key: cord-0844148-t6am4sw5 authors: Clark, Ian Albert title: Chronic cerebral aspects of long COVID, post‐stroke syndromes and similar states share their pathogenesis and perispinal etanercept treatment logic date: 2022-02-16 journal: Pharmacol Res Perspect DOI: 10.1002/prp2.926 sha: 89d55ee5f6c16780a4910872b3d99700fad98d82 doc_id: 844148 cord_uid: t6am4sw5 The chronic neurological aspects of traumatic brain injury, post‐stroke syndromes, long COVID‐19, persistent Lyme disease, and influenza encephalopathy having close pathophysiological parallels that warrant being investigated in an integrated manner. A mechanism, common to all, for this persistence of the range of symptoms common to these conditions is described. While TNF maintains cerebral homeostasis, its excessive production through either pathogen‐associated molecular patterns or damage‐associated molecular patterns activity associates with the persistence of the symptoms common across both infectious and non‐infectious conditions. The case is made that this shared chronicity arises from a positive feedback loop causing the persistence of the activation of microglia by the TNF that these cells generate. Lowering this excess TNF is the logical way to reducing this persistent, TNF‐maintained, microglial activation. While too large to negotiate the blood‐brain barrier effectively, the specific anti‐TNF biological, etanercept, shows promise when administered by the perispinal route, which allows it to bypass this obstruction. done about the syndromes observed in post-stroke patients, who, permanently deprived of the neurons that died, can expect to have limited function for the rest of their lives. Similarly, pathogens are often deemed to kill cells directly. Thus teaching and learning about infectious diseases runs the risk of being largely limited to the specific diagnosis of the pathogen in order to select the anti-pathogen agent with the best contemporary reputation. Despite the advent of awareness of pathogen-associated molecular patterns (PAMPs) and damage (or danger) associated molecular patterns (DAMPs), and the activities of the cytokines thus induced through Toll-like, and functionally similar pattern recognition receptors in increased concentrations (discussed below), these earlier mindsets can also dominate research into both these non-infectious and infectious conditions, preventing them being discussed together, using a common knowledge base. As the title infers, this commentary is intended to foster this cytokine approach becoming widespread enough to allow a broader understanding of shared features of what are, traditionally, seen as profoundly different illnesses, with very different causes. In particular, discussion focusses on how a number of chronic neurological changes can be common to certain conditions, be they infectious or non-infectious. It seems appropriate to expand here on the above PAMP and DAMP terminology, since it is central to understanding the pathophysiology of both infectious and non-infectious diseases, including their influence on neurological function. Further detail of this concept, attributable to Janeway 2 and Matzinger, 3 has been reviewed before from our group, albeit in an Alzheimer's disease context. 4 These insights illuminate a useful concept for explaining how not only molecular patterns on pathogens (grouped as PAMPs), but also patterns revealed by host tissue damage or hypoxia (grouped as DAMPs), act to release TNF and related cytokines. In brief, they do so by these molecular patterns recognizing sites such as Toll-like receptors (TLRs) expressed on or in many cells, but most strongly on macrophages and microglia, for which they act as agonists. This terminology facilitates explaining the natural history of two infectious diseases, one bacterial (Lyme disease) one viral , both triggered by PAMPs, in the same terms as non-infectious states such as post-stroke syndrome and TBI triggered by DAMPs, as is done here. Despite this difference in what initiates them, the illness they cause can often be very similar, with diverse and complex cerebral symptoms persisting after the pathogen in the case of Lyme disease, or the acute hypoxia in the case of stroke, or tissue damage in TBI, have long gone. Clinical similarities within acute infectious diseases can also be rationalized within this framework. At the Dana-Faber Institute in Boston, when rTNF was being tested for killing tumors in patients, 5 and before the PAMP/DAMP concept had been elucidated, these authors and I exchanged views on the side effects thus generated. To these researchers this toxicity mimicked influenza, whereas it reminded me more of severe malaria. Two other groups have since independently reported influenza mimicry by rTNF administration. 6, 7 Years later this striking diagnostic overlap became official in a blinded trial conducted by clinical experts in influenza and falciparum malaria. 8 This is strikingly consistent with two different PAMPs, one on a virus, the other on a protozoan, generating the one illness by inducing TNF, an activity for which they have both been on record for some time. It also rationalizes why the syndromes seen in acute COVID-19, bacterial sepsis, and Lyme disease all have influenza high on their differential diagnosis list. To illustrate the DAMP principle more broadly, the wide spectrum of involvement of high-mobility group box 1 (HMGB1) warrants brief discussion. This DNA-binding protein is evolutionarily conserved (e.g. is present in shellfish 9 as well as humans). Indeed, it is constitutively expressed within the nucleus of virtually all types of cells. As a nuclear protein released from damaged cells or those undergoing non-lethal physiological stress 10 and secreted by activated leukocytes, HMGB1 is a common intermediary between cellular stress, or physical damage, and TNF release. Moreover, hypoxia, 11 and TNF itself, 12 can induce HMGB1 release. Thus it synergizes with both DAMPs and PAMPs. 13 HMGB1 is reported to function as such not only during the pathogenesis of Lyme disease, 14 but also COVID-19, 15 influenza, 16 TBI, 17, 18 and the cognitive loss that can follow post-operative cognitive deficiency (POCD). 19 Ischemia also induces HMGB1, 20 with its importance implied by the ability of an anti-HMGB1 antibody to improve brain dysfunction in a rat model of cardiac arrest. 21 Thus HMGB1 illustrates how closely interwoven are the signaling pathways that drive disease pathogenesis in both infectious and non-infectious states. The capacity to generate the cytokine TNF evidently appeared extremely early in biological evolution, and has been scrupulously retained. Remarkably, the human TNF molecule, or at least its receptor-binding portion, pre-dates bilateral symmetry, in that Acropora spp., the major reef-builder corals, generate a TNF whose receptor recognizes human TNF. 22 It is not surprising that a molecule so rigorously preserved has proven to be widely and essentially involved in physiology 23 and disease 24 of more complex creatures such as insects and fish, as well as the physiology and disease in all vertebrates so far examined. It also has roles in mediating innate immunity. Most TNF is generated by macrophages stimulated by PAMPs or DAMPs, with microglia, the cerebral equivalent of macrophages, taking over the role within the blood-brain barrier. Reducing its excess levels in chronic non-cerebral inflammatory diseases such as rheumatic arthritis, Crohn's disease and psoriasis has proved to be an enormous clinical success, but its application in neurological conditions is as yet in its infancy. This is partly because its physiological roles in the brain are so subtle and complex, but commercial sparring within a highly competitive field also plays a large role in preventing this being broadly appreciated. TNF has an astonishing number of essential roles in normal brain tissue. This is reviewed in some detail here in order to demonstrate how dependent normal brain function is the widespread homeostatic roles of this cytokine, for example through controlling neuronal plasticity. 25 TNF and other members of the TNF superfamily of cytokines 26 mediate neurite outgrowth, normal fetal development of nociception, and the survivability, excitability and cell differentiation mediated by nerve growth factor. 27 Its biological influence spans generations, with a requirement for adequate maternal TNF to induce, in milk, the chemokines needed for normal hippocampal development and memory in offspring. 28 TNF released during physiological neuronal activity plays a crucial role in regulating the strength of normal synaptic transmission. 29 Moreover, there has been evidence for some time now that TNF governs behavioral phenotypes in physiological ageing, without immunological challenge. 30 As we have reviewed, 31 free synaptic glutamate, which is central to synaptic function, is largely regulated by TNF's control over both glutaminase and certain key glutamate re-uptake transporters. Thus TNF, one of the few cytokines styled as gliotransmitters, has, as reviewed, 32 subtle but effective control over synaptic physiology, influencing AMPA receptors on synapses, synaptic plasticity (considered, by Hebbian theory, to be an important foundation of memory and learning), and long-term potentiation, a paradigm for how memory may be consolidated at the molecular level. In excess it can lead to glutamate excitotoxicity, which is discussed later. In other words, the brain requires low levels of properly orchestrated TNF for normal physiological function. Clearly this level has to fluctuate as physiology requires. Normal physiological neuronal activity therefore requires TNF to be released in homeostatically controlled quantities from microglia, astrocytes and neurons before it is cleared by TNF receptors. TNF is also involved in normal neurotransmission via modulating excitatory inputs, 32 trafficking of AMPA receptors, 33 homeostatic synaptic scaling, 34 and long-term potentiation. 35 Furthermore, it maintains normal background levels of neurogenesis. 36 Mitochondrial function depends on TNF, 37 as does regulation of the neurotransmitter, orexin, 38 which, as we recently reviewed, 39 controls sleep, motor control, focused effort, appetite and water intake. TNF also regulates neuronal type-1 inositol trisphosphate receptors (IP3R), which are central to neuronal Ca ++ homeostasis, and thus the ionic signaling cascades on which normal function of neurons depends. 40 This large functional overlap between the brain and the innate immune system described in the infectious disease literature is reinforced in a comprehensive review by others. 41 Through this bigger picture, the subtle but widespread functional changes detected by administering the then newest revisions of the WAIS-III and WMS-III mental performance tests to chronic Lyme disease patients 42 can be better appreciated. Equally, it is small wonder that a recent study of a relatively young cohort of COVID-19 survivors reported that a substantial proportion exhibited cognitive dysfunction. 43 Clearly, all the above functions are vulnerable to TNF being outside its physiological range when it is over-induced by either PAMPs or DAMPs that can enter, or be generated in, the brain. The capacity of microglia, via the cytokines these cells generate, to influence neuronal function, are regularly reviewed. 44 Thus, increased cerebral levels of microglial-origin TNF, whether arising from induction by PAMPs on pathogens such as SARS-CoV-2, influenza viruses or Borrelia spp., by DAMPs (e.g. post-stroke syndromes) or, as discussed below, non-infectious influences such as hypoxia or trauma, can alter brain function in the same way. 45 Understanding the physiological roles of TNF in biology gives us the opportunity to visualize how the incredibly wide range of subtle function loss reported during persistent Lyme disease, 46 long COVID-19, 47 and post-stroke neurological changes, might arise. This arguably bewildering range and degree of changes is, nevertheless, consistent with what can seem, at first sight, an equally bewildering array of pleiotropic functions of TNF in neurophysiology, as outlined above. This degree of complexity is entirely compatible with the subtle intellectual and memory deficits documented to affect many performance tasks in persistent Lyme disease. 42 It is telling of that period that these authors should have inserted the caveat that further work was needed to definitively rule out non-specific illness effects on cerebral function. Nowadays, in contrast, we can infer from the literature in this and the above paragraph that little if anything specific is involved in persistent Lyme disease. By rationalizing this non-specificity, this approach may well shed new light on settling the controversies that have plagued our understanding of this disease for decades. Once TNF, IL-1β, IL-6, IL-4 and other host-origin cytokines were identified, interacting pathways proposed, and laboratory reagents available, an awareness of their roles in states such as infectious and inflammatory disease, sickness behavior, ageing, and neurodegeneration dominated their literatures. Consequently, the infectious disease world often demonstrates little awareness that, at lower concentrations, these cytokines have many essential roles in brain physiology. Homeostatic normal synaptic plasticity and scaling as well as associated glutamate control, neurogenesis, neuromodulation, synaptic function, memory, 29 learning, 48 and cognitive function 49 are particularly important examples. We discussed these aspects of TNF further in 2010, 50 and similar roles of other cytokines have been updated since. 41 Importantly, all of this low-level fluctuating cerebral cytokine activity is also necessary for homeostasis in healthy brains. As summarized earlier, such homeostasis depends on these cytokines being appropriately and autonomously orchestrated, both in precise locations and minute concentrations, within the bounds of neurophysiology. The next challenge is to understand why such neurophysiological change can be rendered chronic, whether in post-stroke syndromes, Lyme disease, COVID-19, and influenza encephalopathy survivors, whereas systemic cytokine levels and associated acute symptoms do not persist. In 2007 it was reported that TNF generation in the mouse, after intraperitoneal injection of bacterial LPS, persists in the CSF for very much longer (at least 10 months) than in the serum (6 h). 51 It is plausible, therefore, that the microglia whose cytokines flow into this CSF do not become LPS-tolerant, as do mouse macrophages after exposure to LPS. 52 Human macrophages also prove to react to LPS in this way. 53 These intriguing sets of observations and proposals are consistent with the report 54 of a positive feedback loop in the activation of microglia by the TNF that these microglia generate. In other words, an autocrine loop exists whereby TNF can prolong the activation of the cerebral microglia that generate it. Others have since invoked an intermediary role for brain-derived neurotropic factor (BDNF) in this feedback microglial activation. 55 These cellular interactions have been further investigated from another angle by Puntener and co-workers in Southampton. 56 In brief, they concluded that the in- A precedent for the existence of long-term microglial activation, with a DAMP rather than a PAMP the original cause, has been reported in ischemic stroke, 58 subarachnoid hemorrhage, 59 and TBI. 60, 61 Since this was demonstrated in vivo in two of these three quoted studies, it seems pressing for the techniques in the previous paragraph to be applied to influenza, persistent Lyme disease and long COVID brains. This concept is also consistent with the endotoxin tolerance being observed systemically but not in the brain. Importantly, it implies that perispinally administered etanercept (discussed later) can deactivate these microglia by inactivating this excess TNF, hence removing the positive feedback loop that causes its production to persist. In this way these PAMP and DAMP-induced cytokines, and not the pathogen, ischemia or trauma directly, are argued here to be the essential cause of most of the commonly seen neurological dysfunction in the persistent stage of all these illnesses. Logically, therefore, this principle applies to the persistent cerebral phases of COVID-19, influenza or Lyme disease as well as to encephalopathies seen post-stroke or in TBI survivors. Having already covered similar ground with long COVID, 62 here we discuss the wider literature that has accumulated with influenza encephalopathy. A convenient point to start recounting it is the work of Jurgens and co-authors, who reported that a mouse model exhibited increased microglial activity and increased inflammatory cytokine generation in the hippocampus, and impaired reversal learning in the Morris water maze. 63 Others 64 subsequently confirmed these cytokine and microglial observations, as well as an accompanying deficit in spatial learning. Importantly, others 65 have recently discussed the concept of post-acute sequelae caused by non-persistent viruses causing chronic inflammation. Since these are non-infectious conditions, their pathophysiology is exclusively DAMP-driven, mostly by hypoxia and the products of tissue damage. Non-infectious inflammatory conditions such as poststroke syndromes 66 provide a convenient way to demonstrate the importance of TNF, however it is generated, in conditions that share this clinical picture. Thirty years ago Ghezzi and co-workers reported that hypoxia -the basis of cerebral stroke -greatly increases TNF generation by human monocytes, which are closely related to microglia. 67 Consequences include mitochondrial DNA and HMGB1 escaping from damaged mitochondria 68 and cell nuclei. 69 For example, persistent HMGB1 release has been confirmed in patients who have recently experienced stroke, 70 TBI, 18 and cardiac arrest. 71 The cytokines thereby secreted by these cerebrally-released DAMPs inevitably generate the same characteristic group of symptoms as seen in long COVID-19, persistent Lyme disease, and influenza encephalopathy. Examples are given in the next paragraph. A cluster of chronic symptoms form the syndrome observed when cerebral TNF becomes inappropriately excessive for the normal function of a particular cerebral site. Below, we discuss fatigue, neurogenic pain, delirium, aggressiveness, and suicide tendency as examples. The ingredients in a particular mix can be expected to depend on the site and pattern of excessive induction of TNF, independent of whether the driving force is a PAMP or a DAMP ( Figure 1 ). Even within a single syndrome, further differences might well also result from different genetic backgrounds or epigenetic modifications in individuals. In short, from a pathogenesis perspective these symptoms are all under the one TNF umbrella, whether infectious or non-infectious in origin. Thus there is a rationale for excessive cerebral TNF, and thus the same chronic symptoms, to persist, either when a pathogen is no longer present (e.g. long COVID and persistent Lyme disease), or is an irrelevancy (e.g. post-stroke syndromes and TBI). When an acute excess of TNF extends to chronic intracerebral TNF An illustration of how a range of chronic external stimuli that present pathogen-associated molecular patterns or pathogen-associated molecular patterns to various Toll-like receptors on different cerebral cellular components, and thus induce TNF and TNF-induced cytokines. In contrast to activated macrophages, activated microglia maintain their TNF-generating state, generating a series of distinctive clinical consequences Fatigue Delirium In contrast to macrophages, activated microglia are kept in this TNF-generating state by the excess TNF they produce. The case was subsequently made 81 for excess TNF diminishing the intracellular movement of mitochondria required for optimal ATP generation, leading to their disintegration and a resultant diminished capacity of the organism to utilize oxygen. 82 The chronically activated microglia that can be expected to be the source of this chronic excess TNF -see two Sections back, and in Figure 1 -have already been documented in chronic fatigue patients. 83 Delirium is variously described as an acute and profound disturbance of thinking, memory, orientation, perception and emotion. Transient delirium is common in intensive care units, and has been perceptively described as an extreme manifestation of the sickness behavior caused by TNF and the other cytokines it induces. 95 Although described in Lyme disease, 96 Behavioral changes, such as aggressiveness, are also recorded in persistent Lyme disease. 107 As with delirium, aggressiveness has also been observed in the persistent neurological impairments after cerebral malaria in African children. The syndrome often includes persistent deficits in cognition, learning ability at school, attention, memory, visuo-spatial and language skills that often begin after recovery from acute cerebral malaria. 108 The relevant literature on suicidal tendencies, plausibly termed self-aggression in this context, also warrants summarizing here. Risk of suicide is reportedly increased in many of the conditions being discussed here, both infectious (Lyme disease 112 and long COVID-19 113 ) and non-infectious states with tissue damage and consequent DAMP release (TBI 114 and post-traumatic stress disorder (PTSD)). The latter is widely accepted to be a tragedy of a particular magnitude for returned service personnel. 115 It is well-recognized that pro-inflammatory cytokines such as TNF are chronically raised in PTSD. 116 The nature of this subsection of this review also draws attention to the literature that focusses on TNF in order to gain an understanding of mechanisms of psychiatric conditions. [117] [118] Therefore, should cerebral TNF, the initiator of the above cytokine cascade, and mostly generated in the brain by microglia, become inappropriately increased through the advent of an infectious disease such as COVID-19 119 or Lyme disease, 120 we can expect that mental health, including memory, cognition, and alertness, would be harmed. Circulating cytokine levels can inform about the immune response and the acute disease, but tell us little about why the neurological symptoms seen in the chronic stage of these acute infections occur. Importantly, these postacute sequelae also can be said to occur in non-infectious disease states where the same cytokines are generated by DAMPs. In these circumstances, such as in post-stroke and TBI syndromes, the acute stage consisted of trauma (TBI) or ischemia (post-stroke syndromes), and pathogens are clearly irrelevant. In other words, chronically increased intracerebral cytokines causes these chronic changes in which pathogens play no part. This is consistent with the argument that antibiotics are inappropriate treatments for persistent Lyme disease. 85 Above, the case is made that the neuropathophysiology that brings about the symptoms of post-stroke syndromes, TBI, long COVID-19, persistent influenza and Lyme disease are essentially identical. This may seem to be counterintuitive, since the first two have non-infectious origins, while the other three are initiated by infectious agents. As we have indicated, their commonality arises because DAMPs and PAMPs both activate the same intracerebral cytokine TNF-driven pathways. These regulatory pathways are essential in the homeostatically managed low, appropriately fluctuating TNF concentrations required to maintain brain health, but when over-stimulated they throw neurophysiology out of kilter. Inevitably, the harmful changes that ensue are common across the board, irrespective of whether they were initiated by DAMPs (as with hypoxia or trauma) or PAMPs (as with virus, bacteria or protozoan). As discussed earlier, changes in cerebral function, often subtle, but sometimes disabling, can become persistent. Inactivating chronically activated microglia by removing the excess TNF that maintains this state 54 is a logical explanation for this fundamental and rapid reversal to normal seen in post-stroke case studies 75 and the one random control trial to date. 76 As referenced earlier in the present text, post-stroke syndromes, as well as other non-infectious neurodegenerative diseases, typically exhibit the same fatigue, neurogenic pain, delirium and aggressiveness as are recorded in persistent Lyme and long COVID-19. This is consistent with the chronic cerebral TNF generation that maintains microglia in a chronically activated state, rather than the initial pathogen, being a plausible therapeutic target in persistent Lyme disease. Gabapentin, generally regarded as an anticonvulsant, is a synthetic analogue of gamma aminobutyric acid. Hampered by dose-limiting side effects, it has nevertheless been used, with some success, against chronic neurogenic pain, including in Lyme disease. 121 It has been demonstrated to reduce TNF levels, 122 arguably because of its capacity to enhance IL-10, a recognized inhibitor of TNF generation. 123 Glutamate is a key physiological excitatory neurotransmitter in virtually all activities of the nervous system, yet in excess it is extremely harmful. As we expanded upon 5 years ago 31 this combination of functional importance and potentially high toxicity demands tight control over its release and re-uptake. Crucially for understanding the outcome of etanercept therapy, both glutamate release [124] [125] and subsequent re-uptake 126 are inhibited by excessive cerebral TNF. Various alphaviruses cause the same chronic neurological sequelae as seen in COVID-19 and influenza. The glutamine analogue 6-diazo-5-oxo-L-norleucine (DON), albeit too toxic to be a practical drug, is a useful experimental agent. As does TNF, 124 This implies that specific anti-TNF agents, such as etanercept, can be expected to be more effective than DON, which has only one string to its bow, at treating this condition. With the realization that TNF is as central to normal signaling and pathophysiology in the central nervous system, as we have reviewed, 50 the therapeutic challenge, because of its molecular size, has been to get a specific anti-TNF biological through, or around, the blood-brain barrier in pharmaceutical amounts. Intriguingly, after The perispinal route consists of a shallow injection of the dose into the cerebrospinal venous system. 131 This is followed by a headdown tilt for 5 min 132,133 on the grounds that this allows, through reverse flow, rapid entry of large molecules into the CSF through the valveless veins that usually drain the cerebrospinal fluid. 134, 135 Hence the blood-brain barrier is bypassed, rather than penetrated. The history of appreciating the details of this drainage is welldocumented, 136 54 This can be expected to switch off chronic TNF production by these microglia, and explain the permanent restoration of much cerebral normality in case studies 75, 141 and the initial RCT 76 (Figure 2 ). The specific intention is to remove, permanently, the chronic symptoms that can be expected to persist indefinitely, long after the acute effects of PAMPS and DAMPs have passed. It has already been proposed as an argument for reversing the neurological aspects of long COVID-19 62 and post-cerebral malaria cognitive impairment. 142 The initial observation that perispinal treatment with a specific anti-TNF biological typically causes such a rapid return to normal neurological function was as striking as it was unexpected across the field. Certainly, large players can find the phenomenon more convenient to dismiss than to attempt to understand. My invited 2017 editorial to comment on a Clinical Advisory issued by the American Academy of Neurology (AAN) 130 In reality, once the initial PAMP or DAMP event arising from a viral or bacterial infection (PAMP-driven), or from trauma or hypoxia (DAMP driven), has occurred, the microglia begin to generate a stock range of cytokines. Some of these, such as IL-1β, will inevitably be present F I G U R E 2 Predictable effects of perispinal anti-TNF biologicals on these chronic neurological common events The distinctive clinical consequences of chronic cerebral excessive production. These symptoms can be predicted to dissipate once cerebral TNF levels are restored to its normal homeostatic levels Fatigue Delirium Removing this excess cerebral TNF deactivates microglia rapidly and long-term, thus reducing symptoms caused by inappropriately and chronically increased cerebral TNF Loss of ability to taste and smell • Post-stroke syndromes • Traumatic brain injury whether a PAMP or DAMPs induces the TNF, which has activated the caspase-1 that converts pre-IL-1 to IL-1β, a process for which indirect evidence was first reported in decades ago. 145 Subsequently, in an in vitro cerebral context, exposure of a human neuroblastoma cell line to TNF was shown to promote oxygen radical-mediated caspase-1 activation and thence IL-1β secretion. 146 This was largely the basis of developing the inflammasome concept. Not surprisingly, therefore, anti-TNF inhibits IL-1 generation. 147, 148 Moreover, infliximab, the first of the clinical anti-TNF biological agents, has been reported to reduce functionally similar cytokines, for example IL-1 as well as TNF levels. [149] [150] [151] It also reduces IL-6 148,150 and IL-8. 150 Hence every time TNF is mentioned in this text one can infer that IL-1β is a downstream fellow traveler and collaborator. Indeed in the publication, forty years ago, the first to argue for such cytokines causing disease as well as innate immunity, we assayed for IL-1 -then termed lymphocyte activating factor (LAF) -in addition to TNF, and discussed them together. 1 In the same decade, before it was appreciated that one induced the other, Carl spective. 153 Unfortunately these authors did not stray into the realm of neurologists. The author is most grateful to Sharyn Wragg for her artwork in the Figures. The author declares no conflict of interest or financial involvement in the research discussed here. No data available in the study. Ian Albert Clark https://orcid.org/0000-0003-1945-0370 Possible importance of macrophage-derived mediators in acute malaria Pillars article: approaching the asymptote? Evolution and revolution in immunology Danger signals: SOS to the immune system Amyloid beta: one of three danger-associated molecules that are secondary inducers of the proinflammatory cytokines that mediate Alzheimer's disease Recombinant human tumor necrosis factor administered as a 24-hour intravenous infusion. A phase 1 and pharmacologic study Phase 1 study of 24-hour continuous intravenous infusion of recombinant human tumor necrosis factor Recombinant tumour necrosis factor in the local therapy of malignant pleural effusion Distinguishing malaria and influenza: early clinical features in controlled human experimental infection studies A high mobility group box 1 (HMGB1) gene from Chlamys farreri and the DNA-binding ability and pro-inflammatory activity of its recombinant protein Alarmins: feel the stress Hypoxia-induced HMGB1 in wound tissues promotes the osteoblast cell proliferation via activating ERK/JNK signaling TNF-α induced secretion of HMGB1 from non-immune canine mammary epithelial cells (MTH53A) The alarmin HMGB1 acts in synergy with endogenous and exogenous danger signals to promote inflammation Assessment of HMGB-1 concentration in tick-borne encephalitis and neuroborreliosis Extracellular HMGB1: a therapeutic target in severe pulmonary inflammation including COVID-19 Systemic release of high mobility group box 1 protein during severe murine influenza Systemic HMGB1 neutralization prevents postoperative neurocognitive dysfunction in aged rats The danger zone: systematic review of the role of HMGB1 danger signalling in traumatic brain injury Surgery upregulates high mobility group box-1 and disrupts the blood-brain barrier causing cognitive dysfunction in aged rats High mobility group box 1 protein is released by neural cells upon different stresses and worsens ischemic neurodegeneration in vitro and in vivo Inhibiting high-mobility group box 1 (HMGB1) attenuates inflammatory cytokine expression and neurological deficit in ischemic brain injury following cardiac arrest in rats Evolution of TNF-induced apoptosis reveals 550 My of functional conservation Two types of TNF-alpha exist in teleost fish: phylogeny, expression, and bioactivity analysis of Type-II TNF-alpha3 in rainbow trout Oncorhynchus mykiss The expression of two novel orangespotted grouper (Epinephelus coioides) TNF genes in peripheral blood leukocytes, various organs, and fish larvae Inflammatory' cytokines: neuromodulators in normal brain? Developmental regulation of sensory neurite growth by the tumor necrosis factor superfamily member LIGHT TNF-alpha/TNFR1 signaling is required for the development and function of primary nociceptors Maternal hematopoietic TNF, via milk chemokines, programs hippocampal development and memory Learning and memory … and the immune system Tumor necrosis factor alpha and its receptors in behaviour and neurobiology of adult mice, in the absence of an immune challenge Excess cerebral TNF causing glutamate excitotoxicity rationalizes treatment of neurodegenerative diseases and neurogenic pain by anti-TNF agents Actions of TNF-alpha on glutamatergic synaptic transmission in the central nervous system Cell death after spinal cord injury is exacerbated by rapid TNFalpha-induced trafficking of GluR2-lacking AMPARs to the plasma membrane Synaptic scaling mediated by glial TNFalpha Evidence for a role for the group I metabotropic glutamate receptor in the inhibitory effect of tumor necrosis factor-alpha on long-term potentiation Tumor necrosis factor-alpha modulates survival, proliferation, and neuronal differentiation in neonatal subventricular zone cell cultures Tumor necrosis factor-alpha increases the steady-state reduction of cytochrome b of the mitochondrial respiratory chain in metabolically inhibited L929 cells Tumor necrosis factor-alpha regulates the hypocretin system via mRNA degradation and ubiquitination Inflammation-sleep interface in brain disease: TNF, insulin, orexin Tumor necrosis factor-alpha potentiates intraneuronal Ca2+ signaling via regulation of the inositol 1,4,5-trisphosphate receptor Immune modulation of learning, memory, neural plasticity and neurogenesis WAIS-III and WMS-III performance in chronic Lyme disease Assessment of cognitive function in patients after COVID-19 infection Neuroimmune activation drives multiple brain states Beyond host defense: emerging functions of the immune system in regulating complex tissue physiology Neuropsychiatric Lyme Borreliosis: an overview with a focus on a specialty psychiatrist's clinical practice. Healthcare The emerging spectrum of COVID-19 neurology: clinical, radiological and laboratory findings Cognitive dysfunction in mice deficient for TNF-and its receptors Evidence for a cytokine model of cognitive function The roles of TNF in brain dysfunction and disease Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration Reduced release of TNF and PCA from macrophages of tolerant mice Effects of gamma interferon on release of tumor necrosis factor-alpha from lipopolysaccharide-tolerant human monocyte-derived macrophages Autocrine activation of microglia by tumor necrosis factoralpha Positive feedback loop of autocrine BDNF from microglia causes prolonged microglia activation Long-term impact of systemic bacterial infection on the cerebral vasculature and microglia Dysregulation of brain and choroid plexus cell types in severe COVID-19 Thalamic microglial activation in ischemic stroke detected in vivo by PET and [11C] PK1195 Long-term functional consequences and ongoing cerebral inflammation after subarachnoid hemorrhage in the rat Inflammation after trauma: microglial activation and traumatic brain injury Dissemination of brain inflammation in traumatic brain injury Background to new treatments for COVID-19, including its chronicity, through altering elements of the cytokine storm Influenza infection induces neuroinflammation, alters hippocampal neuron morphology, and impairs cognition in adult mice Respiratory viral infection in neonatal piglets causes marked microglia activation in the hippocampus and deficits in spatial learning Implications of innate immunity in post-acute sequelae of non-persistent viral Infections Global brain inflammation in stroke Hypoxia increases production of interleukin-1 and tumor necrosis factor by human mononuclear cells Circulating mitochondrial DAMPs cause inflammatory responses to injury Expression analysis of HMGB1 in histological samples of malignant pleural mesothelioma Severe stroke induces long-lasting alterations of high-mobility group box 1 Systemic impact on secondary brain aggravation due to ischemia/reperfusion injury in postcardiac arrest syndrome: a prospective observational study using high-mobility group box 1 protein Regulation of bitter taste responses by tumor necrosis factor The role of TNF-α in inflammatory olfactory loss Inflammation arising from obesity reduces taste bud abundance and inhibits renewal Selective TNF inhibition for chronic stroke and traumatic brain injury: an observational study involving 629 consecutive patients treated with perispinal etanercept Phase I/II parallel double-blind randomized controlled clinical trial of perispinal etanercept for chronic stroke: improved mobility and pain alleviation Neuropsychiatric problems after traumatic brain injury: unraveling the silent epidemic Exploratory cohort study of associations between serum C -reactive protein and fatigue after stroke TNF alpha suppresses the expression of clock genes by interfering with E-box-mediated transcription Tumor necrosis factor-induced cerebral insulin resistance in Alzheimer's disease links numerous treatment rationales TNFα decreases mitochondrial movement in human airway smooth muscle TNFα induces mitochondrial fragmentation and biogenesis in human airway smooth muscle Neuroinflammation in patients with chronic fatigue syndrome/myalgic encephalomyelitis: An ¹¹C-(R)-PK11195 PET Study Long-COVID" by a Prospective Multimodal Evaluation of Patients with Long-Term Symptoms Attributed to SARS-CoV-2 Infection. Infectious diseases and therapy Commentary: 'What's in a name? That which we call a rose by any other name would smell as sweet Central post-stroke pain: clinical characteristics, pathophysiology, and management Recent findings on how proinflammatory cytokines cause pain: peripheral mechanisms in inflammatory and neuropathic hyperalgesia Chronic constriction injury-induced nociception is relieved by nanomedicinemediated decrease of rat hippocampal tumor necrosis factor Perispinal injection of a TNF blocker directed to the brain of rats alleviates the sensory and affective components of chronic constriction injury-induced neuropathic pain Randomized controlled trial validating the use of perispinal etanercept to reduce post-stroke disability has wide-ranging implications Outcomes of children treated for Lyme arthritis: results of a large pediatric cohort Discrepancies between patients and physicians in their perceptions of rheumatoid arthritis disease activity Brain TNF drives postinflammation depression-like behavior and persistent pain in experimental arthritis End of the debate? At the extreme end of the psychoneuroimmunological spectrum: delirium as a maladaptive sickness behaviour response Delirium and Lyme disease Delirium is common in patients hospitalized with COVID-19 Neurologic features in severe SARS-CoV-2 infection Human cerebral malaria: 2019 mini review Delirious behavior in children with influenza: its clinical features and EEG findings Elevated plasma levels of galectin-3 binding protein are associated with post-stroke delirium -A pilot study Delirium after a traumatic brain injury: predictors and symptom patterns Congestive heart failure as a determinant of postoperative delirium Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: a randomised, double-blind, placebo-controlled trial The inflammatory nature of post-surgical delirium predicts benefit of agents with anti-TNF effects, such as Dexmedetomidine Protective role of dexmedetomidine in unmethylated CpG-induced inflammation responses in BV2 microglia cells A clinical diagnostic system for late-stage neuropsychiatric lyme borreliosis based upon an analysis of 100 patients Severe neurological sequelae and behaviour problems after cerebral malaria in Ugandan children We can't handle things we don't know about": perceived neurorehabilitation challenges for Malawian paediatric cerebral malaria survivors Elevated cerebrospinal fluid tumour necrosis factor is associated with acute and long-term neurocognitive impairment in cerebral malaria Institutionalized stroke patients: status of functioning of an under researched population Suicide and Lyme and associated diseases Post-COVID syndrome and suicide risk Traumatic brain injury and suicidal behavior: a review Factors associated with multiple suicide attempts in a nationally representative study of U.S. military veterans Proinflammatory milieu in combat-related PTSD is independent of depression and early life stress Modulatory effects of antidepressant classes on the innate and adaptive immune system in depression Inflammatory pathways in psychiatric disorders: the case of schizophrenia and depression The many faces of the anti-COVID immune response Propensity to excessive proinflammatory response in chronic Lyme borreliosis Gabapentin for the symptomatic treatment of chronic neuropathic pain in patients with late-stage lyme borreliosis: a pilot study Gabapentin, a synthetic analogue of gamma aminobutyric acid, reverses systemic acute inflammation and oxidative stress in mice Gabapentin enhances the morphine anti-nociceptive effect in neuropathic pain via the interleukin-10-heme oxygenase-1 signalling pathway in rats IL-1beta and TNF-alpha induce neurotoxicity through glutamate production: a potential role for neuronal glutaminase Tumor necrosis factor-alpha induces neurotoxicity via glutamate release from hemichannels of activated microglia in an autocrine manner Tumor necrosis factor alpha inhibits glutamate uptake by primary human astrocytes -implications for pathogenesis of HIV-1 dementia Glutamate immunoreactivity in rat cerebral cortex is reversibly abolished by 6-diazo-5-oxo-L-norleucine (DON), an inhibitor of phosphate-activated glutaminase Neurological sequelae induced by alphavirus infection of the CNS are attenuated by treatment with the glutamine antagonist 6-diazo-5-oxo-l-norleucine Therapeutic evaluation of etanercept in a model of traumatic brain injury Editorial: an unsound AAN practice advisory on poststroke etanercept The function of the vertebral veins and their role in the spread of metastases TNF-alpha modulation for treatment of Alzheimer's disease: A 6-month pilot study Perispinal etanercept: a new therapeutic paradigm in neurology Visualization of the cerebrospinal fluid drainage into the Galen's vein Direct venous spinal reabsorption of cerebrospinal fluid: a new concept with serial magnetic resonance cisternography in rabbits Perispinal delivery of CNS drugs Revisiting the vertebral venous plexus-A comprehensive review of the literature The cerebrospinal venous system: anatomy, physiology, and clinical implications Protein accumulation in cerebrospinal fluid during -90 degrees head-down tilt in rabbit Rapid intracerebroventricular delivery of Cu-DOTA-etanercept after peripheral administration demonstrated by PET imaging Rapid improvement of chronic stroke deficits after perispinal etanercept: three consecutive cases How diseases caused by parasites allowed a wider understanding of disease in general: my encounters with parasitology in Australia and elsewhere over the last 50 years Tumor necrosis factor is a brain damaging cytokine in cerebral ischemia Infectious diseaseassociated encephalopathies Tumor necrosis factor (cachectin) is an endogenous pyrogen and induces production of interleukin 1 TNF-Α may mediate inflammasome activation in the absence of bacterial infection in more than one way Cytokines in autoimmunity Regulation of cytokines, cytokine inhibitors, and acute-phase proteins following anti-TNF-alpha therapy in rheumatoid arthritis Inhibitory effect of TNF alpha antibodies on synovial cell interleukin-1 production in rheumatoid arthritis Modulation of proinflammatory cytokine release in rheumatoid synovial membrane cell cultures. Comparison of monoclonal anti TNF-alpha antibody with the interleukin-1 receptor antagonist In vivo blockade of TNFalpha by intravenous infusion of a chimeric monoclonal TNF-alpha antibody in patients with rheumatoid arthritis. Short term cellular and molecular effects Secretory products of macrophages Immunemediated inflammation across disease boundaries: breaking down research silos How to cite this article: Clark IA. Chronic cerebral aspects of long COVID, post-stroke syndromes and similar states share their pathogenesis and perispinal etanercept treatment logic