Skip to main content
Daily Report

Daily Sepsis Research Analysis

01/11/2026
3 papers selected
31 analyzed

Analyzed 31 papers and selected 3 impactful papers.

Summary

Analyzed 31 papers and selected 3 impactful articles.

Selected Articles

1. HDAC3 Mediates Hippocampal Microglial Pyroptosis Via the STING/NLRP3 Pathway and Contributes To Cognitive Impairment in Sepsis-Associated Encephalopathy.

84Level VCase-control
Inflammation · 2026PMID: 41518423

Using CLP-induced sepsis models with microglia-specific genetic manipulation and a selective HDAC3 inhibitor, the study shows that microglial HDAC3 activates pyroptosis via STING/NLRP3, driving oxidative stress, neural dysfunction, and cognitive deficits. Pharmacologic inhibition with RGFP966 mitigated these effects, positioning HDAC3 as a therapeutic target in sepsis-associated encephalopathy.

Impact: Identifies a cell type–specific epigenetic driver of neuroinflammation and cognitive impairment in sepsis, supported by both genetic and pharmacologic interventions. This mechanistic clarity opens a tractable pathway for therapeutic development in SAE.

Clinical Implications: While preclinical, targeting microglial HDAC3 could inform future trials of HDAC3 inhibitors or epigenetic modulators to prevent or treat sepsis-associated encephalopathy, particularly if effective in delayed treatment windows.

Key Findings

  • Microglial HDAC3 activates pyroptosis via STING/NLRP3, increasing oxidative stress and impairing neural activity in CLP-induced sepsis.
  • Microglia-specific overexpression of HDAC3 reproduces cognitive and pathological deficits characteristic of SAE.
  • The selective HDAC3 inhibitor RGFP966 attenuates pyroptosis, inflammatory signaling, and cognitive deficits when administered for 14 days.

Methodological Strengths

  • Combined cell type–specific genetic manipulation with selective pharmacologic inhibition to establish causality.
  • Use of a clinically relevant CLP sepsis model with behavioral and molecular readouts.

Limitations

  • Preemptive dosing started 2 hours before CLP may overestimate translatability to clinical settings.
  • Sample size and sex balance were not specified; human validation is lacking.

Future Directions: Test delayed/post-insult HDAC3 inhibition, validate HDAC3/STING/NLRP3 signaling in human biospecimens, and assess target engagement and cognitive outcomes in early phase clinical trials.

Microglial pyroptosis-mediated neuroinflammation emerges as a critical pathogenic mechanism underlying sepsis-associated encephalopathy (SAE). Epigenetic modifications, especially histone acetylation states, exert fundamental regulatory effects on microglial pyroptosis. Among these, histone deacetylase 3 (HDAC3) has been identified as a central epigenetic regulator orchestrating these processes. This study investigates the functional role of HDAC3 in microglial pyroptosis and its underlying mechanisms contributing to SAE-related cognitive impairment. To explore this, male C57BL/6 mice subjected to cecal ligation and puncture (CLP) served as the SAE model. We employed RGFP966, a selective HDAC3 inhibitor, administered at 20 mg/kg/day via daily subcutaneous injections for 14 days starting 2 h prior to CLP surgery. To specifically examine HDAC3's role in microglia, we bilaterally injected recombinant adeno-associated virus (rAAV)-expressing rEGFP under the control of a DIO promoter into the hippocampus of Cx3cr1-Cre mice to achieve selective overexpression. Our data demonstrate that HDAC3 in microglia activates pyroptosis through the STING/NLRP3 pathway, exacerbating oxidative stress responses and impairing neural activity, ultimately leading to cognitive deficits in SAE. Furthermore, HDAC3 overexpression in microglia recapitulates these pathological changes, underscoring its central role in driving disease progression. Conversely, RGFP966 treatment effectively attenuates these abnormalities by suppressing HDAC3 expression and downstream inflammatory pathways. These findings highlight the therapeutic potential of targeting microglial HDAC3 to mitigate neuroinflammation and cognitive dysfunction in SAE, offering a novel direction for future clinical applications.

2. Programmed death-ligand 1 (PD-L1) Modulates Chemokine Production Via the TLR4/TRAF6 Signaling Axis During LPS + IFN-γ-Induced Endotoxemia-mimicked Sepsis.

78.5Level VCase-control
Inflammation · 2026PMID: 41518433

Integrating clinical data mining with RNA-seq and protein interaction analyses, the authors reveal a macrophage-intrinsic, proinflammatory role of PD-L1 that increases CCL8 and CXCL9 via the TLR4/TRAF6 axis under septic conditions. This reframes PD-L1 from a purely inhibitory checkpoint to a context-dependent amplifier of innate chemokine responses.

Impact: Defines a novel, macrophage-centered PD-L1 signaling mechanism that could be druggable and shifts the paradigm of immune checkpoint biology in sepsis.

Clinical Implications: Therapies modulating PD-L1 or the TLR4/TRAF6 axis may need to consider macrophage-intrinsic proinflammatory functions to avoid exacerbating chemokine-driven tissue injury in sepsis.

Key Findings

  • Macrophage PD-L1 upregulates CCL8 and CXCL9 under septic-like stimulation (LPS + IFN-γ) per RNA-seq and clinical data integration.
  • IP–MS, molecular docking, and mutagenesis support PD-L1 regulation of chemokines via the TLR4/TRAF6 signaling axis.
  • Findings suggest PD-L1 has context-dependent proinflammatory roles beyond PD-1–mediated T-cell suppression.

Methodological Strengths

  • Multi-omics approach combining clinical dataset mining, RNA-seq, and protein interaction assays.
  • Mechanistic triangulation with IP–MS, docking, and mutagenesis increases causal plausibility.

Limitations

  • Limited in vivo validation of therapeutic modulation and absence of survival or organ-injury outcomes.
  • Endotoxemia-mimic may not capture full complexity of polymicrobial sepsis.

Future Directions: Test macrophage-targeted PD-L1 modulation in vivo in CLP models with organ and survival endpoints; map PD-L1 interactome under sepsis to refine drug targets.

As a pivotal immune checkpoint molecule, programmed death-ligand 1 (PD-L1) is anchored primarily on the membrane surface of immune cells, where it exerts immunosuppressive effects, thereby facilitating tumor immune evasion. Macrophages, which serve as essential sentinels of the innate immune system, play dual regulatory roles in inflammatory pathologies, particularly during sepsis progression. While their secreted chemokines mediate inflammatory cell recruitment for pathogen clearance, excessive chemokine production can paradoxically induce organ damage and immune cell exhaustion, necessitating precise regulatory mechanisms. Conventional understanding suggests that PD-L1 on macrophages engages with programmed cell death protein 1 (PD-1) on T lymphocytes to suppress T-cell proliferation, cytokine secretion (e.g., IFN-γ and IL-2), and cytotoxic functions, thereby negatively modulating adaptive immunity. However, emerging evidence also suggests that PD-L1 has context-dependent proinflammatory functions. Given this context, we hypothesized that macrophage-intrinsic PD-L1 plays a poorly understood role in directly regulating chemokine production during sepsis. Our integrative analysis incorporating clinical database mining and RNA sequencing (RNA-seq) revealed a less defined proinflammatory property of PD-L1 under septic conditions-an ability to increase CCL8 and CXCL9 chemokine expression in inflammatory macrophages. Through combinatorial approaches, including immunoprecipitation‒mass spectrometry (IP‒MS), molecular docking, and site-directed mutagenesis, we preliminarily elucidated that PD-L1 likely governs the chemotactic mediators CCL8 and CXCL9 via the TLR4/TRAF6 signaling axis. These findings collectively establish the previously unappreciated regulatory capacity of macrophage-intrinsic PD-L1 in chemokine modulation during sepsis, potentially informing the development of innovative therapeutic strategies targeting immune dysregulation in critical care settings.

3. Association of lactate to albumin ratio with the severity and prognosis of patients with sepsis admitted to the emergency intensive care unit: A prospective cohort study.

69.5Level IICohort
Medicine · 2026PMID: 41517778

In a single-center prospective cohort of adult sepsis patients, higher lactate-to-albumin ratio independently predicted septic shock, SA-AKI, and 28-day mortality. Discrimination was strongest for mortality (AUC 0.863) with clinically actionable cutoffs around 0.10.

Impact: Provides a simple, early, and readily available biomarker ratio that stratifies risk for key sepsis outcomes with strong discrimination, supporting bedside implementation.

Clinical Implications: Incorporating LAR into triage and early management may improve identification of patients at high risk for shock, SA-AKI, and death, informing monitoring intensity and resource allocation.

Key Findings

  • LAR independently predicted septic shock, SA-AKI, and 28-day mortality in multivariable analyses.
  • Risk increased progressively across higher LAR quartiles.
  • ROC performance: AUC 0.705 for shock (cutoff 0.106), 0.762 for SA-AKI (cutoff 0.097), and 0.863 for 28-day mortality (cutoff 0.098).

Methodological Strengths

  • Prospective design with predefined outcomes and multivariable logistic modeling.
  • ROC analyses provide clinically actionable cutoffs with confidence intervals.

Limitations

  • Single-center study limits generalizability; sample size not reported in the abstract.
  • External validation and assessment of additive value over established scores (e.g., SOFA) are needed.

Future Directions: Multicenter validation, integration with early warning systems/EHRs, and impact evaluations on clinical decision-making and outcomes.

The aim of this study is to evaluate the prognostic value of the lactate to albumin ratio (LAR) in predicting morbidity, acute kidney injury associated with sepsis (SA-AKI) and mortality in sepsis patients. This was a single-center prospective cohort study. All adult patients above the age of 18 with a diagnosis of sepsis who presented between January 1, 2024, and June 1, 2025, were included. The primary outcome was 28-day mortality, septic shock and SA-AKI. The patients were divided into non-shock group and shock group, non-acute kidney injury group and acute kidney injury group, survival group and death group for comparison. Group differences were significant based on LAR quartiles. Univariate and Logistic regression analyses showed that LAR was an independent risk factor for septic shock, septic acute kidney injury, and 28-day mortality. Elevated LAR stratification was associated with a significantly increased risk of shock, acute kidney injury, and 28-day mortality. LAR remained an independent risk factor for shock, acute kidney injury, and death when used as a continuous variable. Receiver operating characteristic (ROC) curve showed that the area under the curve (AUC) of septic shock was 0.705 (95% CI = 0.611-0.800) when the cutoff value of LAR was 0.106. When the cutoff value was 0.097, the AUC of sepsis-induced acute kidney injury was 0.762 (95% CI = 0.669-0.854). When the cutoff value was 0.098, the AUC of 28-day mortality was 0.863 (95% CI = 0.796-0.931). As the quartile of LAR layers increases, the risks of septic shock, SA-AKI and death gradually increased significantly. Early LAR has a certain predictive value for septic shock, SA-AKI and death outcomes.