Microglia in Sepsis-Associated Encephalopathy: Comparison
Please note this is a comparison between Version 1 by Joana D'Avila and Version 2 by Lily Guo.

Multiple organ dysfunction is a hallmark of sepsis pathogenesis, and neurological manifestations are a frequent and underestimated symptom. Sepsis induces an acute brain dysfunction that is not related to direct brain infection and is characterized by clinical and electroencephalographic changes that range from sickness behavior to altered consciousness, varying from confusion to delirium and coma.

  • Microglia
  • Sepsis-Associated Encephalopathy
  • Neuroinflammation

1. Introduction

Sepsis is defined as a life-threatening organic dysfunction caused by a dysregulated host response to infection [1]. It was estimated that in 2017, there were 48.9 million cases and 11 million sepsis-related deaths worldwide, with a mortality rate that varies from 26% to 60% depending on the severity [2]. Despite all the efforts to improve sepsis treatment in the last decades, there is no specific therapy for sepsis, and mortality remains high, especially in low- and middle-income countries.
Sepsis is a clinical syndrome characterized by a maladaptive host response to infection that may be significantly amplified by endogenous factors. The pathogenesis of sepsis goes beyond an uncontrolled inflammatory response and includes major modifications in nonimmunologic systems such as cardiovascular, autonomic, neuroendocrine, coagulation, bioenergetic, and metabolic alterations [1]. While the inflammatory response in sepsis is relatively well-understood, the mechanisms driving metabolic deregulation and multiple organ dysfunction remain puzzling.
Multiple organ dysfunction is a hallmark of sepsis pathogenesis, and neurological manifestations are a frequent and underestimated symptom. Sepsis induces an acute brain dysfunction that is not related to direct brain infection and is characterized by various clinical and electroencephalographic changes [3][4][3,4]. This acute brain dysfunction is known as sepsis-associated encephalopathy (SAE), and it ranges from sickness behavior to altered consciousness, varying from confusion to delirium and, in worse cases, even coma [5]. SAE is strongly associated with higher mortality [6] and long-term cognitive impairments [7][8][7,8] affecting patients’ quality of life. To date, no curative or preventive treatment for SAE exists, and some pharmacological attempts have proved ineffective or even dangerous [9]. The absence of an effective therapeutic strategy is explained by the lack of precise knowledge of pathophysiological mechanisms. Some authors even suggest that the neurocognitive sequelae identified in the patients who had stayed in an ICU could be a risk factor for progressive neurodegenerative disorders of higher functions [10][11][10,11].
The mechanisms involved in SAE are diverse and include neurotransmitter dysfunction, inflammatory and ischemic lesions to the brain, microglia activation, changes in the blood–brain barrier (BBB) permeability, oxidative stress due to inflammation and mitochondrial dysfunction, and bioenergetic shifts that are part of metabolic adaptations to systemic inflammation [12][13][12,13]. Evidence suggests that alterations of the BBB resulting from both microglial and endothelial cell activation [14] could, in part, explain the acute neurological dysfunction by increasing the entry of inflammatory mediators and neurotoxic substances within the CNS [15]. Local production of pro-inflammatory markers and damage-associated molecular patterns (DAMPs) by the cells maintaining BBB (activated endothelial cells and astrocytes and perivascular macrophages) also contribute to neuroinflammation. Dysregulated cytokine responses are significant contributors to tissue injury and neurological deficits [16][17][18][16,17,18]. Inflammatory cytokines are involved in the pathophysiology of intensive care unit-acquired weakness, mostly, of neuropathies and myopathies [19]. All these mechanisms constitute a process known as neuroinflammation, which is the focus of this review.
To some extent, neuroinflammation is essential for maintaining brain homeostasis by inducing repair mechanisms. However, non-resolving neuroinflammation is a potentially harmful mechanism of brain damage. In this respect, microglia are major players in neuroinflammation, and exacerbated microglia activation has been associated with neurodegenerative diseases. As neurodegenerative diseases progress, microglia switch from a helpful role to a dysfunctional phenotype that becomes detrimental to neurons [20]. It is becoming clear that microglia surrounding Aβ plaques or Lewy bodies, for example, are not activated, but are nonfunctional [21]. There is a delicate balance determining whether microglia—and neuroinflammation in general—have beneficial or detrimental effects on the brain. This dual role is covered in the following sections, with a focus on sepsis and SAE.

2. Microglia in Sepsis-Associated Encephalopathy

Activated microglia exhibit rapid and profound morphological, phenotypic, bioenergetic, and functional changes in pathological conditions, broadly defined as “microglia activation” [22][23][30,57]. Microglia activation is a transient response characterized by an altered secretory profile, increased phagocytic activity, and deviation from the homeostatic phenotype.
Microglia activation is consistently observed acutely in sepsis, both in experimental models and septic patients [13][24][13,58]. Activated microglia express high levels of the 18-kDa translocator protein that can be measured in the human brain with the positron emission tomography (PET) radiotracer [11C] PBR28. LPS administration increased [11C] PBR28 binding, demonstrating microglia activation in human brains. This activation was associated with increased blood levels of inflammatory cytokines, vital sign changes, and sickness symptoms [25][59].
In a case-control study of 13 patients who died of sepsis, there was an increase in CD68 expression in the cortex compared to the control, and more amoeboid microglia were observed [26][60]. Similarly, in a postmortem case-control study of patients with delirium, there was an increase in microglial markers CD68 and HLA-DR compared to age-matched controls, suggesting that microglia activation could be related to delirium [27][61]. In a prospective study of 17 patients who died from septic shock, hippocampal tissue was assessed for a neuropathological study that observed increased microglia activation and apoptosis in septic patients with hyperglycemia [28][62].
The pathologic outcome of infections is a direct consequence of the extent of metabolic dysfunction and damage imposed on tissues that sustain host homeostasis [29][63]. Experimental models provide a deeper understanding of cellular response and the molecular mechanisms associated with organ dysfunction in sepsis. Oxidative stress and microglia activation have been consistently detected acutely in sepsis [12][24][30][12,58,64]. Genetic manipulation or pharmacological inhibition of pathways contributing to neuroinflammation and oxidative stress can protect survivors from neurocognitive impairments, indicating that neuroinflammation and oxidative stress are critical brain dysfunction mechanisms in SAE [31][32][33][41,65,66].
Video Production Service