Skip to main content

Daily Sepsis Research Analysis

3 papers

Mechanistic and clinical sepsis research converged on three fronts: a multi-omics/mechanistic study uncovered a PIK3C3–MAPK14 axis linking impaired autophagy to M1 macrophage polarization in sepsis-induced acute lung injury; an ICU cohort showed hyperdynamic left ventricular ejection fraction (EF ≥70%) strongly predicts 28-day mortality in septic shock; and a proteomics analysis proposed a compact two-protein panel (ANXA6 + FIBG) to predict sepsis among trauma-induced SIRS. Together, these studi

Summary

Mechanistic and clinical sepsis research converged on three fronts: a multi-omics/mechanistic study uncovered a PIK3C3–MAPK14 axis linking impaired autophagy to M1 macrophage polarization in sepsis-induced acute lung injury; an ICU cohort showed hyperdynamic left ventricular ejection fraction (EF ≥70%) strongly predicts 28-day mortality in septic shock; and a proteomics analysis proposed a compact two-protein panel (ANXA6 + FIBG) to predict sepsis among trauma-induced SIRS. Together, these studies advance pathophysiology, risk stratification, and early diagnosis.

Research Themes

  • Macrophage autophagy and polarization in sepsis-induced lung injury
  • Cardiac echocardiographic risk markers in septic shock
  • Proteomics-based early sepsis prediction in trauma-induced SIRS

Selected Articles

1. The PIK3C3/MAPK14 axis drives M1 polarization via autophagy Inhibition to exacerbate Sepsis-Induced acute lung injury.

68.5Level VBasic/Mechanistic ResearchScientific reports · 2025PMID: 41318541

Integrating transcriptomics, single-cell data, molecular docking, and wet-lab validation, the study identifies a PIK3C3–MAPK14 signaling axis that impairs autophagy and drives M1 macrophage polarization in sepsis-induced ALI. PIK3C3 downregulation upregulates MAPK14, reduces autophagic flux, and promotes pro-inflammatory phenotypes, positioning this axis as a druggable target.

Impact: This work offers a coherent mechanistic pathway connecting autophagy disruption to macrophage-driven lung injury in sepsis, advancing targetable biology beyond descriptive associations.

Clinical Implications: Although preclinical, modulating the PIK3C3–MAPK14 axis could underpin therapies aiming to restore autophagy and rebalance macrophage polarization in sepsis-induced lung injury.

Key Findings

  • MAPK14 was identified as a core ALI gene with increased expression localized to pro-inflammatory macrophages by single-cell analysis.
  • PIK3C3 downregulation increased MAPK14, impaired autophagic flux (LC3-II/I↓, TOM20↑, P62↑, HSP60↑), and promoted M1 polarization after LPS stimulation.
  • RNA pull-down captured a PIK3C3–MAPK14 complex; molecular docking showed high-affinity interaction (ΔG-bind ≈ −128 kJ/mol), suggesting a functional axis.

Methodological Strengths

  • Multi-omics integration with single-cell localization and machine-learning-guided gene prioritization
  • Orthogonal wet-lab validations (WB, qRT-PCR, flow cytometry, ELISA, RNA pull-down) supporting a mechanistic axis

Limitations

  • Predominantly in vitro without in vivo sepsis/ALI validation
  • Reliance on molecular docking and correlation analyses may not fully capture causal dynamics in complex in vivo contexts

Future Directions: Validate the axis in animal models of sepsis-induced lung injury and test pharmacologic modulators of MAPK14 or PIK3C3 to assess therapeutic efficacy and safety.

2. Hyperdynamic left ventricular ejection fraction as a predictor of mortality in intensive care unit patients with septic shock.

65.5Level IIICohortHeart & lung : the journal of critical care · 2025PMID: 41319356

In a 235-patient ICU cohort with septic shock, hyperdynamic EF (≥70%) was significantly associated with 28-day mortality (OR 4.822). Age, lower mean arterial pressure, higher SOFA scores, and elevated lactate independently predicted death; male sex had higher mortality. The study was preregistered and used standardized echocardiography.

Impact: Identifies a pragmatic echocardiographic metric (hyperdynamic EF) for early risk stratification in septic shock, linking cardiac function phenotype to mortality.

Clinical Implications: Incorporating EF ≥70% as a high-risk flag could prompt closer hemodynamic monitoring, refined fluid/vasopressor strategies, and earlier consultation for cardiovascular dysfunction in sepsis.

Key Findings

  • Hyperdynamic EF was more prevalent in non-survivors, strongly associated with 28-day mortality (OR 4.822, 95% CI 1.467–8.852).
  • Independent mortality predictors included older age, lower mean arterial pressure, higher SOFA scores, and elevated serum lactate.
  • Male patients exhibited significantly higher mortality; echocardiography was performed by accredited operators within a preregistered protocol (NCT06993948).

Methodological Strengths

  • Prospective daily SOFA assessment with standardized echocardiography by accredited clinicians
  • Multivariable modeling identifying independent predictors; preregistration at ClinicalTrials.gov

Limitations

  • Single-center observational design limits causal inference and generalizability
  • EF measurements in the context of vasopressors and dynamic preload may confound interpretation; diastolic metrics were not detailed

Future Directions: Multicenter validation and incorporation of comprehensive diastolic and deformation indices to evaluate whether hyperdynamic EF-guided management improves outcomes.

3. Identifying Predictive Biomarkers for Sepsis in Trauma-Induced Systemic Inflammatory Response Syndrome Using Proteomics Data.

59Level IIICase-controlThe Journal of surgical research · 2025PMID: 41317596

Retrospective analysis of plasma proteomics from 62 severe trauma patients identified 15 proteins distinguishing SIRS patients who developed sepsis versus those who did not. A minimal two-protein panel (ANXA6 + FIBG) achieved high predictive performance (AUC 0.9652), suggesting a feasible early risk stratification tool.

Impact: Proposes a compact biomarker panel with excellent discrimination to flag high-risk trauma patients before overt sepsis, potentially enabling earlier prevention and intervention.

Clinical Implications: If validated prospectively, an ANXA6+FIBG assay could guide targeted monitoring, antibiotic stewardship, and preemptive supportive care in trauma-induced SIRS.

Key Findings

  • Identified 15 differentially expressed proteins between SIRS patients who developed sepsis (n=15) and those who did not (n=23).
  • Top single markers showed strong discrimination: ANXA6 (AUC 0.881), FA12 (0.873), TRY2 (0.868), FIBG (0.759), DOPD (0.745).
  • A two-protein panel (ANXA6 + FIBG) balanced parsimony and performance with AUC 0.9652; a five-marker combination reached AUC 0.9913.

Methodological Strengths

  • Unbiased proteomics with ROC/AUC-based evaluation and model parsimony optimization
  • Clear case definition within trauma-induced SIRS enabling clinically relevant comparison

Limitations

  • Small, retrospective single-cohort analysis without external validation increases overfitting risk
  • Timing of sampling and calibration/clinical utility metrics (e.g., decision-curve analysis) not reported

Future Directions: Prospective multicenter validation, assay development with clinical lab platforms, and assessment of clinical impact via decision-curve and implementation studies.