Skip to main content

Sepsis-associated brain injury: underlying mechanisms and potential therapeutic strategies for acute and long-term cognitive impairments

Abstract

Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis causes cerebral dysfunction in the short and long term and induces disruption of the blood–brain barrier (BBB), neuroinflammation, hypoperfusion, and accumulation of amyloid β (Aβ) and tau protein in the brain. White matter changes and brain atrophy can be detected using brain imaging, but unfortunately, there is no specific treatment that directly addresses the underlying mechanisms of cognitive impairments in sepsis. Here, we review the underlying mechanisms of sepsis-associated brain injury, with a focus on BBB dysfunction and Aβ and tau protein accumulation in the brain. We also describe the neurological manifestations and imaging findings of sepsis-associated brain injury, and finally, we propose potential therapeutic strategies for acute and long-term cognitive impairments associated with sepsis. In the acute phase of sepsis, we suggest using antibiotics (such as rifampicin), targeting proinflammatory cytokines, and preventing ischemic injuries and hypoperfusion. In the late phase of sepsis, we suggest targeting neuroinflammation, BBB dysfunction, Aβ and tau protein phosphorylation, glycogen synthase kinase-3 beta (GSK3β), and the receptor for advanced glycation end products (RAGE). These proposed strategies are meant to bring new mechanism-based directions for future basic and clinical research aimed at preventing or ameliorating acute and long-term cognitive impairments in patients with sepsis.

Introduction

Sepsis is a systemic inflammatory disease defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection [1]. Sepsis is a global health problem affecting 49 million people and causing 11 million deaths each year worldwide [2]. The mortality rate of sepsis has been declining, but it is still remarkably high, ranging between 15 and 25%. In-hospital mortality of patients with septic shock could be as high as 30% to 50% [3]. Studies have shown that many patients with sepsis experience acute and long-term cognitive impairments [4,5,6]. However, no specific treatment that directly addresses the underlying mechanisms of cognitive impairments in sepsis currently exists, and many controversies surround the optimal treatment approach for these patients. Advances in therapy for sepsis are urgently needed to improve clinical outcomes [7]. Here, we review the underlying mechanisms of sepsis-associated brain injury, with a focus on blood–brain barrier (BBB) dysfunction, neuroinflammation, and amyloid β (Aβ) and tau protein accumulation in the brain. We also describe the neurological manifestations and imaging findings of sepsis-associated brain injury, and finally, we propose potential therapeutic strategies for acute and long-term cognitive impairments associated with sepsis.

Underlying mechanisms of sepsis-associated brain injury

Multiple mechanisms contribute to brain damage in sepsis, including systemic inflammation, neuroinflammation, BBB dysfunction, ischemia/hypoxia due to hypoperfusion, Aβ and tau protein accumulation, and direct damage to the brain (Fig. 1 and Table 1). Notably, systemic inflammation in sepsis can exacerbate BBB dysfunction, hypoperfusion, as well as, accumulation of both Aβ and tau protein. On the other hand, BBB disruption induces brain inflammation and may ultimately cause white matter changes and cerebral dysfunction.

Fig. 1
figure 1

Schematic representation of underlying mechanisms of sepsis-related brain injuries. Inflammation caused by sepsis affects a wide range of processes. In the short term, it induces dysfunction of endothelial cells (EC) leading to blood–brain barrier (BBB) leakage. Proinflammatory cytokines can lead to BBB leakage and contribute to hypoperfusion. Hypoperfusion and dysfunction of coagulation induce ischemia/hypoxia and microhemorrhage. These factors ultimately lead to changes in the white matter in the brain. Sepsis-related inflammation may have a long-term effect. Sepsis may lead to amyloid β (Aβ) and tau protein in the brain. Ultimately, these processes lead to cognitive impairment

Table 1 Summary of the main topics discussed in this review

Sepsis modeling in animals

Although no one model reflects all the clinical complexities of sepsis, animal modeling is a valuable way to study its underlying mechanisms and to develop new therapeutic strategies [8, 9]. The main two animal models used in sepsis research are the cecal ligation and puncture (CLP) and lipopolysaccharide (LPS) models. The CLP model mimics the nature and evolution of severe sepsis in humans [10]. Following a simple procedure to ligate and puncture the cecum, the CLP model induces sepsis as a result of stercoral peritonitis, followed by polymicrobial translocation in the blood circulation [8]. During gram-negative bacterial infection and other acute illnesses, LPS is a major mediator in sepsis [11]. In the LPS model, endotoxins are infused intravascularly, which results in an early and transient peak of cytokines, whereas in the CLP model, the pro-inflammatory response is delayed and persists over a longer period [9]. Due to the effects of the intense inflammatory response on the cardiovascular system, mortality in the LPS model occurs early, whereas in the CLP model, the mortality is delayed and occurs due to multiple organ failure as a complication of the induced peritonitis [8, 12].

From systemic inflammation to neuroinflammation and BBB dysfunction

Systemic inflammation

Sepsis is fundamentally a systemic inflammatory disease mediated by the activation of the innate immune system [3]. The activation of the innate immune response is mediated by pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), nucleotide-binding oligomerization domain (NOD)-like receptors, retinoic acid-inducible gene (RIG)-like receptors, mannose-binding lectin, and scavenger receptors [3, 13]. PRRs detect both pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), which initiate the upregulation of inflammatory-related genes [14] and induce a complex intracellular signaling system [15]. The transcription complex nuclear factor kappa B (NF-κB) is triggered in response to numerous extracellular inflammatory stimuli [16]. The activation of NF-κB induces the expression of early activation genes, including tumor necrosis factor alpha (TNF-α), interleukin (IL)‑1, IL‑12, IL‑18, and type I interferons (IFNs), among others. These cytokines initiate a cascade of other inflammatory cytokines and chemokines, including IL‑6, IL‑8, IFNγ, CC–chemokine ligand 2 (CCL2), CCL3, and CXC–chemokine ligand 10 (CXCL10), exacerbating the inflammatory response [3, 17]. The activation of these inflammatory networks begins within minutes of PAMP or DAMP recognition [3]. Maciel et al. reported that plasma levels of IL-6 and IL-10 in patients at hospital discharge from the intensive care unit (ICU) correlated with long-term functional and cognitive performance (mean follow-up, 4 years) [18], suggesting that the effects of proinflammatory cytokines are not limited to the acute phase of sepsis.

Neuroinflammation

Proinflammatory cytokines enter the brain during sepsis by several mechanisms. LPS can activate TLRs through areas that lack BBB, such as the choroid plexus, circumventricular organ, and leptomeninges [19]. Many cytokines enter the brain through receptor-mediated endocytosis on brain endothelial cells. Receptors for IL-1β, IL-6, and TNF-α are expressed on cerebral endothelium [20,21,22] and systemic IL-1β and TNF-α cause cerebral endothelial activation [23]. Activation of cytokine receptors such as IL-1 receptor (IL-1R1) and TNF receptor (TNFR) elevates cytokine levels in the brain [24, 25]. Once these inflammatory mediators enter the brain tissue, microglial cells get activated [26]. Animal [27] and human [28] studies during sepsis consistently show microglial activation without any known cerebral infection [27, 29]. Immunostaining of a marker for microglia (CD68) was higher in brain tissues of patients who died of sepsis, supporting that neuroinflammation occurs after the onset of sepsis [28, 30]. Once microglia are activated, they most likely induce brain damage during sepsis [29, 31]. Sustained microglia activation enhances the production of inflammatory cytokines and reactive oxygen species (ROS), which creates a vicious cycle of increased BBB permeability and increased neuronal apoptosis [32, 33].

BBB dysfunction

The BBB is a specialized multicellular structure that selectively separates the brain interstitium from the contents of the blood vessels [34, 35]. The BBB is a dynamic barrier, with active communication between cells of the BBB and the brain parenchymal cells [36]. The innermost layer of the BBB is composed of endothelial cells, and the BBB regulates the paracellular permeability of endothelial cells through junctional proteins, including tight junction (TJ) proteins [37].

Increasing evidence suggests that BBB permeability is increased during sepsis. Using contrast-enhanced MRI of the brain, Towner et al. showed increased BBB leakage at 24 h in LPS-exposed rat brains in the cerebral cortex, hippocampus, thalamus, and perirhinal cortex regions [38]. Patients with sepsis-associated encephalopathy (SAE) frequently have vasogenic edema and white matter hyperintensities on MRI, indicating BBB disruption [39, 40]. How sepsis disrupts BBB is not entirely understood, but several mechanisms have been postulated.

An important pathway that could mediate BBB dysfunction is the disruption of TJ proteins induced by sepsis. TJ-associated proteins, mainly occludin, contain a putative matrix metalloproteinase (MMP) cleavage site [41]. After CLP in rats, MMP-2 and MMP-9 were detected in cerebral microvessels, and the increase in detected BBB permeability was time-related to the increase in MMP-2 and MMP-9 activation [41]. In humans, a reduction in TJ proteins expression was detected in the brain tissue of patients who died due to sepsis, indicating BBB damage [42].

Sepsis may also disrupt the BBB through the endothelial sphingosine 1-phosphate (S1P) receptor pathway, which maintains the BBB integrity by regulating the proper localization of TJ proteins [43]. It has been shown that in patients during sepsis, serum S1P levels were dramatically decreased and inversely associated with disease severity [44]. Additionally, the upregulation of endothelial caveolin-1 causes S1P to interact with intercellular adhesion molecule 1 (ICAM-1) and increases its binding affinity for peripheral immune cells [45]. Wu et al. found that caveolin-1 facilitates T-cell trafficking into the CNS via ICAM-1-mediated signaling [46].

Research suggests that TNF-α is another important mediator of BBB dysfunction in sepsis. TNF-α induces depolymerization of actin and the generation of intercellular gaps in the endothelial cytoskeleton [47]. In cultured brain endothelial cells, both TNF-α and IL-1β increased the expression of leukocyte adhesion molecules, such as ICAM-1, resulting in increased barrier permeability as measured by transendothelial electrical resistance [48]. TNF-α also increases barrier permeability through activating protein-kinase-6 and the internalization of VE-cadherin [49]. TNF-α has been reported to play a crucial role in the development of brain edema in acute liver failure, and increased BBB permeability was associated with loss of the TJ-associated protein occludin [50].

Ischemia/hypoperfusion

Hypoxic or ischemic changes are the most common abnormalities found on brain autopsies of patients with sepsis [51]. Decreased cerebral flow and hypercoagulability are the main factors behind the development of cerebral ischemia in patients with sepsis [52]. Not surprisingly, impairment of cerebral blood flow autoregulation is also common in patients with septic shock, especially in the presence of hypercapnia [53].

The acute dysregulated inflammatory response that is meant to eradicate the infectious agent in patients with sepsis may cause hypercoagulability and vasculopathy of cerebral blood vessels leading to cerebral ischemia. Acute vascular endothelial dysfunction is a central event in the pathogenesis of sepsis, contributing to vascular permeability and impaired autoregulation of cerebral blood flow [52, 54]. Infiltration of immune cells into the brain tissue and increased proinflammatory cytokines release activate endothelial cells, resulting in the activation of the coagulation cascade. Microthrombi may then form in blood vessels and can cause cerebral ischemia [5].

Available studies suggest that hypoperfusion and ischemia can induce white matter (WM) changes and may contribute to long-term cognitive impairments in patients with ischemic stroke. For example, microstructural changes to the WM have been reported in patients with hyperacute ischemic stroke (less than 6 h) [55]. It has also been proposed that hypoperfusion may also cause long-term cognitive impairments in stroke through the accumulation of tau pathology [56, 57]. Studies investigating the role of ischemia/hypoperfusion in sepsis-associated cognitive impairments are warranted.

Amyloid β and tau protein accumulation

Available preclinical and clinical data indicate that sepsis-associated brain injury is associated with molecular alterations that are shared with those seen in patients with Alzheimer’s disease (AD). It is well established that neuroinflammation in age-related brain diseases, such as AD, induces accumulation of Aβ and phosphorylated tau (p-tau) [58,59,60,61,62]. Data from animal models show that Aβ plaques and p-tau also accumulate acutely after experimental sepsis [59, 61]. It has also been shown that 30 days after the induction of sepsis, the brains of survivor animals have increased accumulation of Aβ and p-tau and this was associated with cognitive impairment [63]. In an animal sepsis model, LPS exposure in the rat resulted in hippocampal accumulation of Aβ plaques and intracellular p-tau, accompanied by behavioral deficits attributable to the dorsal dentate gyrus [61]. In another study, it was shown that both sepsis and aging induce brain inflammation, oxidative damage, and accumulation of Aβ, as well as impaired behavioral learning and memory function [62].

Although most of the data that show that sepsis triggers the accumulation of Aβ and p-tau in the brain come from animal research, there are several pieces of evidence that support the hypothesis that sepsis also triggers Aβ and p-tau accumulation in the brain of patients. In patients with sepsis, serum levels of tau protein are higher in those who had SAE than those who did not, and tau levels are independently associated with SAE [64]. In a prospective observational study, Orhun et al. investigated the association between neuroimaging features of sepsis-induced brain dysfunction and neuroinflammation and neurodegeneration factors [65]. The authors found that brain atrophy on MRI brain was associated with an increased serum level of p-tau. Regression analysis also identified an association between C5a levels and the presence of lesions on MRI and p-tau levels and the presence of atrophy on MRI [65].

Ehler et al. showed that in postmortem brain tissue from 5 patients who died as a result of sepsis there was diffuse staining of axons for Aβ precursor protein (β-APP). The authors also found amyloid plaque in a 79-year-old patient whose medical history did not suggest cognitive impairment [40]. Rogne et al. used 18F-flutemetamol positron emission tomography to visualize Aβ accumulation in patients with brain abscess and found that in 3 patients out of 17 there was an accumulation of Aβ [66]. Kramer et al. also found that periodontal infection was associated with higher Aβ load in the brain of normal elderly as measured by PIB–PET scan [67]. Kantonen et al. detected strong APP immunoreactivity in autopsies of patients with COVID-19 [68]. In a more recent study, published as a preprint, Rhodes et al. found numerous Aβ deposits in the neocortex of patients under the age of 60 who died with COVID-19 [69].

Excessive phosphorylation of tau protein is the main cause of tauopathy in AD, and glycogen synthase kinase-3β (GSK3β) is one of the most important kinases of the tau protein [70]. In addition, GSK3β is a powerful regulator of inflammation. The NF-κB pathway is a key player for the proinflammatory actions of GSK3β [71,72,73,74,75]. GSK3β is necessary for full stimulation of the production of several pro-inflammatory cytokines, including IL-6, IL-1β, and TNFα, and inhibition of GSK3β greatly reduces the production of pro-inflammatory cytokines [73, 75]. GSK3β regulates the inflammatory response by differentially affecting the nuclear amounts of transcription factors NF-κB subunit p65 and cAMP response element-binding protein (CREB) interacting with the coactivator CREB-binding protein (CBP) [73]. Signaling of rapamycin complex 1 (mTORC1) and GSK3β converge, and the capacity of mTORC1 to regulate the inflammatory response is related to the inactivation of GSK3β [76]. In addition, GSK3 participates in modulating the downstream signaling induced by IFNγ and the JAK/STAT pathway [77].

The receptor for advanced glycation end products (RAGE) has been identified as an important link between inflammation, amyloidogenesis, and apoptosis during AD progression [78, 79]. RAGE binds to several DAMPs, including advanced glycation end products (AGEs) [80], S100 [81, 82], and DNA [83]. In patients with AD, the plasma levels of soluble RAGE (sRAGE) are lower than in normal elderly and associated with cognitive deterioration [84]. Analysis of brain tissue from patients with AD compared to individuals without dementia showed upregulation of RAGE expression in microglia and neurons in the hippocampus, entorhinal cortex, and superior frontal gyrus [85]. By employing transgenics with targeted neuronal overexpression of RAGE and mutant APP, Arancio et al. showed that RAGE is a cofactor for Aβ-induced neuronal perturbation in a model of AD-type pathology [86]. Fang et al. showed that transgenic mice expressing human mutant APP in neurons and RAGE in microglia displayed enhanced IL-1β and TNF-α production, increased infiltration of microglia and astrocytes, accumulation of Aβ, reduced acetylcholine esterase activity, and accelerated deterioration of spatial learning/memory [87].

Available data indicate that RAGE is also involved in the pathogenesis of sepsis-associated brain injury. In the CLP animal model of sepsis, the levels of RAGE and p-tau are increased in the brain [63]. In addition, injection of RAGE antibodies into the hippocampus reduces Aβ and p-tau accumulation [60]. It has also been shown that RAGE-dependent signaling pathway regulates β- and ɤ-secretase cleavage of APP to generate Aβ, at least in part through activation of GSK3β and p38 MAP kinase [88].

Direct brain damage

Patients with sepsis may have focal lesions that cause direct damage to the brain. Sharshar et al. reported various focal lesions in 23 patients who died in the ICU from septic shock: ischemia (100%), hemorrhages (26%), hypercoagulability syndrome (9%), micro-abscesses (9%), and multifocal necrotizing leukoencephalopathy (9%) [89]. Severe sepsis can induce life-threatening coagulopathy, including disseminated intravascular coagulation (DIC) and intracerebral hemorrhage [90]. Focal brain lesions should be considered in patients with sepsis who show signs of cognitive impairment.

Other mechanisms, such as mitochondrial and neurotransmitter dysfunction, age-related changes, and iatrogenic factors may also be involved in the pathogenesis of brain damage in sepsis. For a thorough discussion of these mechanisms, please refer to previous publications by Mazeraud et al. [29], Annane et al. [91], and Manabe et al. [92].

Neurological manifestations of sepsis-associated brain injury

The spectrum of acute cognitive impairments in sepsis

In the acute phase, sepsis causes several clinical syndromes that are associated with cognitive impairments, including SAE, delirium, sickness behavior, and cerebral ischemia and hemorrhage [93]. For unclear reasons, acute cognitive impairments may sometimes persist for a long time even after sepsis has improved. Currently, the management of acute sepsis-associated brain injury is still focused on treating sepsis as the underlying disease, with no specific treatments to address brain dysfunction. While this is the case, it is encouraging that appropriate management of sepsis can help prevent cognitive impairments [91, 94].

Sepsis-associated encephalopathy

SAE is a key manifestation of sepsis, ranging from delirium to coma [29]. SAE is defined as cognitive dysfunction associated with sepsis, in the absence of infection in the central nervous system, structural brain injury, or other metabolic abnormalities [94]. Clinically, SAE is manifested by confusion, anxiety, irritability, depression, anhedonia, decreased social communication and environmental interest, and other cognitive changes, including decreased concentration and learning capacity and impaired memory [94]. SAE is a relatively common cause of altered mental status in critically ill patients admitted to the ICU, and its prevalence varies from 8 to 70% depending on the inclusion and exclusion criteria [95]. Sonneville et al. reported that 53% of 2513 patients with sepsis admitted to the ICU had SAE, which was associated with a higher mortality rate, higher use of ICU resources, and a longer hospital stay [96]. The pathophysiology of SAE is diverse, and the mechanisms by which sepsis induces cognitive dysfunction in patients with SAE are complex and may include cerebrovascular damage, BBB disruption, neuroinflammation, neurotransmitter, and mitochondrial dysfunction, oxidative stress, and severe glial (microglia and astrocytes) activation [29, 91]. For a thorough discussion of the roles of systemic inflammation, neuroinflammation, BBB dysfunction, ischemia/hypoperfusion, and Aβ and tau accumulation in sepsis-associated cognitive dysfunction, please refer to “Underlying mechanisms of sepsis-associated brain injury” section of this manuscript. For additional information on other mechanisms, such as mitochondrial and neurotransmitter dysfunction and iatrogenic factors, please refer to published reviews by Mazeraud et al. [29] and Annane et al. [91].

Delirium

Delirium has many shared characteristics and symptoms with the acute phase of SAE: agitation, hallucinations, reduced concentration, and alteration of the sleep–wake cycle [97]. Delirium is commonly seen in critically ill patients and is clinically diagnosed using screening tools, such as the Confusion Assessment Method for the ICU (CAM–ICU) or the Intensive Care Delirium Screening Checklist (ISDSC). Zhang et al. reported sepsis as a risk factor for delirium in ICU patients; others reported risk factors include a history of hypertension, hypoxemia, use of benzodiazepines, deep sedation, and mechanical ventilation [98].

There is still much to learn about the mechanisms underlying sepsis-associated delirium; however, the current understanding is that its pathophysiology involves a combination of neuroinflammation and disturbances in cerebral perfusion, BBB, and neurotransmission [99]. Girard et al. identified four clinical delirium phenotypes: hypoxic, septic, sedative-associated, and metabolic delirium, which were defined a priori based on clinical judgment as well as common risk factors and hypothesized mechanisms for delirium during critical illness [100]. The authors found that longer durations of each one of these phenotypes predicted worse long-term cognitive impairment. Sedative-associated delirium was the most common phenotype and predicted more profound long-term cognitive impairment [100].

Sickness behavior

Patients who experience infection show common symptoms of sickness, such as loss of appetite, sleepiness, withdrawal from normal social activities, fever, aching joints, and fatigue. Collectively, this clinical syndrome is described as sickness behavior, and it has been shown that physiological concentrations of proinflammatory cytokines that occur after infection affect the brain and induce these symptoms. Sickness behavior is now recognized as part of a motivational system that reorganizes the organism’s priorities to promote recovery from infection [101, 102]. Among proinflammatory cytokines, IL-6 and IL-1β have significant relationships with sickness behavior [103]. In the CLP animal model, the severity of sickness behavior correlated with sepsis severity, mortality, increased permeability of the BBB, high level of IL-6 and oxidative stress, as well as cognitive and memory impairments [104].

Long-term cognitive impairments in sepsis

Sepsis has various long-term effects on brain function and neurological outcomes, including alteration in mood, motor function, and cognitive impairments [4,5,6]. More than half of sepsis survivors have long-term cognitive impairments [91], which are still inadequately characterized, but may include loss of verbal fluency, memory, and attention deficits [4,5,6, 91]. Although their prevalence and temporal trajectory differ depending on the cognitive instruments used [105], cognitive impairments among ICU survivors appear to be common, severe, and persistent [105]. Furthermore, sepsis is associated with an accelerated long-term decline in global cognitive function. Compared with pre-sepsis slopes, the long-term rates of cognitive decline are faster after sepsis [106]. SAE has long been considered a reversible syndrome; however, many patients with SAE experience long-term cognitive impairments [5]. Mild to moderate neurological symptoms, including memory changes, depression, anxiety, or cognitive disorders persist in 20% to 40% of these patients 1 year after hospital discharge [94]. Acute delirium in patients with critical illness is also associated with cognitive impairments that persist long after hospital discharge [6, 107].

Imaging findings in sepsis-associated brain injury

WM changes have a direct effect on brain dysfunction and cognitive impairments [108, 109]. Patients with sepsis-associated brain injury tend to have decreased volumes of cerebral and cerebellar WM [110]. Global WM changes on CT head imaging have been reported in adult cases of SAE [111], with no specific locations or patterns for these changes [39]. WM changes tend to worsen with a longer duration of septic shock and worse Glasgow Coma Scale scores [112].

Atrophy has also often been described in long-term observational studies of brain function after recovery from sepsis [113], and early brain atrophy may have long-term effects on the cognitive function of sepsis survivors [113]. Gunther et al. reported that a longer duration of sepsis-related delirium in ICU patients was independently associated with increased global brain atrophy and smaller hippocampal volume [114]. Furthermore, greater brain atrophy at 3 months was associated with worse cognitive performance at 12 months [114].

Apart from WM changes and atrophy, other types of pathological intracranial findings may also be present in patients with sepsis, including hemorrhages, micro abscess formation, central pontine myelinolysis, multifocal necrotizing leukoencephalopathy, and ischemic changes [115].

Although it appears that sepsis can cause widespread damage to the brain, specific anatomical locations such as the medial temporal and frontal cortex may correlate with a specific cognitive domain that has been affected in patients with sepsis [92]. Neuroimaging studies show reduced volumes of the hippocampus, amygdala, and cortex during hospitalization due to sepsis [110]. In post-mortem brain tissues from patients who died from sepsis, Sharshar et al. found that apoptotic neurons were found in the amygdala, as well as in the hypothalamus and medulla [89, 116].

New imaging modalities, such as magnetic resonance spectroscopy, diffusion tensor imaging, and positron emission tomography are promising options for early detection of sepsis-associated brain changes [39, 112, 117].

Potential therapeutic strategies for acute and long-term cognitive impairments associated with sepsis

Based on the underlying mechanisms of sepsis-associated brain injury, we propose the following brain-directed therapeutic strategies in the acute and late phases of sepsis to help prevent or ameliorate cognitive impairments (Table 1).

Acute phase therapeutic strategies

Antibiotics

Appropriate use of antimicrobial drugs is critical for the treatment of bacterial sepsis. Fluoroquinolones (e.g., ciprofloxacin, moxifloxacin), sulfonamide, rifampicin (i.e., rifampin), metronidazole, and chloramphenicol readily enter the brain, regardless of the BBB state. In contrast, more hydrophilic and larger drugs, such as vancomycin and members of the β-lactam class of antibiotics, cannot enter the brain, unless the meninges are inflamed [5]. Rifampicin should be noted in particular, because it inhibits Aβ1–40 protein aggregation and neurotoxicity in vitro [118], attenuates the inhibition of autophagosome formation and suppresses the accumulation of Aβ1–42 in vivo [119]. Furthermore, in a mouse model of LPS-induced sepsis, rifampicin protected hippocampal neurons and ameliorated cognitive impairments [119]. Iizuka et al. retrospectively reviewed FDG–PET findings of elderly patients with mycobacterium infection who were treated with rifampicin and did not have dementia at the start of treatment to examine the preventive effects of rifampicin on the progression to AD [120]. The authors found that rifampicin therapy (450 mg/day for at least 12 months) before the onset of dementia improved or stabilized AD-type hypometabolism and made metabolic decline milder in long-term follow-up after completion of therapy. Multiple regression analysis revealed that rifampicin dose and treatment duration significantly influenced FDG uptake in the posterior cingulate cortex [120].

Thus, we propose that the use of rifampicin for the acute treatment of sepsis may have the potential to prevent long-term cognitive dysfunction. However, further research is needed to test this hypothesis. Rifampicin was initially approved for the treatment of tuberculosis. Because of its low toxicity, broad-spectrum activity, and good bioavailability, rifampicin is now commonly used as part of a combination antimicrobial therapy for various infections caused by bacterial infections other than mycobacteriosis (such as acute bacterial meningitis, infective endocarditis and bacteremia, pneumonia, and biofilm-related infections) [121]. Further studies to explore the effects of rifampicin on cognitive function in sepsis should be considered. Any benefits will need to be balanced against the known complications of rifampicin use and its toxicity, including the risk for adverse drug–drug interactions and the emergence of rifampicin resistance during treatment [121].

Targeting proinflammatory cytokines

Proinflammatory cytokines, such as IL-6 and IL-1, are important in the early phase of systemic inflammation [4] and have been associated with cognitive impairments in sepsis [103]. Another type of brain injury caused by severe inflammation is chimeric antigen receptor (CAR)-T cell-related encephalopathy syndrome (CRES), a treatment-related complication that may occur in patients with cancer after receiving genetically modified T cells. Cytokine release syndrome (CRS) and its associated CRES neurotoxicity are caused by the release of IL-1 and IL-6, among other cytokines [122]. To suppress this inflammation, both IL-6 and IL-1 are considered viable therapeutic targets, and monoclonal antibodies (mAbs) are already established for this purpose (anakinra against IL-1R, siltuximab against IL-6, and tocilizumab and sarilumab against IL-6R). Targeting IL-1 abolished both CRS and neurotoxicity, whereas targeting IL-6 has failed to suppress delayed lethal neurotoxicity in humanized mice models [123]. In a clinical trial, anakinra was associated with significant improvement in the survival of patients with sepsis [124]. However, cognitive impairments were not assessed in this trial. Given the known role of IL-1 in sepsis, the use of mAbs against IL-1 may represent a new promising approach to suppress neuroinflammation in patients with sepsis and prevent long-term cognitive impairments, but further studies are required. TNF-α is another promising target in sepsis. In a clinical trial, TNF-α blockade therapy in patients with sepsis significantly reduced mortality 3 days after infusion, and at 28 days following treatment, a trend toward reduced mortality continued, but the difference in mortality was not significant. Neurologic outcomes were not included as part of this trial [125].

Prevention of hypovolemia and cerebral ischemia

Fluid replacement in sepsis is necessary to treat hypotension and hypovolemia and to prevent ischemic changes that may occur. Several studies have been conducted to determine what is the best fluid replacement strategy for sepsis. As of today, fluid replacement with crystalloid solutions is recommended [126, 127], whereas the use of colloid solutions is discouraged [128,129,130]. Further studies are needed to determine the efficacy of albumin-containing fluid replacement therapy [127]. Adequate fluid replacement could help prevent ischemia/hypoxia, subsequently preventing future cognitive impairments in patients with sepsis.

As noted earlier, sepsis induces hypoperfusion in the brain in a similar fashion to ischemic stroke. MRI and CT imaging studies showed that patients with SAE have similar imaging characteristics to those found in patients with stroke [39]. If neuroprotective agents (e.g., PSD-95 inhibitors [131, 132]) currently being investigated in acute ischemic stroke are proven to be beneficial, their use for early ischemic changes in patients with sepsis may help prevent long-term cognitive sequelae.

Long-term therapeutic strategies

Targeting neuroinflammation

Neuroinflammation starts acutely after sepsis and persists for months [133]. Olivieri et al. showed that brain inflammation persists up to 3 months after sepsis in rats and is associated with increased Aβ level, activated microglia, and long-term cognitive impairment [133]. Microglia is the main inflammatory cell that gets activated in sepsis [26] in animals [27] and humans [28]. Thus, modulating activated microglia may be a promising approach to prevent chronic neuroinflammation and long-term cognitive impairment associated with sepsis. Recent research revealed that IL-17A/IL-17R might serve as a checkpoint in microglia-mediated neuroinflammation [134, 135]. In the CLP animal model, neutralizing anti-IL-17A or anti-IL-17R antibodies mitigated neuroinflammation and microglia activation, and alleviated cognitive dysfunction [136]. In another research using a traditional medicinal herb that is widely used for treating inflammatory conditions in South‑East Asia [137], it was shown that attractylone treatment attenuates sepsis-associated encephalopathy and cognitive dysfunction by inhibiting microglial activation and neuroinflammation [138]. Despite these encouraging data, targeting microglial activation might have drawbacks, as microglia also have neuroprotective effects [139], and inhibiting their function might have deleterious effects. More research is needed to examine targeting microglia as a therapeutic approach to help alleviate cognitive impairments associated with sepsis.

Targeting BBB dysfunction

BBB breakdown is an early biomarker of cognitive dysfunction in humans [140]. In patients with sepsis and SAE, vasogenic edema and WM hyperintensities on MRI are found frequently, indicating BBB disruption [39, 40]. Brain tissues of patients who died due to sepsis show a reduction of TJ proteins, indicating BBB damage [42]. Sepsis may disrupt the BBB through the S1P pathway, which maintains BBB integrity [43, 141]. Serum-S1P levels are dramatically decreased and inversely associated with disease severity in patients with sepsis [44] and in LPS-induced SAE in mice [142]. Fingolimod (FTY720) is an S1P modulator that is approved for the treatment of relapsing–remitting multiple sclerosis [143]. Fingolimod was also shown to modulate multiple neuroinflammatory markers and to decrease Aβ plaque density in a mouse model of AD [144]. Fingolimod and other S1P modulators have also shown promising results in modulating neuroinflammation [145] and BBB integrity [146] after ischemic stroke. Thus, targeting BBB disruption through modulation of the S1P pathway may be a promising direction to alleviate sepsis-induced BBB disruption and cognitive impairment, but more research is needed to test this approach.

Targeting amyloid β and tau phosphorylation

Growing evidence suggests that targeting Aβ oligomers (soluble aggregates of Aβ) and plaques (insoluble extracellular aggregates of fibrillar Aβ) with selective mAbs (e.g., aducanumab) can slow disease progression in patients with AD [58, 147]. Available data from animal [59, 61,62,63] and human research [64,65,66,67,68,69] suggest that similar to AD, Aβ and p-tau accumulation may be involved in long-term cognitive dysfunction associated with sepsis. Thus, targeting Aβ and p-tau with mAbs may also be a promising new approach to prevent or treat long-term cognitive impairments in patients with sepsis, but more research to test this hypothesis is warranted.

Targeting GSK3β

Excessive phosphorylation of tau is the main cause of tauopathy. GSK3β is one of the most important kinases of the tau protein [70] and a powerful regulator of inflammation [71]. This makes GSK3β another potential target for sepsis-associated brain injury. Lithium is a known inhibitor of GSK3β that has been mainly used to treat bipolar disorders and major depression. In a meta-analysis of randomized placebo-controlled trials investigating lithium as a treatment for mild cognitive impairment and AD dementia, it was suggested that lithium might have beneficial effects on cognitive performance [148, 149]. Taken together, these data and the known underlying mechanisms of sepsis-related cognitive impairments suggest that lithium might have the potential to ameliorate long-term cognitive impairments in sepsis survivors.

Targeting RAGE

RAGE is upregulated in microglia and neurons in the brains of patients with AD [85], and it has been identified as an important link between inflammation, amyloidogenesis, and apoptosis during AD progression [78, 79]. In animal models of sepsis, the levels of RAGE and p-tau are increased in the rat brain [63], and the injection of RAGE antibodies into the hippocampus reduces Aβ and p-tau accumulation [60]. High mobility group box 1 (HMGB1) plays an important role in inflammation and operates mainly through RAGE and TLR4 receptors [150]. In sepsis survivors, serum HMGB1 levels remain elevated at the time of hospital discharge [151], and administration of neutralizing anti-HMGB1 antibody beginning at 1 week [152] after the onset of sepsis in animals provided significant protection against cognitive impairment [152]. These data suggest that targeting RAGE directly or through HMGB1 might be a promising approach to regulate inflammation and decrease cognitive impairment in sepsis; however, more research is needed to further confirm the findings of these preliminary studies.

For discussion of other therapeutic strategies that were not mentioned in this article, please refer to Barichello et al. [4], Jarczak et al.[153], and Slikke et al. [154].

Conclusions

In this review, we have described the spectrum of cognitive brain injuries associated with sepsis. Although the diverse molecular mechanisms underlying brain damage in sepsis are intertwined and have not been fully characterized, it is evident that local and systemic inflammation plays an important role in both the short and long term. Our proposed strategies here are meant to bring new mechanism-based directions for future basic and clinical research aimed at preventing or ameliorating acute and long-term cognitive impairments in patients with sepsis. New imaging modalities are also needed for clinical detection and characterization of sepsis-associated brain injuries.

Availability of data and materials

Not applicable.

Abbreviations

Aβ:

Amyloid-beta

AD:

Alzheimer’s disease

BBB:

Blood–brain barrier

CRES:

CAR-T-related encephalopathy syndrome

CRS:

Cytokine release syndrome

GSK3β:

Glycogen synthase kinase-3 beta

ICU:

Intensive care unit

IL:

Interleukin

LPS:

Lipopolysaccharide

MRI:

Magnetic resonance imaging

SAE:

Sepsis-associated encephalopathy

TJ:

Tight junction

WM:

White matter

References

  1. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: analysis for the Global Burden of Disease Study. Lancet. 2020;395(10219):200–11.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Hotchkiss RS, Moldawer LL, Opal SM, Reinhart K, Turnbull IR, Vincent JL. Sepsis and septic shock. Nat Rev Dis Primers. 2016;2:16045.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Barichello T, Sayana P, Giridharan VV, Arumanayagam AS, Narendran B, Della Giustina A, Petronilho F, Quevedo J, Dal-Pizzol F. Long-term cognitive outcomes after sepsis: a translational systematic review. Mol Neurobiol. 2019;56(1):186–251.

    Article  CAS  PubMed  Google Scholar 

  5. Nwafor DC, Brichacek AL, Mohammad AS, Griffith J, Lucke-Wold BP, Benkovic SA, Geldenhuys WJ, Lockman PR, Brown CM. Targeting the blood-brain barrier to prevent sepsis-associated cognitive impairment. J Cent Nerv Syst Dis. 2019;11:1179573519840652.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Pandharipande PP, Girard TD, Jackson JC, Morandi A, Thompson JL, Pun BT, Brummel NE, Hughes CG, Vasilevskis EE, Shintani AK, et al. Long-term cognitive impairment after critical illness. N Engl J Med. 2013;369(14):1306–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rhodes A, Evans LE, Alhazzani W, Levy MM, Antonelli M, Ferrer R, Kumar A, Sevransky JE, Sprung CL, Nunnally ME, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock: 2016. Crit Care Med. 2017;45(3):486–552.

    Article  PubMed  Google Scholar 

  8. Ruiz S, Vardon-Bounes F, Merlet-Dupuy V, Conil JM, Buleon M, Fourcade O, Tack I, Minville V. Sepsis modeling in mice: ligation length is a major severity factor in cecal ligation and puncture. Intensive Care Med Exp. 2016;4(1):22.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Buras JA, Holzmann B, Sitkovsky M. Animal models of sepsis: setting the stage. Nat Rev Drug Discov. 2005;4(10):854–65.

    Article  CAS  PubMed  Google Scholar 

  10. Raven K. Rodent models of sepsis found shockingly lacking. Nat Med. 2012;18(7):998.

    Article  CAS  PubMed  Google Scholar 

  11. Gabarin RS, Li M, Zimmel PA, Marshall JC, Li Y, Zhang H. Intracellular and extracellular lipopolysaccharide signaling in sepsis: avenues for novel therapeutic strategies. J Innate Immun. 2021;13(6):323–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Remick DG, Newcomb DE, Bolgos GL, Call DR. Comparison of the mortality and inflammatory response of two models of sepsis: lipopolysaccharide vs. cecal ligation and puncture. Shock. 2000;13(2):110–6.

    Article  CAS  PubMed  Google Scholar 

  13. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11(5):373–84.

    Article  CAS  PubMed  Google Scholar 

  14. Raymond SL, Holden DC, Mira JC, Stortz JA, Loftus TJ, Mohr AM, Moldawer LL, Moore FA, Larson SD, Efron PA. Microbial recognition and danger signals in sepsis and trauma. Biochim Biophys Acta Mol Basis Dis. 2017;1863(10 Pt B):2564–73.

    Article  CAS  PubMed  Google Scholar 

  15. Tang D, Kang R, Coyne CB, Zeh HJ, Lotze MT. PAMPs and DAMPs: signal 0s that spur autophagy and immunity. Immunol Rev. 2012;249(1):158–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 1999;18(49):6853–66.

    Article  CAS  PubMed  Google Scholar 

  17. Nedeva C, Menassa J, Puthalakath H. Sepsis: inflammation is a necessary evil. Front Cell Dev Biol. 2019;7:108.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Maciel M, Benedet SR, Lunardelli EB, Delziovo H, Domingues RL, Vuolo F, Tomasi CD, Walz R, Ritter C, Dal-Pizzol F. Predicting long-term cognitive dysfunction in survivors of critical illness with plasma inflammatory markers: a retrospective cohort study. Mol Neurobiol. 2019;56(1):763–7.

    Article  CAS  PubMed  Google Scholar 

  19. Laflamme N, Echchannaoui H, Landmann R, Rivest S. Cooperation between toll-like receptor 2 and 4 in the brain of mice challenged with cell wall components derived from gram-negative and gram-positive bacteria. Eur J Immunol. 2003;33(4):1127–38.

    Article  CAS  PubMed  Google Scholar 

  20. Ericsson A, Liu C, Hart RP, Sawchenko PE. Type 1 interleukin-1 receptor in the rat brain: distribution, regulation, and relationship to sites of IL-1-induced cellular activation. J Comp Neurol. 1995;361(4):681–98.

    Article  CAS  PubMed  Google Scholar 

  21. Vallieres L, Rivest S. Regulation of the genes encoding interleukin-6, its receptor, and gp130 in the rat brain in response to the immune activator lipopolysaccharide and the proinflammatory cytokine interleukin-1beta. J Neurochem. 1997;69(4):1668–83.

    Article  CAS  PubMed  Google Scholar 

  22. Nadeau S, Rivest S. Regulation of the gene encoding tumor necrosis factor alpha (TNF-alpha) in the rat brain and pituitary in response in different models of systemic immune challenge. J Neuropathol Exp Neurol. 1999;58(1):61–77.

    Article  CAS  PubMed  Google Scholar 

  23. Skelly DT, Hennessy E, Dansereau MA, Cunningham C. A systematic analysis of the peripheral and CNS effects of systemic LPS, IL-1beta, [corrected] TNF-alpha and IL-6 challenges in C57BL/6 mice. PLoS ONE. 2013;8(7):e69123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Liu X, Nemeth DP, McKim DB, Zhu L, DiSabato DJ, Berdysz O, Gorantla G, Oliver B, Witcher KG, Wang Y, et al. Cell-type-specific interleukin 1 receptor 1 signaling in the brain regulates distinct neuroimmune activities. Immunity. 2019;50(3):764–6.

    Article  CAS  PubMed  Google Scholar 

  25. Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol. 1993;47(2):169–76.

    Article  CAS  PubMed  Google Scholar 

  26. van Gool WA, van de Beek D, Eikelenboom P. Systemic infection and delirium: when cytokines and acetylcholine collide. Lancet. 2010;375(9716):773–5.

    Article  PubMed  CAS  Google Scholar 

  27. Hoogland IC, Houbolt C, van Westerloo DJ, van Gool WA, van de Beek D. Systemic inflammation and microglial activation: systematic review of animal experiments. J Neuroinflamm. 2015;12:114.

    Article  CAS  Google Scholar 

  28. Zrzavy T, Hoftberger R, Berger T, Rauschka H, Butovsky O, Weiner H, Lassmann H. Pro-inflammatory activation of microglia in the brain of patients with sepsis. Neuropathol Appl Neurobiol. 2019;45(3):278–90.

    Article  CAS  PubMed  Google Scholar 

  29. Mazeraud A, Righy C, Bouchereau E, Benghanem S, Bozza FA, Sharshar T. Septic-associated encephalopathy: a comprehensive review. Neurotherapeutics. 2020;17(2):392–403.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Lemstra AW, Groen in’t Woud JC, Hoozemans JJ, van Haastert ES, Rozemuller AJ, Eikelenboom P, van Gool WA. Microglia activation in sepsis: a case-control study. J Neuroinflamm. 2007;4:4.

    Article  CAS  Google Scholar 

  31. Moraes CA, Santos G, de Sampaio e Spohr TC, D’Avila JC, Lima FR, Benjamim CF, Bozza FA, Gomes FC. Activated microglia-induced deficits in excitatory synapses through IL-1beta: implications for cognitive impairment in sepsis. Mol Neurobiol. 2015;52(1):653–63.

    Article  CAS  PubMed  Google Scholar 

  32. Akrout N, Sharshar T, Annane D. Mechanisms of brain signaling during sepsis. Curr Neuropharmacol. 2009;7(4):296–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hughes CG, Patel MB, Pandharipande PP. Pathophysiology of acute brain dysfunction: what’s the cause of all this confusion? Curr Opin Crit Care. 2012;18(5):518–26.

    Article  PubMed  Google Scholar 

  34. Haley MJ, Lawrence CB. The blood-brain barrier after stroke: structural studies and the role of transcytotic vesicles. J Cereb Blood Flow Metab. 2017;37(2):456–70.

    Article  PubMed  Google Scholar 

  35. Bernardo-Castro S, Sousa JA, Bras A, Cecilia C, Rodrigues B, Almendra L, Machado C, Santo G, Silva F, Ferreira L, et al. Pathophysiology of blood-brain barrier permeability throughout the different stages of ischemic stroke and its implication on hemorrhagic transformation and recovery. Front Neurol. 2020;11:594672.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Stokum JA, Gerzanich V, Simard JM. Molecular pathophysiology of cerebral edema. J Cereb Blood Flow Metab. 2016;36(3):513–38.

    Article  CAS  PubMed  Google Scholar 

  37. Paris L, Tonutti L, Vannini C, Bazzoni G. Structural organization of the tight junctions. Biochim Biophys Acta. 2008;1778(3):646–59.

    Article  CAS  PubMed  Google Scholar 

  38. Towner RA, Saunders D, Smith N, Towler W, Cruz M, Do S, Maher JE, Whitaker K, Lerner M, Morton KA. Assessing long-term neuroinflammatory responses to encephalopathy using MRI approaches in a rat endotoxemia model. Geroscience. 2018;40(1):49–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Stubbs DJ, Yamamoto AK, Menon DK. Imaging in sepsis-associated encephalopathy–insights and opportunities. Nat Rev Neurol. 2013;9(10):551–61.

    Article  CAS  PubMed  Google Scholar 

  40. Ehler J, Barrett LK, Taylor V, Groves M, Scaravilli F, Wittstock M, Kolbaske S, Grossmann A, Henschel J, Gloger M, et al. Translational evidence for two distinct patterns of neuroaxonal injury in sepsis: a longitudinal, prospective translational study. Crit Care. 2017;21(1):262.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Dal-Pizzol F, Rojas HA, dos Santos EM, Vuolo F, Constantino L, Feier G, Pasquali M, Comim CM, Petronilho F, Gelain DP, et al. Matrix metalloproteinase-2 and metalloproteinase-9 activities are associated with blood-brain barrier dysfunction in an animal model of severe sepsis. Mol Neurobiol. 2013;48(1):62–70.

    Article  CAS  PubMed  Google Scholar 

  42. Erikson K, Tuominen H, Vakkala M, Liisanantti JH, Karttunen T, Syrjala H, Ala-Kokko TI. Brain tight junction protein expression in sepsis in an autopsy series. Crit Care. 2020;24(1):385.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Yanagida K, Liu CH, Faraco G, Galvani S, Smith HK, Burg N, Anrather J, Sanchez T, Iadecola C, Hla T. Size-selective opening of the blood-brain barrier by targeting endothelial sphingosine 1-phosphate receptor 1. Proc Natl Acad Sci USA. 2017;114(17):4531–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Winkler MS, Nierhaus A, Holzmann M, Mudersbach E, Bauer A, Robbe L, Zahrte C, Geffken M, Peine S, Schwedhelm E, et al. Decreased serum concentrations of sphingosine-1-phosphate in sepsis. Crit Care. 2015;19:372.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Lopes Pinheiro MA, Kroon J, Hoogenboezem M, Geerts D, van Het Hof B, van der Pol SM, van Buul JD, de Vries HE. Acid sphingomyelinase-derived ceramide regulates ICAM-1 function during T cell transmigration across brain endothelial cells. J Immunol. 2016;196(1):72–9.

    Article  CAS  PubMed  Google Scholar 

  46. Wu H, Deng R, Chen X, Wong WC, Chen H, Gao L, Nie Y, Wu W, Shen J. Caveolin-1 is critical for lymphocyte trafficking into central nervous system during experimental autoimmune encephalomyelitis. J Neurosci. 2016;36(19):5193–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Goldblum SE, Ding X, Campbell-Washington J. TNF-alpha induces endothelial cell F-actin depolymerization, new actin synthesis, and barrier dysfunction. Am J Physiol. 1993;264(4 Pt 1):C894-905.

    Article  CAS  PubMed  Google Scholar 

  48. O’Carroll SJ, Kho DT, Wiltshire R, Nelson V, Rotimi O, Johnson R, Angel CE, Graham ES. Pro-inflammatory TNFalpha and IL-1beta differentially regulate the inflammatory phenotype of brain microvascular endothelial cells. J Neuroinflamm. 2015;12:131.

    Article  CAS  Google Scholar 

  49. Haines RJ, Beard RS Jr, Wu MH. Protein tyrosine kinase 6 mediates TNFalpha-induced endothelial barrier dysfunction. Biochem Biophys Res Commun. 2015;456(1):190–6.

    Article  CAS  PubMed  Google Scholar 

  50. Lv S, Song HL, Zhou Y, Li LX, Cui W, Wang W, Liu P. Tumour necrosis factor-alpha affects blood-brain barrier permeability and tight junction-associated occludin in acute liver failure. Liver Int. 2010;30(8):1198–210.

    Article  CAS  PubMed  Google Scholar 

  51. Janz DR, Abel TW, Jackson JC, Gunther ML, Heckers S, Ely EW. Brain autopsy findings in intensive care unit patients previously suffering from delirium: a pilot study. J Crit Care. 2010;25(3):538.e537-512.

    Article  Google Scholar 

  52. Pfister D, Siegemund M, Dell-Kuster S, Smielewski P, Rüegg S, Strebel SP, Marsch SC, Pargger H, Steiner LA. Cerebral perfusion in sepsis-associated delirium. Crit Care. 2008;12(3):R63.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Taccone FS, Castanares-Zapatero D, Peres-Bota D, Vincent JL, Berre J, Melot C. Cerebral autoregulation is influenced by carbon dioxide levels in patients with septic shock. Neurocrit Care. 2010;12(1):35–42.

    Article  CAS  PubMed  Google Scholar 

  54. Bermejo-Martin JF, Martin-Fernandez M, Lopez-Mestanza C, Duque P, Almansa R. Shared features of endothelial dysfunction between sepsis and its preceding risk factors (aging and chronic disease). J Clin Med. 2018;7(11):400.

    Article  CAS  PubMed Central  Google Scholar 

  55. Nael K, Trouard TP, Lafleur SR, Krupinski EA, Salamon N, Kidwell CS. White matter ischemic changes in hyperacute ischemic stroke: voxel-based analysis using diffusion tensor imaging and MR perfusion. Stroke. 2015;46(2):413–8.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Miners S, Kehoe PG, Love S. Cognitive impact of COVID-19: looking beyond the short term. Alzheimers Res Ther. 2020;12(1):170.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wen Y, Yang S, Liu R, Simpkins JW. Transient cerebral ischemia induces site-specific hyperphosphorylation of tau protein. Brain Res. 2004;1022(1–2):30–8.

    Article  CAS  PubMed  Google Scholar 

  58. Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, Cummings J, van der Flier WM. Alzheimer’s disease. Lancet. 2021;397:1577–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Brown GC. The endotoxin hypothesis of neurodegeneration. J Neuroinflamm. 2019;16(1):180.

    Article  CAS  Google Scholar 

  60. Gasparotto J, Girardi CS, Somensi N, Ribeiro CT, Moreira JCF, Michels M, Sonai B, Rocha M, Steckert AV, Barichello T, et al. Receptor for advanced glycation end products mediates sepsis-triggered amyloid-β accumulation, Tau phosphorylation, and cognitive impairment. J Biol Chem. 2018;293(1):226–44.

    Article  CAS  PubMed  Google Scholar 

  61. Kirk RA, Kesner RP, Wang LM, Wu Q, Towner RA, Hoffman JM, Morton KA. Lipopolysaccharide exposure in a rat sepsis model results in hippocampal amyloid-β plaque and phosphorylated tau deposition and corresponding behavioral deficits. Geroscience. 2019;41(4):467–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Milioli MVM, Burger H, Olivieri R, Michels M, Ávila P, Abatti M, Indalécio A, Ritter C, Dal-Pizzol F. The impact of age on long-term behavioral and neurochemical parameters in an animal model of severe sepsis. Neurosci Lett. 2019;708:134339.

    Article  CAS  PubMed  Google Scholar 

  63. Gasparotto J, Girardi CS, Somensi N, Ribeiro CT, Moreira JCF, Michels M, Sonai B, Rocha M, Steckert AV, Barichello T, et al. Receptor for advanced glycation end products mediates sepsis-triggered amyloid-beta accumulation, Tau phosphorylation, and cognitive impairment. J Biol Chem. 2018;293(1):226–44.

    Article  CAS  PubMed  Google Scholar 

  64. Zhao T, Xia Y, Wang D, Pang L. Association between elevated serum tau protein level and sepsis-associated encephalopathy in patients with severe sepsis. Can J Infect Dis Med Microbiol. 2019;2019:1876174.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Orhun G, Esen F, Ozcan PE, Sencer S, Bilgic B, Ulusoy C, Noyan H, Kucukerden M, Ali A, Barburoglu M, et al. Neuroimaging findings in sepsis-induced brain dysfunction: association with clinical and laboratory findings. Neurocrit Care. 2019;30(1):106–17.

    Article  CAS  PubMed  Google Scholar 

  66. Rogne AG, Muller EG, Udnaes E, Sigurdardottir S, Raudeberg R, Connelly JP, Revheim ME, Hassel B, Dahlberg D. beta-Amyloid may accumulate in the human brain after focal bacterial infection: an (18) F-flutemetamol positron emission tomography study. Eur J Neurol. 2021;28(3):877–83.

    Article  PubMed  Google Scholar 

  67. Kamer AR, Pirraglia E, Tsui W, Rusinek H, Vallabhajosula S, Mosconi L, Yi L, McHugh P, Craig RG, Svetcov S, et al. Periodontal disease associates with higher brain amyloid load in normal elderly. Neurobiol Aging. 2015;36(2):627–33.

    Article  CAS  PubMed  Google Scholar 

  68. Kantonen J, Mahzabin S, Mayranpaa MI, Tynninen O, Paetau A, Andersson N, Sajantila A, Vapalahti O, Carpen O, Kekalainen E, et al. Neuropathologic features of four autopsied COVID-19 patients. Brain Pathol. 2020;30(6):1012–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Rhodes CH, Priemer DS, Esma K, Perl DP, James G. Β-Amyloid deposits in young COVID patients. Preprints with THE LANCET. 2022.

  70. Wang JZ, Xia YY, Grundke-Iqbal I, Iqbal K. Abnormal hyperphosphorylation of tau: sites, regulation, and molecular mechanism of neurofibrillary degeneration. J Alzheimers Dis. 2013;33(Suppl 1):S123-139.

    PubMed  Google Scholar 

  71. Golpich M, Amini E, Hemmati F, Ibrahim NM, Rahmani B, Mohamed Z, Raymond AA, Dargahi L, Ghasemi R, Ahmadiani A. Glycogen synthase kinase-3 beta (GSK-3β) signaling: Implications for Parkinson’s disease. Pharmacol Res. 2015;97:16–26.

    Article  CAS  PubMed  Google Scholar 

  72. Gong R, Ge Y, Chen S, Liang E, Esparza A, Sabo E, Yango A, Gohh R, Rifai A, Dworkin LD. Glycogen synthase kinase 3beta: a novel marker and modulator of inflammatory injury in chronic renal allograft disease. Am J Transplant. 2008;8(9):1852–63.

    Article  CAS  PubMed  Google Scholar 

  73. Martin M, Rehani K, Jope RS, Michalek SM. Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol. 2005;6(8):777–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Hoeflich KP, Luo J, Rubie EA, Tsao MS, Jin O, Woodgett JR. Requirement for glycogen synthase kinase-3beta in cell survival and NF-kappaB activation. Nature. 2000;406(6791):86–90.

    Article  CAS  PubMed  Google Scholar 

  75. Beurel E. Regulation by glycogen synthase kinase-3 of inflammation and T cells in CNS diseases. Front Mol Neurosci. 2011;4:18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Wang H, Brown J, Gu Z, Garcia CA, Liang R, Alard P, Beurel E, Jope RS, Greenway T, Martin M. Convergence of the mammalian target of rapamycin complex 1- and glycogen synthase kinase 3-beta-signaling pathways regulates the innate inflammatory response. J Immunol. 2011;186(9):5217–26.

    Article  CAS  PubMed  Google Scholar 

  77. Beurel E, Jope RS. Differential regulation of STAT family members by glycogen synthase kinase-3. J Biol Chem. 2008;283(32):21934–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Yu Y, Ye RD. Microglial Abeta receptors in Alzheimer’s disease. Cell Mol Neurobiol. 2015;35(1):71–83.

    Article  CAS  PubMed  Google Scholar 

  79. Cai Z, Liu N, Wang C, Qin B, Zhou Y, Xiao M, Chang L, Yan LJ, Zhao B. Role of RAGE in Alzheimer’s disease. Cell Mol Neurobiol. 2016;36(4):483–95.

    Article  CAS  PubMed  Google Scholar 

  80. Wautier MP, Tessier FJ, Wautier JL. Advanced glycation end products: a risk factor for human health. Ann Pharm Fr. 2014;72(6):400–8.

    Article  CAS  PubMed  Google Scholar 

  81. Paudel YN, Angelopoulou E, Piperi C, Othman I, Aamir K, Shaikh MF. Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s disease (AD): from risk factors to therapeutic targeting. Cells. 2020;9(2):383.

    Article  CAS  PubMed Central  Google Scholar 

  82. Leclerc E, Fritz G, Vetter SW, Heizmann CW. Binding of S100 proteins to RAGE: an update. Biochim Biophys Acta. 2009;1793(6):993–1007.

    Article  CAS  PubMed  Google Scholar 

  83. Sirois CM, Jin T, Miller AL, Bertheloot D, Nakamura H, Horvath GL, Mian A, Jiang J, Schrum J, Bossaller L, et al. RAGE is a nucleic acid receptor that promotes inflammatory responses to DNA. J Exp Med. 2013;210(11):2447–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Li K, Dai D, Zhao B, Yao L, Yao S, Wang B, Yang Z. Association between the RAGE G82S polymorphism and Alzheimer’s disease. J Neural Transm (Vienna). 2010;117(1):97–104.

    Article  CAS  Google Scholar 

  85. Lue LF, Walker DG, Brachova L, Beach TG, Rogers J, Schmidt AM, Stern DM, Yan SD. Involvement of microglial receptor for advanced glycation endproducts (RAGE) in Alzheimer’s disease: identification of a cellular activation mechanism. Exp Neurol. 2001;171(1):29–45.

    Article  CAS  PubMed  Google Scholar 

  86. Arancio O, Zhang HP, Chen X, Lin C, Trinchese F, Puzzo D, Liu S, Hegde A, Yan SF, Stern A, et al. RAGE potentiates Abeta-induced perturbation of neuronal function in transgenic mice. EMBO J. 2004;23(20):4096–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Fang F, Lue LF, Yan S, Xu H, Luddy JS, Chen D, Walker DG, Stern DM, Yan S, Schmidt AM, et al. RAGE-dependent signaling in microglia contributes to neuroinflammation, Abeta accumulation, and impaired learning/memory in a mouse model of Alzheimer’s disease. FASEB J. 2010;24(4):1043–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Fang F, Yu Q, Arancio O, Chen D, Gore SS, Yan SS, Yan SF. RAGE mediates Abeta accumulation in a mouse model of Alzheimer’s disease via modulation of beta- and gamma-secretase activity. Hum Mol Genet. 2018;27(6):1002–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Sharshar T, Annane D, de la Grandmaison GL, Brouland JP, Hopkinson NS, Francoise G. The neuropathology of septic shock. Brain Pathol. 2004;14(1):21–33.

    Article  PubMed  Google Scholar 

  90. Valderrama EV, Humbert K, Lord A, Frontera J, Yaghi S. Severe acute respiratory syndrome coronavirus 2 infection and ischemic stroke. Stroke. 2020;51(7):e124–7.

    Article  CAS  PubMed  Google Scholar 

  91. Annane D, Sharshar T. Cognitive decline after sepsis. Lancet Respir Med. 2015;3(1):61–9.

    Article  CAS  PubMed  Google Scholar 

  92. Manabe T, Heneka MT. Cerebral dysfunctions caused by sepsis during ageing. Nat Rev Immunol. 2021. https://doi.org/10.1038/s41577-021-00643-7.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Czempik PF, Pluta MP, Krzych ŁJ. Sepsis-associated brain dysfunction: a review of current literature. Int J Environ Res Public Health. 2020;17(16):5852.

    Article  CAS  PubMed Central  Google Scholar 

  94. Catarina AV, Branchini G, Bettoni L, De Oliveira JR, Nunes FB. Sepsis-associated encephalopathy: from pathophysiology to progress in experimental studies. Mol Neurobiol. 2021. https://doi.org/10.1007/s12035-021-02303-2.

    Article  PubMed  Google Scholar 

  95. Chaudhry N, Duggal AK. Sepsis associated encephalopathy. Adv Med. 2014;2014:762320.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Sonneville R, de Montmollin E, Poujade J, Garrouste-Orgeas M, Souweine B, Darmon M, Mariotte E, Argaud L, Barbier F, Goldgran-Toledano D, et al. Potentially modifiable factors contributing to sepsis-associated encephalopathy. Intensive Care Med. 2017;43(8):1075–84.

    Article  PubMed  Google Scholar 

  97. Chung HY, Wickel J, Brunkhorst FM, Geis C. Sepsis-associated encephalopathy: from delirium to dementia? J Clin Med. 2020;9(3):703.

    Article  PubMed Central  Google Scholar 

  98. Zhang H, Yuan J, Chen Q, Cao Y, Wang Z, Lu W, Bao J. Development and validation of a predictive score for ICU delirium in critically ill patients. BMC Anesthesiol. 2021;21(1):37.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Atterton B, Paulino MC, Povoa P, Martin-Loeches I. Sepsis associated delirium. Medicina (Kaunas). 2020;56(5):240.

    Article  Google Scholar 

  100. Girard TD, Thompson JL, Pandharipande PP, Brummel NE, Jackson JC, Patel MB, Hughes CG, Chandrasekhar R, Pun BT, Boehm LM, et al. Clinical phenotypes of delirium during critical illness and severity of subsequent long-term cognitive impairment: a prospective cohort study. Lancet Respir Med. 2018;6(3):213–22.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Dantzer R, Kelley KW. Twenty years of research on cytokine-induced sickness behavior. Brain Behav Immun. 2007;21(2):153–60.

    Article  CAS  PubMed  Google Scholar 

  102. Kelley KW, Bluthé RM, Dantzer R, Zhou JH, Shen WH, Johnson RW, Broussard SR. Cytokine-induced sickness behavior. Brain Behav Immun. 2003;17(Suppl 1):S112-118.

    Article  CAS  PubMed  Google Scholar 

  103. Shattuck EC, Muehlenbein MP. Towards an integrative picture of human sickness behavior. Brain Behav Immun. 2016;57:255–62.

    Article  CAS  PubMed  Google Scholar 

  104. Pereira de Souza Goldim M, Della Giustina A, Mathias K, de Oliveira Junior A, Fileti ME, De Carli R, Zarbato G, Garbossa L, da Rosa N, Oliveira J, et al. Sickness behavior score is associated with neuroinflammation and late behavioral changes in polymicrobial sepsis animal model. Inflammation. 2020;43(3):1019–34.

    Article  CAS  PubMed  Google Scholar 

  105. Honarmand K, Lalli RS, Priestap F, Chen JL, McIntyre CW, Owen AM, Slessarev M. Natural history of cognitive impairment in critical illness survivors. A systematic review. Am J Respir Crit Care Med. 2020;202(2):193–201.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Wang HE, Kabeto MM, Gray M, Wadley VG, Muntner P, Judd SE, Safford MM, Kempker J, Levine DA. Trajectory of cognitive decline after sepsis. Crit Care Med. 2021. https://doi.org/10.1097/CCM.0000000000004897.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Mart MF, Williams Roberson S, Salas B, Pandharipande PP, Ely EW. Prevention and management of delirium in the intensive care unit. Semin Respir Crit Care Med. 2021;42(1):112–26.

    Article  PubMed  Google Scholar 

  108. Bennett IJ, Madden DJ. Disconnected aging: cerebral white matter integrity and age-related differences in cognition. Neuroscience. 2014;276:187–205.

    Article  CAS  PubMed  Google Scholar 

  109. Hase Y, Horsburgh K, Ihara M, Kalaria RN. White matter degeneration in vascular and other ageing-related dementias. J Neurochem. 2018;144(5):617–33.

    Article  CAS  PubMed  Google Scholar 

  110. Orhun G, Tuzun E, Bilgic B, Ergin Ozcan P, Sencer S, Barburoglu M, Esen F. Brain volume changes in patients with acute brain dysfunction due to sepsis. Neurocrit Care. 2020;32(2):459–68.

    Article  PubMed  Google Scholar 

  111. Luitse MJ, van Asch CJ, Klijn CJ. Deep coma and diffuse white matter abnormalities caused by sepsis-associated encephalopathy. Lancet. 2013;381(9884):2222.

    Article  PubMed  Google Scholar 

  112. Sharshar T, Carlier R, Bernard F, Guidoux C, Brouland JP, Nardi O, de la Grandmaison GL, Aboab J, Gray F, Menon D, et al. Brain lesions in septic shock: a magnetic resonance imaging study. Intensive Care Med. 2007;33(5):798–806.

    Article  PubMed  Google Scholar 

  113. Seidel G, Gaser C, Götz T, Günther A, Hamzei F. Accelerated brain ageing in sepsis survivors with cognitive long-term impairment. Eur J Neurosci. 2020;52(10):4395–402.

    Article  PubMed  Google Scholar 

  114. Gunther ML, Morandi A, Krauskopf E, Pandharipande P, Girard TD, Jackson JC, Thompson J, Shintani AK, Geevarghese S, Miller RR 3rd, et al. The association between brain volumes, delirium duration, and cognitive outcomes in intensive care unit survivors: the VISIONS cohort magnetic resonance imaging study*. Crit Care Med. 2012;40(7):2022–32.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Garofalo AM, Lorente-Ros M, Goncalvez G, Carriedo D, Ballén-Barragán A, Villar-Fernández A, Peñuelas Ó, Herrero R, Granados-Carreño R, Lorente JA. Histopathological changes of organ dysfunction in sepsis. Intensive Care Med Exp. 2019;7(Suppl 1):45.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Sharshar T, Gray F, Lorin de la Grandmaison G, Hopkinson NS, Ross E, Dorandeu A, Orlikowski D, Raphael JC, Gajdos P, Annane D. Apoptosis of neurons in cardiovascular autonomic centres triggered by inducible nitric oxide synthase after death from septic shock. Lancet. 2003;362(9398):1799–805.

    Article  CAS  PubMed  Google Scholar 

  117. Bozza FA, Garteiser P, Oliveira MF, Doblas S, Cranford R, Saunders D, Jones I, Towner RA, Castro-Faria-Neto HC. Sepsis-associated encephalopathy: a magnetic resonance imaging and spectroscopy study. J Cereb Blood Flow Metab. 2010;30(2):440–8.

    Article  PubMed  Google Scholar 

  118. Tomiyama T, Shoji A, Kataoka K, Suwa Y, Asano S, Kaneko H, Endo N. Inhibition of amyloid beta protein aggregation and neurotoxicity by rifampicin. Its possible function as a hydroxyl radical scavenger. J Biol Chem. 1996;271(12):6839–44.

    Article  CAS  PubMed  Google Scholar 

  119. Zhu L, Yuan Q, Zeng Z, Zhou R, Luo R, Zhang J, Tsang CK, Bi W. Rifampicin suppresses amyloid-β accumulation through enhancing autophagy in the hippocampus of a lipopolysaccharide-induced mouse model of cognitive decline. J Alzheimers Dis. 2021;79(3):1171–84.

    Article  CAS  PubMed  Google Scholar 

  120. Iizuka T, Morimoto K, Sasaki Y, Kameyama M, Kurashima A, Hayasaka K, Ogata H, Goto H. Preventive effect of rifampicin on alzheimer disease needs at least 450 mg daily for 1 year: an FDG-PET follow-up study. Dement Geriatr Cogn Dis Extra. 2017;7(2):204–14.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Lee CY, Huang CH, Lu PL, Ko WC, Chen YH, Hsueh PR. Role of rifampin for the treatment of bacterial infections other than mycobacteriosis. J Infect. 2017;75(5):395–408.

    Article  PubMed  Google Scholar 

  122. Liu D, Zhao J. Cytokine release syndrome: grading, modeling, and new therapy. J Hematol Oncol. 2018;11(1):121.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, Sanvito F, Ponzoni M, Doglioni C, Cristofori P, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 2018;24(6):739–48.

    Article  CAS  PubMed  Google Scholar 

  124. Shakoory B, Carcillo JA, Chatham WW, Amdur RL, Zhao H, Dinarello CA, Cron RQ, Opal SM. Interleukin-1 receptor blockade is associated with reduced mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior phase III trial. Crit Care Med. 2016;44(2):275–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Abraham E, Wunderink R, Silverman H, Perl TM, Nasraway S, Levy H, Bone R, Wenzel RP, Balk R, Allred R, et al. Efficacy and safety of monoclonal antibody to human tumor necrosis factor alpha in patients with sepsis syndrome. A randomized, controlled, double-blind, multicenter clinical trial. TNF-alpha MAb Sepsis Study Group. JAMA. 1995;273(12):934–41.

    Article  CAS  PubMed  Google Scholar 

  126. Avila AA, Kinberg EC, Sherwin NK, Taylor RD. The use of fluids in sepsis. Cureus. 2016;8(3):e528.

    PubMed  PubMed Central  Google Scholar 

  127. Semler MW, Rice TW. Sepsis resuscitation: fluid choice and dose. Clin Chest Med. 2016;37(2):241–50.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Myburgh JA, Finfer S, Bellomo R, Billot L, Cass A, Gattas D, Glass P, Lipman J, Liu B, McArthur C, et al. Hydroxyethyl starch or saline for fluid resuscitation in intensive care. N Engl J Med. 2012;367(20):1901–11.

    Article  CAS  PubMed  Google Scholar 

  129. Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, et al. Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med. 2008;358(2):125–39.

    Article  CAS  PubMed  Google Scholar 

  130. Perner A, Haase N, Guttormsen AB, Tenhunen J, Klemenzson G, Åneman A, Madsen KR, Møller MH, Elkjær JM, Poulsen LM, et al. Hydroxyethyl starch 130/0.42 versus Ringer’s acetate in severe sepsis. N Engl J Med. 2012;367(2):124–34.

    Article  CAS  PubMed  Google Scholar 

  131. Tymianski M. Combining neuroprotection with endovascular treatment of acute stroke: is there hope? Stroke. 2017;48(6):1700–5.

    Article  PubMed  Google Scholar 

  132. Hill MD, Goyal M, Menon BK, Nogueira RG, McTaggart RA, Demchuk AM, Poppe AY, Buck BH, Field TS, Dowlatshahi D, et al. Efficacy and safety of nerinetide for the treatment of acute ischaemic stroke (ESCAPE-NA1): a multicentre, double-blind, randomised controlled trial. Lancet. 2020;395(10227):878–87.

    Article  CAS  PubMed  Google Scholar 

  133. Olivieri R, Michels M, Pescador B, Avila P, Abatti M, Cucker L, Burger H, Dominguini D, Quevedo J, Dal-Pizzol F. The additive effect of aging on sepsis-induced cognitive impairment and neuroinflammation. J Neuroimmunol. 2018;314:1–7.

    Article  CAS  PubMed  Google Scholar 

  134. Waisman A, Hauptmann J, Regen T. The role of IL-17 in CNS diseases. Acta Neuropathol. 2015;129(5):625–37.

    Article  CAS  PubMed  Google Scholar 

  135. Kawanokuchi J, Shimizu K, Nitta A, Yamada K, Mizuno T, Takeuchi H, Suzumura A. Production and functions of IL-17 in microglia. J Neuroimmunol. 2008;194(1–2):54–61.

    Article  CAS  PubMed  Google Scholar 

  136. Ye B, Tao T, Zhao A, Wen L, He X, Liu Y, Fu Q, Mi W, Lou J. Blockade of IL-17A/IL-17R pathway protected mice from sepsis-associated encephalopathy by inhibition of microglia activation. Mediators Inflamm. 2019;2019:8461725.

    PubMed  PubMed Central  Google Scholar 

  137. Cheng Y, Mai JY, Hou TL, Ping J, Chen JJ. Antiviral activities of atractylon from atractylodis rhizoma. Mol Med Rep. 2016;14(4):3704–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Tian M, Qingzhen L, Zhiyang Y, Chunlong C, Jiao D, Zhang L, Li W. Attractylone attenuates sepsis-associated encephalopathy and cognitive dysfunction by inhibiting microglial activation and neuroinflammation. J Cell Biochem. 2019. https://doi.org/10.1002/jcb.27983.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Chen Z, Trapp BD. Microglia and neuroprotection. J Neurochem. 2016;136(Suppl 1):10–7.

    Article  CAS  PubMed  Google Scholar 

  140. Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, Sepehrband F, Nelson AR, Buennagel DP, Harrington MG, et al. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med. 2019;25(2):270–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Zhang SQ, Xiao J, Chen M, Zhou LQ, Shang K, Qin C, Tian DS. Sphingosine-1-phosphate signaling in ischemic stroke: from bench to bedside and beyond. Front Cell Neurosci. 2021;15:781098.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Vutukuri R, Brunkhorst R, Kestner RI, Hansen L, Bouzas NF, Pfeilschifter J, Devraj K, Pfeilschifter W. Alteration of sphingolipid metabolism as a putative mechanism underlying LPS-induced BBB disruption. J Neurochem. 2018;144(2):172–85.

    Article  CAS  PubMed  Google Scholar 

  143. Willis MA, Cohen JA. Fingolimod therapy for multiple sclerosis. Semin Neurol. 2013;33(1):37–44.

    Article  PubMed  Google Scholar 

  144. Aytan N, Choi JK, Carreras I, Brinkmann V, Kowall NW, Jenkins BG, Dedeoglu A. Fingolimod modulates multiple neuroinflammatory markers in a mouse model of Alzheimer’s disease. Sci Rep. 2016;6:24939.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Gaire BP, Lee CH, Sapkota A, Lee SY, Chun J, Cho HJ, Nam TG, Choi JW. Identification of sphingosine 1-phosphate receptor subtype 1 (S1P1) as a pathogenic factor in transient focal cerebral ischemia. Mol Neurobiol. 2018;55(3):2320–32.

    Article  CAS  PubMed  Google Scholar 

  146. Nitzsche A, Poittevin M, Benarab A, Bonnin P, Faraco G, Uchida H, Favre J, Garcia-Bonilla L, Garcia MCL, Leger PL, et al. Endothelial S1P1 signaling counteracts infarct expansion in ischemic stroke. Circ Res. 2021;128(3):363–82.

    Article  CAS  PubMed  Google Scholar 

  147. Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–6.

    Article  CAS  PubMed  Google Scholar 

  148. Hampel H, Lista S, Mango D, Nisticò R, Perry G, Avila J, Hernandez F, Geerts H, Vergallo A. Lithium as a treatment for Alzheimer’s disease: the systems pharmacology perspective. J Alzheimers Dis. 2019;69(3):615–29.

    Article  PubMed  Google Scholar 

  149. Matsunaga S, Kishi T, Annas P, Basun H, Hampel H, Iwata N. Lithium as a treatment for Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2015;48(2):403–10.

    Article  CAS  PubMed  Google Scholar 

  150. Yang H, Wang H, Andersson U. Targeting inflammation driven by HMGB1. Front Immunol. 2020;11:484.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Angus DC, Yang L, Kong L, Kellum JA, Delude RL, Tracey KJ, Weissfeld L, Gen IMSI. Circulating high-mobility group box 1 (HMGB1) concentrations are elevated in both uncomplicated pneumonia and pneumonia with severe sepsis. Crit Care Med. 2007;35(4):1061–7.

    Article  PubMed  Google Scholar 

  152. Chavan SS, Huerta PT, Robbiati S, Valdes-Ferrer SI, Ochani M, Dancho M, Frankfurt M, Volpe BT, Tracey KJ, Diamond B. HMGB1 mediates cognitive impairment in sepsis survivors. Mol Med. 2012;18:930–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Jarczak D, Kluge S, Nierhaus A. Sepsis-pathophysiology and therapeutic concepts. Front Med (Lausanne). 2021;8:628302.

    Article  Google Scholar 

  154. van der Slikke EC, An AY, Hancock REW, Bouma HR. Exploring the pathophysiology of post-sepsis syndrome to identify therapeutic opportunities. EBioMedicine. 2020;61:103044.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Haifa Kassis, MD, for her editorial assistance.

Funding

The work was supported by NIH grant R01AG055559.

Author information

Authors and Affiliations

Authors

Contributions

Conceived and designed the manuscript: NS and AS; Wrote the main manuscript: NS and AS; Revised the manuscript: AS, NS and MS; All authors reviewed the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Amjad Shehadah.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors consented for publication.

Competing interests

The authors have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sekino, N., Selim, M. & Shehadah, A. Sepsis-associated brain injury: underlying mechanisms and potential therapeutic strategies for acute and long-term cognitive impairments. J Neuroinflammation 19, 101 (2022). https://doi.org/10.1186/s12974-022-02464-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-022-02464-4

Keywords