, Dorsey, Durrant, Bollman, Soung, Yu, Perez-Torres, Frouin, Wilton, Funk, DeMasters, Jiang, Bowen, Mennerick, Robinson, Garbow, Tyler, Suthar, Schmidt, Stevens and Klein2016; Liddelow et al., Reference Liddelow, Guttenplan, Clarke, Bennett, Bohlen, Schirmer, Bennett, Münch, Chung, Peterson, Wilton, Frouin, Napier, Panicker, Kumar, Buckwalter, Rowitch, Dawson, Dawson, Stevens and Barres2017; Xu et al., Reference Xu, He and Bai2016). These changes contribute to excitotoxicity and long-term neuronal damage and may start a neurodegenerative process. Inflammatory neuropsychiatric disorders have been previously reviewed in more detail (Leonard & Wegener, Reference Leonard and Wegener2020; Myint Reference Myint2013; Tang et al., Reference Tang, Helmeste and Leonard2021).

The expression of the neuropsychiatric symptoms of long COVID differs in different age groups. Delirium from hypoxia and metabolic complications are more likely in the vulnerable aged patients and those with dementia (Butler & Barrientos, Reference Butler and Barrientos2020; Garg, Reference Garg2020; Toniolo et al., Reference Toniolo, Scarioni, Di Lorenzo, Hort, Georges, Tomic, Nobili and Frederiksen2021), whereas encephalopathy and encephalitis, together with acute neuropsychiatric symptoms, were more likely to occur in younger patients (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). Severe fatigue is a common feature in most age groups. Elevated creatine kinase indicates severe myopathy which accounts for the debilitating fatigue and myalgia (Garg, Reference Garg2020; Orsucci et al., Reference Orsucci, Trezzi, Anichini, Blanc, Barontini, Biagini, Capitanini, Comeglio, Corsini, Gemignani, Giannecchini, Giusti, Lombardi, Marrucci, Natali, Nenci, Vannucci and Volpi2021).

Long COVID neurodegeneration

Normal CNS neuronal mitochondrial function requires high oxygen levels. SARS-CoV-2 virus can hijack mitochondrial function to cause long-lasting metabolic problems. Coupled with the inflammatory process discussed above, neurodegeneration or exacerbation of pre-existing dementia may begin (ElBini Dhouib, Reference ElBini Dhouib2021; Ge et al., Reference Ge, Zivadinov, Wang, Charidimou and Haacke2021; Roman et al., Reference Roman, Egert and Di Benedetto2021; Stefano et al., Reference Stefano, Büttiker, Weissenberger, Martin, Ptacek and Kream2021a, b; Stuckey et al., Reference Stuckey, Ong, Collins-Praino and Turner2021; Tang et al., Reference Tang, Helmeste and Leonard2017, Reference Tang, Helmeste and Leonard2021).

COVID-19 neuroinflammation may lead to decreased neurogenesis, as shown in a reduction in the size of the hippocampus, dentate gyrus and fewer granule neurons and neural progenitor cells (Mahajan et al., Reference Mahajan, Vallender, Garrett, Challagundla, Overholser, Jurjus, Dieter, Syed, Romero, Benghuzzi and Stockmeier2018; Boldrini et al., Reference Boldrini, Galfalvy, Dwork, Rosoklija, Trencevska-Ivanovska, Pavlovski, Hen, Arango and Mann2019; Klein et al., Reference Klein, Soung, Sissoko, Nordvig, Canoll, Mariani, Jiang, Bricker, Goldman, Rosoklija, Arango, Underwood, Mann, Boon, Dowrk and Boldrini2021). It is already well-established that cognitive dysfunction may persist for many months after patients have apparently recovered from COVID-19 (Troyer et al., Reference Troyer, Kohn and Hong2020; Zhou et al., Reference Zhou, Lu, Chen, Wei, Wang, Lyu, Shi and Hu2020). This points to the possibility that there is long-term damage to the neuronal networks initiated by the virus and extended and sustained by chronic neuroinflammation and the disruption of brain metabolic homeostasis. Stroke, which is common in COVID-19, is also known to be associated with the development of dementia (Kalaria et al., Reference Kalaria, Akinyemi and Ihara2016) which could be up to 18.4 %, 1 year after stroke (Craig et al., Reference Craig, Hoo, Yan, Wardlaw and Quinn2021) or 28.5% mostly at 6 months after stroke (Hénon et al., Reference Hénon, Durieu, Guerouaou, Lebert, Pasquier and Leys2001). Impaired glucose tolerance and asymptomatic hyperglycaemia are common in the elderly (Wargny et al., Reference Wargny, Potier, Gourdy, Pichelin, Amadou, Benhamou, Bonnet, Bordier, Bourron, Chaumeil, Chevalier, Darmon, Delenne, Demarsy, Dumas, Dupuy, Flaus-Furmaniuk, Gautier, Guedj, Jeandidier, Larger, Le Berre, Lungo, Montanier, Moulin, Plat, Rigalleau, Robert, Seret-Bégué, Sérusclat, Smati, Thébaut, Tramunt, Vatier, Velayoudom, Vergès, Winiszewski, Zabulon, Gourraud, Roussel, Cariou and Hadjadj2021), and the role of brain glucose metabolism in depression and neurodegeneration has been studied (Leonard & Wegener, Reference Leonard and Wegener2020).

COVID-19, diabetes and brain energy metabolism

Diabetes and obesity are cofactors which are frequently associated with vulnerability to SARS-CoV-2 infection and death.

A report summarised 9 studies in China, involving a total of 1070 patients with diabetes, out of 8807 COVID-19 case. It showed that comorbid diabetes was associated with an increased risk of disease severity or death (Guo et al., Reference Guo, Shi, Zhang, Wang, Do Vale Moreira, Zuo and Hussain2020). Another study from China demonstrated that elevated blood glucose levels led to the rapid progression and high death rates, based on data obtained from 2433 COVID-19 patients (Wang et al., Reference Wang, Shen, Tao, Fairley, Zhong, Li, Chen, Ong, Zhang, Zhang, Xing, Guo, Qin, Guan, Yang, Zhang, Zhang and He2021). It was reported that in patients of 60 years or older, the elevated blood glucose levels correlated with the respiratory rate, fever, blood CRP, lactic dehydrogenase, low serum albumin and low lymphocyte counts; these were significant factors in the progression and the severity of the disease. In addition, elevated glucose, fibrinogen and creatine kinase levels were significant risk factors for death. The authors concluded that patients with elevated blood glucose were 58% more likely to progress to hospitalisation and 3.22 times more likely to die from the infection.

Another report showed that in 952 patients with pre-existing type 2 diabetes, well-controlled blood glucose was associated with markedly lower mortality compared to individuals with poorly controlled blood glucose (Zhu et al., Reference Zhu, Jin, Wu, Hu, Wang, Bu, Sun, Wang, Qu and Chen2020a, b). Similar findings were reported by others (Aggarwal et al., Reference Aggarwal, Lippi, Lavie, Henry and Sanchis-Gomar2020; Kumar et al., Reference Kumar, Arora, Sharma, Anikhindi, Bansal, Singla, Khare and Srivastava2020; Singh & Singh, Reference Singh and Singh2020; Wang et al., Reference Wang, Ma, Zhang, Song, Wang, Ma, Xu, Wu, Duan, Yin, Luo, Xiong, Xu, Zeng and Jin2020b, c; Zhang et al., Reference Zhang, Kong, Xia, Xu, Li, Li, Yang, Wei, Wang, Li, Zheng, Sun, Xia, Liu, Zhong, Qiu, Li, Wang, Wang, Song, Liu, Xiong, Liu, Cui, Hu, Chen, Pan and Zeng2020b) and even in non-diabetics (Singh & Singh, Reference Singh and Singh2020b; Lin et al., Reference Lin, Chen, Ding, Liu, Zhou, Huang, Zhang, Liu, Zhang, Yuan, Zhang, Zhang, She, Cai, Chen and Li2021).

In a study involving 5700 patients hospitalised in the New York City area, the most common comorbidities were hypertension, obesity and diabetes. Of the patients who died, those with diabetes were more likely to have received invasive mechanical ventilation or care in the ICU compared with those who did not have diabetes (Richardson et al., Reference Richardson, Hirsch, Narasimhan, Crawford, McGinn, Davidson, Barnaby, Becker, Chelico, Cohen, Cookingham, Coppa, Diefenbach, Dominello, Duer-Hefele, Falzon, Gitlin, Hajizadeh, Harvin, Hirschwerk, Kim, Kozel, Marrast, Mogavero, Osorio, Qiu and Zanos2020).

This suggests that brain glucose metabolism might be a factor in the spread of the SARS-CoV-2 virus in the brain (Morand et al., Reference Morand, Campion, Lepine, Bosdure, Luciani, Cammilleri, Chabrol and Guedj2021).

SARS-CoV-2 infection induces the expression of glucose transporters and enhances the uptake of glucose into the tissues it infects. It also increases the activity of the glycolytic pathway enzymes. These changes in glucose metabolism have been shown to be a feature of other types of viruses such as the influenza virus (Reading et al., Reference Reading, Allison, Crouch and Anders1998) and more recently for SARS-CoV-2 (Codo et al., Reference Codo, Davanzo, Monteiro, de Souza, Muraro, Virgilio-da-Silva, Prodonoff, Carregari, de Biagi Junior, Crunfli, Jimenez Restrepo, Vendramini, Reis-de-Oliveira, Bispo Dos Santos, Toledo-Teixeira, Parise, Martini, Marques, Carmo, Borin, Coimbra, Boldrini, Brunetti, Vieira, Mansour, Ulaf, Bernardes, Nunes, Ribeiro, Palma, Agrela, Moretti, Sposito, Pereira, Velloso, Vinolo, Damasio, Proença-Módena, Carvalho, Mori, Martins-de-Souza, Nakaya, Farias and Moraes-Vieira2020). There is experimental and clinical evidence that significant change in brain glucose metabolism is a prelude to neurodegenerative changes (Leonard & Wegener, Reference Leonard and Wegener2020), a situation that is enhanced by the increased energy demands of the activated microglia (Pailla et al., Reference Pailla, El-Mir, Cynober and Blonde-Cynober2001) and, in the case of chronic major depression, by insulin receptor desensitisation, oxidative stress and hypercortisolemia.

One of the consequences of inflammation is insulin and glucocorticoid receptor resistance (Miller et al., Reference Miller, Chen, Sze, Marin, Arevalo, Doll, Ma and Cole2008; Shelton & Miller, Reference Shelton and Miller2010; Leonard, Reference Leonard and Kim2018). Functional insulin receptor insensitivity may occur as a consequence of stress-induced cortisol elevation and decrease the insulin-mediated expression of the GLUT 4 glucose transporter. Increase in TNF-alpha also contributes to the desensitisation of the insulin receptors (Solomon et al., Reference Solomon, Mishra, Palazzolo, Postlethwaite and Seyer1997). These changes result in a reduction of glucose availability to peripheral tissues and the brain (Weinstein et al., Reference Weinstein, Paquin, Pritsker and Haber1995). Such changes would be particularly relevant following the recovery from the acute stage while the inflammation remains.

The brain is a unique organ which requires glucose as the main energy source. The glucose transporters located on the BBB are vital to ensuring that sufficient glucose is available for optimal brain activity. GLUT1 is produced in brain microvasculature and ensures glucose transport across the blood–brain barrier (BBB) (Jurcovicova, Reference Jurcovicova2014). Once it enters the brain, glucose is further transported by GLUT 1 and GLUT 3 to astrocytes and neurons, respectively (Freemerman et al., Reference Freemerman, Johnson, Sacks, Milner, Kirk, Troester, Macintyre, Goraksha-Hicks, Rathmell and Makowski2014; Wang et al., Reference Wang, Pavlou, Du, Bhuckory, Xu and Chen2019).

Glucose, glutamate, lactate, fatty acid and amino acid transporters are involved in the regulation of macrophage polarisation. Metabolite transporters required for the uptake of metabolites (such as glucose, glutamate, fatty acid and amino acids) are important regulators of macrophage polarisation. They may represent novel drug targets for the treatment of disorders in which macrophages play a part, such as seen in long COVID (Cheng et al., Reference Cheng, Cai, Zong, Yu and Wei2021).

Logette et al. (Reference Logette, Lorin, Favreau, Oshurko, Coggan, Casalegno, Sy, Monney, Bertschy, Delattre, Fonta, Krepl, Schmidt, Keller, Kerrien, Scantamburlo, Kaufmann and Markram2021) provided evidence that elevated glucose in the pulmonary airway surface liquid facilitates the major entry point for the virus. This breaks down the primary innate antiviral defence in the lungs and facilitates the viral infection, stimulates the release of pro-inflammatory cytokines and causing the acute respiratory distress syndrome. In diabetes, there is endothelial dysfunction or leaky endothelium, resulting in hypercoagulation, thrombosis and vascular complications. SARS-CoV-2 can gain entry into endothelial cells via the endothelial cell surface ACE2 receptors (Varghese et al., Reference Varghese, Samuel, Liskova, Kubatka and Büsselberg2021). Cumulative evidence suggests that a glycolytic trait can influence the course of the disease by promoting viral tropism and negatively modulate the immune response and functional integrity of tissues, including endothelium. Finally, elevated blood glucose acts synergistically with COVID-19 to inactivate ACE2 which dysregulates glycaemic control in all those cell types that are infected by the virus.

Once SARS-Cov-2 virus enters the cell, like other viruses, it switches the cellular energy metabolism from the aerobic to the anaerobic state thereby ensuring ATP synthesis provided by glycolysis without the requirement of molecular oxygen. At the same time, glucose transport is increased and coupled with the increased activity of hexokinase and lactate dehydrogenase (Fontaine et al., Reference Fontaine, Sanchez, Camarda and Lagunoff2015; Ritter et al., Reference Ritter, Wahl, Freund, Genzel and Reichl2010; Sanchez & Lagunoff, Reference Sanchez and Lagunoff2015). The replication of COVID-19 is entirely dependent on ATP provided by the elevated glucose (Codo et al., Reference Codo, Davanzo, Monteiro, de Souza, Muraro, Virgilio-da-Silva, Prodonoff, Carregari, de Biagi Junior, Crunfli, Jimenez Restrepo, Vendramini, Reis-de-Oliveira, Bispo Dos Santos, Toledo-Teixeira, Parise, Martini, Marques, Carmo, Borin, Coimbra, Boldrini, Brunetti, Vieira, Mansour, Ulaf, Bernardes, Nunes, Ribeiro, Palma, Agrela, Moretti, Sposito, Pereira, Velloso, Vinolo, Damasio, Proença-Módena, Carvalho, Mori, Martins-de-Souza, Nakaya, Farias and Moraes-Vieira2020). While the data supporting these mechanisms are largely dependent on in vitro studies, there is clinical evidence demonstrating that elevated blood glucose explains the variance and the severity of the COVID-19 infection as discussed above. Epidemiological, clinical and in-depth experimental studies have identified an increase in blood glucose as a key factor in the spread of the virus throughout the body including the brain. Controlling the blood glucose level in infected patients could therefore be a practical way to reduce the severity of the disease and contribute to a reduction in the death rate. It could form the basis for treatment with anti-hyperglycaemic drugs such as metformin, supported by a low carbohydrate diet or ketone diet as an alternative energy source to glucose. More extensive and detailed studies are essential to validate, or invalidate, this hypothesis.

Psychotropics, sigma-1 receptor and antihistamines in long COVID

Whether to continue on psychotropics in patients suffering from long COVID is an important clinical decision. In this regard, the sporadic reports on the protective action of antidepressants and antipsychotics on COVID-19 patients suggest that this is an important area for further investigation.

Antidepressant drug’s protective effect on long COVID

In Missouri, USA, 115 outpatients completed a randomised trial. Patients treated with fluvoxamine, compared with placebo, had a lower likelihood of clinical deterioration over 15 days. Clinical deterioration occurred in none of the 80 patients in the fluvoxamine group but in 6 of 72 patients in the placebo group (Lenze et al., Reference Lenze, Mattar, Zorumski, Stevens, Schweiger, Nicol, Miller, Yang, Yingling, Avidan and Reiersen2020).

In a cell culture model to test small molecule acting on the homeostasis of the endolysosomal host-pathogen interface, fluoxetine impaired endolysosomal acidification and the accumulation of cholesterol within the endosomes. Fluoxetine, an inhibitor of acid sphingomyelinase (FIASMA), was found to inhibit the entry and propagation of SARS-CoV-2. It also showed potent antiviral activity against two influenza A virus subtypes. The FIASMAs amiodarone and imipramine also showed similar effect. Thus, the FIASMA group of small molecules may offer opportunities for the development of host-directed therapy to counteract enveloped viruses, including SARS-CoV-2 (Schloer et al., Reference Schloer, Brunotte, Goretzko, Mecate-Zambrano, Korthals, Gerke, Ludwig and Rescher2020).

There are earlier reports of anti-microbial/anti-parasitic (Hewlett et al., Reference Hewlett, Pearson, Zilberstein and Dwyer1985) or cytotoxic effect of antidepressant drugs. Tricyclic antidepressant drugs, such as clomipramine and imipramine, were reported to be cytotoxic against human protozoan parasites Leishmania donovani and Leishmania major. The mechanism of action was hypothesised to cause cellular death by non-specific mechanisms, probably involving a general increase in membrane permeability (Zilberstein & Dwyer, Reference Zilberstein and Dwyer1984; Zilberstein et al., Reference Zilberstein, Liveanu and Gepstein1990).

Amitriptyline was also reported to be antibacterial (against 254 strains, with 72 gram-positive and 181 gram-negative), anti-fungal and anti-virulent strains of Salmonella typhimurium. It inhibited both Cryptococcus and Candida albicans as well (Mandal et al., Reference Mandal, Sinha, Kumar Jena, Ghosh and Samanta2010). In a pre- and post-infection H5N1-infection mouse model, significant alleviation of acute lung injury by amitriptyline was reported (Huang et al., Reference Huang, Zhang, Liu, Zhao, Zhang, Qin, Li, Li, Zhou, Jin and Jiang2020). Imipramine was reported to alter the sterol profile in Leishmania amazonensis and increases its sensitivity to miconazole (Andrade-Neto et al., Reference Andrade-Neto, Pereira, do Canto-Cavalheiro and Torres-Santos2016). In Leishmaniasis, clomipramine, in µM concentrations, stimulated nitric oxide production in host macrophages and led to mitochondrial depolarisation in the parasites. Coupled with the inhibition of trypanothione reductase induced strong oxidative stress in the parasites, it induced programmed cell death (da Silva Rodrigues et al., Reference da Silva Rodrigues, Miranda, Volpato, Ueda-Nakamura and Nakamura2019).

Microglia detect and subsequently clear microbial pathogens and injured tissue. They adapt their phenotype depending on whether they participate in acute defence against pathogenic organisms (’M1’-phenotype) or in clearing damaged tissues and performing repair activities (’M2’-phenotype). Stimulation of pattern recognition receptors by viruses or vaccines, presence of bacterial membrane components such as bacterial lipopolysaccharides (LPS), promotes M1 polarisation. A less known action of antidepressant treatment and agents, electroconvulsive shock (ECT), and vagus nerve stimulation (VNS), inhibit LPS-induced microglia/macrophage M1 polarisation and inflammation (Kalkman & Feuerbach, Reference Kalkman and Feuerbach2016). How much the protective effect of antidepressant agents against COVID-19 and long COVID is related to their inhibition of microglial M1 polarisation would require further research.

An important issue which needs to be resolved is that in many of these in vitro experiments, high concentrations of antidepressant drugs (in mM and high µM) were used. A concentration of 30 mg per Kg body weight of amitriptyline (Zilberstein & Dwyer, Reference Zilberstein and Dwyer1984) is equivalent to 1500 mg dosage. This dosage is fatal in human.

Antipsychotic’s protective effect in long COVID

Canal-Rivero et al. (Reference Canal-Rivero, Catalán-Barragán, Rubio-García, Garrido-Torres, Crespo-Facorro and Ruiz-Veguilla2021) found patients with severe mental disorders and good compliance on antipsychotics were less likely to contract COVID-19. They also had better outcomes following infection, than the general population.

In Romania, Moga et al. (Reference Moga, Teodorescu, Ifteni, Gavris and Petric2021) reported no deaths in their patients with schizophrenia. The patient group had a higher number of cases with pulmonary and metabolic comorbidities but there were fewer severe cases compared to the control group. Some markers of inflammation (CRP and fibrinogen) were significantly lower in the patients also.

These reports led to the hypothesis that psychotropic drugs have a prophylactic action against SARS-CoV-2 or have an antiviral action.

Some psychotropics have indeed been investigated for their anti-microbe properties.

Chlorpromazine (CPZ) was claimed to be antiviral (Hewlett et al., Reference Hewlett, Lee, Molnar, Foldeak, Pine, Weaver and Aszalos1997; Plaze et al., Reference Plaze, Attali, Petit, Blatzer, Simon-Loriere, Vinckier, Cachia, Chrétien and Gaillard2020, Reference Plaze, Attali, Prot, Petit, Blatzer, Vinckier, Levillayer, Chiaravalli, Perin-Dureau, Cachia, Friedlander, Chrétien, Simon-Loriere and Gaillard2021), maybe via the inhibition of clathrin-mediated endocytosis (Pho et al., Reference Pho, Ashok and Atwood2000). Inhibition of HIV infection of H9 cells by chlorpromazine derivatives was also reported (Hewlett et al., Reference Hewlett, Lee, Molnar, Foldeak, Pine, Weaver and Aszalos1997). Recent in vitro studies have reported that CPZ exhibits anti-MERS-CoV and anti-SARS-CoV-1 activity in monkey VeroE6 cells, with an IC50 (half maximal inhibitory concentration) of 8.2 µm, half maximal cytotoxic concentration (CC50) of 13.5 µm. In human A549-ACE2 cells, CPZ was found to have anti-SARS-CoV-2 activity, with IC50 of 11.3 µm and CC50 of 23.1 µm. However, similar to the case of antidepressant drugs, such high drug concentration in the µM range is unlikely to be achieved clinically. There were arguments that a high chlorpromazine concentration could be achieved in the human saliva and in the brain (Tsuneizumi et al., Reference Tsuneizumi, Babb and Cohen1992; Wiesel & Alfredsson, Reference Wiesel and Alfredsson1976), much higher than in the plasma (May et al., Reference May, Van Putten, Jenden and Cho1978). However, this requires further validation.

Other studies reported the immunomodulatory effects of CPZ, such as increasing blood levels of IgM (Zucker et al., Reference Zucker, Zarrabi, Schubach, Varma, Derman, Lysik, Habicht and Seitz1990). In the mouse septic shock model, CPZ caused a decrease in IL-2, IL-4, IFN alpha, TNF and GM-CSF pro-inflammatory cytokines, an increase in IL-10 and anti-inflammatory cytokine (Bertini et al., Reference Bertini, Garattini, Delgado and Ghezzi1993; Mengozzi et al., Reference Mengozzi, Fantuzzi, Faggioni, Marchant, Goldman, Orencole, Clark, Sironi, Benigni and Ghezzi1994; Tarazona et al., Reference Tarazona, González-García, Zamzami, Marchetti, Frechin, Gonzalo, Ruiz-Gayo, van Rooijen, Martínez and Kroemer1995). In a recent meta-analysis of 12 studies, consisting of 961 patients with schizophrenia vs 729 controls, on the impact of antipsychotics on the production of serum interleukin-6 (IL-6) (Kappelmann et al., Reference Kappelmann, Dantzer and Khandaker2021) a pro-inflammatory cytokine, it was found that antipsychotic treatment was associated with a decrease of IL-6 in patients (Zhou et al., Reference Zhou, Tian and Han2021).

For other antipsychotics, trifluoperazine was also found to reduce inflammatory response by suppressing pro-inflammatory cytokines in mice (Park et al., Reference Park, Park, Lee, Kim, Jang, Han, Jung, Shin, Bae, Kang and Park2019) while a transcriptomic analysis revealed that aripiprazole could revert effects induced by COVID-19 on gene expression in patients (Crespo-Facorro et al., Reference Crespo-Facorro, Ruiz-Veguilla, Vázquez-Bourgon, Sánchez-Hidalgo, Garrido-Torres, Cisneros, Prieto and Sainz2021; Dratcu & Boland, Reference Dratcu and Boland2021).

May et al. (Reference May, Slitzky, Rostama, Barlow and Houseknecht2020), on the other hand, cautioned the use of antipsychotics in COVID-19, citing the anti-inflammatory action as risky in viral infections.

Sigma-1 receptor

The endoplasmic reticulum (ER) resident multi-functional protein sigma-1 receptor is an essential inhibitor of cytokine production (Rosen et al., Reference Rosen, Seki, Fernández-Castañeda, Beiter, Eccles, Woodfolk and Gaultier2019). Sigma-1 receptors play crucial roles in cellular signal transduction and interact with receptors, ion channels, lipids and kinases. Changes in their functions and expression may lead to various neuropsychiatric disorders, including affective and cognitive disorders (Salaciak & Pytka, Reference Salaciak and Pytka2021), pain (Gris et al., Reference Gris, Cobos, Zamanillo and Portillo-Salido2015; Ruiz-Cantero et al., Reference Ruiz-Cantero, González-Cano, Tejada, Santos-Caballero, Perazzoli, Nieto and Cobos2021), neurodegeneration and neuro-restoration (Ruscher & Wieloch, Reference Ruscher and Wieloch2015), addiction and DA function (Hong et al., Reference Hong, Yano, Hiranita, Chin, McCurdy, Su, Amara and Katz2017), memory impairments (Sałaciak and Pytka 2021). When stimulated by ligands or undergoing prolonged stress, sigma-1 receptors translocate from the mitochondrion-associated ER membrane to the ER reticular network and plasma membrane to regulate a variety of functional proteins such as ion channels, receptors and kinases. Sigma-1 receptors thus serve as inter-organelle signalling modulators and coordinators locally at the mitochondrion-associated ER membrane and remotely at the plasmalemma/plasma membrane (Su et al., Reference Su, Hayashi, Maurice, Buch and Ruoho2010, Reference Su, Su, Nakamura and Tsai2016).

Sigma-1 receptors modulate a number of neurotransmitters and neurotransmission processes, including glutamate-NMDA, 5HT, NE and DA and BDNF signalling (Skuza, Reference Skuza2012), pain (Sánchez-Fernández et al., Reference Sánchez-Fernández, Entrena, Baeyens and Cobos2017) neuroplasticity, neuroinflammation (Kourrich et al., Reference Kourrich, Su, Fujimoto and Bonci2012; Jerčić et al., Reference Jerčić, Kostić, Vitlov Uljević, Vukušić Pušić, Vukojević and Filipović2019; Jia et al., Reference Jia, Cheng, Wang and Zhen2018) and neurorepair (Lisak et al., Reference Lisak, Nedelkoska and Benjamins2020).

Some psychoactive drugs show high to moderate affinity for sigma-1 receptors, including haloperidol, fluvoxamine and sertraline, cocaine and methamphetamine, whereas phenytoin allosterically modulates sigma-1 receptors (Cobos et al., Reference Cobos, Entrena, Nieto, Cendán and Del Pozo2008). We have previously reported the affinity of DA agents to brain sigma receptors and the decrease of sigma receptors in post-mortem schizophrenia brains, implying that sigma receptors may play a role in psychiatric disorders (Helmeste et al., Reference Helmeste, Tang, Fang and Li1996a, b, Reference Helmeste, Tang, Li and Fang1997, Reference Helmeste, Shioiri, Mitsuhashi and Tang1999; Tang et al., Reference Tang, Helmeste, Fang, Li, Vu, Bunney, Potkin and Jones1997). The decrease in sigma-1 receptor density in mental disorders has also been observed by other research groups (Reynolds et al., Reference Reynolds, Brown and Middlemiss1991; Weissman et al., Reference Weissman, Casanova, Kleinman, London and De Souza1991). What needs to be determined is whether this is a result of drug treatment or whether reduced sigma receptor numbers are also consistently seen in drug-naïve subjects. Considering that knockdown/knockout of sigma-1 receptors reduces SARS-CoV-2 replication, we also need to know if reduced sigma-1 receptor expression in schizophrenic patients makes these subjects less sensitive to SARS-CoV-2 infection (Brimson et al., Reference Brimson, Prasanth, Malar, Brimson, Thitilertdecha and Tencomnao2021; Gordon et al., Reference Gordon, Hiatt, Bouhaddou, Rezelj, Ulferts, Braberg, Jureka, Obernier, Guo, Batra, Kaake, Weckstein, Owens, Gupta, Pourmal, Titus, Cakir, Soucheray, McGregor, Cakir, Jang, O’Meara, Tummino, Zhang, Foussard, Rojc, Zhou, Kuchenov, Hüttenhain, Xu, Eckhardt, Swaney, Fabius, Ummadi, Tutuncuoglu, Rathore, Modak, Haas, Haas, Naing, Pulido, Shi, Barrio-Hernandez, Memon, Petsalaki, Dunham, Marrero, Burke, Koh, Vallet, Silvas, Azumaya, Billesbølle, Brilot, Campbell, Diallo, Dickinson, Diwanji, Herrera, Hoppe, Kratochvil, Liu, Merz, Moritz, Nguyen, Nowotny, Puchades, Rizo, Schulze-Gahmen, Smith, Sun, Young, Zhao, Asarnow, Biel, Bowen, Braxton, Chen, Chio, Chio, Deshpande, Doan, Faust, Flores, Jin, Kim, Lam, Li, Li, Li, Li, Liu, Lo, Lopez, Melo, Moss, Nguyen, Paulino, Pawar, Peters, Pospiech, Safari, Sangwan, Schaefer, Thomas, Thwin, Trenker, Tse, Tsui, Wang, Whitis, Yu, Zhang, Zhang, Zhou, Saltzberg, Hodder, Shun-Shion, Williams, White, Rosales, Kehrer, Miorin, Moreno, Patel, Rihn, Khalid, Vallejo-Gracia, Fozouni, Simoneau, Roth, Wu, Karim, Ghoussaini, Dunham, Berardi, Weigang, Chazal, Park, Logue, McGrath, Weston, Haupt, Hastie, Elliott, Brown, Burness, Reid, Dorward, Johnson, Wilkinson, Geyer, Giesel, Baillie, Raggett, Leech, Toth, Goodman, Keough, Lind, Klesh, Hemphill, Carlson-Stevermer, Oki, Holden, Maures, Pollard, Sali, Agard, Cheng, Fraser, Frost, Jura, Kortemme, Manglik, Southworth, Stroud, Alessi, Davies, Frieman, Ideker, Abate, Jouvenet, Kochs, Shoichet, Ott, Palmarini, Shokat, García-Sastre, Rassen, Grosse, Rosenberg, Verba, Basler, Vignuzzi, Peden, Beltrao and Krogan2020; Hashimoto, Reference Hashimoto2021). Note however that sigma-1 receptor agonists stimulate brain-derived neurotrophic factor (BDNF) levels in the brain (Dalwadi et al., Reference Dalwadi, Kim, Schetz, Schreihofer and Kim2021; Hashimoto, Reference Hashimoto2013) and may be useful in treating COVID-19 infection irrespective of their effects on viral replication (Hashimoto, Reference Hashimoto2021).

Also important is whether the reduced sigma-1 receptor number seen in schizophrenic patient brain is likewise reduced in peripheral body tissues of the same subjects. Besides being expressed in the brain, sigma-1 receptors are widely expressed in the lung, liver, adrenal glands, testis, kidney and heart (Abdullah et al., Reference Abdullah, Alam, Aishwarya, Miriyala, Panchatcharam, Bhuiyan, Peretik, Orr, James, Osinska, Robbins, Lorenz and Bhuiyan2018; Dalwadi et al., Reference Dalwadi, Kim, Schetz, Schreihofer and Kim2021; Hashimoto, Reference Hashimoto2013; Lever et al., Reference Lever, Litton and Fergason-Cantrell2015; Patone et al., Reference Patone, Mei, Handunnetthi, Dixon, Zaccardi, Shankar-Hari, Watkinson, Khunti, Harnden, Coupland, Channon, Mills, Sheikh and Hippisley-Cox2021; Shen et al., Reference Shen, Kapfhamer, Minnella, Kim, Won, Chen, Huang, Low, Massa and Swanson2017; Su et al., Reference Su, Su, Nakamura and Tsai2016; Vela, Reference Vela2020).

As a chaperone protein, the sigma-1 receptor does not appear to exhibit biological functions independent of other protein partners (Jia et al., Reference Jia, Cheng, Wang and Zhen2018). However, cardiac dysfunction is seen in sigma-1 receptor knockout mice and is associated with impaired mitochondrial dynamics and bioenergetics (Abdullah et al., Reference Abdullah, Alam, Aishwarya, Miriyala, Panchatcharam, Bhuiyan, Peretik, Orr, James, Osinska, Robbins, Lorenz and Bhuiyan2018). Considering that myocarditis has been reported in a small number of younger patients after COVID-19 vaccination or infection (Patone et al., Reference Patone, Mei, Handunnetthi, Dixon, Zaccardi, Shankar-Hari, Watkinson, Khunti, Harnden, Coupland, Channon, Mills, Sheikh and Hippisley-Cox2021), would sigma-1 receptor levels in these subjects predict or predispose these subjects to certain types of treatment? Lung tissue also expresses sigma-1 receptors (Lever et al., Reference Lever, Litton and Fergason-Cantrell2015) but how this affects COVID-19 therapeutics is not known yet.

SARS-CoV-2 enters cells via the spike glycoprotein through a process called endocytosis. Subsequent SARS-CoV-2 replication takes place in an ER-derived intermediate compartment in the ER-Golgi (Harrison et al., Reference Harrison, Lin and Wang2020). Substantial evidence suggests that the sigma-1 receptor plays a role in the pathophysiology of a number of psychiatric and neurodegenerative disorders. Fluvoxamine’s agonistic effects at the sigma-1 receptor (S1R) may reduce damaging effects of the inflammatory response.

In other studies, using the comparative viral-human protein–protein interaction map, it was revealed that the sigma-1 receptor in the ER plays an important role in SARS-CoV-2 replication in cells. Knockout and knockdown of SIGMAR1 (sigma-1 receptor, encoded by SIGMAR1) caused robust reductions in SARS-CoV-2 replication. This indicates that the sigma-1 receptor may be a key therapeutic target for SARS-CoV-2 replication and lead to the proposed repurposing of traditional CNS drugs that have a high affinity at the sigma-1 receptor (i.e. fluvoxamine, donepezil, ifenprodil) for the treatment of SARS-CoV-2-infected patients, and also other agents such as cutamesine and arketamine. (Hashimoto, Reference Hashimoto2021).

Of the common antidepressant drugs, only fluvoxamine possesses agonist and sertraline antagonist action at the sigma receptor with clinically relevant affinity (Ki) at the receptor. Imipramine, escitalopram and fluoxetine are only agonists at high concentrations as their affinity (Ki) are all in the 200–300 nm range, well above the concentration seen in clinical situation (Narita et al., Reference Narita, Hashimoto, Tomitaka and Minabe1996; Ishima et al., Reference Ishima, Fujita and Hashimoto2014).

Antipsychotics such as haloperidol (Ki = 4 nm), perphenazine (Ki = 12 nm), fluphenazine (Ki = 17 nm), trifluoperazine (Ki = 67 nm), pimozide (Ki = 144 nm), chlorpromazine (Ki = 180 nm) and triflupromazine (Ki = 214 nm) all possess high to moderate affinity at the sigma-1 receptor (Tam & Cook, Reference Tam and Cook1984). These agents given at normal clinical doses would tag on the sigma receptors.

In summary, sigma receptors agents (Brimson et al., Reference Brimson, Brimson, Chomchoei and Tencomnao2020) are beginning to emerge as potential modulators of neuroplasticity and the neuroinflammatory process, both of which are important in the development of effective treatment for long COVID.

Antihistamine

Histamine is an important mediator of the immune response to infection. Antihistamines thus play an important role in the management of inflammatory diseases and the cytokine storm of COVID-19 (Eldanasory et al., Reference Eldanasory, Eljaaly, Memish and Al-Tawfiq2020). Usage of diphenhydramine, hydroxyzine and azelastine was associated with reduced incidence of SARS-CoV-2 positivity in subjects greater than age 61. Diphenhydramine, hydroxyzine and azelastine were found to exhibit direct antiviral activity against SARS-CoV-2 in vitro. Mechanisms by which specific antihistamines exert antiviral effects is not clear, but hydroxyzine binds to (ACE2) and the sigma-1 receptor (Reznikov et al., Reference Reznikov, Norris, Vashisht, Bluhm, Li, Liao, Brown, Butte and Ostrov2021).

Unpublished Chinese data claiming that the mortality rate for patients with COVID-19 taking famotidine was 14% compared with 27% for those not taking the drug triggered off the rapid launch of a 21 million study (Borrell, Reference Borrell2020; Ghosh et al., Reference Ghosh, Chatterjee, Dubey and Lavie2020). Use of famotidine was associated with a decreased risk of in-hospital mortality (Mather et al., Reference Mather, Seip and McKay2020). High-dose oral famotidine is associated with improved patient-reported outcomes in non-hospitalised patients with COVID-19 (Janowitz et al., Reference Janowitz, Gablenz, Pattinson, Wang, Conigliaro, Tracey and Tuveson2020). Another cohort study of cetirizine and famotidine found them to be safe and effective in reducing the progression in symptom severity, presumably by minimising the histamine-mediated cytokine storm (Hogan Ii et al., Reference Hogan Ii, Hogan Iii, Cannon, Rappai, Studdard, Paul and Dooley2020).

The use of Nigella sativa (black cumin seeds) to treat the patients with COVID-19 was being analysed, as it has been shown to possess antihistaminic action, in addition to being antiviral, antioxidant, anti-inflammatory, anticoagulant, immunomodulatory, bronchodilatory, antipyretic and analgesic (Maideen, Reference Maideen2020).

An Electronic health record (EHR) review suggested that 3 allergy medications (cetirizine, diphenhydramine and hydroxyzine) could prevent SARS-CoV-2 infection. It found that only use of diphenhydramine was associated with a negative SARS-CoV-2 test. Selection bias was cautioned, citing the observation that increasing age and public insurance were associated with a higher adjusted odds of test negativity, while being Black or Hispanic was significantly associated with test positivity. (Thompson et al., Reference Thompson, Gurka, Filipp, Schatz, Mercado, Ostrov, Atkinson and Rasmussen2021).

It is important to remember that early psychotropic drugs were derived from the atropine molecule and many, such as the tricyclic antidepressant drugs (TCAs), retain significant antihistamine properties (Tang & Tang, Reference Tang and Tang2019). Some of them, for example, doxepine, is a potent antihistamine, and more potent than the commonly used antihistamine diphenhydramine. 11C-doxepin was a popular ligand used to study histamine H1 receptor occupancy (Tashiro et al., Reference Tashiro, Duan, Kato, Miyake, Watanuki, Ishikawa, Funaki, Iwata, Itoh and Yanai2008).

CYP enzyme

Anderson (Reference Anderson2021) argued that fluvoxamine might also exert beneficial effects in COVID patients through its high ability to substantially increase (∼ 2–3-fold) night-time plasma levels of melatonin through inhibition of the melatonin-metabolising liver enzymes CYP1A2 and CYP2C19 (von Bahr et al., Reference von Bahr, Ursing, Yasui, Tybring, Bertilsson and Röjdmark2000). Many other psychotropic drugs also inhibit CYP enzymes but fluvoxamine, specifically possesses a strong action against CYP2C19 and CYP1A2. Paroxetine and fluoxetine also possess significant inhibition against both CYP1A2 and CYP2C19. Whether only CYP2C19 and CYP1A2, and no other CYP enzyme inhibition (such as CYP2D6) is related to protection against COVID-19 would require further confirmation.

Stress from isolation and the vulnerable

Although depression, anxiety, insomnia and PTSD are observed to be common in long COVID, the mechanism is still unclear. In many of the reports concerning neuropsychiatric symptoms in long COVID, quality of the diagnostic and inclusion criteria were often unclear. Telephone, questionnaires, self-reporting, review of records were often used in time of a pandemic and social distancing. With the quarantine measures imposed in many parts of the world, quarantine/travel ban related economic downturn and job losses, social isolations and many other adversaries, stress-related symptoms and emotional complaints could be difficult to separate from true depression, anxiety and PTSD cases that met strict criteria. This may explain the relative low numbers of psychosis in long COVID.

Health-care professionals are particularly under tremendous stress, especially those caring for COVID-19 patients in the frontline. They suffered heavy mental workload (Ching et al., Reference Ching, Ng, Lee, Yee, Lim, Ranita, Devaraj, Ooi and Cheong2021; Pollock et al., Reference Pollock, Campbell, Cheyne, Cowie, Davis, McCallum, McGill, Elders, Hagen, McClurg, Torrens and Maxwell2020; Mo et al., Reference Mo, Deng, Zhang, Lang, Liao, Wang, Qin and Huang2020; Zhan et al., Reference Zhan, Zhao, Yuan, Liu, Liu, Gui, Zheng, Zhou, Qiu, Chen, Yu and Li2020; Wu et al., Reference Wu, Li, Geng, Wang, Wang and Zhang2021; Shan et al., Reference Shan, Shang, Yan, Lu, Hu and Ye2021), facing shortage of protective equipment, worked long hours and serviced high numbers of critical patients. It is also stressful facing dying COVID-19 patients, feeling helpless and unable to help. There is the fear of carrying virus back to family. Thus, anxiety, depression, burnout, addiction and PTSD could be the outcome of a stressful profession (El-Hage et al., Reference El-Hage, Hingray, Lemogne, Yrondi, Brunault, Bienvenu, Etain, Paquet, Gohier, Bennabi, Birmes, Sauvaget, Fakra, Prieto, Bulteau, Vidailhet, Camus, Leboyer, Krebs and Aouizerate2020; Li et al., Reference Li, Hu, Chen, Zhu, Wu, Li, Zhang and Liu2021). A digital learning package developed to mitigate the psychological impact of COVID on frontline health workers was accessed 17,633 times within 7 days of completion (Blake et al., Reference Blake, Bermingham, Johnson and Tabner2020). The contents included practical items for health-care workers such as psychological first aid, self-care strategies (e.g. rest, work breaks, sleep, shift work, fatigue, healthy lifestyle behaviours), managing emotions (e.g. moral injury, coping, guilt, grief, fear, anxiety, depression, preventing burnout and psychological trauma) (Blake et al., Reference Blake, Bermingham, Johnson and Tabner2020). It is understandable that this is a group that would be highly vulnerable for long COVID neuropsychiatric disorders.

The isolated and quarantined subjects consisted of another group with high stress and vulnerability for long COVID. Apart from isolation stress, there is reduction in physical activity (Burtscher et al., Reference Burtscher, Burtscher and Millet2020; Razai et al., Reference Razai, Oakeshott, Kankam, Galea and Stokes-Lampard2020; Rivers & Ihle, Reference Rivers and Ihle2020) producing or accelerating sarcopenia, a deterioration of muscle mass and function and increases in body fat (Kirwan et al., Reference Kirwan, McCullough, Butler, Perez de Heredia, Davies and Stewart2020; Simpson & Katsanis, Reference Simpson and Katsanis2020). Many also experienced the problem of obtaining accurate and reliable news and associated fear and anxiety (Nowak et al., Reference Nowak, Miedziarek, Pełczyński and Rzymski2021). A stressful life could produce the vague health symptoms in long COVID.

Being a woman and severity of illness were risk factors for persistent psychological symptoms in long COVID. Female COVID or SARS survivors appeared to have higher stress levels and higher levels of depression and anxiety and contributed to the high incidence of depression and anxiety in long COVID (Huang et al., Reference Huang, Huang, Wang, Li, Ren, Gu, Kang, Guo, Liu, Zhou, Luo, Huang, Tu, Zhao, Chen, Xu, Li, Li, Peng, Li, Xie, Cui, Shang, Fan, Xu, Wang, Wang, Zhong, Wang, Wang, Zhang and Cao2021).

Long COVID in the young age groups may also be stress-related. Stress could be from prolonged school absence, concerns about loss of the family, isolation/quarantine related loss of friendships, loss of peer supports, domestic violence and child maltreatment. Children and adolescents with disabilities, existing mental health problems, migrant background and poverty are especially vulnerable (Fegert et al., Reference Fegert, Vitiello, Plener and Clemens2020).

The elderly and live-alone, with or without dementia, plus their care givers, are definitely a high-risk group for long COVID (Liu et al., Reference Liu, Liu, Zhang, An and Zhao2021). Research has shown that people suffering from dementia have a relatively high risk of contracting severe COVID-19. They are also at risk of additional neuropsychiatric disturbances as a result of quarantine/lockdown measures and stringent social isolation (Cations et al., Reference Cations, Day, Laver, Withall and Draper2021; Giebel et al., Reference Giebel, Cannon, Hanna, Butchard, Eley, Gaughan, Komuravelli, Shenton, Callaghan, Tetlow, Limbert, Whittington, Rogers, Rajagopal, Ward, Shaw, Corcoran, Bennett and Gabbay2021; Numbers & Brodaty, Reference Numbers and Brodaty2021; Ryoo et al., Reference Ryoo, Pyun, Baek, Suh, Kang, Wang, Youn, Yang, Kim, Park and Kim2020). Lack of social engagements with families and friends, cancelled day care centre programmes may worsen the cognitive, physical and neuropsychological condition of the patients with dementia. Being confined at home without contact with the outside or without updated news may increase levels of stress, anxiety and a feeling of loneliness and depression (Xu & Liu, Reference Xu and Liu2021). This is critical for dementia patients, as stress is known to be detrimental to patients with cognitive impairments. Maintaining physical activity during isolation is important (Morrison et al., Reference Morrison, Jurak and Starc2020; Oren et al., Reference Oren, Gersh and Blumenthal2020). In addition to the patients, we must also focus on the well-being of families and caregivers who may be suffering from reduced public health-care support or home care services during the COVID-19 outbreak.

Mohamed-Hussein et al. (Reference Mohamed-Hussein, Amin, Makhlouf, Makhlouf, Galal, Abd-Elaal, Abdeltawab, Kholief and Hashem2021) found that during the acute phase, hospitalised patients had more respiratory symptoms, while non-hospitalised patients had more neuropsychiatric symptoms (84.4% vs. 69.5%). This implies long COVID may not be related to the severity of COVID-19 infection, in contrast to some other studies, which showed that long COVID was related to severity of infection.

Thus, there are specific groups of COVID-19 patients who were living a highly stressful life during COVID. Managing stress in those infected obviously is an urgent public health task (Hagger et al., Reference Hagger, Keech and Hamilton2020). It would be interesting to investigate if incidence of long COVID neuropsychiatric disorders (such as depression, anxiety, PTSD, psychosomatic and other stress-related disorders) would be lower in communities which implemented effective stress management programmes (Cheng et al., Reference Cheng, Xia, Pang, Wu, Jiang, Li, Wang, Ling, Chang, Wang, Dai, Lin and Bi2020b).

Areas of further research in long COVID

In the shortage of truly effective agents against the SARS-CoV-2 virus, repurposing/trials of existing drugs are taking place in many parts of the world. These included the controversial animal anti-parasite drug ivermectin (Ortega-Guillén et al., Reference Ortega-Guillén, Meneses and Coila2021; Rajter et al., Reference Rajter, Sherman, Fatteh, Vogel, Sacks and Rajter2021), the commonly used antimalarials chloroquine (CQ) and hydroxychloroquine (HCQ) (Abena et al., Reference Abena, Decloedt, Bottieau, Suleman, Adejumo, Sam-Agudu, Muyembe TamFum, Seydi, Eholie, Mills, Kallay, Zumla and Nachega2020) and psychotropics described above. These two areas are important for future research in the fight against COVID-19.

Targeting glucose metabolism

There are a number of possible strategies to reverse the changes in brain glucose metabolism particularly in the early stages of the infection. Reducing neuroinflammation, re-sensitising the glucose transporters and increasing brain energy metabolism by dietary manipulations are practical approaches which seem worthy of consideration. A reduction in glucose during the acute infective stage would not only reduce viral replication but also help to restore the innate immune defence mechanisms. However, during the post-infective long COVID stage, the adverse changes in the brain are mainly due to neuroinflammation which has a major impact on brain glucose metabolism. The anti-diabetic agent metformin would appear to have advantages in treating long COVID patients. The mechanisms of action of metformin and its potential advantages make it a viable candidate drug for repurposing against SARS-CoV-2 infection (Ibrahim et al., Reference Ibrahim, Lowe, Bramante, Shah, Klatt, Sherwood, Aronne, Puskarich, Tamariz, Palacio, Bomberg, Usher, King, Benson, Vojta, Tignanelli and Ingraham2021; Samuel et al., Reference Samuel, Varghese and Büsselberg2021; Malhotra et al., Reference Malhotra, Hepokoski, McCowen and Shyy2020).

Besides its efficacy in regulating blood glucose levels without inducing hypoglycaemia, metformin is an effective anti-inflammatory agent (Dehkordi et al., Reference Dehkordi, Sattari, Khoshdel and Kasiri2016). Metformin inhibits the formation of advanced glycation products which are required for the formation of the glycan tree structure which are essential for viral pathogenesis. An added advantage is the reduction in mitochondrial synthesis of ROS which would further contribute to the beneficial effects of metformin in treating long COVID patients (Beisswenger & Ruggiero-Lopez, Reference Beisswenger and Ruggiero-Lopez2003; Bellin et al., Reference Bellin, de Wiza, Wiernsperger and Rösen2006).

So far, the clinical benefit of metformin in COVID-19 patients is limited but there is evidence that diabetic patients with COVID infection have benefited (Scheen, Reference Scheen2020); similar results have been recorded in patients with heart failure (Cheng et al., Reference Cheng, Liu, Li, Zhang, Lei, Qin, Chen, Deng, Lin, Chen, Song, Xia, Huang, Liu, Cai, Zhang, Zhou, Zhang, Wang, Ma, Xu, Yang, Ye, Mao, Huang, Xia, Zhang, Guo, Zhu, Lu, Yuan, Wei, She, Ji and Li2020a). Metformin’s anti-thrombotic effects, its potential attenuation of endothelial dysfunction, inhibition of viral entry and infection and modulation of inflammatory and immune responses would also be added advantages (Samuel et al., Reference Samuel, Varghese and Büsselberg2021). However, only appropriate controlled and randomised clinical trials will establish if metformin is an effective treatment both at the acute stage and in long COVID.

Regulation of the carbohydrate content of the diet could be a practical approach to limiting the virus. A ketone rich diet, and a low carbohydrate diet, could provide ketones as an alternative energy source for neurons. Unlike other organs, the brain requires acetoacetate and gamma-hydroxybutyrate to compensate for the lack of glucose. In addition, there is evidence that ketones activate the protective gamma-delta T-cell responses involved in antiviral protection against the influenza virus (Goldberg et al., Reference Goldberg, Molony, Kudo, Sidorov, Kong, Dixit and Iwasaki2019). There are several clinical studies in progress exploring the protective value of low carbohydrate diets in those with COVID-19 infection.

Targeting sigma receptor and histamine receptor

The role of the sigma receptor and histamine in both inflammation and viral infection is an important area for future research in long COVID. The reported decrease in sigma receptor numbers in post-mortem schizophrenic brains, the reported protective role of certain psychotropics against COVID-19 infection and the observed reduction in COVID-19 infection in patients with schizophrenia, taken together, suggests that sigma receptor agents should be tested for their potential anti-COVID-19 effect, in properly designed experiments and clinical trials.

Conclusion

As of this date, there appears to be no well-established or accepted treatment to reduce, or prevent, long COVID. Understandably, clinical attention has been directed primarily at the impact of COVID-19 on the potentially lethal acute infection stage. However, there is now increasing concern regarding the pathological changes and health problems which occur in a significant number of patients following their apparent recovery from the acute infection, the ‘long COVID’. The neuropsychiatric symptoms of long COVID are obstacles for returning to a normal life and a potentially heavy burden on the health system of all countries. Whether the long COVID neuropsychiatric symptoms reported are stress-related and psychosomatic, or metabolic and neurodegeneration-related are all important areas for further research. The suspected protective action of some psychotropics against COVID-19, amid contradictory reports on the higher/lower mortality rates of patients with severe mental disorders infected, is also an important area to pursue. The neuropsychopharmacology of continuing neuroinflammation, glucose metabolism, the role of ACE2, sigma and histamine receptors are all important areas for further research and may lead to repurposing or novel therapeutics against COVID-19. Meanwhile, cognitive and neuropsychological function, and signs of neurodegeneration, should be closely monitored in COVID-19 survivors (Serrano-Castro et al., Reference Serrano-Castro, Estivill-Torrús, Cabezudo-García, Reyes-Bueno, Ciano Petersen, Aguilar-Castillo, Suárez-Pérez, Jiménez-Hernández, Moya-Molina, Oliver-Martos, Arrabal-Gómez and Rodríguez de Fonseca2020).

In this review, we have reviewed the spectrum of long-term neuropsychiatric and psychological impacts of COVID-19. We have also attempted to identify some areas of long COVID that might provide useful paths to research for new treatments of long COVID.

References

Abdullah, CS, Alam, S, Aishwarya, R, Miriyala, S, Panchatcharam, M, Bhuiyan, MAN, Peretik, JM, Orr, AW, James, J, Osinska, H, Robbins, J, Lorenz, JN, Bhuiyan, MS (2018) Cardiac dysfunction in the sigma 1 receptor knockout mouse associated with impaired mitochondrial dynamics and bioenergetics. Journal of the American Heart Assoc 7(20), e009775. doi: 10.1161/JAHA.118.009775.CrossRefGoogle ScholarPubMed
Abena, PM, Decloedt, EH, Bottieau, E, Suleman, F, Adejumo, P, Sam-Agudu, NA, Muyembe TamFum, JJ, Seydi, M, Eholie, SP, Mills, EJ, Kallay, O, Zumla, A, Nachega, JB (2020) Chloroquine and hydroxychloroquine for the prevention or treatment of COVID-19 in Africa: caution for inappropriate off-label use in healthcare settings. American Journal of Tropical Medicine and Hygiene 102(6), 11841188.CrossRefGoogle ScholarPubMed
Aggarwal, G, Lippi, G, Lavie, CJ, Henry, BM and Sanchis-Gomar, F (2020) Diabetes mellitus association with coronavirus disease 2019 (COVID-19) severity and mortality: a pooled analysis. Journal of Diabetes 12(11), 851855.CrossRefGoogle ScholarPubMed
Ahmed, ZM, Khalil, MF, Kohail, AM, Eldesouky, IF, Elkady, A and Shuaib, A (2020) The prevalence and predictors of post-stroke depression and anxiety during COVID-19 pandemic. Journal of Stroke and Cerebrovascular Diseases 29(12), 105315.CrossRefGoogle ScholarPubMed
Anderson, GM (2021) Fluvoxamine, melatonin and COVID-19. Psychopharmacology 238(2), 611. doi: 10.1007/s00213-020-05753-z.CrossRefGoogle ScholarPubMed
Andrade-Neto, VV, Pereira, TM, do Canto-Cavalheiro, M and Torres-Santos, EC (2016) Imipramine alters the sterol profile in Leishmania amazonensis and increases its sensitivity to miconazole. Parasit Vectors 9, 183.CrossRefGoogle ScholarPubMed
Asadi-Pooya, AA, Akbari, A, Emami, A, Lotfi, M, Rostamihosseinkhani, M, Nemati, H, Barzegar, Z, Kabiri, M, Zeraatpisheh, Z, Farjoud-Kouhanjani, M, Jafari, A, Sasannia, F, Ashrafi, S, Nazeri, M, Nasiri, S, Shahisavandi, M (2021) Long COVID syndrome-associated brain fog. Journal of Medical Virology. doi: 10.1002/jmv.27404.Google ScholarPubMed
Banerjee, D and Viswanath, B (2020) Neuropsychiatric manifestations of COVID-19 and possible pathogenic mechanisms: insights from other coronaviruses. Asian Journal of Psychiatry 54(6), 102350. doi: 10.1016/j.ajp.2020.102350.CrossRefGoogle ScholarPubMed
Barlati, S, Nibbio, G and Vita, A (2021) Schizophrenia during the COVID-19 pandemic. Current Opinion in Psychiatry 34(3), 203210.CrossRefGoogle ScholarPubMed
Batlle, D, Wysocki, J and Satchell, K (2020) Soluble angiotensin-converting enzyme 2: a potential approach for coronavirus infection therapy? Clinical Science 134(5), 543545.CrossRefGoogle ScholarPubMed
Beisswenger, P and Ruggiero-Lopez, D (2003) Metformin inhibition of glycation processes. Diabetes & Metabolism 2(4), 6S956S103.Google Scholar
Bellin, C, de Wiza, DH, Wiernsperger, NF and Rösen, P (2006) Generation of reactive oxygen species by endothelial and smooth muscle cells: influence of hyperglycemia and metformin. Hormone and Metabolic Research 38(11), 732739.CrossRefGoogle ScholarPubMed
Bertini, R, Garattini, S, Delgado, R and Ghezzi, P (1993) Pharmacological activities of chlorpromazine involved in the inhibition of tumour necrosis factor production in vivo in mice. Immunology 79(2), 217219.Google ScholarPubMed
Beyerstedt, S, Casaro, EB and Rangel, É.B (2021) COVID-19: angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. European Journal of Clinical Microbiology & Infectious Diseases 40(5), 905919.CrossRefGoogle ScholarPubMed
Bilinska, K, Jakubowska, P, Von Bartheld, CS and Butowt, R (2020) Expression of the SARS-CoV-2 entry proteins, ACE2 and TMPRSS2, in cells of the olfactory epithelium: identification of cell types and trends with age. ACS Chemical Neuroscience 11(11), 15551562.CrossRefGoogle ScholarPubMed
Blake, H, Bermingham, F, Johnson, G and Tabner, A (2020) Mitigating the psychological impact of COVID-19 on healthcare workers: a digital learning package. International Journal of Environmental Research and Public Health 17(9), 2997. doi: 10.3390/ijerph17092997.CrossRefGoogle ScholarPubMed
Boldrini, M, Canoll, PD and Klein, RS (2021) How COVID-19 affects the brain. JAMA Psychiatry 78(6), 682683.CrossRefGoogle ScholarPubMed
Boldrini, M, Galfalvy, H, Dwork, AJ, Rosoklija, GB, Trencevska-Ivanovska, I, Pavlovski, G, Hen, R, Arango, V and Mann, JJ (2019) Resilience is associated with larger dentate gyrus, while suicide decedents with major depressive disorder have fewer granule neurons. Biological Psychiatry 85(10), 850862.CrossRefGoogle ScholarPubMed
Borrell, B (2020) New York clinical trial quietly tests heartburn remedy against coronavirus. Science. https://www.sciencemag.org/news/2020/04/new-york-clinical-trial-quietly-tests-heartburn-remedy-against-coronavirus CrossRefGoogle Scholar
Brimson, JM, Brimson, S, Chomchoei, C and Tencomnao, T (2020) Using sigma-ligands as part of a multi-receptor approach to target diseases of the brain. Expert Opinion on Therapeutic Targets 24(10), 10091028.CrossRefGoogle ScholarPubMed
Brimson, JM, Prasanth, MI, Malar, DS, Brimson, S, Thitilertdecha, P and Tencomnao, T (2021) Drugs that offer the potential to reduce hospitalization and mortality from SARS-CoV-2 infection. the possible role of the sigma 1 receptor and autophagy. Expert Opinion on Therapeutic Targets 25(6), 435449. doi: 10.1080/14728222.2021.1952987.CrossRefGoogle ScholarPubMed
Burtscher, J, Burtscher, M and Millet, GP (2020) (Indoor) isolation, stress, and physical inactivity: vicious circles accelerated by COVID-19? Scandinavian Journal of Medicine & Science in Sports 30(8), 15441545.CrossRefGoogle ScholarPubMed
Butler, MJ and Barrientos, RM (2020) The impact of nutrition on COVID-19 susceptibility and long-term consequences. Brain Behavior and Immunity 87, 5354.CrossRefGoogle ScholarPubMed
Butowt, R and Bilinska, K (2020) SARS-CoV-2: olfaction, brain infection, and the urgent need for clinical samples allowing earlier virus detection. ACS Chemical Neuroscience 11(9), 12001203.CrossRefGoogle ScholarPubMed
Canal-Rivero, M, Catalán-Barragán, R, Rubio-García, A, Garrido-Torres, N, Crespo-Facorro, B, Ruiz-Veguilla, M and IBIS Translational Psychiatry Group (2021) Lower risk of SARS-CoV2 infection in individuals with severe mental disorders on antipsychotic treatment: a retrospective epidemiological study in a representative Spanish population. Schizophrenia Research 229, 5354.CrossRefGoogle Scholar
Carey, RM (2005) Update on the role of the AT2 receptor. Current Opinion in Nephrology and Hypertension 14(1), 6771.CrossRefGoogle ScholarPubMed
Cations, M, Day, S, Laver, K, Withall, A and Draper, B (2021) People with young-onset dementia and their families experience distinctive impacts of the COVID-19 pandemic and associated restrictions. International Psychogeriatrics 33(8), 839841.CrossRefGoogle ScholarPubMed
Chen, R, Wang, K, Yu, J, Howard, D, French, L, Chen, Z, Wen, C and Xu, Z (2021) The spatial and cell-type distribution of SARS-CoV-2 receptor ACE2 in the human and mouse brains. Frontiers in Neurology 11, 573095. doi: 10.3389/fneur.2020.573095.CrossRefGoogle ScholarPubMed
Cheng, J, Cai, W, Zong, S, Yu, Y and Wei, F (2021) Metabolite transporters as regulators of macrophage polarization. Naunyn-Schmiedeberg’s Archives of Pharmacology. doi: 10.1007/s00210-021-02173-4.Google ScholarPubMed
Cheng, P, Xia, G, Pang, P, Wu, B, Jiang, W, Li, YT, Wang, M, Ling, Q, Chang, X, Wang, J, Dai, X, Lin, X, Bi, X (2020b) COVID-19 epidemic peer support and crisis intervention via social media. Community Mental Health Journal 56(5), 786792.CrossRefGoogle ScholarPubMed
Cheng, X, Liu, YM, Li, H, Zhang, X, Lei, F, Qin, JJ, Chen, Z, Deng, KQ, Lin, L, Chen, MM, Song, X, Xia, M, Huang, X, Liu, W, Cai, J, Zhang, XJ, Zhou, F, Zhang, P, Wang, Y, Ma, X, Xu, Q, Yang, J, Ye, P, Mao, W, Huang, X, Xia, J, Zhang, BH, Guo, J, Zhu, L, Lu, Z, Yuan, Y, Wei, X, She, ZG, Ji, YX and Li, H (2020a) Metformin is associated with higher incidence of acidosis, but not mortality, in individuals with COVID-19 and pre-existing type 2 diabetes. Cell Metabolism 32(4), 537547.CrossRefGoogle Scholar
Ching, SM, Ng, KY, Lee, KW, Yee, A, Lim, PY, Ranita, H, Devaraj, NK, Ooi, PB and Cheong, AT (2021) Psychological distress among healthcare providers during COVID-19 in Asia: systematic review and meta-analysis. PLoS One 16(10), e0257983. doi: 10.1371/journal.pone.0257983.CrossRefGoogle ScholarPubMed
Chippa, V, Aleem, A and Anjum, F (2021) Post acute coronavirus (COVID-19) syndrome, oct 1, StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing, 2021Google Scholar
Cobos, EJ, Entrena, JM, Nieto, FR, Cendán, CM and Del Pozo, E (2008) Pharmacology and therapeutic potential of sigma(1) receptor ligands. Current Neuropharmacology 6(4), 344366.CrossRefGoogle ScholarPubMed
Codo, AC, Davanzo, GG, Monteiro, LB, de Souza, GF, Muraro, SP, Virgilio-da-Silva, JV, Prodonoff, JS, Carregari, VC, de Biagi Junior, CAO, Crunfli, F, Jimenez Restrepo, JL, Vendramini, PH, Reis-de-Oliveira, G, Bispo Dos Santos, K, Toledo-Teixeira, DA, Parise, PL, Martini, MC, Marques, RE, Carmo, HR, Borin, A, Coimbra, LD, Boldrini, VO, Brunetti, NS, Vieira, AS, Mansour, E, Ulaf, RG, Bernardes, AF, Nunes, TA, Ribeiro, LC, Palma, AC, Agrela, MV, Moretti, ML, Sposito, AC, Pereira, FB, Velloso, LA, Vinolo, MAR, Damasio, A, Proença-Módena, JL, Carvalho, RF, Mori, MA, Martins-de-Souza, D, Nakaya, HI, Farias, AS and Moraes-Vieira, PM (2020) Elevated glucose levels favor SARS-CoV-2 infection and monocyte response through a HIF-1α/glycolysis-dependent axis. Cell Metabolism 32(3), 437446.CrossRefGoogle ScholarPubMed
Collantes, M, Espiritu, AI, Sy, M, Anlacan, V and Jamora, R (2021) Neurological manifestations in COVID-19 infection: a systematic review and meta-analysis. The Canadian Journal of Neurological Sciences. Le journal canadien des sciences neurologiques 48(1), 6676.CrossRefGoogle ScholarPubMed
Craig, L, Hoo, ZL, Yan, TZ, Wardlaw, J and Quinn, TJ (2021) Prevalence of dementia in ischaemic or mixed stroke populations: systematic review and meta-analysis. Journal of Neurology, Neurosurgery, and Psychiatry. doi: 10.1136/jnnp-2020-325796.Google ScholarPubMed
Crespo-Facorro, B, Ruiz-Veguilla, M, Vázquez-Bourgon, J, Sánchez-Hidalgo, AC, Garrido-Torres, N, Cisneros, JM, Prieto, C and Sainz, J (2021) Aripiprazole as a candidate treatment of COVID-19 identified through genomic analysis. Frontiers in Pharmacology 12, 646701. doi: 10.3389/fphar.2021.646701.CrossRefGoogle ScholarPubMed
da Silva Rodrigues, JH, Miranda, N, Volpato, H, Ueda-Nakamura, T and Nakamura, CV (2019) The antidepressant clomipramine induces programmed cell death in Leishmania amazonensis through a mitochondrial pathway. Parasitology Research 118(3), 977989.CrossRefGoogle ScholarPubMed
Dalwadi, DA, Kim, S, Schetz, J, Schreihofer, DA and Kim, S (2021) Brain-derived neurotrophic factor for high throughput evaluation of selective sigma-1 receptor ligands. Journal of Pharmacological and Toxicological Methods. 10.1016/j.vascn.2021.107129.Google ScholarPubMed
Das, J and Rajanikant, GK (2018) Post stroke depression: the sequelae of cerebral stroke. Neuroscience Biobehavioral Reviews 90(1), 104114.CrossRefGoogle ScholarPubMed
Davis, HE, Assaf, GS, McCorkell, L, Wei, H, Low, RJ, Re’em, Y, Redfield, S, Austin, JP and Akrami, A (2021) Characterizing long COVID in an international cohort: 7 months of symptoms and their impact. EClinicalMedicine 38(10252), 101019. doi: 10.1016/j.eclinm.2021.101019.CrossRefGoogle Scholar
Dehkordi, EH, Sattari, F, Khoshdel, A and Kasiri, K (2016) Effect of folic acid and metformin on insulin resistance and inflammatory factors of obese children and adolescents. Journal of Research in Medical Sciences 21(1), 71. doi: 10.4103/1735-1995.189669.CrossRefGoogle ScholarPubMed
Delaitre, C, Boisbrun, M, Lecat, S and Dupuis, F (2021) Targeting the angiotensin II type 1 receptor in cerebrovascular diseases: biased signaling raises new hopes. International Journal of Molecular Sciences 22(13), 6738. doi: 10.3390/ijms22136738.CrossRefGoogle ScholarPubMed
Delorme, C, Paccoud, O, Kas, A, Hesters, A, Bombois, S, Shambrook, P, Boullet, A, Doukhi, D, Le Guennec, L, Godefroy, N, Maatoug, R, Fossati, P, Millet, B, Navarro, V, Bruneteau, G, Demeret, S and Pourcher V CoCo-Neurosciences study group and COVID SMIT PSL Study Group (2020) COVID-19-related encephalopathy: a case series with brain FDG-positron-emission tomography/computed tomography findings. European Journal of Neurology 27(12), 26512657.CrossRefGoogle ScholarPubMed
Dewanjee, S, Vallamkondu, J, Kalra, RS, Puvvada, N, Kandimalla, R and Reddy, PH (2021) Emerging COVID-19 neurological manifestations: present outlook and potential neurological challenges in COVID-19 pandemic. Molecular Neurobiology 58(9), 46944715.CrossRefGoogle ScholarPubMed
Dratcu, L and Boland, X (2021) Can antipsychotic use protect from COVID-19? Schizophrenia Research 236(3), 12. doi: 10.1016/j.schres.2021.07.035.CrossRefGoogle ScholarPubMed
El-Hage, W, Hingray, C, Lemogne, C, Yrondi, A, Brunault, P, Bienvenu, T, Etain, B, Paquet, C, Gohier, B, Bennabi, D, Birmes, P, Sauvaget, A, Fakra, E, Prieto, N, Bulteau, S, Vidailhet, P, Camus, V, Leboyer, M, Krebs, MO and Aouizerate, B (2020) Les professionnels de santé face à la pandémie de la maladie à coronavirus (COVID-19) : quels risques pour leur santé mentale ? [Health professionals facing the coronavirus disease 2019 (COVID-19) pandemic: What are the mental health risks?]. Encephale 46(3S), S73S80.CrossRefGoogle Scholar
ElBini Dhouib, I (2021) Does coronaviruses induce neurodegenerative diseases? A systematic review on the neurotropism and neuroinvasion of SARS-CoV-2. Drug Discoveries & Therapeutics 14(6), 262272.CrossRefGoogle ScholarPubMed
Eldanasory, OA, Eljaaly, K, Memish, ZA and Al-Tawfiq, JA (2020) Histamine release theory and roles of antihistamine in the treatment of cytokines storm of COVID-19, travel med infect dis 2020 Sep-Oct, Vol. 37, pp. 101874.Google Scholar
Fegert, JM, Vitiello, B, Plener, PL and Clemens, V (2020) Challenges and burden of the Coronavirus 2019 (COVID-19) pandemic for child and adolescent mental health: a narrative review to highlight clinical and research needs in the acute phase and the long return to normality. Child and Adolescent Psychiatry and Mental Health 14(1), 20.CrossRefGoogle Scholar
Fond, G, Pauly, V, Orleans, V, Antonini, F, Fabre, C, Sanz, M, Klay, S, Jimeno, MT, Leone, M, Lancon, C, Auquier, P, Boyer, L (2021) Increased in-hospital mortality from COVID-19 in patients with schizophrenia. Encephale 47(2), 8995.CrossRefGoogle ScholarPubMed
Fontaine, KA, Sanchez, EL, Camarda, R and Lagunoff, M (2015) Dengue virus induces and requires glycolysis for optimal replication. Journal of Virology 89(4), 23582366.CrossRefGoogle ScholarPubMed
Freemerman, AJ, Johnson, AR, Sacks, GN, Milner, JJ, Kirk, EL, Troester, MA, Macintyre, AN, Goraksha-Hicks, P, Rathmell, JC, Makowski, L (2014) Metabolic reprogramming of macrophages: glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. Journal of Biological Chemistry 289(11), 78847896.CrossRefGoogle ScholarPubMed
Garber, C, Vasek, MJ, Vollmer, LL, Sun, T, Jiang, X and Klein, RS (2018) Astrocytes decrease adult neurogenesis during virus-induced memory dysfunction via IL-1. Nature Immunology 19(2), 151161.CrossRefGoogle ScholarPubMed
Garcia-Ribera, C, Carrasco, M and Ruiz-Ripoll, A (2021) Covid-19, hypercoagulability and risk of mortality in schizophrenia. Actas Españolas de Psiquiatría 49(4), 194195.Google Scholar
Garg, M, Maralakunte, M, Garg, S, Dhooria, S, Sehgal, I, Bhalla, AS, Vijayvergiya, R, Grover, S, Bhatia, V, Jagia, P, Bhalla, A, Suri, V, Goyal, M, Agarwal, R, Puri, GD, Sandhu, MS (2021) The conundrum of ‘Long-COVID-19’: a narrative review. International Journal of General Medicine 14, 24912506.CrossRefGoogle ScholarPubMed
Garg, RK (2020) Spectrum of neurological manifestations in Covid-19: a review. Neurology India 68(3), 560572.CrossRefGoogle ScholarPubMed
Ge, Y, Zivadinov, R, Wang, M, Charidimou, A and Haacke, EM (2021) Editorial: update on vascular contributions to age-related neurodegenerative diseases and cognitive impairment - research of ISNVD 2020 meeting. Frontiers in Neurology 12, 797486. doi: 10.3389/fneur.2021.797486.CrossRefGoogle ScholarPubMed
Ghosh, R, Chatterjee, S, Dubey, S and Lavie, CJ (2020) Famotidine against SARS-CoV2: A hope or hype? Mayo Clinic Proceedings 95(8), 17971799.CrossRefGoogle ScholarPubMed
Giebel, C, Cannon, J, Hanna, K, Butchard, S, Eley, R, Gaughan, A, Komuravelli, A, Shenton, J, Callaghan, S, Tetlow, H, Limbert, S, Whittington, R, Rogers, C, Rajagopal, M, Ward, K, Shaw, L, Corcoran, R, Bennett, K, Gabbay, M (2021) Impact of COVID-19 related social support service closures on people with dementia and unpaid carers: a qualitative study. Aging & Mental Health 25(7), 12811288.CrossRefGoogle ScholarPubMed
Goldberg, EL, Molony, RD, Kudo, E, Sidorov, S, Kong, Y, Dixit, VD and Iwasaki, A (2019) Ketogenic diet activates protective γδ T cell responses against influenza virus infection. Science Immunology 4(41), eaav2026. doi: 10.1126/sciimmunol.aav2026.CrossRefGoogle ScholarPubMed
Gordon, DE, Hiatt, J, Bouhaddou, M, Rezelj, VV, Ulferts, S, Braberg, H, Jureka, AS, Obernier, K, Guo, JZ, Batra, J, Kaake, RM, Weckstein, AR, Owens, TW, Gupta, M, Pourmal, S, Titus, EW, Cakir, M, Soucheray, M, McGregor, M, Cakir, Z, Jang, G, O’Meara, MJ, Tummino, TA, Zhang, Z, Foussard, H, Rojc, A, Zhou, Y, Kuchenov, D, Hüttenhain, R, Xu, J, Eckhardt, M, Swaney, DL, Fabius, JM, Ummadi, M, Tutuncuoglu, B, Rathore, U, Modak, M, Haas, P, Haas, KM, Naing, ZZC, Pulido, EH, Shi, Y, Barrio-Hernandez, I, Memon, D, Petsalaki, E, Dunham, A, Marrero, MC, Burke, D, Koh, C, Vallet, T, Silvas, JA, Azumaya, CM, Billesbølle, C, Brilot, AF, Campbell, MG, Diallo, A, Dickinson, MS, Diwanji, D, Herrera, N, Hoppe, N, Kratochvil, HT, Liu, Y, Merz, GE, Moritz, M, Nguyen, HC, Nowotny, C, Puchades, C, Rizo, AN, Schulze-Gahmen, U, Smith, AM, Sun, M, Young, ID, Zhao, J, Asarnow, D, Biel, J, Bowen, A, Braxton, JR, Chen, J, Chio, CM, Chio, US, Deshpande, I, Doan, L, Faust, B, Flores, S, Jin, M, Kim, K, Lam, VL, Li, F, Li, J, Li, YL, Li, Y, Liu, X, Lo, M, Lopez, KE, Melo, AA, Moss, FR 3rd, Nguyen, P, Paulino, J, Pawar, KI, Peters, JK, Pospiech, TH Jr, Safari, M, Sangwan, S, Schaefer, K, Thomas, PV, Thwin, AC, Trenker, R, Tse, E, Tsui, TKM, Wang, F, Whitis, N, Yu, Z, Zhang, K, Zhang, Y, Zhou, F, Saltzberg, D, QCRG Structural Biology Consortium, Hodder, AJ, Shun-Shion, AS, Williams, DM, White, KM, Rosales, R, Kehrer, T, Miorin, L, Moreno, E, Patel, AH, Rihn, S, Khalid, MM, Vallejo-Gracia, A, Fozouni, P, Simoneau, CR, Roth, TL, Wu, D, Karim, MA, Ghoussaini, M, Dunham, I, Berardi, F, Weigang, S, Chazal, M, Park, J, Logue, J, McGrath, M, Weston, S, Haupt, R, Hastie, CJ, Elliott, M, Brown, F, Burness, KA, Reid, E, Dorward, M, Johnson, C, Wilkinson, SG, Geyer, A, Giesel, DM, Baillie, C, Raggett, S, Leech, H, Toth, R, Goodman, N, Keough, KC, Lind, AL, Zoonomia Consortium, Klesh, RJ, Hemphill, KR, Carlson-Stevermer, J, Oki, J, Holden, K, Maures, T, Pollard, KS, Sali, A, Agard, DA, Cheng, Y, Fraser, JS, Frost, A, Jura, N, Kortemme, T, Manglik, A, Southworth, DR, Stroud, RM, Alessi, DR, Davies, P, Frieman, MB, Ideker, T, Abate, C, Jouvenet, N, Kochs, G, Shoichet, B, Ott, M, Palmarini, M, Shokat, KM, García-Sastre, A, Rassen, JA, Grosse, R, Rosenberg, OS, Verba, KA, Basler, CF, Vignuzzi, M, Peden, AA, Beltrao, P, Krogan, NJ. (2020) Comparative host-coronavirus protein interaction networks reveal pan-viral disease mechanisms. Science 370(6521), eabe9403. doi: 10.1126/science.abe9403.CrossRefGoogle ScholarPubMed
Gris, G, Cobos, EJ, Zamanillo, D and Portillo-Salido, E (2015) Sigma-1 receptor and inflammatory pain. Inflammation Research 64(6), 377381.CrossRefGoogle ScholarPubMed
Guedj, E, Campion, JY, Dudouet, P, Kaphan, E, Bregeon, F, Tissot-Dupont, H, Guis, S, Barthelemy, F, Habert, P, Ceccaldi, M, Million, M, Raoult, D, Cammilleri, S, Eldin, C (2021a) 18F-FDG brain PET hypometabolism in patients with long COVID. European Journal of Nuclear Medicine and Molecular Imaging 48(9), 28232833.CrossRefGoogle ScholarPubMed
Guedj, E, Million, M, Dudouet, P, Tissot-Dupont, H, Bregeon, F, Cammilleri, S and Raoult, D (2021b) 18F-FDG brain PET hypometabolism in post-SARS-CoV-2 infection: substrate for persistent/delayed disorders? European Journal of Nuclear Medicine and Molecular Imaging 48, 592595.CrossRefGoogle ScholarPubMed
Guo, L, Shi, Z, Zhang, Y, Wang, C, Do Vale Moreira, NC, Zuo, H and Hussain, A (2020) Comorbid diabetes and the risk of disease severity or death among 8807 COVID-19 patients in China: a meta-analysis. Diabetes Research and Clinical Practice 166, 108346. doi: 10.1016/j.diabres.2020.108346.CrossRefGoogle ScholarPubMed
Hagger, MS, Keech, JJ and Hamilton, K (2020) Managing stress during the coronavirus disease 2019 pandemic and beyond: reappraisal and mindset approaches. Stress and Health 36(3), 396401.CrossRefGoogle ScholarPubMed
Haghighi, MM, Kakhki, EG, Sato, C, Ghani, M and Rogaeva, E (2020) The intersection between COVID-19, the gene family of ACE2 and Alzheimer’s disease. Neuroscience Insights 15, 2633105520975743. doi: 10.1177/2633105520975743.CrossRefGoogle ScholarPubMed
Han, T, Kang, J, Li, G, Ge, J and Gu, J (2020) Analysis of 2019-nCoV receptor ACE2 expression in different tissues and its significance study. Annals of Translational Medicine 8(17), 1077. doi: 10.21037/atm-20-4281.CrossRefGoogle ScholarPubMed
Harrison, AG, Lin, T and Wang, P (2020) Mechanisms of SARS-CoV-2 transmission and pathogenesis. Trends in Immunology 41(12), 11001115.CrossRefGoogle ScholarPubMed
Hashimoto, K (2013) Sigma-1 receptor chaperone and brain-derived neurotrophic factor: emerging links between cardiovascular disease and depression. Progress in Neurobiology 100, 1529.CrossRefGoogle ScholarPubMed
Hashimoto, K (2021) Repurposing of CNS drugs to treat COVID-19 infection: targeting the sigma-1 receptor. European Archives of Psychiatry and Clinical Neuroscience 271(2), 249258.CrossRefGoogle ScholarPubMed
Hassan, L, Peek, N, Lovell, K, Carvalho, AF, Solmi, M, Stubbs, B and Firth, J (2021) Disparities in COVID-19 infection, hospitalisation and death in people with schizophrenia, bipolar disorder, and major depressive disorder: a cohort study of the UK Biobank. Molecular Psychiatry 6, 441. doi: 10.1038/s41380-021-01344-2.Google Scholar
Helmeste, D, Tang, SW, Fang, H and Li, M (1996a) Brain sigma receptors labelled by [3H]nemonapride. European Journal of Pharmacology 301(1-3), R13.CrossRefGoogle Scholar
Helmeste, DM, Shioiri, T, Mitsuhashi, M and Tang, SW (1999) Binding of [3H]U-101958 to sigma1 receptor-like sites in human cerebellum and neuroblastoma cells. European Journal of Pharmacology 370(2), 205209.CrossRefGoogle Scholar
Helmeste, DM, Tang, SW, Bunney, WE Jr, Potkin, SG and Jones, EG (1996b) Decrease in sigma but no increase in striatal dopamine D4 sites in schizophrenic brains. European Journal of Pharmacology 314(3), R3R5.CrossRefGoogle ScholarPubMed
Helmeste, DM, Tang, SW, Li, M and Fang, H (1997) Multiple [3H]-nemonapride binding sites in calf brain. Naunyn Schmiedeberg’s Archives of Pharmacology 356(1), 1721.CrossRefGoogle ScholarPubMed
Helms, J, Kremer, S, Merdji, H, Clere-Jehl, R, Schenck, M, Kummerlen, C, Collange, O, Boulay, C, Fafi-Kremer, S, Ohana, M, Anheim, M, Meziani, F (2020) Neurologic features in severe SARS-CoV-2 infection. New England Journal of Medicine 382(23), 22682270.CrossRefGoogle ScholarPubMed
Hénon, H, Durieu, I, Guerouaou, D, Lebert, F, Pasquier, F and Leys, D (2001) Poststroke dementia: incidence and relationship to prestroke cognitive decline. Neurology 57(7), 12161222.CrossRefGoogle ScholarPubMed
Hewlett, EL, Pearson, RD, Zilberstein, D and Dwyer, DM (1985) Antiprotozoal activity of tricyclic compounds. Science 230(4729), 10631064.CrossRefGoogle ScholarPubMed
Hewlett, I, Lee, S, Molnar, J, Foldeak, S, Pine, PS, Weaver, JL and Aszalos, A (1997) Inhibition of HIV infection of H9 cells by chlorpromazine derivatives. Journal of Acquired Immune Deficiency Syndromes and Human Retrovirology 15(1), 1620.CrossRefGoogle ScholarPubMed
Hogan Ii, RB, Hogan Iii, RB, Cannon, T, Rappai, M, Studdard, J, Paul, D and Dooley, TP (2020) Dual-histamine receptor blockade with cetirizine - famotidine reduces pulmonary symptoms in COVID-19 patients. Pulmonary Pharmacology & Therapeutics 63(4), 101942. doi: 10.1016/j.pupt.2020.101942.CrossRefGoogle ScholarPubMed
Hong, WC, Yano, H, Hiranita, T, Chin, FT, McCurdy, CR, Su, T-P, Amara, SG and Katz, JL (2017) The sigma-1 receptor modulates dopamine transporter conformation and cocaine binding and may therefore potentiate cocaine self-administration in rats. Journal of Biological Chemistry 292(27), 1125011261.CrossRefGoogle ScholarPubMed
Huang, C, Huang, L, Wang, Y, Li, X, Ren, L, Gu, X, Kang, L, Guo, L, Liu, M, Zhou, X, Luo, J, Huang, Z, Tu, S, Zhao, Y, Chen, L, Xu, D, Li, Y, Li, C, Peng, L, Li, Y, Xie, W, Cui, D, Shang, L, Fan, G, Xu, J, Wang, G, Wang, Y, Zhong, J, Wang, C, Wang, J, Zhang, D and Cao, B (2021) 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. The Lancet 397(10270), 220232.CrossRefGoogle ScholarPubMed
Huang, F, Zhang, C, Liu, Q, Zhao, Y, Zhang, Y, Qin, Y, Li, X, Li, C, Zhou, C, Jin, N, Jiang, C (2020) Identification of amitriptyline HCl, flavin adenine dinucleotide, azacitidine and calcitriol as repurposing drugs for influenza A H5N1 virus-induced lung injury. PLoS Pathogens 16(3), e1008341. doi: 10.1371/journal.ppat.1008341.CrossRefGoogle ScholarPubMed
Huang, Y, Tan, C and Wu, J (2019) Impact of coronavirus disease 2019 on pulmonary function in early convalescence phase. Respiratory Research 21(1), 163.CrossRefGoogle Scholar
Ibrahim, S, Lowe, JR, Bramante, CT, Shah, S, Klatt, NR, Sherwood, N, Aronne, L, Puskarich, M, Tamariz, L, Palacio, A, Bomberg, E, Usher, M, King, S, Benson, B, Vojta, D, Tignanelli, C, Ingraham, N (2021) Metformin and Covid-19: Focused Review of Mechanisms and Current Literature Suggesting Benefit. Frontiers in Endocrinology 12, 587801. doi: 10.3389/fendo.2021.587801.CrossRefGoogle ScholarPubMed
Ishima, T, Fujita, Y and Hashimoto, K (2014) Interaction of new antidepressants with sigma-1 receptor chaperones and their potentiation of neurite outgrowth in PC12 cells. European Journal of Pharmacology 727, 167173.CrossRefGoogle ScholarPubMed
Janowitz, T, Gablenz, E, Pattinson, D, Wang, TC, Conigliaro, J, Tracey, K and Tuveson, D (2020) Famotidine use and quantitative symptom tracking for COVID-19 in non-hospitalised patients: a case series. Gut 69(9), 15921597.CrossRefGoogle ScholarPubMed
Jerčić, L, Kostić, S, Vitlov Uljević, M, Vukušić Pušić, T, Vukojević, K and Filipović, N (2019) Sigma-1 receptor expression in DRG neurons during a carrageenan-provoked inflammation. The Anatomical Record (Hoboken) 302(9), 16201627.CrossRefGoogle ScholarPubMed
Jha, NK, Ojha, S, Jha, SK, Dureja, H, Singh, SK, Shukla, SD, Chellappan, DK, Gupta, G, Bhardwaj, S, Kumar, N, Jeyaraman, M, Jain, R, Muthu, S, Kar, R, Kumar, D, Goswami, VK, Ruokolainen, J, Kesari, KK, Singh, SK and Dua, K (2021) Evidence of coronavirus (CoV) pathogenesis and emerging pathogen SARS-CoV-2 in the nervous system: A review on neurological impairments and manifestations. Journal of Molecular Neuroscience 71(11), 21922209.CrossRefGoogle ScholarPubMed
Jia, J, Cheng, J, Wang, C and Zhen, X (2018) Sigma-1 receptor-modulated neuroinflammation in neurological diseases. Frontiers in Cellular Neuroscience 12, 14. doi: 10.3389/fncel.2018.00314.CrossRefGoogle ScholarPubMed
Jozuka, R, Kimura, H, Uematsu, T, Fujigaki, H, Yamamoto, Y, Kobayashi, M, Kawabata, K, Koike, H, Inada, T, Saito, K, Katsuno, M, Ozaki, N (2021) Severe and long-lasting neuropsychiatric symptoms after mild respiratory symptoms caused by COVID-19: a case report. Neuropsychopharmacology Reports 19(9), 767. doi: 10.1002/npr2.12222.Google Scholar
Jurcovicova, J (2014) Glucose transport in brain - effect of inflammation. Endocrine Regulations 48(1), 3548.CrossRefGoogle ScholarPubMed
Kalaria, RN, Akinyemi, R and Ihara, M (2016) Stroke injury, cognitive impairment and vascular dementia. Biochimica Et Biophysica Acta 1862(5), 915925.CrossRefGoogle ScholarPubMed
Kalkman, HO and Feuerbach, D (2016) Antidepressant therapies inhibit inflammation and microglial M1-polarization. Pharmacology & Therapeutics 163(3), 8293.CrossRefGoogle ScholarPubMed
Kappelmann, N, Dantzer, R and Khandaker, GM (2021) Interleukin-6 as potential mediator of long-term neuropsychiatric symptoms of COVID-19. Psychoneuroendocrinology 131, 105295. doi: 10.1016/j.psyneuen.2021.105295.CrossRefGoogle ScholarPubMed
Karaoulanis, SE and Christodoulou, NG (2021) Do patients with schizophrenia have higher infection and mortality rates due to COVID-19? A systematic review. Psychiatriki 32(3), 219223.Google ScholarPubMed
Kas, A, Soret, M, Pyatigoskaya, N, Habert, MO, Hesters, A, Le Guennec, L, Paccoud, O, Bombois, S, Delorme, C and on the behalf of CoCo-Neurosciences study group and COVID SMIT PSL study group (2021) The cerebral network of COVID-19-related encephalopathy: a longitudinal voxel-based 18F-FDG-PET study. European Journal of Nuclear Medicine and Molecular Imaging 48(8), 25432557.CrossRefGoogle ScholarPubMed
Kirwan, R, McCullough, D, Butler, T, Perez de Heredia, F, Davies, IG and Stewart, C (2020) Sarcopenia during COVID-19 lockdown restrictions: long-term health effects of short-term muscle loss. Geroscience 42(6), 15471578.CrossRefGoogle ScholarPubMed
Klein, R, Soung, A, Sissoko, C, Nordvig, A, Canoll, P, Mariani, M, Jiang, X, Bricker, T, Goldman, J, Rosoklija, G, Arango, V, Underwood, M, Mann, JJ, Boon, A, Dowrk, A, Boldrini, M (2021) COVID-19 induces neuroinflammation and loss of hippocampal neurogenesis. Research Square. doi: 10.21203/rs.3.rs-1031824/v1.Google ScholarPubMed
Kourrich, S, Su, TP, Fujimoto, M and Bonci, A (2012) The sigma-1 receptor: roles in neuronal plasticity and disease. Trends in Neurosciences 35(12), 762771.CrossRefGoogle ScholarPubMed
Kumar, A, Arora, A, Sharma, P, Anikhindi, SA, Bansal, N, Singla, V, Khare, S and Srivastava, A (2020) Is diabetes mellitus associated with mortality and severity of COVID-19? A meta-analysis. Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14(4), 535545.CrossRefGoogle ScholarPubMed
Lee, AC, Tang, SW, Yu, GK and Cheung, RT (2007) Incidence and predictors of depression after stroke (DAS). International Journal of Psychiatry in Clinical Practice 11(3), 200206.CrossRefGoogle Scholar
Lee, AC, Tang, SW, Yu, GK and Cheung, RT (2008) The smiley as a simple screening tool for depression after stroke: a preliminary study. International Journal of Nursing Studies 45(7), 10811089.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) Fluvoxamine vs placebo and clinical deterioration in outpatients with symptomatic COVID-19: a randomized clinical trial. JAMA 324(22), 22922300.CrossRefGoogle ScholarPubMed
Leonard, BE (2018) Chronic inflammation and resulting neuroprogression in major depression. In Kim, Y-K (ed), Understanding Depression, Vol. 1. Singapore: Springer, pp. 191196.CrossRefGoogle Scholar
Leonard, BE and Wegener, G (2020) Inflammation, insulin resistance and neuroprogression in depression. Acta Neuropsychiatrica 32(1), 19. doi: 10.1017/neu.2019.17.CrossRefGoogle ScholarPubMed
Letko, M, Marzi, A and Munster, V (2020) Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. Nature Microbiology 5(4), 562569.CrossRefGoogle ScholarPubMed
Lever, JR, Litton, TP and Fergason-Cantrell, EA (2015) Characterization of pulmonary sigma receptors by radioligand binding. European Journal of Pharmacology 762(14), 118126.CrossRefGoogle ScholarPubMed
Li, D, Hu, Y, Chen, H, Zhu, X, Wu, X, Li, J, Zhang, Z and Liu, S (2021) Identifying the subtypes and characteristics of mental workload among Chinese physicians in outpatient practice: a latent profile analysis. Frontiers in Public Health 9, 779262. doi: 10.3389/fpubh.2021.779262.CrossRefGoogle ScholarPubMed
Li, MY, Li, L, Zhang, Y and Wang, XS (2020) Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infectious Diseases of Poverty 9(1), 45. doi: 10.1186/s40249-020-00662-x.CrossRefGoogle Scholar
Li, Y, Chen, B, Hong, Z, Sun, Q, Dai, Y, Basta, M, Tang, X and Qin, Q (2021b) Insomnia symptoms during the early and late stages of the COVID-19 pandemic in China: a systematic review and meta-analysis. Sleep Medicine 16(8), S1389-9457(21)00494-9. doi: 10.1016/j.sleep.2021.09.014.Google Scholar
Liddelow, SA, Guttenplan, KA, Clarke, LE, Bennett, FC, Bohlen, CJ, Schirmer, L, Bennett, ML, Münch, AE, Chung, WS, Peterson, TC, Wilton, DK, Frouin, A, Napier, BA, Panicker, N, Kumar, M, Buckwalter, MS, Rowitch, DH, Dawson, VL, Dawson, TM, Stevens, B and Barres, BA (2017) Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541(7638), 481487.CrossRefGoogle ScholarPubMed
Lin, L, Chen, Z, Ding, T, Liu, H, Zhou, F, Huang, X, Zhang, X, Liu, W, Zhang, BH, Yuan, Y, Zhang, P, Zhang, XJ, She, ZG, Cai, J, Chen, W, Li, H (2021) Newly-diagnosed diabetes and sustained hyperglycemia are associated with poorer outcomes in COVID-19 inpatients without pre-existing diabetes. Diabetes, Metabolic Syndrome and Obesity 14, 44694482.CrossRefGoogle ScholarPubMed
Lisak, RP, Nedelkoska, L and Benjamins, JA (2020) Sigma-1 receptor agonists as potential protective therapies in multiple sclerosis. Journal of Neuroimmunology 342(2), 577188. doi: 10.1016/j.jneuroim.2020.577188.CrossRefGoogle ScholarPubMed
Liu, Q, Liu, Y, Zhang, C, An, Z and Zhao, P (2021) Elderly mobility during the COVID-19 pandemic: a qualitative exploration in Kunming. China Journal of Transport Geography 96(1), 103176.CrossRefGoogle ScholarPubMed
Logette, E, Lorin, C, Favreau, C, Oshurko, E, Coggan, JS, Casalegno, F, Sy, MF, Monney, C, Bertschy, M, Delattre, E, Fonta, PA, Krepl, J, Schmidt, S, Keller, D, Kerrien, S, Scantamburlo, E, Kaufmann, AK, Markram, H (2021) A machine-generated view of the role of blood glucose levels in the severity of COVID-19. Frontiers in Public Health 9, 695139. doi: 10.3389/fpubh.2021.695139.CrossRefGoogle ScholarPubMed
Lubbe, L, Cozier, GE, Oosthuizen, D, Acharya, KR and Sturrock, ED (2020) ACE2 and ACE: structure-based insights into mechanism, regulation and receptor recognition by SARS-CoV. Clinical Science (London) 134(21), 28512871.CrossRefGoogle ScholarPubMed
Mahajan, GJ, Vallender, EJ, Garrett, MR, Challagundla, L, Overholser, JC, Jurjus, G, Dieter, L, Syed, M, Romero, DG, Benghuzzi, H, Stockmeier, CA (2018) Altered neuro-inflammatory gene expression in hippocampus in major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 82, 177186.CrossRefGoogle ScholarPubMed
Mahalaxmi, I, Kaavya, J, Mohana Devi, S and Balachandar, V (2021) COVID-19 and olfactory dysfunction: a possible associative approach towards neurodegenerative diseases. Journal of Cellular Physiology 236(2), 763770.CrossRefGoogle ScholarPubMed
Maideen, NMP (2020) Prophetic medicine-Nigella sativa (black cumin seeds) - Potential herb for COVID-19? Journal of Pharmacopuncture 23(2), 6270.CrossRefGoogle ScholarPubMed
Malhotra, A, Hepokoski, M, McCowen, KC and Shyy, JY-J (2020) ACE2, metformin, and COVID-19. iScience 23(9), 101425. doi: 10.1016/j.isci.2020.101425.CrossRefGoogle ScholarPubMed
Mandal, A, Sinha, C, Kumar Jena, A, Ghosh, S and Samanta, A (2010) An investigation on in vitro and in vivo antimicrobial properties of the antidepressant: amitriptyline hydrochloride. Brazilian Journal of Microbiology 41(3), 635645.CrossRefGoogle ScholarPubMed
Mao, L, Jin, H, Wang, M, Hu, Y, Chen, S, He, Q, Chang, J, Hong, C, Zhou, Y, Wang, D, Miao, X, Li, Y, Hu, B (2020) Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurology 77(6), 683690.CrossRefGoogle ScholarPubMed
Mather, JF, Seip, RL and McKay, RG (2020) Impact of famotidine use on clinical outcomes of hospitalized patients with COVID-19. American Journal of Gastroenterology 115(10), 16171623.CrossRefGoogle ScholarPubMed
May, M, Slitzky, M, Rostama, B, Barlow, D and Houseknecht, KL (2020) Antipsychotic-induced immune dysfunction: a consideration for COVID-19 risk. Brain, Behavior, & Immunity - Health 6(1), 100097. doi: 10.1016/j.bbih.2020.100097.CrossRefGoogle ScholarPubMed
May, PR, Van Putten, T, Jenden, DJ and Cho, AK (1978) Test dose response in schizophrenia: chlorpromazine blood and saliva levels. Archives of General Psychiatry 35(9), 10911097.CrossRefGoogle ScholarPubMed
Medeiros, GC, Roy, D, Kontos, N and Beach, SR (2020) Post-stroke depression: a 2020 updated review. General Hospital Psychiatry 66(1), 7080.CrossRefGoogle ScholarPubMed
Meinhardt, J, Radke, J, Dittmayer, C, Franz, J, Thomas, C, Mothes, R, Laue, M, Schneider, J, Brünink, S, Greuel, S, Lehmann, M, Hassan, O, Aschman, T, Schumann, E, Chua, RL, Conrad, C, Eils, R, Stenzel, W, Windgassen, M, Rößler, L, Goebel, HH, Gelderblom, HR, Martin, H, Nitsche, A, Schulz-Schaeffer, WJ, Hakroush, S, Winkler, MS, Tampe, B, Scheibe, F, Körtvélyessy, P, Reinhold, D, Siegmund, B, Kühl, AA, Elezkurtaj, S, Horst, D, Oesterhelweg, L, Tsokos, M, Ingold-Heppner, B, Stadelmann, C, Drosten, C, Corman, VM, Radbruch, H and Heppner, FL (2021) Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nature Neuroscience 24(2), 168175.CrossRefGoogle ScholarPubMed
Méndez, R, Balanzá-Martínez, V, Luperdi, SC, Estrada, I, Latorre, A, González-Jiménez, P, Bouzas, L, Yépez, K, Ferrando, A, Reyes, S, Menéndez, R (2021) Long-term neuropsychiatric outcomes in COVID-19 survivors: a 1-year longitudinal study. Internal Medicine. doi: 10.1111/joim.13389.CrossRefGoogle ScholarPubMed
Mengozzi, M, Fantuzzi, G, Faggioni, R, Marchant, A, Goldman, M, Orencole, S, Clark, BD, Sironi, M, Benigni, F, Ghezzi, P (1994) Chlorpromazine specifically inhibits peripheral and brain TNF production, and up-regulates IL-10 production, in mice. Immunology 82(2), 207210.Google ScholarPubMed
Miller, GE, Chen, E, Sze, J, Marin, T, Arevalo, JM, Doll, R, Ma, R and Cole, SW (2008) A functional genomic fingerprint of chronic stress in humans: blunted glucocorticoid and increased NF-kappa B signaling. Biological Psychiatry 64(4), 266272.CrossRefGoogle Scholar
Mo, Y, Deng, L, Zhang, L, Lang, Q, Liao, C, Wang, N, Qin, M and Huang, H (2020) Work stress among Chinese nurses to support Wuhan in fighting against COVID-19 epidemic. Journal of Nursing Management 28(5), 10021009.CrossRefGoogle ScholarPubMed
Moga, S, Teodorescu, A, Ifteni, P, Gavris, C and Petric, PS (2021) Inflammatory response in SARS-CoV-2 infection of patients with schizophrenia and long-term antipsychotic treatment. Neuropsychiatric Disease and Treatment 17, 30533060. doi: 10.2147/NDT.S325062.CrossRefGoogle ScholarPubMed
Mohamed-Hussein, AAR, Amin, MT, Makhlouf, HA, Makhlouf, NA, Galal, I, Abd-Elaal, HK, Abdeltawab, D, Kholief, KMS and Hashem, MK (2021) Non-hospitalised COVID-19 patients have more frequent long COVID symptoms. The International Journal of Tuberculosis and Lung Disease 25, 732737.CrossRefGoogle Scholar
Morand, A, Campion, JY, Lepine, A, Bosdure, E, Luciani, L, Cammilleri, S, Chabrol, B and Guedj, E (2021) Similar patterns of [18F]-FDG brain PET hypometabolism in paediatric and adult patients with long COVID: a paediatric case series. European Journal of Nuclear Medicine and Molecular Imaging, 18. doi: 10.1007/s00259-021-05528-4.Google Scholar
Morrison, SA, Jurak, G and Starc, G (2020) Responding to a global pandemic: republic of Slovenia on maintaining physical activity during self-isolation. The Scandinavian Journal of Medicine & Science in Sports 30(8), 15461548.CrossRefGoogle ScholarPubMed
Myint, AM (2013) Inflammation, neurotoxins and psychiatric disorders. Modern Trends in Pharmacopsychiatry 28, 6174.CrossRefGoogle ScholarPubMed
Narita, N, Hashimoto, K, Tomitaka, S and Minabe, Y (1996) Interactions of selective serotonin reuptake inhibitors with subtypes of sigma receptors in rat brain. European Journal of Pharmacology 307(1), 117119.CrossRefGoogle ScholarPubMed
Nowak, BM, Miedziarek, C, Pełczyński, S and Rzymski, P (2021) Misinformation, fears and adherence to preventive measures during the early phase of COVID-19 pandemic: a cross-sectional study in Poland. International Journal of Environmental Research and Public Health 18(22), 12266. doi: 10.3390/ijerph182212266.CrossRefGoogle ScholarPubMed
Numbers, K and Brodaty, H (2021) The effects of the COVID-19 pandemic on people with dementia. Nature Reviews Neurology 17(2), 6970.CrossRefGoogle ScholarPubMed
Oren, O, Gersh, BJ and Blumenthal, RS (2020) Anticipating and curtailing the cardiometabolic toxicity of social isolation and emotional stress in the time of COVID-19. American Heart Journal 226(13), 13.CrossRefGoogle ScholarPubMed
Orsucci, D, Trezzi, M, Anichini, R, Blanc, P, Barontini, L, Biagini, C, Capitanini, A, Comeglio, M, Corsini, P, Gemignani, F, Giannecchini, R, Giusti, M, Lombardi, M, Marrucci, E, Natali, A, Nenci, G, Vannucci, F, Volpi, G (2021) Increased creatine kinase may predict a worse COVID-19 outcome. Journal of Clinical Medicine 10(8), 1734. doi: 10.3390/jcm10081734.CrossRefGoogle ScholarPubMed
Ortega-Guillén, E, Meneses, G and Coila, E (2021) Remarks about retrospective analysis of ivermectin effectiveness on coronavirus disease 2019 (ICON study). Chest 159(5), 21102111.CrossRefGoogle Scholar
Ortelli, P, Ferrazzoli, D, Sebastianelli, L, Engl, M, Romanello, R, Nardone, R, Bonini, I, Koch, G, Saltuari, L, Quartarone, A, Oliviero, A, Kofler, M, Versace, V (2021) Neuropsychological and neurophysiological correlates of fatigue in post-acute patients with neurological manifestations of COVID-19: insights into a challenging symptom. Journal of the Neurological Sciences 420(6), 117271. doi: 10.1016/j.jns.2020.117271.CrossRefGoogle ScholarPubMed
Otero-Losada, M, Kobiec, T, Udovin, L, Chevalier, G, Quarracino, C, Maissonave, CM, Bordet, S, Capani, F and Perez-Lloret, S (2020) Parkinson’s disease in the era of a novel respiratory virus pandemic. Frontiers in Neurology 11, 995. doi: 10.3389/fneur.2020.00995.CrossRefGoogle ScholarPubMed
Pagliaro, P and Penna, C (2020) ACE/ACE2 ratio: a key also in 2019 coronavirus disease (Covid-19)? Frontiers in Medicine 7, 335. doi: 10.3389/fmed.2020.00335.CrossRefGoogle ScholarPubMed
Pailla, K, El-Mir, MY, Cynober, L and Blonde-Cynober, F (2001) Cytokinemediated inhibition of ketogenesis is unrelated to nitric oxide or protein synthesis. Clinical Nutrition 20(4), 313317.CrossRefGoogle ScholarPubMed
Park, JH, Park, HJ, Lee, SE, Kim, YS, Jang, GY, Han, HD, Jung, ID, Shin, KC, Bae, YM, Kang, TH, Park, YM (2019) Repositioning of the antipsychotic drug TFP for sepsis treatment. Journal of Molecular Medicine (Berlin) 97(5), 647658.CrossRefGoogle ScholarPubMed
Paterson, RW, Brown, RL, Benjamin, L, Nortley, R, Wiethoff, S, Bharucha, T, Jayaseelan, DL, Kumar, G, Raftopoulos, RE, Zambreanu, L, Vivekanandam, V, Khoo, A, Geraldes, R, Chinthapalli, K, Boyd, E, Tuzlali, H, Price, G, Christofi, G, Morrow, J, McNamara, P, McLoughlin, B, Lim, ST, Mehta, PR, Levee, V, Keddie, S, Yong, W, Trip, SA, Foulkes, AJM, Hotton, G, Miller, TD, Everitt, AD, Carswell, C, Davies, NWS, Yoong, M, Attwell, D, Sreedharan, J, Silber, E, Schott, JM, Chandratheva, A, Perry, RJ, Simister, R, Checkley, A, Longley, N, Farmer, SF, Carletti, F, Houlihan, C, Thom, M, Lunn, MP, Spillane, J, Howard, R, Vincent, A, Werring, DJ, Hoskote, C, Jäger, HR, Manji, H and Zandi, MS (2020) The emerging spectrum of COVID-19 neurology: clinical, radiological and laboratory findings. Brain 143(10), 31043120.CrossRefGoogle ScholarPubMed
Patone, M, Mei, XW, Handunnetthi, L, Dixon, S, Zaccardi, F, Shankar-Hari, M, Watkinson, P, Khunti, K, Harnden, A, Coupland, CAC, Channon, KM, Mills, NL, Sheikh, A, Hippisley-Cox, J (2021) Risks of myocarditis, pericarditis, and cardiac arrhythmias associated with COVD-19 vaccination or SARS-CoV-2 infection. Nature Medicine 354, i4515. doi: 10.1038/s41591-021-01630-0.Google Scholar
Pezzini, A and Padovani, A (2020) Lifting the mask on neurological manifestations of COVID-19. Nature Reviews. Neurology 16(11), 636644.CrossRefGoogle ScholarPubMed
Pho, MT, Ashok, A and Atwood, WJ (2000) JC virus enters human glial cells by clathrin-dependent receptor-mediated endocytosis. Journal of Virology 74(5), 22882292.CrossRefGoogle ScholarPubMed
Pilotto, A, Cristillo, V, Cotti Piccinelli, S, Zoppi, N, Bonzi, G, Sattin, D, Schiavolin, S, Raggi, A, Canale, A, Gipponi, S, Libri, I, Frigerio, M, Bezzi, M, Leonardi, M, Padovani, A (2021) Long-term neurological manifestations of COVID-19: prevalence and predictive factors. Neurological Sciences 42(12), 49034907.CrossRefGoogle ScholarPubMed
Plaze, M, Attali, D, Petit, AC, Blatzer, M, Simon-Loriere, E, Vinckier, F, Cachia, A, Chrétien, F and Gaillard, R (2020) Repurposing chlorpromazine to treat COVID-19: the reCoVery study. Encephale 46(3), 169172.CrossRefGoogle ScholarPubMed
Plaze, M, Attali, D, Prot, M, Petit, AC, Blatzer, M, Vinckier, F, Levillayer, L, Chiaravalli, J, Perin-Dureau, F, Cachia, A, Friedlander, G, Chrétien, F, Simon-Loriere, E, Gaillard, R (2021) Inhibition of the replication of SARS-CoV-2 in human cells by the FDA-approved drug chlorpromazine. International Journal of Antimicrobial Agents 57(3), 106274. doi: 10.1016/j.ijantimicag.2020.106274.CrossRefGoogle ScholarPubMed
Pollock, A, Campbell, P, Cheyne, J, Cowie, J, Davis, B, McCallum, J, McGill, K, Elders, A, Hagen, S, McClurg, D, Torrens, C, Maxwell, M (2020) Interventions to support the resilience and mental health of frontline health and social care professionals during and after a disease outbreak, epidemic or pandemic: a mixed methods systematic review. Cochrane Database of Systematic Reviews 11(11), CD013779. doi: 10.1002/14651858.CD013779.Google ScholarPubMed
Porrello, ER, Delbridge, LM and Thomas, WG (2009) The angiotensin II type 2 (AT2) receptor: an enigmatic seven transmembrane receptor. Frontiers in Bioscience 14, 958972.CrossRefGoogle ScholarPubMed
Povlsen, AL, Grimm, D, Wehland, M, Infanger, M and Krüger, M (2020) The vasoactive mas receptor in essential hypertension. Journal of Clinical Medicine 9(1), 267. doi: 10.3390/jcm9010267.CrossRefGoogle ScholarPubMed
Rajter, JC, Sherman, MS, Fatteh, N, Vogel, F, Sacks, J and Rajter, JJ (2021) Use of ivermectin is associated with lower mortality in hospitalized patients with coronavirus disease 2019: the ivermectin in COVID nineteen study. Chest 159(1), 8592.CrossRefGoogle ScholarPubMed
Razai, MS, Oakeshott, P, Kankam, H, Galea, S and Stokes-Lampard, H (2020) Mitigating the psychological effects of social isolation during the COVID-19 pandemic. BMJ 369, m1904. doi: 10.1136/bmj.m1904.CrossRefGoogle ScholarPubMed
Reading, PC, Allison, J, Crouch, EC and Anders, EM (1998) Increased susceptibility of diabetic mice to influenza virus infection: compromise of collectin-mediated host defense of the lung by glucose? Journal of Virology 72(8), 68846887.CrossRefGoogle Scholar
Reynolds, GP, Brown, JE and Middlemiss, DN (1991) [3H]ditolylguanidine binding to human brain sigma sites is diminished after haloperidol treatment. European Journal of Pharmacology 194(2-3), 235236.CrossRefGoogle ScholarPubMed
Reznikov, LR, Norris, MH, Vashisht, R, Bluhm, AP, Li, D, Liao, YJ, Brown, A, Butte, AJ and Ostrov, DA (2021) Identification of antiviral antihistamines for COVID-19 repurposing. Biochemical and Biophysical Research Communications 538(2), 173179.CrossRefGoogle ScholarPubMed
Richardson, S, Hirsch, JS, Narasimhan, M, Crawford, JM, McGinn, T, Davidson, KW, the Northwell COVID-19 Research Consortium, Barnaby, DP, Becker, LB, Chelico, JD, Cohen, SL, Cookingham, J, Coppa, K, Diefenbach, MA, Dominello, AJ, Duer-Hefele, J, Falzon, L, Gitlin, J, Hajizadeh, N, Harvin, TG, Hirschwerk, DA, Kim, EJ, Kozel, ZM, Marrast, LM, Mogavero, JN, Osorio, GA, Qiu, M and Zanos, TP (2020) Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City area. JAMA 323(20), 20522059.CrossRefGoogle ScholarPubMed
Ritter, JB, Wahl, AS, Freund, S, Genzel, Y and Reichl, U (2010) Metabolic effects of influenza virus infection in cultured animal cells: intra- and extracellular metabolite profiling. BMC Systems Biology 4, 61. doi: 10.1186/1752-0509-4-61.CrossRefGoogle ScholarPubMed
Rivas-Ramírez, Á.R, Tendilla-Beltrán, H, Gómez-Mendoza, LE, Loaiza, G and Flores, G (2021) Patients with schizophrenia have decreased COVID-19 prevalence among hospitalized patients with psychiatric and neurological diseases: a retrospective analysis in Mexican population. International Journal of Clinical Practice 75(10), e14528. doi: 10.1111/ijcp.14528.CrossRefGoogle ScholarPubMed
Rivers, J and Ihle, JF (2020) COVID-19 social isolation-induced takotsubo cardiomyopathy. Medical Journal of Australia 213(7), 336336.CrossRefGoogle ScholarPubMed
Robinson, RG and Jorge, RE (2016) Post-stroke depression: a review. The American Journal of Psychiatry 173(3), 221231.CrossRefGoogle ScholarPubMed
Rodríguez-Alfonso, B, Ruiz Solís, S, Silva-Hernández, L, Pintos Pascual, I, Aguado Ibáñez, S and Salas Antón, C (2021) 18F-FDG-PET/CT in SARS-CoV-2 infection and its sequelae. Revista Española de Medicina Nuclear e Imagen Molecular (English Edition) 40(5), 299309.CrossRefGoogle ScholarPubMed
Rogers, JP, Watson, CJ, Badenoch, J, Cross, B, Butler, M, Song, J, Hafeez, D, Morrin, H, Rengasamy, ER, Thomas, L, Ralovska, S, Smakowski, A, Sundaram, RD, Hunt, CK, Lim, MF, Aniwattanapong, D, Singh, V, Hussain, Z, Chakraborty, S, Burchill, E, Jansen, K, Holling, H, Walton, D, Pollak, TA, Ellul, M, Koychev, I, Solomon, T, Michael, BD, Nicholson, TR and Rooney, AG (2021) Neurology and neuropsychiatry of COVID-19: a systematic review and meta-analysis of the early literature reveals frequent CNS manifestations and key emerging narratives. Journal of Neurology, Neurosurgery & Psychiatry 92(9), 932941.Google ScholarPubMed
Roman, C, Egert, L and Di Benedetto, B (2021) Astrocytic-neuronal crosstalk gets jammed: alternative perspectives on the onset of neuropsychiatric disorders. European Journal of Neuroscience 54(5), 57175729.CrossRefGoogle ScholarPubMed
Rosen, DA, Seki, SM, Fernández-Castañeda, A, Beiter, RM, Eccles, JD, Woodfolk, JA and Gaultier, A (2019) Modulation of the sigma-1 receptor-IRE1 pathway is beneficial in preclinical models of inflammation and sepsis. Science Translational Medicine 11(478), eaau5266. doi: 10.1126/scitranslmed.aau5266.CrossRefGoogle Scholar
Rudroff, T, Fietsam, AC, Deters, JR, Bryant, AD and Kamholz, J (2020) Post-COVID-19 fatigue: potential contributing factors. Brain Sciences 10(12), 1012. doi: 10.3390/brainsci10121012.CrossRefGoogle ScholarPubMed
Ruiz-Cantero, MC, González-Cano, R, Tejada, MÁ., Santos-Caballero, M, Perazzoli, G, Nieto, FR and Cobos, EJ (2021) Sigma-1 receptor: a drug target for the modulation of neuroimmune and neuroglial interactions during chronic pain. Pharmacological Research 163(3), 105339. doi: 10.1016/j.phrs.2020.105339.CrossRefGoogle ScholarPubMed
Ruscher, K and Wieloch, T (2015) The involvement of the sigma-1 receptor in neurodegeneration and neurorestoration. Journal of Pharmacological Sciences 127(1), 3035.CrossRefGoogle ScholarPubMed
Ryoo, N, Pyun, JM, Baek, MJ, Suh, J, Kang, MJ, Wang, MJ, Youn, YC, Yang, DW, Kim, SY, Park, YH, Kim, S (2020) Coping with dementia in the middle of the COVID-19 pandemic. Journal of Korean Medical Science 35(42), e383. doi: 10.3346/jkms.2020.35.e383.CrossRefGoogle ScholarPubMed
Salaciak, K and Pytka, K (2021) Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neuroscience and Biobehavioral Reviews. 10.1016/j.neubiorev.Google ScholarPubMed
Samavati, L and Uhal, BD (2020) ACE2, much more than just a receptor for SARS-COV-2. Frontiers in Cellular and Infection Microbiology 10, 317. doi: 10.3389/fcimb.2020.00317.CrossRefGoogle ScholarPubMed
Samuel, SM, Varghese, E and Büsselberg, D (2021) Therapeutic potential of metformin in COVID-19: reasoning for its protective role. Trends in Microbiology 29(10), 894907.CrossRefGoogle ScholarPubMed
Sanchez, EL and Lagunoff, M (2015) Viral activation of cellular metabolism. Virology 479-480, 609618. doi: 10.1016/j.virol.2015.02.038.CrossRefGoogle ScholarPubMed
Sánchez-Fernández, C, Entrena, JM, Baeyens, JM and Cobos, EJ (2017) Sigma-1 receptor antagonists: a new class of neuromodulatory analgesics. Advances in Experimental Medicine and Biology 964, 109132.CrossRefGoogle ScholarPubMed
Saponaro, F, Rutigliano, G, Sestito, S, Bandini, L, Storti, B, Bizzarri, R and Zucchi, R (2020) ACE2 in the era of SARS-CoV-2: controversies and novel perspectives. Frontiers in Molecular Biosciences 7, 588618. doi: 10.3389/fmolb.2020.588618.CrossRefGoogle ScholarPubMed
Scheen, AJ (2020) Metformin and COVID-19: from cellular mechanisms to reduced mortality. Diabetes & Metabolism 46(6), 423426.CrossRefGoogle ScholarPubMed
Schloer, S, Brunotte, L, Goretzko, J, Mecate-Zambrano, A, Korthals, N, Gerke, V, Ludwig, S and Rescher, U (2020) Targeting the endolysosomal host-SARS-CoV-2 interface by clinically licensed functional inhibitors of acid sphingomyelinase (FIASMA) including the antidepressant fluoxetine. Emerging Microbes & Infections 9(1), 22452255. doi: 10.1080/22221751.2020.1829082.CrossRefGoogle ScholarPubMed
Schöttke, H and Giabbiconi, CM (2015) Post-stroke depression and post-stroke anxiety: prevalence and predictors. International Psychogeriatrics 27(11), 18051812.CrossRefGoogle ScholarPubMed
Schou, TM, Joca, S, Wegener, G and Bay-Richter, C (2021) Psychiatric and neuropsychiatric sequelae of COVID-19 - a systematic review. Brain, Behavior, and Immunity 97, 328348.CrossRefGoogle ScholarPubMed
Scialo, F, Daniele, A, Amato, F, Pastore, L, Matera, MG, Cazzola, M, Castaldo, G and Bianco, A (2020) ACE2: the major cell entry receptor for SARS-CoV-2. Lung 198(6), 867877.CrossRefGoogle ScholarPubMed
Serrano-Castro, PJ, Estivill-Torrús, G, Cabezudo-García, P, Reyes-Bueno, JA, Ciano Petersen, N, Aguilar-Castillo, MJ, Suárez-Pérez, J, Jiménez-Hernández, MD, Moya-Molina, MÁ., Oliver-Martos, B, Arrabal-Gómez, C, Rodríguez de Fonseca, F (2020) Impact of SARS-CoV-2 infection on neurodegenerative and neuropsychiatric diseases: a delayed pandemic? Neurologia (English Edition) 35(4), 245251.CrossRefGoogle ScholarPubMed
Shan, Y, Shang, J, Yan, Y, Lu, G, Hu, D and Ye, X (2021) Mental workload of frontline nurses aiding in the COVID-19 pandemic: a latent profile analysis. Journal of Advanced Nursing 77(5), 23742385.CrossRefGoogle ScholarPubMed
Sharma, R, Mallick, D, Llinas, RH and Marsh, EB (2020) Early post-stroke cognition: in-hospital predictors and the association with functional outcome. Frontiers in Neurology 11, 613607. doi: 10.3389/fneur.2020.613607.CrossRefGoogle ScholarPubMed
Shelton, RC and Miller, AH (2010) Eating ourselves to death (and despair): the contribution of adiposity and inflammation to depression. Progress in Neurobiology 91(4), 275299.CrossRefGoogle ScholarPubMed
Shen, Y, Kapfhamer, D, Minnella, AM, Kim, J-E, Won, SJ, Chen, Y, Huang, Y, Low, LH, Massa, SM, Swanson, RA (2017) Bioenergetic state regulates innate inflammatory responses through the transcriptional co-repressor CtBP. Nature Communications 8(1), 624. doi: 10.1038/s41467-017-00707-0.CrossRefGoogle ScholarPubMed
Simões e Silva, AC, Silveira, KD, Ferreira, AJ and Teixeira, MM (2013) ACE2, angiotensin-(1-7) and Mas receptor axis in inflammation and fibrosis. British Journal of Pharmacology 169(3), 477492.CrossRefGoogle ScholarPubMed
Simpson, RJ and Katsanis, E (2020) The immunological case for staying active during the COVID-19 pandemic. Brain, Behavior, and Immunity 87, 67.CrossRefGoogle ScholarPubMed
Singh, AK and Singh, R (2020) Does poor glucose control increase the severity and mortality in patients with diabetes and COVID-19? Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14(5), 725727.CrossRefGoogle ScholarPubMed
Singh, AK and Singh, R (2020b) Hyperglycemia without diabetes and new-onset diabetes are both associated with poorer outcomes in COVID-19. Diabetes Research and Clinical Practice 167(6), 108382. doi: 10.1016/j.diabres.2020.108382.CrossRefGoogle ScholarPubMed
Sisó-Almirall, A, Brito-Zerón, P, Conangla Ferrín, L, Kostov, B, Moragas Moreno, A, Mestres, J, Sellarès, J, Galindo, G, Morera, R, Basora, J, Trilla, A, Ramos-Casals, M and On Behalf Of The CAMFiC Long COVID-Study Group (2021) Long COVID: proposed primary care clinical guidelines for diagnosis and disease management. International Journal of Environmental Research and Public Health 18(8), 4350. doi: 10.3390/ijerph18084350.CrossRefGoogle ScholarPubMed
Skuza, G (2012) Pharmacology of sigma (σ) receptor ligands from a behavioral perspective. Current Pharmaceutical Design 18(7), 863874.CrossRefGoogle ScholarPubMed
Sollini, M, Morbelli, S, Ciccarelli, M, Cecconi, M, Aghemo, A, Morelli, P, Chiola, S, Gelardi, F and Chiti, A (2021) Long COVID hallmarks on [18F]FDG-PET/CT:a case-control study. European Journal of Nuclear Medicine and Molecular Imaging 48(10), 31873197.CrossRefGoogle Scholar
Solomon, SS, Mishra, SK, Palazzolo, MR, Postlethwaite, AE and Seyer, JM (1997) Identification of specific sites in the TNF-alpha molecule promoting insulin resistance in H-411E cells. Journal of Laboratory and Clinical Medicine 130(2), 139146.CrossRefGoogle ScholarPubMed
Song, J, Li, Y, Huang, X, Chen, Z, Li, Y, Liu, C, Chen, Z and Duan, X (2020) Systematic analysis of ACE2 and TMPRSS2 expression in salivary glands reveals underlying transmission mechanism caused by SARS-CoV-2. Journal of Medical Virology 92(11), 25562566.CrossRefGoogle ScholarPubMed
Stefano, GB, Büttiker, P, Weissenberger, S, Martin, A, Ptacek, R and Kream, RM (2021a) Editorial: the pathogenesis of long-term neuropsychiatric COVID-19 and the role of microglia, mitochondria, and persistent neuroinflammation: a hypothesis. Medical Science Monitor 27, e933015. doi: 10.12659/MSM.933015.CrossRefGoogle ScholarPubMed
Stefano, GB, Ptacek, R, Ptackova, H, Martin, A and Kream, RM (2021b) Selective neuronal mitochondrial targeting in SARS-CoV-2 infection affects cognitive processes to induce ‘brain fog’ and results in behavioral changes that favor viral survival. Medical Science Monitor 27, e930886. doi: 10.12659/MSM.930886.CrossRefGoogle ScholarPubMed
Stoll, M and Unger, T (2001) Angiotensin and its AT2 receptor: new insights into an old system. Regulatory Peptides 99(2-3), 175182.CrossRefGoogle ScholarPubMed
Stuckey, SM, Ong, LK, Collins-Praino, LE and Turner, RJ (2021) Neuroinflammation as a key driver of secondary neurodegeneration following stroke? International Journal of Molecular Sciences 22(23), 13101. doi: 10.3390/ijms222313101.CrossRefGoogle ScholarPubMed
Su, T-P, Su, T-C, Nakamura, Y and Tsai, S-Y (2016) Sigma-1 receptor as a pluripotent modulator in the living system. Trends Pharmacological Sciences 37(4), 262278.CrossRefGoogle Scholar
Su, TP, Hayashi, T, Maurice, T, Buch, S and Ruoho, AE (2010) The sigma-1 receptor chaperone as an inter-organelle signaling modulator. Trends in Pharmacological Sciences 31(12), 557566.CrossRefGoogle ScholarPubMed
Tai, W, He, L, Zhang, X, Pu, J, Voronin, D, Jiang, S, Zhou, Y and Du, L (2020) Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cellular & Molecular Immunology 17(6), 613620.CrossRefGoogle ScholarPubMed
Tam, SW and Cook, L (1984) Sigma opiates and certain antipsychotic drugs mutually inhibit (+)-[3H] SKF 10,047 and [3H]haloperidol binding in guinea pig brain membranes. Proceedings of the National Academy of Sciences 81(17), 56185621.CrossRefGoogle Scholar
Tang, SW, Helmeste, D and Leonard, B (2021) Inflammatory neuropsychiatric disorders and COVID-19 neuroinflammation. Acta Neuropsychiatrica 33(4), 165177.CrossRefGoogle ScholarPubMed
Tang, SW, Helmeste, DM, Fang, H, Li, M, Vu, R, Bunney, W Jr, Potkin, S and Jones, EG (1997) Differential labeling of dopamine and sigma sites by [3H]nemonapride and [3H]raclopride in postmortem human brains. Brain Research 765(1), 712.CrossRefGoogle Scholar
Tang, SW, Helmeste, DM and Leonard, BE (2017) Neurodegeneration, neuroregeneration, and neuroprotection in psychiatric disorders. Modern Trends in Pharmacopsychiatry 31, 107123.CrossRefGoogle ScholarPubMed
Tang, SW and Tang, WH (2019) Opportunities in novel psychotropic drug design from natural compounds. International Journal of Neuropsychopharmacology 22(9), 601607.CrossRefGoogle ScholarPubMed
Tansey, CM, Louie, M and Loeb, M (2007) One-year outcomes and health care utilization in survivors of severe acute respiratory syndrome. Archives of Internal Medicine 167(12), 13121320.CrossRefGoogle ScholarPubMed
Taquet, M, Luciano, S, Geddes, JR and Harrison, PJ (2021) Bidirectional associations between COVID-19 and psychiatric disorder: retrospective cohort studies of 62 354 COVID-19 cases in the USA. Lancet Psychiatry 8(2), 130140.CrossRefGoogle ScholarPubMed
Tarazona, R, González-García, A, Zamzami, N, Marchetti, P, Frechin, N, Gonzalo, JA, Ruiz-Gayo, M, van Rooijen, N, Martínez, C, Kroemer, G (1995) Chlorpromazine amplifies macrophage-dependent IL-10 production in vivo . Journal of Immunology 154(2), 861870.Google ScholarPubMed
Tashiro, M, Duan, X, Kato, M, Miyake, M, Watanuki, S, Ishikawa, Y, Funaki, Y, Iwata, R, Itoh, M, Yanai, K (2008) Brain histamine H1 receptor occupancy of orally administered antihistamines, bepotastine and diphenhydramine, measured by PET with 11C-doxepin. British Journal of Clinical Pharmacology 65(6), 811821.CrossRefGoogle ScholarPubMed
Tawakul, AA, Alharbi, AH, Basahal, AM, Almalki, AM, Alharbi, B, Almaghrabi, M and Imam, A (2021) Neurological symptoms and complications of COVID-19 among patients in a tertiary hospital in Saudi Arabia. Cureus 13(11), e19200. doi: 10.7759/cureus.19200.Google Scholar
Teixeira, AL, Krause, TM, Ghosh, L, Shahani, L, Machado-Vieira, R, Lane, SD, Boerwinkle, E and Soares, JC (2021) Analysis of COVID-19 infection and mortality among patients with psychiatric disorders, 2020. JAMA Network Open 4(11), e2134969. doi: 10.1001/jamanetworkopen.2021.34969.CrossRefGoogle ScholarPubMed
Thompson, LA, Gurka, MJ, Filipp, SL, Schatz, DA, Mercado, RE, Ostrov, DA, Atkinson, MA and Rasmussen, SA (2021) The influence of selection bias on identifying an association between allergy medication use and SARS-CoV-2 infection. EClinicalMedicine 37(1), 100936. doi: 10.1016/j.eclinm.2021.100936.CrossRefGoogle ScholarPubMed
Toniolo, S, Scarioni, M, Di Lorenzo, F, Hort, J, Georges, J, Tomic, S, Nobili, F, Frederiksen, KS and Management Group of the EAN Dementia and Cognitive Disorders Scientific Panel (2021) Dementia and COVID-19, a bidirectional liaison: risk factors, biomarkers, and optimal health care. Journal of Alzheimers Disease 82(3), 883898.CrossRefGoogle ScholarPubMed
Troyer, EA, Kohn, JN and 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
Tsuneizumi, T, Babb, SM and Cohen, BM (1992) Drug distribution between blood and brain as a determinant of antipsychotic drug effects. Biological Psychiatry 32(9), 817824.CrossRefGoogle ScholarPubMed
Tzur Bitan, D, Krieger, I, Kridin, K, Komantscher, D, Scheinman, Y, Weinstein, O, Cohen, AD, Cicurel, AA and Feingold, D (2021) COVID-19 prevalence and mortality among schizophrenia patients: a large-scale retrospective cohort study. Schizophrenia Bulletin 47(5), 12111217.CrossRefGoogle ScholarPubMed
Varatharaj, A, Thomas, N, Ellul, MA, Davies, NWS, 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 and CoroNerve Study Group (2020) Neurological and neuropsychiatric complications of COVID-19 in 153 patients: a UK-wide surveillance study. The Lancet Psychiatry 7(10), 875882.CrossRefGoogle ScholarPubMed
Varghese, E, Samuel, SM, Liskova, A, Kubatka, P and Büsselberg, D (2021) Diabetes and coronavirus (SARS-CoV-2): molecular mechanism of metformin intervention and the scientific basis of drug repurposing. PLoS Pathogens 17(6), e1009634. doi: 10.1371/journal.ppat.1009634.CrossRefGoogle ScholarPubMed
Vasek, MJ, Garber, C, Dorsey, D, Durrant, DM, Bollman, B, Soung, A, Yu, J, Perez-Torres, C, Frouin, A, Wilton, DK, Funk, K, DeMasters, BK, Jiang, X, Bowen, JR, Mennerick, S, Robinson, JK, Garbow, JR, Tyler, KL, Suthar, MS, Schmidt, RE, Stevens, B and Klein, RS (2016) A complement-microglial axis drives synapse loss during virus-induced memory impairment. Nature 534(7608), 538543.CrossRefGoogle ScholarPubMed
Vela, JM (2020) Repurposing sigma-1 receptor ligands for COVID-19 therapy? Frontiers in Pharmacology 11, 582310.CrossRefGoogle ScholarPubMed
Verdecchia, P, Cavallini, C, Spanevello, A and Angeli, F (2020) The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. European Journal of Internal Medicine 76, 1420.CrossRefGoogle ScholarPubMed
Viszlayová, D, Sojka, M, Dobrodenková, S, Szabó, S, Bilec, O, Turzová, M, Ďurina, J, Baloghová, B, Borbély, Z, Kršák, M (2021) SARS-CoV-2 RNA in the cerebrospinal fluid of a patient with long COVID. Therapeutic Advances in Infectious Disease 8, 20499361211048572. doi: 10.1177/20499361211048572.CrossRefGoogle ScholarPubMed
von Bahr, C, Ursing, C, Yasui, N, Tybring, G, Bertilsson, L and Röjdmark, S (2000) Fluvoxamine but not citalopram increases serum melatonin in healthy subjects-- an indication that cytochrome P450 CYP1A2 and CYP2C19 hydroxylate melatonin. European Journal of Clinical Pharmacology 56(2), 123127.CrossRefGoogle Scholar
Wang, F, Kream, RM and Stefano, GB (2020) Long-term respiratory and neurological sequelae of COVID-19. Medical Science Monitor 26, e928996. doi: 10.12659/MSM.928996.Google ScholarPubMed
Wang, L, Pavlou, S, Du, X, Bhuckory, M, Xu, H and Chen, M (2019) Glucose transporter 1 critically controls microglial activation through facilitating glycolysis. Molecular Neurodegeneration 14(1), 2. doi: 10.1186/s13024-019-0305-9.CrossRefGoogle ScholarPubMed
Wang, Q, Xu, R and Volkow, ND (2021b) Increased risk of COVID-19 infection and mortality in people with mental disorders: analysis from electronic health records in the United States. World Psychiatry 20(1), 124130.CrossRefGoogle ScholarPubMed
Wang, S, Ma, P, Zhang, S, Song, S, Wang, Z, Ma, Y, Xu, J, Wu, F, Duan, L, Yin, Z, Luo, H, Xiong, N, Xu, M, Zeng, T, Jin, Y (2020b) Fasting blood glucose at admission is an independent predictor for 28-day mortality in patients with COVID-19 without previous diagnosis of diabetes: a multi-centre retrospective study. Diabetologia 63(10), 21022111.CrossRefGoogle ScholarPubMed
Wang, W, Shen, M, Tao, Y, Fairley, CK, Zhong, Q, Li, Z, Chen, H, Ong, JJ, Zhang, D, Zhang, K, Xing, N, Guo, H, Qin, E, Guan, X, Yang, F, Zhang, S, Zhang, L, He, K (2021) Elevated glucose level leads to rapid COVID-19 progression and high fatality. BMC Pulmonary Medicine 21(1), 64. doi: 10.1186/s12890-021-01413-w.CrossRefGoogle ScholarPubMed
Wang, X, Wang, S, Sun, L and Qin, G (2020c) Prevalence of diabetes mellitus in 2019 novel coronavirus: a meta-analysis. Diabetes Research and Clinical Practice 164(4), 108200. doi: 10.1016/j.diabres.2020.108200.CrossRefGoogle ScholarPubMed
Wargny, M, Potier, L, Gourdy, P, Pichelin, M, Amadou, C, Benhamou, PY, Bonnet, JB, Bordier, L, Bourron, O, Chaumeil, C, Chevalier, N, Darmon, P, Delenne, B, Demarsy, D, Dumas, M, Dupuy, O, Flaus-Furmaniuk, A, Gautier, JF, Guedj, AM, Jeandidier, N, Larger, E, Le Berre, JP, Lungo, M, Montanier, N, Moulin, P, Plat, F, Rigalleau, V, Robert, R, Seret-Bégué, D, Sérusclat, P, Smati, S, Thébaut, JF, Tramunt, B, Vatier, C, Velayoudom, FL, Vergès, B, Winiszewski, P, Zabulon, A, Gourraud, PA, Roussel, R, Cariou, B and Hadjadj, S (2021) CORONADO investigators. Predictors of hospital discharge and mortality in patients with diabetes and COVID-19: updated results from the nationwide CORONADO study. Diabetologia. 64(4), 778794.CrossRefGoogle ScholarPubMed
Weinstein, SP, Paquin, T, Pritsker, A and Haber, RS (1995) Glucocorticoidinduced insulin-resistance – dexamethasone inhibits the activation of glucose-transport in rat skeletal-muscle by both insulin-related and non-insulin-related stimuli. Diabetes 44(4), 441445.CrossRefGoogle Scholar
Weissman, AD, Casanova, MF, Kleinman, JE, London, ED and De Souza, EB (1991) Selective loss of cerebral cortical sigma, but not PCP binding sites in schizophrenia. Biological Psychiatry 29(1), 4154.CrossRefGoogle ScholarPubMed
Wiesel, FA and Alfredsson, G (1976) The distribution and metabolism of chlorpromazine in rats and the relationship to effects on cerebral monoamine metabolism. European Journal of Pharmacology 40(2), 263272.CrossRefGoogle ScholarPubMed
Woo, MS, Malsy, J, Pöttgen, J, Seddiq Zai, S, Ufer, F, Hadjilaou, A, Schmiedel, S, Addo, MM, Gerloff, C, Heesen, C, Schulze Zur Wiesch, J, Friese, MA (2020) Frequent neurocognitive deficits after recovery from mild COVID-19. Brain Communications 2(2), fcaa205. doi: 10.1093/braincomms/fcaa205.CrossRefGoogle ScholarPubMed
Wrapp, D, Wang, N, Corbett, KS, Goldsmith, JA, Hsieh, CL, Abiona, O, Graham, BS and McLellan, JS (2020) Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367(6483), 12601263.CrossRefGoogle ScholarPubMed
Wu, J, Li, H, Geng, Z, Wang, Y, Wang, X and Zhang, J (2021) Subtypes of nurses’ mental workload and interaction patterns with fatigue and work engagement during coronavirus disease 2019 (COVID-19) outbreak: a latent class analysis. BMC Nursing 20(1), 206. doi: 10.1186/s12912-021-00726-9.CrossRefGoogle ScholarPubMed
Xu, J and Liu, C (2021) Infodemic vs. pandemic factors associated to public anxiety in the early stage of the COVID-19 outbreak: a cross-sectional study in China. Frontiers in Public Health 9, 723648. doi: 10.3389/fpubh.2021.723648.CrossRefGoogle ScholarPubMed
Xu, L, He, D and Bai, Y (2016) Microglia-mediated inflammation and neurodegenerative disease. Molecular Neurobiology 53(10), 67096715.CrossRefGoogle ScholarPubMed
Zhan, YX, Zhao, SY, Yuan, J, Liu, H, Liu, YF, Gui, LL, Zheng, H, Zhou, YM, Qiu, LH, Chen, JH, Yu, JH, Li, SY (2020) Prevalence and influencing factors on fatigue of first-line nurses combating with COVID-19 in China: a descriptive cross-sectional study. Current Medical Science 40(4), 625635.CrossRefGoogle ScholarPubMed
Zhand, N and Joober, R (2021) Implications of the COVID-19 pandemic for patients with schizophrenia spectrum disorders: narrative review. BJPsych Open 7(1), e35. doi: 10.1192/bjo.2020.157.CrossRefGoogle ScholarPubMed
Zhang, J, Kong, W, Xia, P, Xu, Y, Li, L, Li, Q, Yang, L, Wei, Q, Wang, H, Li, H, Zheng, J, Sun, H, Xia, W, Liu, G, Zhong, X, Qiu, K, Li, Y, Wang, H, Wang, Y, Song, X, Liu, H, Xiong, S, Liu, Y, Cui, Z, Hu, Y, Chen, L, Pan, A and Zeng, T (2020b) Impaired fasting glucose and diabetes are related to higher risks of complications and mortality among patients with coronavirus disease 2019. Frontiers in Endocrinology 11, 525.CrossRefGoogle ScholarPubMed
Zhang, P, Li, J and Liu, H (2020) Long-term bone and lung consequences associated with hospital-acquired severe acute respiratory syndrome: a 15-year follow-up from a prospective cohort study. Bone Research 8(1), 8.CrossRefGoogle ScholarPubMed
Zhao, YM, Shang, YM and Song, WB (2020) Follow-up study of the pulmonary function and related physiological characteristics of COVID-19 survivors three months after recovery. EClinicalMedicine 25, 100463.CrossRefGoogle ScholarPubMed
Zhou, H, Lu, S, Chen, J, Wei, N, Wang, D, Lyu, H, Shi, C and Hu, S (2020) The landscape of cognitive function in recovered COVID-19 patients. Journal of Psychiatric Research 129, 98102.CrossRefGoogle ScholarPubMed
Zhou, X, Tian, B and Han, HB (2021) Serum interleukin-6 in schizophrenia: a system review and meta-analysis. Cytokine 141(6), 155441. doi: 10.1016/j.cyto.2021.155441.CrossRefGoogle ScholarPubMed
Zhu, B, Jin, S, Wu, L, Hu, C, Wang, Z, Bu, L, Sun, H, Wang, X, Qu, S, Chen, D (2020a) J-shaped association between fasting blood glucose levels and COVID-19 severity in patients without diabetes. Diabetes Research and Clinical Practice 168, 108381. doi: 10.1016/j.diabres.2020.108381.CrossRefGoogle ScholarPubMed
Zhu, L, She, ZG, Cheng, X, Qin, JJ, Zhang, XJ, Cai, J, Lei, F, Wang, H, Xie, J, Wang, W, Li, H, Zhang, P, Song, X, Chen, X, Xiang, M, Zhang, C, Bai, L, Xiang, D, Chen, MM, Liu, Y, Yan, Y, Liu, M, Mao, W, Zou, J, Liu, L, Chen, G, Luo, P, Xiao, B, Zhang, C, Zhang, Z, Lu, Z, Wang, J, Lu, H, Xia, X, Wang, D, Liao, X, Peng, G, Ye, P, Yang, J, Yuan, Y, Huang, X, Guo, J, Zhang, BH and Li, H (2020b) Association of blood glucose control and outcomes in patients with COVID-19 and pre-existing type 2 diabetes. Cell Metabolism 31(6), 10681077.CrossRefGoogle ScholarPubMed
Zilberstein, D and Dwyer, DM (1984) Antidepressants cause lethal disruption of membrane function in the human protozoan parasite Leishmania. Science 226(4677), 977979.CrossRefGoogle ScholarPubMed
Zilberstein, D, Liveanu, V and Gepstein, A (1990) Tricyclic drugs reduce proton motive force in Leishmania donovani promastigotes. Biochemical Pharmacology 39(5), 935940.CrossRefGoogle ScholarPubMed
Zucker, S, Zarrabi, HM, Schubach, WH, Varma, A, Derman, R, Lysik, RM, Habicht, G and Seitz, PM (1990) Chlorpromazine-induced immunopathy: progressive increase in serum IgM. Medicine (Baltimore) 69(2), 92100.CrossRefGoogle ScholarPubMed