Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Review
. 2025 Jun 3;23(1):622.
doi: 10.1186/s12967-025-06635-8.

Convergence of sepsis-associated encephalopathy pathogenesis onto microglia

Affiliations
Review

Convergence of sepsis-associated encephalopathy pathogenesis onto microglia

Dan Xiao et al. J Transl Med. .

Abstract

Sepsis-associated encephalopathy (SAE) is a neurological dysfunction induced by sepsis, with symptoms ranging from mild delirium to deep coma. About 70% of patients with severe systemic infection develop SAE and with more than half of surviving patients suffering from long-term cognitive deficits, which seriously damaged the quality of their daily life and brought a heavy burden to society. The pathogenesis of SAE is multifactorial, including activated inflammation, blood- brain barrier (BBB) disruption, cerebral blood flow impairment, and neurotransmitter disturbances. Microglia mediate multiple SAE pathologies. In this review, we summarized the most recent findings in the roles of microglia in every stage of SAE pathogenesis, focusing on the molecular pathways in microglia activation and downstream effects. We also demonstrated the novel therapeutic studies targeting microglia in SAE. Deep insight into the role of microglia in SAE is of great importance in exploring pathogenesis and developing effective remedies of SAE.

Keywords: Glycometabolism; Lipid metabolism; Microglia activation; Neuroinflammation; Neurological dysfunction; Sepsis-associated encephalopathy (SAE).

PubMed Disclaimer

Conflict of interest statement

Declarations. Ethics approval and consent to participate: Not applicable. Consent for publication: Not applicable. Competing interests: The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Pathogenesis of SAE. The pathogenesis of SAE is multifactorial, including blood-brain barrier disruption, neuroinflammation, cerebral ischemia, mitochondrial dysfunction, and neurotransmitter disruption. These factors involve in parallel and contribute to a varying degree to the development of SAE
Fig. 2
Fig. 2
Pathways involved in microglia activation. Activation of microglia is related to multiple signaling pathways, including NF-κB, JAK-STAT, inflammasome, pyroptosis, et al. During sepsis, TLRs play a pivotal role in inducing the polarization of microglia toward M1 by activating NF-kB. SIRT1 and Foxc1 can induce anti-inflammatory responses by directly inhibiting NF-κB signaling. The expression of TRAM1, who is an adaptor of TLR4, is highly induced by LPS/ IFN-γ stimulation and significantly enhances M1 polarization of microglia. Sepsis induced activation of JAK/STAT3 signaling promotes the binding of STAT3 to the promoter of TERT and leads to increased TERT expression. The activation of NF-κB further induces the expression of NLRP3, Pro-IL-1β and Pro-IL-18, which are necessary for inflammasome activation. The activated NLRP3 inflammasome triggers self-cleavage and generates active caspase-1, which then cleaves pro-IL-1β and pro-IL-18, causing the release of the pro-inflammatory cytokines IL-1β/18. Caspase-1 also cleaves GSDMD into N-GSDMD, which causes the formation of pores in the cell membrane and triggers pyroptosis
Fig. 3
Fig. 3
Glucose and lipid metabolism during microglia activation. Under neuroinflammatory conditions, microglia undergo metabolic reprogramming, especially in glucose and lipids metabolism, which is necessary for proper immune actions and cytokine release in the brain. Left: Microglia can recognize PAMPs (e.g., LPS) and other cellular inflammatory signals. Some inflammatory stimulus such as LPS and BAFF can cause increased glycolysis in microglia, promoting the expression of inflammatory genes, which are mainly through the activation of mTOR signal pathway. HIF-1α can enhance glycolysis through Akt/mTOR/HIF-1α pathway. The expression of Glut-1 was increased to uptake more glucose to meet the energy demand under inflammatory conditions. When inhibiting microglia glycolysis with glycolysis inhibitors 2-DG and 3-BPA, or by silencing GLUT1, NF-κB pathway is inhibited and thereby blocking the activation of microglia activation. Right: Microglia also recognize and phagocytose ApoE, myelin debris, lipoproteins, and other substances through certain receptors. Microglia recognize and phagocytose myelin debris through CD36 pathway. Myelin internalization increases CD36 expression through NRF2, skewing microglia to a less-inflammatory phenotype. While recognition of oxidized low-density lipoprotein (oxLDL) and amyloid-β by CD36 prime microglia for pro-inflammation phenotype by triggering the assembly of a heterotrimeric complex, which composed of CD36-TLR4-TLR6 and results in IL-1β transcription. ApoE bind to LRP1 and TREM2 on the microglial surface to inhibit microglial activation and reduce neuroinflammation. The phagocytosed lipid components can be hydrolyzed by PLA2 to release DHA and ARA. DHA and ARA can be oxidized to produce 4-HNE and 4-HHE, both of which can increase HO-1 expression and activate the NRF2 pathway, exerting anti-inflammatory effects
Fig. 4
Fig. 4
Role of microglia activation in SAE. Microglial activation is strongly associated with the pathogenesis of SAE, especially in BBB disruption, neuroinflammation and neurotransmitter disruption. Under the systemic inflammatory condition, changes in BBB permeability occur, trigger the infiltration of peripheral immune cells into the brain parenchyma, as well as the entry of peripheral inflammatory factors, including cytokines, complements, NO, and chemokines. These inflammatory factors enter the brain and activate microglia. After activation, microglia perpetuate further inflammatory response by releasing inflammatory factors and inducing (by the release of TNF-α IL-1α, and C1q) the transformation of astrocytes into a neurotoxic subtype. Meanwhile, the secretion of neurotransmitters and the expression of their receptors in microglia are changed after microglia activation, leading to the disruption of neurotransmitter in the brain, such as glutamate accumulation. Microglia release large amounts of glutamate after activation. Increased glutamate release and impaired glutamate reuptake lead to extracellular glutamate accumulation and neurotoxicity. All of these pathogeneses induced by microglia ultimately result in neuronal death and cognitive impairment

Similar articles

References

    1. 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;395:200–11. - PMC - PubMed
    1. Danielski LG, Giustina AD, Badawy M, Barichello T, Quevedo J, Dal-Pizzol F, Petronilho F. Brain barrier breakdown as a cause and consequence of neuroinflammation in Sepsis. Mol Neurobiol. 2018;55:1045–53. - PubMed
    1. Lelubre C, Vincent JL. Mechanisms and treatment of organ failure in sepsis. Nat Rev Nephrol. 2018;14:417–27. - PubMed
    1. Trzeciak A, Lerman YV, Kim TH, Kim MR, Mai N, Halterman MW, Kim M. Long-Term microgliosis driven by acute systemic inflammation. J Immunol. 2019;203:2979–89. - PMC - PubMed
    1. Tauber SC, Djukic M, Gossner J, Eiffert H, Bruck W, Nau R. Sepsis-associated encephalopathy and septic encephalitis: an update. Expert Rev Anti Infect Ther. 2021;19:215–31. - PubMed

LinkOut - more resources