1
|
Cusack RAF, Rodríguez A, Cantan B, Garduno A, Connolly E, Zilahi G, Coakley JD, Martin-Loeches I. Microcirculation properties of 20 % albumin in sepsis; a randomised controlled trial. J Crit Care 2025; 87:155039. [PMID: 40020556 DOI: 10.1016/j.jcrc.2025.155039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Sepsis and septic shock are associated with microcirculatory dysfunction, significantly impacting patient outcomes. This study aimed to evaluate the effects of a 20 % albumin bolus on microcirculation compared to crystalloid resuscitation in fluid-responsive patients (ClinicalTrials.govID:NCT05357339). METHODS We conducted a single-centre randomised controlled trial, enrolling 103 patients (Albumin n = 52, Control n = 51). Fluid responsiveness was assessed, and fluid was administered in boluses of 100 ml to clinical effect. Microcirculation was measured using the Side stream Dark Field camera and AVA 4.3 software. Baseline characteristics, macrohaemodynamics, and microcirculation parameters were recorded. Three patients were excluded from analysis. RESULTS The final cohort comprised 100 patients, 35 (35 %) females with a mean age of 58 years (range: 18-86). The mean APACHE score was 28 (range: 7-45), and the mean SOFA score was 9.4 (range: 1-17). No significant differences in APACHE (26.24 vs. 29.4, p = 0.069) or SOFA (9.08 vs. 9.78, p = 0.32) scores were found for albumin and control group respectively. The albumin group had worse microcirculation at baseline but demonstrated significant improvements in microvascular density and activity at 15 min and 60 min (p < 0.005), while the control group exhibited no significant changes. Additionally, both groups were fluid responsive, with a mean pulse pressure variability of 17 % at admission. There were no significant differences in overall fluid balances, vasopressor days, length of ICU stay, or mortality between groups. CONCLUSION This study demonstrates that a 20 % albumin bolus significantly enhances microcirculation in fluid-responsive patients with septic shock. These findings underscore the potential benefits of targeted microcirculation therapy in critically ill patients.
Collapse
Affiliation(s)
- Rachael A F Cusack
- Trinity College Dublin, School of Medicine, College Green, Dublin, Ireland; Intensive Care Medicine Department, St. James's Hospital, James's Street, Dublin, Ireland
| | - Alejandro Rodríguez
- Critical Care Department, Hospital Universitario Joan XXIII de Tarragona, Rovira & Virgili University, Tarragona, Spain
| | - Ben Cantan
- Intensive Care Medicine Department, St. James's Hospital, James's Street, Dublin, Ireland
| | - Alexis Garduno
- Trinity College Dublin, School of Medicine, College Green, Dublin, Ireland
| | - Elizabeth Connolly
- Intensive Care Medicine Department, St. James's Hospital, James's Street, Dublin, Ireland
| | - Gabor Zilahi
- Intensive Care Medicine Department, St. James's Hospital, James's Street, Dublin, Ireland
| | - John Davis Coakley
- Intensive Care Medicine Department, St. James's Hospital, James's Street, Dublin, Ireland
| | - Ignacio Martin-Loeches
- Trinity College Dublin, School of Medicine, College Green, Dublin, Ireland; Intensive Care Medicine Department, St. James's Hospital, James's Street, Dublin, Ireland; Hospital Clinic, Universitat de Barcelona, IDIBAPS, CIBERES, Barcelona, Spain.
| |
Collapse
|
2
|
Dasgupta A, Jones TK, Giannini H, Bennett R, Emre G, Ittner CAG, Turner A, Esperanza M, Housel K, Miano T, Erlich M, Anderson BJ, Shashaty MGS, Meyer NJ, Reilly JP. Identifying a unique signature of sepsis in patients with pre-existing cirrhosis. Crit Care 2025; 29:199. [PMID: 40390062 DOI: 10.1186/s13054-025-05423-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/19/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND The pre-existing diagnosis of cirrhosis is a complicating factor in the progression and prognosis of sepsis; however, the unique epidemiology, sepsis characteristics, and underlying mechanisms of immune dysregulation in sepsis among patients with cirrhosis remain incompletely understood. Our primary objective was to identify clinical outcomes and biological characteristics that differ between patients with and without cirrhosis among critically ill patients with sepsis. METHODS We analyzed data from a prospective cohort of critically ill patients presenting to single center with sepsis. Subjects were followed for 6 days for the development of acute respiratory distress syndrome (ARDS) and acute kidney injury (AKI), and 30 days for mortality. Inflammatory, endothelial, and coagulopathic proteins were measured in plasma collected at ICU admission in a subset of patients. We determined associations of cirrhosis with outcomes using multivariable logistic regression adjusting for pre-specified confounders. We tested differences in plasma protein levels by cirrhosis diagnosis using the Wilcoxon Rank-sum test. RESULTS We enrolled 2962 subjects, 371 (13%) of whom had a pre-existing diagnosis of cirrhosis. Patients with cirrhosis had higher severity of illness scores, were more likely to have an abdominal source of sepsis, and had more significant clinically measured coagulation abnormalities relative to patients without cirrhosis. In multivariate analysis, cirrhosis was associated with higher AKI risk (adjusted OR 1.65; 95% CI 1.21 to 2.26; P = 0.002), and 30-day mortality (adjusted OR 1.38; 95% CI 1.05 to 1.82; P = 0.022). There was no significant difference in risk for ARDS (adjusted OR 1.02; 95% CI 0.69 to 1.50; P = 0.92). Cirrhosis was associated with higher plasma levels of angiopoietin-2 (P < 0.001), von Willebrand factor (P < 0.001), and soluble thrombomodulin (P < 0.001), as well as lower levels of interleukin (IL)-10 (P < 0.001), IL-1β (P = 0.008), and IL-1RA (P = 0.036). There were no significant differences in levels of IL-6 (P = 0.30). CONCLUSIONS We identified associations between pre-existing cirrhosis and endothelial injury, AKI, and mortality in sepsis. Patients with pre-existing cirrhosis who develop sepsis may display a unique phenotype of endothelial dysfunction that requires unique targeted approaches.
Collapse
Affiliation(s)
- Anushka Dasgupta
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Tiffanie K Jones
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Translational Lung Biology, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather Giannini
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Translational Lung Biology, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Bennett
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Translational Lung Biology, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gulus Emre
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline A G Ittner
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Alexandra Turner
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Mika Esperanza
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Kaitlyn Housel
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Todd Miano
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Erlich
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Brian J Anderson
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Michael G S Shashaty
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Translational Lung Biology, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nuala J Meyer
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Translational Lung Biology, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John P Reilly
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
- Center for Translational Lung Biology, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Puthiyottil S, Skaria T. Paracrine signaling mediators of vascular endothelial barrier dysfunction in sepsis: implications for therapeutic targeting. Tissue Barriers 2025:2503523. [PMID: 40376886 DOI: 10.1080/21688370.2025.2503523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/25/2025] [Accepted: 04/30/2025] [Indexed: 05/18/2025] Open
Abstract
Vascular endothelial barrier disruption is a critical determinant of morbidity and mortality in sepsis. Whole blood represents a key source of paracrine signaling molecules inducing vascular endothelial barrier disruption in sepsis. This study analyzes whole-genome transcriptome data from sepsis patients' whole blood available in the NCBI GEO database to identify paracrine mediators of vascular endothelial barrier dysfunction, uncovering novel insights that may guide drug repositioning strategies. This study identifies the regulated expression of paracrine signaling molecules TFPI, MMP9, PROS1, JAG1, S1PR1, and S1PR5 which either disrupt or protect vascular endothelial barrier function in sepsis and could serve as potential targets for repositioning existing drugs. Specifically, TFPI (barrier protective), MMP9 (barrier destructive), PROS1 (barrier protective), and JAG1 (barrier destructive) are upregulated, while S1PR1 (barrier protective) and S1PR5 (barrier protective) are downregulated. Our observations highlight the importance of considering both protective and disruptive mediators in the development of therapeutic strategies to restore endothelial barrier integrity in septic patients. Identifying TFPI, MMP9, PROS1, JAG1, S1PR1, and S1PR5 as druggable paracrine regulators of vascular endothelial barrier function in sepsis could pave the way for precision medicine approaches, enabling personalized treatments that target specific mediators of endothelial barrier disruption to improve patient outcomes in sepsis.
Collapse
Affiliation(s)
- Shahid Puthiyottil
- Department of Bioscience and Engineering, National Institute of Technology Calicut, Calicut, India
| | - Tom Skaria
- Department of Bioscience and Engineering, National Institute of Technology Calicut, Calicut, India
| |
Collapse
|
4
|
He J, Shao Z, Li Z, He Y, Zhang J, Zhong H, Li J, Liu Q, Shao Y. Mechanistic Insights into HOTAIR-Driven ADAM17/NF-Κb Activation and Endothelial Dysfunction in LPS-Challenged HUVECs. Immunol Invest 2025:1-27. [PMID: 40366882 DOI: 10.1080/08820139.2025.2503174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
INTRODUCTION HOX transcript antisense intergenic RNA (HOTAIR) has been implicated in inflammation and vascular pathology, but its role in regulation of ADAM17 and sepsis-induced endothelial injury remains unclear. METHODS LPS-treated human umbilical vein endothelial cells (HUVECs) modeled sepsis-induced endothelial injury, which were assessed via qRT-PCR, western blot and immunofluorescence. HOTAIR-knockout mice were treated with cecal ligation and perforation to establish sepsis model. RESULTS LPS-stimulation increased expression of HOTAIR and ADAM17 and decreased miR-326 levels in HUVECs. HOTAIR-knockdown by antisense oligonucleotides (ASOs) decreased ADAM17, TNF-α production and NF-κB activities; it also alleviated endothelial inflammation, VE-cadherin integrity damage, apoptosis and barrier dysfunction, while miR-326 inhibition reversed these effects. MiR-326 inhibited TNF-α/NF-κB via targeting ADAM17. Further experiments demonstrated recombinant TNF-α reversed the inhibitory effect of HOTAIR-ASOs on LPS-triggered TNF-α/NF-κB activation and downstream endothelial injury, which were further mitigated by NF-κB or p38 MAPK inhibitors. In-vivo experiments in HOTAIR-knockout mice confirmed the role of HOTAIR/miR-326/ADAM17 in regulating NF-κB and p38 MAPK inflammation, with improved lung injury and survival following sepsis. DISCUSSION The HOTAIR/miR-326/ADAM17 axis is a key regulator of inflammation, endothelial injury and barrier dysfunction during sepsis via modulation of TNF-α/NF-κB signaling, providing new insights into the mechanisms underlying endothelial injury in sepsis.
Collapse
Affiliation(s)
- Junbing He
- The Department of Emergency, The First Affiliated Hospital, Jinan University, Guangzhou, GD, China
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, GD, China
| | - Zixuan Shao
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| | - Zhuoji Li
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| | - Yufu He
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, GD, China
| | - Jingqi Zhang
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| | - Haotian Zhong
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| | - Jiekai Li
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, GD, China
| | - Qinghua Liu
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, GD, China
- The Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GD, China
| | - Yiming Shao
- The Department of Emergency, The First Affiliated Hospital, Jinan University, Guangzhou, GD, China
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| |
Collapse
|
5
|
Feng A, Liang Y, Fu P, Dong Y, Black SM, Wang T. Endotoxin-induced m6A RNA methylation landscape in lung endothelial cells: role of METTL3 in regulating inflammation and injury during acute lung injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167907. [PMID: 40379220 DOI: 10.1016/j.bbadis.2025.167907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/31/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Acute Lung Injury (ALI) involves diffuse alveolar damage, neutrophil infiltration, and pulmonary edema, with unacceptable mortality. Bacterial lipopolysaccharide (LPS) activates inflammatory pathways in ALI, which are then regulated by transcriptional and post-transcriptional pathways to affect gene expression. RNA methylation, N6-methyladenosine, is the main m6A mRNA modification that controls the expression of various genes in different environments. There are very few facts about LPS's effect on m6A RNA methylation. This study will explore the m6A RNA methylation landscape in lung endothelial cells (ECs) to understand its role in lung inflammation. In this study, lung endothelial cells were treated with LPS, and the dynamics of mRNA m6A methylation were examined through m6A-methylated RNA sequencing. RNA abundance was measured with RNA-seq, and global protein expression and m6A-binding proteins were identified using mass spectrometry (MS). Following LPS treatment, global m6A methylation levels increased along with the upregulation and nuclear translocation of METTL3 protein, while demethylase activity remained unchanged. METTL3 drove LPS-induced m6A methylation and endothelial injury, as shown by selective METTL3 siRNA and the inhibitor STM2457. MeRIP-seq analyses revealed increased m6A sites near the 5' UTR in LPS-treated cells, with m6A methylation correlating positively with gene expression. The metabolic and apoptosis pathways were shown to be more enriched in different types of methylated exons. METTL3-mediated m6A methylation targeted inflammatory genes, enhancing protein expression in chemokine signaling and MAPK pathways. STM2457 effectively mitigated LPS- or CLP-induced experimental ALI. According to this paper, LPS-mediated m6A RNA methylation is described in terms of genomic structure. Modulation of m6A methylation exerts influence over LPS-mediated endothelial gene expression and the ensuing inflammatory response.
Collapse
Affiliation(s)
- Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Yishu Dong
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
6
|
de Jesus Fernandes Martins Lima JF, Moreira da Silva TE, Dos Santos Lopes AC, Ferreira Freire VA, Barros-Pinheiro M, Alpoim PN. Soluble thrombomodulin in preeclampsia: Systematic review and meta-analysis. Clin Chim Acta 2025; 574:120323. [PMID: 40320160 DOI: 10.1016/j.cca.2025.120323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/12/2025]
Abstract
Preeclampsia (PE) is a severe pregnancy complication associated with endothelial dysfunction. Soluble thrombomodulin (sTM) is a biomarker of endothelial injury, but its role in PE remains unclear. Although previous studies suggest elevated sTM levels in PE, inconsistencies in study designs and diagnostic criteria have led to conflicting findings. Our study addresses this gap through a systematic review and meta-analysis using standardized effect sizes and predefined severity groups to clarify the association between sTM and PE. This review followed the Cochrane Handbook and PRISMA guidelines and was registered in PROSPERO (CRD42023456919). A comprehensive search was conducted in MEDLINE/PubMed, EMBASE, and Web of Science for observational studies evaluating sTM levels in PE and normotensive controls. Eligible studies included case-control and cohort designs measuring sTM in plasma or serum. Two reviewers independently conducted data extraction and risk of bias assessment (Newcastle-Ottawa Scale). A random-effects meta-analysis estimated the Standardized Mean Difference (SMD) and 95 % confidence intervals (CI). Of 285 screened studies, 26 met inclusion criteria, and 20 were included in the meta-analysis. sTM levels were significantly higher in PE (SMD = 0.91, 95 % CI 0.3 to 1.53; I2 = 94 %) with greater elevation in severe PE (SMD: 0.86, 95 % CI 0.55 to 1.16; I2 = 50 %) than mild PE (SMD: 0.57, 95 % CI 0.08 to 1.06; I2 = 82 %). High heterogeneity was observed, likely due to variations in inclusion criteria, measurement techniques, and diagnostic definitions. These findings support the potential of sTM as a biomarker for assessing PE severity. However, its clinical use remains limited by methodological heterogeneity and lack of standardized cutoff values. Further prospective studies are needed to validate its diagnostic and prognostic value and to enable its integration into clinical practice.
Collapse
Affiliation(s)
| | - Thaíse Emilia Moreira da Silva
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil
| | - Ana Cristina Dos Santos Lopes
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil
| | - Victor Antonio Ferreira Freire
- Universidade Federal de São João del-Rei, Campus Centro-Oeste Dona Lindu. Divinópolis, MG, Brazil; Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Melina Barros-Pinheiro
- Universidade Federal de São João del-Rei, Campus Centro-Oeste Dona Lindu. Divinópolis, MG, Brazil
| | - Patrícia Nessralla Alpoim
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brazil.
| |
Collapse
|
7
|
Hong H, Wu Y, Li Y, Han Y, Cao X, Wu VWY, Chan TTH, Zhou J, Cao Q, Lui KO, Wong CK, Dai Z, Tian XY. Endothelial PPARδ Ablation Exacerbates Vascular Hyperpermeability via STAT1/CXCL10 Signaling in Acute Lung Injury. Circ Res 2025; 136:735-751. [PMID: 39996324 DOI: 10.1161/circresaha.124.325855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/16/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Vascular hyperpermeability is one of the hallmarks of acute lung injury, contributing to excessive inflammation and respiratory failure. The PPARδ (peroxisome proliferator-activated receptor delta) is an anti-inflammatory transcription factor, although its role in endothelial barrier function remains unclear. Here, we studied the essential role of PPARδ in maintaining vascular endothelial barrier integrity during lung inflammation and investigated the underlying mechanisms. METHODS Endothelial cell (EC)-selective PPARδ knockout mice (PpardEC-KO) and littermate control mice (PpardEC-WT) received lipopolysaccharide injection to induce acute lung injury. Lung inflammation, pulmonary vascular leakage, and mouse mortality were monitored. Single-cell RNA sequencing was performed on sorted mouse lung ECs. RESULTS PpardEC-KO mice exhibited aggravated lung inflammation, characterized by increased leukocyte infiltration, elevated production of proinflammatory cytokines, and higher mortality rates. The enhanced inflammatory responses were associated with increased protein leakage, interstitial edema, and impaired endothelial barrier structure, leading to vascular hyperpermeability in PpardEC-KO mice. Mechanistically, with single-cell RNA sequencing, we identified the emergence of an interferon-activated capillary EC population marked by CXCL10 (C-X-C motif chemokine 10) expression following lipopolysaccharide challenge. PPARδ silencing significantly increased CXCL10 expression in ECs through activating STAT1 (Signal transducer and activator of transcription 1). Notably, CXCL10 treatment induced degradation of tight junction proteins ZO-1 (zonula occludens protein 1) and claudin-5 through the ubiquitin-proteasome system, disrupting membrane junction continuity in ECs. Administration of anti-CXCL10 antibody or CXCL10 receptor antagonist AMG487 suppressed both lipopolysaccharide-induced lung inflammation and vascular leakage in PpardEC-KO mice. CONCLUSIONS These results highlighted a novel anti-inflammatory role of PPARδ in ECs by suppressing CXCL10-mediating vascular hyperpermeability. Targeting the CXCL10 signaling shows therapeutic potential against vascular injury in acute lung injury.
Collapse
Affiliation(s)
- Huiling Hong
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Yalan Wu
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, China (Y.W.)
| | - Yangxian Li
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Yumeng Han
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Xiaoyun Cao
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Vivian Wei Yan Wu
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Thomas Ting Hei Chan
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Jingying Zhou
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Qin Cao
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Kathy O Lui
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Chun-Kwok Wong
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Zhiyu Dai
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona (Z.D.)
| | - Xiao Yu Tian
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| |
Collapse
|
8
|
Musafiri S, Siddig EE, Nkuranga JB, Rukundo A, Mpunga T, Sendegeya A, Twagirumugabe T, Ahmed A, Muvunyi CM. Emerging Strategies and Progress in the Medical Management of Marburg Virus Disease. Pathogens 2025; 14:322. [PMID: 40333077 PMCID: PMC12030108 DOI: 10.3390/pathogens14040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 05/09/2025] Open
Abstract
During the current outbreak of Marburg virus disease (MVD) in Rwanda, we synthesized evidence from the literature to improve case management. Accordingly, experimental treatment was offered to patients under close follow-up. Remdesivir alone or in combination with monoclonal antibody treatment (MBP091) complemented with supportive care has improved the clinical outcomes of patients. Additionally, we have identified several experimental therapies currently under investigation, including antiviral drugs such as favipiravir, galidesivir, obeldesivir, and remdesivir, along with monoclonal and polyclonal antibodies (e.g., polyclonal IgG, monoclonal antibody MR-78-N; MR82-N; MR191-N; monoclonal antibodies MR186-YTE and MBP091). Furthermore, substantial progress is being made in vaccine development, with promising candidates including adenovirus-vectored vaccines, DNA vaccines, and the recombinant vesicular stomatitis virus (rVSV) vaccine. Moreover, innovative preventive and treatment strategies-such as synthetic hormones like estradiol benzoate, small interfering RNA (siRNA), interferon-β therapy, and phosphorodiamidate morpholino oligomers-are emerging as potential options for MVD management. Further investment is needed to accelerate research and optimize these therapeutics and preventive modalities. Additional epidemiological, preclinical, and clinical studies are warranted to generate the evidence required to inform policymaking, resource mobilization, and the implementation of cost-effective interventions for the prevention, control, and treatment of MVD.
Collapse
Affiliation(s)
- Sanctus Musafiri
- University Teaching Hospital of Kigali (CHUK), Kigali KN 4 Ave, Rwanda
- School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Kigali 3900, Rwanda
| | | | | | - Athanase Rukundo
- Department of Clinical Service, Ministry of Health, Kigali 84, Rwanda
| | - Tharcisse Mpunga
- University Teaching Hospital of Kigali (CHUK), Kigali KN 4 Ave, Rwanda
| | | | | | - Ayman Ahmed
- Rwanda Biomedical Centre, Kigali 7162, Rwanda
- Pan-Africa One Health Institute (PAOHI), Kigali 11KG ST203, Rwanda
| | | |
Collapse
|
9
|
Zeng QB, Peng EL, Zhou Y, Lin QW, Zhong LC, He LP, Zhang NQ, Song JC. Explainable machine learning model for predicting septic shock in critically sepsis patients based on coagulation indexes: A multicenter cohort study. Chin J Traumatol 2025:S1008-1275(25)00032-X. [PMID: 40246624 DOI: 10.1016/j.cjtee.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/19/2024] [Accepted: 08/23/2024] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Septic shock is associated with high mortality and poor outcomes among sepsis patients with coagulopathy. Although traditional statistical methods or machine learning (ML) algorithms have been proposed to predict septic shock, these potential approaches have never been systematically compared. The present work aimed to develop and compare models to predict septic shock among patients with sepsis. METHODS It is a retrospective cohort study based on 484 patients with sepsis who were admitted to our intensive care units between May 2018 and November 2022. Patients from the 908th Hospital of Chinese PLA Logistical Support Force and Nanchang Hongdu Hospital of Traditional Chinese Medicine were respectively allocated to training (n=311) and validation (n=173) sets. All clinical and laboratory data of sepsis patients characterized by comprehensive coagulation indexes were collected. We developed 5 models based on ML algorithms and 1 model based on a traditional statistical method to predict septic shock in the training cohort. The performance of all models was assessed using the area under the receiver operating characteristic curve and calibration plots. Decision curve analysis was used to evaluate the net benefit of the models. The validation set was applied to verify the predictive accuracy of the models. This study also used SHapley Additive exPlanations method to assess variable importance and explain the prediction made by a ML algorithm. RESULTS Among all patients, 37.2% experienced septic shock. The characteristic curves of the 6 models ranged from 0.833 to 0.962 and 0.630 to 0.744 in the training and validation sets, respectively. The model with the best prediction performance was based on the support vector machine (SVM) algorithm, which was constructed by age, tissue plasminogen activator-inhibitor complex, prothrombin time, international normalized ratio, white blood cells, and platelet counts. The SVM model showed good calibration and discrimination and a greater net benefit in decision curve analysis. CONCLUSION The SVM algorithm may be superior to other ML and traditional statistical algorithms for predicting septic shock. Physicians can better understand the reliability of the predictive model by SHapley Additive exPlanations value analysis.
Collapse
Affiliation(s)
- Qing-Bo Zeng
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Intensive Care Unit, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, 330038, China
| | - En-Lan Peng
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Ye Zhou
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Qing-Wei Lin
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Lin-Cui Zhong
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Long-Ping He
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Nian-Qing Zhang
- Intensive Care Unit, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, 330038, China
| | - Jing-Chun Song
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China.
| |
Collapse
|
10
|
Fu Y, Gong T, Loughran PA, Li Y, Billiar TR, Liu Y, Wen Z, Fan J. Roles of TLR4 in macrophage immunity and macrophage-pulmonary vascular/lymphatic endothelial cell interactions in sepsis. Commun Biol 2025; 8:469. [PMID: 40119011 PMCID: PMC11928643 DOI: 10.1038/s42003-025-07921-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
In sepsis, acute lung injury (ALI) is a severe complication and a leading cause of death, involving complex mechanisms that include cellular and molecular interactions between immune and lung parenchymal cells. In recent decades, the role of Toll-like receptor 4 (TLR4) in mediating infection-induced inflammation has been extensively studied. However, how TLR4 facilitates interactions between innate immune cells and lung parenchymal cells in sepsis remains to be fully understood. This study aims to explore the role of TLR4 in regulating macrophage immunity and metabolism in greater depth. It also seeks to reveal how changes in these processes affect the interaction between macrophages and both pulmonary endothelial cells (ECs) and lymphatic endothelial cells (LECs). Using TLR4 knockout mice and the combined approaches of single-cell RNA sequencing and experimental validation, we demonstrate that in sepsis, TLR4-deficient macrophages upregulate Abca1, enhance cholesterol efflux, and reduce glycolysis, promoting M2 polarization and attenuating inflammation. These metabolic and phenotypic shifts significantly affect their interactions with pulmonary ECs and LECs. Mechanistically, we uncovered that TLR4 operates through multiple pathways in endothelial dysfunction: macrophage TLR4 mediates inflammatory damage to ECs/LECs, while endothelial TLR4 both directly sensitizes cells to lipopolysaccharide-induced injury and determines their susceptibility to macrophage-derived inflammatory signals. These findings reveal the complex role of TLR4 in orchestrating both immune-mediated and direct endothelial responses during sepsis-induced ALI, supporting that targeting TLR4 on multiple cell populations may present an effective therapeutic strategy.
Collapse
Affiliation(s)
- Yu Fu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Ting Gong
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, Shenzhen, 518110, China
| | - Patricia A Loughran
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Yuehua Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, Shenzhen, 518110, China
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
11
|
Cao X, Ma R, Wang Y, Huang Y, You K, Zhang L, Li H, Feng G, Chen T, Wang D, Sun K, Fang H, Shen X. Paeoniflorin protects the vascular endothelial barrier in mice with sepsis by activating RXRα signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156384. [PMID: 39826282 DOI: 10.1016/j.phymed.2025.156384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Sepsis is a life-threatening condition characterized by organ dysfunction resulting from the body's aberrant response to infection. A primary indicator of early sepsis is vascular leakage due to endothelial injury. The immunomodulatory effects of paeoniflorin are well established. However, its effect on vascular endothelial injury in sepsis remains to be verified. METHODS The sepsis model was established by cecal ligation and puncture (CLP), along with simultaneous administration of paeoniflorin. The therapeutic effectiveness of paeoniflorin was evaluated by assessing the survival rate, the bacterial load in blood and the histopathological lung tissue injury. The pulmonary vascular endothelial barrier integrity was assessed using immunofluorescence, western blot, Evans blue dye, and qPCR. Human umbilical vein endothelial cells (HUVECs) were used for in vitro validation and exploration of the underlying mechanisms. RESULTS The CLP mice exhibited significant damage to pulmonary tissue and breakdown of endothelial barrier. Administration of paeoniflorin markedly improved survival rates, mitigated lung injury, and preserved the integrity of the pulmonary vascular endothelial barrier in CLP mice which was confirmed by in vitro experiments. Pharmacological mechanism studies showed that the protective effects of paeoniflorin on the vascular endothelium was achieved through activation of RXRα signaling, which could be reversed by RXRα knockdown. CONCLUSION Our experiments demonstrates the protective effect of paeoniflorin on the vascular endothelial barrier through activation of the RXRα, thereby offering potential therapeutic options for sepsis treatment. We also identified RXRα as a novel transcription factor for VE-cadherin, providing a potential new intervention target for vascular endothelial barrier damage in sepsis.
Collapse
Affiliation(s)
- Xinyue Cao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Ruihua Ma
- Department of Anesthesiology, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China, 215125
| | - Yirui Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Yuran Huang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Keyuan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Lijie Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Haidong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Guize Feng
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Tongqing Chen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Dong Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Keyu Sun
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, PR China, 201199.
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, PR China, 200032; Department of Anesthesiology, Shanghai Geriatic Medical Center, Shanghai, PR China, 201104; Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, PR China, 201199.
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China.
| |
Collapse
|
12
|
Federspiel JJ. Inpatient pharmacological thromboprophylaxis in the antepartum period: an argument for risk-based thromboprophylaxis. Am J Obstet Gynecol MFM 2025; 7:101567. [PMID: 39586470 PMCID: PMC11955298 DOI: 10.1016/j.ajogmf.2024.101567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
Venous thromboembolism (VTE) is a significant cause of maternal morbidity and mortality in the United States. People hospitalized during pregnancy for reasons other than routine birth (ie, during antepartum admissions) are at increased risk of VTE compared with nonhospitalized obstetric patients, but there is no consensus regarding which patients should receive thromboprophylaxis during antepartum hospitalizations as the absolute event rates are low and anticoagulation can complicate antepartum management. We argue that an approach informed by individualized patient risk assessment is likely to produce the greatest net benefit for patients. Such an approach would avoid the pitfalls of universal pharmacologic prophylaxis (potential to interfere with unplanned delivery or receipt of neuraxial anesthesia) among patients for whom the absolute risk of VTE is low. In contrast, approaches that withhold pharmacologic prophylaxis from all antepartum patients likely place some at significant risk of VTE. We outline the arguments against universal pharmacologic thromboprophylaxis and against universal avoidance of pharmacologic thromboprophylaxis and discuss a risk-based approach proposed at our institution. Finally, we outline a research agenda for identification of optimal antepartum anticoagulation strategies.
Collapse
Affiliation(s)
- Jerome J Federspiel
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Duke University School of Medicine, Durham, NC (Federspiel); Department of Medicine, Duke University School of Medicine, Durham, NC (Federspiel); Department of Population Health Sciences, Duke University School of Medicine, Durham, NC (Federspiel).
| |
Collapse
|
13
|
Yin J, Chi Y, Liu D, Li X, Li X. Unfractionated heparin attenuated histone-induced pulmonary endothelial glycocalyx injury through Ang/Tie2 pathway. J Inflamm (Lond) 2025; 22:9. [PMID: 39962477 PMCID: PMC11834598 DOI: 10.1186/s12950-025-00437-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
PURPOSE This study aimed to investigate the involvement of angiopoietin (Ang)/Tie2 pathway in mediating pulmonary endothelial glycocalyx injury in histone-induced acute lung injury in mice, and the protective mechanism of unfractionated heparin (UFH). METHODS Twenty-four male C57BL/6 mice (20-25 g), 8-12 weeks old, were randomly divided into control, histone, and histone + UFH groups. The histone (50 mg/kg) was administered via tail vein. UFH (400 U/kg) was administered 1 h after histone injection. The control group was administered by an equal amount of sterile saline solution. The lungs of all groups were harvested 4 h after the injection of histones or sterile saline. RESULTS UFH attenuated histone-induced lung histopathological changes and edema. UFH alleviated pulmonary endothelial injury and glycocalyx shedding by reducing histone-induced low expression of thrombomodulin (TM) and decreased lung syndecan-1 levels. UFH improved histone-induced low mRNA expression of TM, syndecan-1, Ang-1, Tie2 and high expression of heparinase (HPA), Ang-2. CONCLUSION UFH may attenuate histone-induced lung injury and pulmonary endothelial glycocalyx degradation via the Ang/Tie2 pathway.
Collapse
Affiliation(s)
- Jia Yin
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China
| | - Yawen Chi
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China
| | - Danyan Liu
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China
| | - Xinghua Li
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China
| | - Xu Li
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
14
|
De Rosa S, Lassola S, Visconti F, De Cal M, Cattin L, Rizzello V, Lampariello A, Zannato M, Danzi V, Marcante S. Acute Kidney Injury in Patients After Cardiac Arrest: Effects of Targeted Temperature Management. Life (Basel) 2025; 15:265. [PMID: 40003674 PMCID: PMC11856830 DOI: 10.3390/life15020265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Cardiac arrest (CA) is a leading cause of mortality and morbidity, with survivors often developing post-cardiac arrest syndrome (PCAS), characterized by systemic inflammation, ischemia-reperfusion injury (IRI), and multiorgan dysfunction. Acute kidney injury (AKI), a frequent complication, is associated with increased mortality and prolonged intensive care unit (ICU) stays. This study evaluates AKI incidence and progression in cardiac arrest patients managed with different temperature protocols and explores urinary biomarkers' predictive value for AKI risk. METHODS A prospective, single-center observational study was conducted, including patients with Return of Spontaneous Circulation (ROSC) post-cardiac arrest. Patients were stratified into three groups: therapeutic hypothermia (TH) at 33 °C, Targeted Temperature Management (TTM) at 35 °C, and no temperature management (No TTM). AKI was defined using KDIGO criteria, with serum creatinine and urinary biomarkers (TIMP-2 and IGFBP7) measured at regular intervals during ICU stay. RESULTS AKI incidence at 72 h was 31%, varying across protocols. It was higher in the No TTM group at 24 h and in the TH and TTM groups during rewarming. Persistent serum creatinine elevation and fluid imbalance were notable in the TH group. Biomarkers indicated moderate tubular stress in the TTM and No TTM groups. CONCLUSIONS AKI is a frequent complication post-cardiac arrest, with the rewarming phase identified as critical for renal vulnerability. Tailored renal monitoring, biomarker-guided risk assessment, and precise temperature protocols are essential to improve outcomes.
Collapse
Affiliation(s)
- Silvia De Rosa
- Department of Anesthesiology and Intensive Care, San Bortolo Hospital, 36100 Vicenza, Italy (L.C.); (V.R.); (A.L.); (M.Z.); (V.D.); (S.M.)
- Centre for Medical Sciences—CISMed, University of Trento, Via S. Maria Maddalena 1, 38122 Trento, Italy
- Anesthesia and Intensive Care, Santa Chiara Regional Hospital, APSS, 38121 Trento, Italy
| | - Sergio Lassola
- Anesthesia and Intensive Care, Santa Chiara Regional Hospital, APSS, 38121 Trento, Italy
| | - Federico Visconti
- Department of Anesthesiology and Intensive Care, San Bortolo Hospital, 36100 Vicenza, Italy (L.C.); (V.R.); (A.L.); (M.Z.); (V.D.); (S.M.)
- Anaesthesia and Intensive Care, Padova University Hospital, 35128 Padua, Italy
| | - Massimo De Cal
- International Renal Research Institute of Vicenza, (IRRIV Foundation), Department of Nephrology, Dialysis and Kidney Transplantation, San Bortolo Hospital, 36100 Vicenza, Italy;
| | - Lucia Cattin
- Department of Anesthesiology and Intensive Care, San Bortolo Hospital, 36100 Vicenza, Italy (L.C.); (V.R.); (A.L.); (M.Z.); (V.D.); (S.M.)
| | - Veronica Rizzello
- Department of Anesthesiology and Intensive Care, San Bortolo Hospital, 36100 Vicenza, Italy (L.C.); (V.R.); (A.L.); (M.Z.); (V.D.); (S.M.)
| | - Antonella Lampariello
- Department of Anesthesiology and Intensive Care, San Bortolo Hospital, 36100 Vicenza, Italy (L.C.); (V.R.); (A.L.); (M.Z.); (V.D.); (S.M.)
| | - Marina Zannato
- Department of Anesthesiology and Intensive Care, San Bortolo Hospital, 36100 Vicenza, Italy (L.C.); (V.R.); (A.L.); (M.Z.); (V.D.); (S.M.)
| | - Vinicio Danzi
- Department of Anesthesiology and Intensive Care, San Bortolo Hospital, 36100 Vicenza, Italy (L.C.); (V.R.); (A.L.); (M.Z.); (V.D.); (S.M.)
| | - Stefano Marcante
- Department of Anesthesiology and Intensive Care, San Bortolo Hospital, 36100 Vicenza, Italy (L.C.); (V.R.); (A.L.); (M.Z.); (V.D.); (S.M.)
| |
Collapse
|
15
|
Zhu L, Dong H, Li L, Liu X. The Mechanisms of Sepsis Induced Coagulation Dysfunction and Its Treatment. J Inflamm Res 2025; 18:1479-1495. [PMID: 39925935 PMCID: PMC11804232 DOI: 10.2147/jir.s504184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/22/2025] [Indexed: 02/11/2025] Open
Abstract
Sepsis is a critical condition characterized by organ dysfunction due to a dysregulated response to infection that poses significant global health challenges. Coagulation dysfunction is nearly ubiquitous among sepsis patients. Its mechanisms involve platelet activation, coagulation cascade activation, inflammatory reaction imbalances, immune dysregulation, mitochondrial damage, neuroendocrine network disruptions, and endoplasmic reticulum (ER) stress. These factors not only interact but also exacerbate one another, leading to severe organ dysfunction. This review illustrates the mechanisms of sepsis-induced coagulopathy, with a focus on tissue factor activation, endothelial glycocalyx damage, and the release of neutrophil extracellular traps (NETs), all of which are potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - He Dong
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Lin Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Xiaojie Liu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| |
Collapse
|
16
|
Yin M, Wang T, Jiang Q, Qu X, Ma J, Xu J, Jin X, Chen X. The association of red blood cell transfusion with mortality in pediatric patients with sepsis, severe sepsis, and septic shock: A single-center retrospective cohort study. Transfus Clin Biol 2025; 32:62-68. [PMID: 39710203 DOI: 10.1016/j.tracli.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND/OBJECTIVES Pediatric patients with sepsis are frequently subjected to red blood cell (RBC) transfusions but yet its association with mortality is still controversial. METHODS We consecutively selected 125 patients with sepsis, severe sepsis, and septic shock admitted to intensive care unit (ICU) in our center from January 2022 to January 2023, and finally 100 patients were included in this retrospective cohort study. The patients were divided into two groups: group I who received RBC transfusion and group II who did not receive RBC transfusion. Logistic regression analysis was used to determine the demographic and clinical factors related to receiving RBC transfusion. The association of RBC transfusion with mortality was determined by the Cox regression model, and the mechanical ventilation rate and length of stay by the logistic regression model. RESULTS Among the 100 patients, 67 and 33 cases belonged to the RBC-transfused and not-transfused groups, respectively. Lower hemoglobin level (OR = 0.918, 95%CI: 0.881-0.957, p < 0.001), increased c-reactive protein level (OR = 1.022, 95%CI: 1.002-1.043, p = 0.034), and lower platelets count (OR = 0.994, 95%CI: 0.988-0.999, p = 0.023) were associated with RBC transfusions. While the associations of RBC transfusion with mortality and mechanical ventilation were not shown to be statistically significant (HR = 3.926, 95%CI: 0.952-16.186, p = 0.058 and OR = 2.588, 95%CI: 0.832-8.046, p = 0.1), RBC transfusion might be associated with increased ICU length of stay (OR = 16.477, 95%CI: 3.86-70.342, p < 0.001). In the overall survival analysis, younger age (HR = 0.093, 95%CI: 0.027-0.320, p < 0.001), the use of mechanical ventilation (HR = 8.893, 95%CI: 1.483-53.336, p = 0.017), and more severe disease (severe sepsis vs. sepsis, HR = 24.531, 95%CI: 1.923-321.914, p = 0.014; septic shock vs. sepsis, HR = 32.187, 95%CI: 2.977-347.949, p = 0.004) were related to increased mortality. CONCLUSIONS RBC transfusions are significantly associated with increased ICU length of stay and not associated with 28-day mortality and mechanical ventilation rate. Other factors affecting mortality in pediatric patients with sepsis, severe sepsis, and septic shock are younger age, use of mechanical ventilation, and more severe disease.
Collapse
Affiliation(s)
- Mingwei Yin
- Department of Blood Transfusion, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, PR China
| | - Ting Wang
- Department of Blood Transfusion, Tai'an Traditional Chinese Medicine Hospital, Tai'an, Shandong Province, PR China
| | - Qian Jiang
- Department of Blood Transfusion, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, PR China
| | - Xinli Qu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, PR China
| | - Jihua Ma
- Department of Blood Transfusion, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, PR China
| | - Jun Xu
- Department of Blood Transfusion, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, PR China
| | - Xiaobo Jin
- Department of Blood Transfusion, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, PR China
| | - Xuejun Chen
- Department of Blood Transfusion, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, PR China.
| |
Collapse
|
17
|
Man C, An Y, Wang GX, Mao EQ, Ma L. Recent Advances in Pathogenesis and Anticoagulation Treatment of Sepsis-Induced Coagulopathy. J Inflamm Res 2025; 18:737-750. [PMID: 39845020 PMCID: PMC11752821 DOI: 10.2147/jir.s495223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025] Open
Abstract
Coagulopathy in sepsis is common and is associated with high mortality. Although immunothrombosis is necessary for infection control, excessive thrombus formation can trigger a systemic thrombo-inflammatory response. Immunothrombosis plays a core role in sepsis-induced coagulopathy, and research has revealed a complex interplay between inflammation and coagulation. Different mechanisms underlying sepsis-related coagulopathy are discussed, including factors contributing to the imbalance of pro- and anticoagulation relevant to endothelial cells. The potential therapeutic implications of anticoagulants on these mechanisms are discussed. This review contributes to our understanding of the pathogenesis of coagulopathy in patients with sepsis. Recent studies suggest that endothelial cells play an important role in immunoregulation and hemostasis. Meanwhile, the non-anticoagulation effects of anticoagulants, especially heparin, which act in the pathogenesis of coagulopathy in septic patients, have been partially revealed. We believe that further insights into the pathogenesis of sepsis-induced coagulopathy will help physicians evaluate patient conditions effectively, leading to advanced early recognition and better decision-making in the treatment of sepsis.
Collapse
Affiliation(s)
- Chit Man
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Yuan An
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Guo-Xin Wang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - En-Qiang Mao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Li Ma
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| |
Collapse
|
18
|
Datta R, Singh S. The endothelium or mitochondrial level therapy: new frontiers in sepsis? Med Intensiva 2025:502130. [PMID: 39799036 DOI: 10.1016/j.medine.2024.502130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 01/15/2025]
Abstract
The host and microbes play complex roles in balancing the pro- and anti-inflammatory pathways that cause sepsis. It is now increasingly recognized as a disorder of the mitochondrial system intrinsically or as a consequence of microcirculatory abnormalities leading to hypoperfusion/hypoxia ("microcirculatory and mitochondrial distress syndrome"). It is expected that improvements in endothelium or mitochondrial level therapy will lower sepsis-related morbidity and mortality. This article aimed to clarify the mitochondrial and microcirculation abnormalities in patients with sepsis and the futuristic research agenda for the management of sepsis.
Collapse
Affiliation(s)
- Rashmi Datta
- Intensive Care Unit, Adesh Medical College and Hospital, NH44, Mohri, Ambala, Haryana-136135, India; Department of Anaesthesiology and Critical Care, Command Hospital (NC), Udhampur 182101, India
| | - Shalendra Singh
- Intensive Care Unit, Adesh Medical College and Hospital, NH44, Mohri, Ambala, Haryana-136135, India; Department of Anaesthesiology and Critical Care, Command Hospital (NC), Udhampur 182101, India.
| |
Collapse
|
19
|
Galtung N, Stein V, Prpic M, Boyraz B, Ulke J, Kurz S, Dernedde J, Diehl-Wiesenecker E, Bauer W, Kappert K. EARLY ANALYSIS OF ENDOTHELIAL MARKERS TO PREDICT SEPSIS IN THE EMERGENCY DEPARTMENT. Shock 2025; 63:72-79. [PMID: 39405404 DOI: 10.1097/shk.0000000000002482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2024]
Abstract
ABSTRACT Background: Acute infections and sepsis are a leading cause of death. These patients are primarily encountered at the emergency department (ED), where early assessment for sepsis is necessary to improve outcome. In sepsis, the inflammatory response causes several characteristic pathophysiological changes, including a dysregulated and generalized activation of the endothelium. This study aimed to analyze endothelial markers released to the blood as diagnostic biomarkers for acute infection and sepsis in the ED, as smaller studies have previously shown promising results in other settings. Methods : Serum samples from n = 312 adult patients with suspected acute infections at presentation to the ED were utilized. Patients' courses of disease and outcomes were assessed by clinical adjudication. E-selectin, P-selectin, ICAM-1, and VCAM-1 were measured by ELISAs. The accuracy of each marker for predicting bacterial infection, sepsis, and in-hospital mortality was evaluated. Results : For sepsis, E-selectin and ICAM-1 both showed an area under the receiver operating characteristic (AUROC) of 0.62, lower than procalcitonin with 0.77 (both P < 0.01) and lactate with 0.73 ( P = 0.030 and 0.046, respectively), but similar to CRP with 0.60 ( P = 0.758 and 0.876, respectively). For 28-day in-hospital mortality among patients with infection, ICAM-1 performed best with an AUROC of 0.75. Conclusions : Despite promising results in small studies and specific cohorts, particularly in intensive care units, this large-scale evaluation of four endothelial biomarkers highlights their limited diagnostic utility in a broader inclusion setup design at the earliest possible time point of evaluation.
Collapse
Affiliation(s)
- Noa Galtung
- Department of Emergency Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vanessa Stein
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Monika Prpic
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Burak Boyraz
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jannis Ulke
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stephan Kurz
- Department of Cardiothoracic and Vascular Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jens Dernedde
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eva Diehl-Wiesenecker
- Department of Emergency Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wolfgang Bauer
- Department of Emergency Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kai Kappert
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
20
|
He X, Liu H. Persistent and severe hypotension during radical transabdominal ovarian cancer surgery: A case report. Medicine (Baltimore) 2024; 103:e40751. [PMID: 39654230 PMCID: PMC11630978 DOI: 10.1097/md.0000000000040751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
RATIONALE In radical surgery for ovarian cancer (OC), hypotension that is difficult to correct is usually rare unless there is significant blood loss. We recently encountered a patient who developed persistent and severe hypotension during radical transabdominal OC surgery. PATIENT CONCERNS A patient was 52 years old with a history of hypertension and well-controlled preoperative blood pressure (BP). A total of 2000 mL of ascites was drained and blood loss was 300 mL when the operation proceeded to 5.5 hours. The patient's cardiopulmonary function and blood gas analysis showed no significant abnormalities. DIAGNOSES persistent and uncorrectable hypotension. INTERVENTIONS There was no significant edema in the patient's head or face, nor did the surgeon observe noticeable edema in her intestinal walls or other organs. No oozing was seen at the surgical site. Fluid resuscitation and vasopressor administration were continued. As BP control further deteriorated, blood counts, coagulation, and biochemical electrolyte analyses revealed severe hypoalbuminemia (13.5 g/L) and coagulation dysfunction. OUTCOMES After intravenous human serum albumin (HSA) and fresh frozen plasma therapy, her hypoalbuminemia and coagulation were gradually corrected. LESSONS Based on this case, we suggest that in OC patients experiencing mild intraoperative bleeding and minimal heart rate variation but persistent refractory hypotension, hypoalbuminemia should be considered even if preoperative biochemical tests (including serum albumin levels) are normal. Confirming hypoalbuminemia warrants HSA administration to alleviate hypovolemic shock symptoms. Additionally, it is important to be cautious of potential coagulation issues with albumin use, possibly requiring plasma infusion to address coagulopathy.
Collapse
Affiliation(s)
- Xinyan He
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Hui Liu
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Wu M, Yan Y, Xie X, Bai J, Ma C, Du X. Effect of endothelial responses on sepsis-associated organ dysfunction. Chin Med J (Engl) 2024; 137:2782-2792. [PMID: 39501810 DOI: 10.1097/cm9.0000000000003342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT Sepsis-related organ dysfunction is associated with increased morbidity and mortality. Previous studies have found that the endothelium plays crucial roles in maintaining the vascular permeability during sepsis, as well as in regulating inflammation and thrombosis. During sepsis, endothelial cells may release cytokines, chemokines, and pro-coagulant factors, as well as express adhesion molecules. In general, endothelial responses during sepsis typically inhibit bacterial transmission and coordinate leukocyte recruitment to promote bacterial clearance. However, excessive or prolonged endothelial activation can lead to impaired microcirculation, tissue hypoperfusion, and organ dysfunction. Given the structural and functional heterogeneity of endothelial cells in different organs, there are potential differences in endothelial responses by organ type, and the risk of organ damage may vary accordingly. This article reviews the endothelial response observed in sepsis and its effects on organ function, summarizes current progress in the development of therapeutic interventions targeting the endothelial response, and discusses future research directions to serve as a reference for researchers in the field.
Collapse
Affiliation(s)
- Miao Wu
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yan Yan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xinyu Xie
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jiawei Bai
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Chengtai Ma
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xianjin Du
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
22
|
Zhou Y, He LP, Qi YH, Huang Y, Hu BQ, Liu JL, Zeng QB, Song JC. Diagnostic value of tissue plasminogen activator-inhibitor complex in sepsis-induced liver injury: A single-center retrospective case-control study. World J Hepatol 2024; 16:1255-1264. [PMID: 39606162 PMCID: PMC11586747 DOI: 10.4254/wjh.v16.i11.1255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/05/2024] [Accepted: 10/15/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Sepsis often causes severe liver injury and leads to poor patient outcomes. Early detection of sepsis-induced liver injury (SILI) and early treatment are key to improving outcomes. AIM To investigate the clinical characteristics of SILI patients and analyze the associated risk factors, to identify potential sensitive biomarkers. METHODS Retrospective analysis of clinical data from 546 patients with sepsis treated in the intensive care unit of the 908th Hospital of Chinese People's Liberation Army Joint Logistic Support Force between May 2018 and December 2022. The patients were divided into the sepsis group (n = 373) and SILI group (n = 173) based on the presence of acute liver injury within 2 hours of admission. We used the random forest algorithm to analyze risk factors and assessed potential diagnostic markers of SILI using the area under the receiver operating characteristic curve, Kaplan-Meier survival curves, subgroup analysis and correlation analysis. RESULTS Compared with the sepsis group, tissue plasminogen activator-inhibitor complex (t-PAIC) levels in serum were significantly higher in the SILI group (P < 0.05). Random forest results showed that t-PAIC was an independent risk factor for SILI, with an area under the receiver operating characteristic curve of 0.862 (95% confidence interval: 0.832-0.892). Based on the optimal cut-off value of 11.9 ng/mL, patients at or above this threshold had significantly higher levels of lactate and Acute Physiology and Chronic Health Evaluation II score. The survival rate of these patients was also significantly worse (hazard ratio = 2.2, 95% confidence interval: 1.584-3.119, P < 0.001). Spearman's correlation coefficients were 0.42 between t-PAIC and lactate, and 0.41 between t-PAIC and aspartate transaminase. Subgroup analysis showed significant differences in t-PAIC levels between patients with different severity of liver dysfunction. CONCLUSION T-PAIC can serve as a diagnostic indicator for SILI, with its elevation correlated with the severity of SILI.
Collapse
Affiliation(s)
- Ye Zhou
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang 330002, Jiangxi Province, China
- Department of Critical Care Medicine, The 908 Hospital of Chinese People's Liberation Army Joint Logistic Support Force, Nanchang 330002, Jiangxi Province, China
| | - Long-Ping He
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang 330002, Jiangxi Province, China
| | - Ying-Han Qi
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang 330002, Jiangxi Province, China
| | - Yu Huang
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang 330002, Jiangxi Province, China
| | - Bing-Qin Hu
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang 330002, Jiangxi Province, China
| | - Jia-Ling Liu
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang 330002, Jiangxi Province, China
| | - Qing-Bo Zeng
- Intensive Care Unit, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang 330002, Jiangxi Province, China
| | - Jing-Chun Song
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang 330002, Jiangxi Province, China
- Department of Critical Care Medicine, The 908 Hospital of Chinese People's Liberation Army Joint Logistic Support Force, Nanchang 330002, Jiangxi Province, China.
| |
Collapse
|
23
|
Tharmaraj D, Mulley WR, Dendle C. Current and emerging tools for simultaneous assessment of infection and rejection risk in transplantation. Front Immunol 2024; 15:1490472. [PMID: 39660122 PMCID: PMC11628869 DOI: 10.3389/fimmu.2024.1490472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/14/2024] [Indexed: 12/12/2024] Open
Abstract
Infection and rejection are major complications that impact transplant longevity and recipient survival. Balancing their risks is a significant challenge for clinicians. Current strategies aimed at interrogating the degree of immune deficiency or activation and their attendant risks of infection and rejection are imprecise. These include immune (cell counts, function and subsets, immunoglobulin levels) and non-immune (drug levels, viral loads) markers. The shared risk factors between infection and rejection and the bidirectional and intricate relationship between both entities further complicate transplant recipient care and decision-making. Understanding the dynamic changes in the underlying net state of immunity and the overall risk of both complications in parallel is key to optimizing outcomes. The allograft biopsy is the current gold standard for the diagnosis of rejection but is associated with inherent risks that warrant careful consideration. Several biomarkers, in particular, donor derived cell-free-DNA and urinary chemokines (CXCL9 and CXCL10), show significant promise in improving subclinical and clinical rejection risk prediction, which may reduce the need for allograft biopsies in some situations. Integrating conventional and emerging risk assessment tools can help stratify the individual's short- and longer-term infection and rejection risks in parallel. Individuals identified as having a low risk of rejection may tolerate immunosuppression wean to reduce medication-related toxicity. Serial monitoring following immunosuppression reduction or escalation with minimally invasive tools can help mitigate infection and rejection risks and allow for timely diagnosis and treatment of these complications, ultimately improving allograft and patient outcomes.
Collapse
Affiliation(s)
- Dhakshayini Tharmaraj
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
| | - William R. Mulley
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
| | - Claire Dendle
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
- Monash Infectious Diseases, Monash Health, Clayton, VIC, Australia
| |
Collapse
|
24
|
Leung G, Middleton EA. The role of platelets and megakaryocytes in sepsis and ARDS. J Physiol 2024; 602:6047-6063. [PMID: 39425883 DOI: 10.1113/jp284879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/06/2024] [Indexed: 10/21/2024] Open
Abstract
Since the global COVID-19 pandemic, there has been a renewed focus on lung injury during infection. Systemic inflammatory responses such as acute respiratory distress syndrome (ARDS) and sepsis are a leading cause of morbidity and mortality for both adults and children. Improvements in clinical care have improved outcomes but mortality remains ∼40% and significant morbidity persists for those patients with severe disease. Mechanistic studies of the underlying biological processes remain essential to identifying therapeutic targets. Furthermore, methods for identifying the underlying drivers of organ failure are key to treating and preventing tissue injury. In this review, we discuss the contribution of megakaryocytes (MKs) and platelets to the pathogenesis of systemic inflammatory syndromes. We explore the role of MKs and the new identification of extramedullary MKs during sepsis. We describe the alterations in the platelet transcriptome during sepsis. Lastly, we explore platelet function as defined by aggregation, activation and the formation of heterotypic aggregates. Much more work is necessary to explore the contribution of platelets to these heterogenous syndromes, but the foundation of platelets as key contributors to inflammation has been laid.
Collapse
Affiliation(s)
- Gabriel Leung
- Division of Pulmonary, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Elizabeth A Middleton
- Division of Pulmonary, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
25
|
Zhang S, Li S, Xie S, Cui L, Gao Y, Wang Y. The Role of Ca 2+/PI3K/Akt/eNOS/NO Pathway in Astragaloside IV-Induced Inhibition of Endothelial Inflammation Triggered by Angiotensin II. Mediators Inflamm 2024; 2024:3193950. [PMID: 39512364 PMCID: PMC11540887 DOI: 10.1155/2024/3193950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
Inflammation induced by angiotensin II (Ang II) is a key event in the progression of numerous cardiovascular diseases. Astragaloside IV (AS-IV), a glycoside extracted from Astragalus membranaceus Bunge, has been shown to inhibit Ang II-induced inflammatory responses in vivo. However, the mechanisms underlying the beneficial effects are still unclear. This study investigated whether AS-IV attenuates endothelial inflammation induced by Ang II via the activation of endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) pathway. Human umbilical vein endothelial cells (HUVECs) were cultured in the presence of AS-IV with or without the specific inhibitor of NOS or Ca2+- and phosphatidylinositol 3-kinase (PI3K)/Akt-dependent cascade prior to Ang II exposure. Incubation of HUVECs with AS-IV enhanced NO production and eNOSser1177 phosphorylation. These responses were abrogated by the inhibition of NOS or Ca2+- and PI3K/Akt-dependent pathway. In addition, preincubation of HUVECs with AS-IV inhibited Ang II-induced cytokine and chemokine production, adhesion molecule expression, monocyte adhesion, and nuclear factor kappa B (NF-κB) activation as evidenced by the attenuation of inhibitor of kappa B alpha phosphorylation and subsequent NF-κB DNA binding. These effects of AS-IV were abolished by the suppression of NOS or Ca2+- and PI3K/Akt-dependent cascade. Our findings indicate that AS-IV attenuates inflammatory responses triggered by Ang II possibly via the activation of Ca2+/PI3K/Akt/eNOS/NO pathway in endothelial cells.
Collapse
Affiliation(s)
- Shiyu Zhang
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Shijie Li
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Shiyang Xie
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Lin Cui
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Yuan Gao
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Youping Wang
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| |
Collapse
|
26
|
Huang B, Tang P, Liu Y, Liu F, Zheng Y, Yang X, Zhang X, Xie H, Lin L, Lin B, Lin B. Xuefu Zhuyu decoction alleviates deep vein thrombosis through inhibiting the activation of platelets and neutrophils via sirtuin 1/nuclear factor kappa-B pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118485. [PMID: 38908490 DOI: 10.1016/j.jep.2024.118485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xuefu Zhuyu Decoction (XZD), a renowned traditional Chinese medicine prescription, is widely employed for the management of conditions characterized by qi-stagnation and blood stasis. Although its anti-thrombotic effect on deep vein thrombosis (DVT) patients has been clinically observed, the underlying mechanism remains largely unexplored. AIM OF THE STUDY Our aim was to investigate the mechanisms by which XZD exerted its effect on DVT. MATERIALS AND METHODS The ultra performance liquid chromatography (UPLC) technique was employed to evaluate quality of XZD. To examine the effect of XZD on DVT, a DVT rat model with inferior vena cava (IVC) stenosis was established. The 4D-label-free proteomics approach was then utilized to uncover the possible mechanisms of XZD against DVT. Based on proteomics, citrullinated histone H3 (CitH3), along with serum levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) were observed the inhibitory activity of XZD on neutrophil activation. Subsequently, the marker of platelet activation, specifically glycoprotein IIb (CD41) and glycoprotein IIIa (CD61), were assessed along with the secretion of von Willebrand factor (vWF) to investigate the inhibitory activity of XZD on platelet activation. Finally, we explored the impact of XZD on the sirtuin 1 (SIRT1)/nuclear factor kappa-B (NF-κB) pathway, which was associated with the activation of platelets and neutrophils. RESULTS Eight distinct components were identified for the quality control of XZD. XZD effectively reduced thrombus weight and length in DVT rats, without affecting the coagulation function or hematological parameters in the systemic circulation. Proteomics analysis revealed that XZD alleviated DVT by inhibiting the activation of platelets and neutrophils. The protein expression of CitH3, along with serum levels of TNF-α and IL-1β, were reduced in XZD-treated DVT rats. Similarly, protein expressions of CD41 and CD61, along with the release of vWF, were markedly down-regulated in XZD-treated DVT rats. Finally, treatment with XZD resulted in an up-regulation of SIRT1 protein expression and a down-regulation of both acetylated NF-κB/p65 and phosphorylated NF-κB/p65 protein expressions in endothelium. CONCLUSIONS XZD alleviates DVT by inhibiting the activation of platelets and neutrophils at the injured endothelium via the regulation of SIRT1/NF-κB pathway.
Collapse
Affiliation(s)
- Boning Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Ping Tang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Youchen Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Fangle Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Yuying Zheng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Xinrong Yang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Xiubing Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Huiyi Xie
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Liuqing Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Bingqing Lin
- School of Mathematical Sciences, Shenzhen University, Shenzhen, Guangdong, China.
| | - Baoqin Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China.
| |
Collapse
|
27
|
Liang P, Zhu M, Sun X, Wang L, Li B, Ming S, Younis M, Yang J, Wu Y, Huang X. LncRNA-mRNA co-expression analysis reveals aquaporin-9-promoted neutrophil extracellular trap formation and inflammatory activation in sepsis. Int Immunopharmacol 2024; 140:112916. [PMID: 39133961 DOI: 10.1016/j.intimp.2024.112916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024]
Abstract
Sepsis is a life-threatening condition caused by an excessive inflammatory response to an infection. However, the precise regulatory mechanism of sepsis remains unclear. Using a strand-specific RNA-sequencing, we identified 115 hub differentially expressed long noncoding RNAs (lncRNAs) and 443 mRNAs in septic patients, primarily participated in crucial pathways including neutrophil extracellular trap (NET) formation and toll-like receptor signaling. Notably, NETs related gene aquaporin-9 (AQP9) and its associated lncRNAs exhibited significant upregulation in septic neutrophils. Functional experiments revealed AQP9 interacts with its lncRNAs to augment the formation of neutrophil NETs. In murine sepsis models, AQP9 inhibition with phloretin reduced proinflammatory cytokine production and lung damage. These findings provide crucial insights into the regulatory role of AQP9 in sepsis, unraveling its interaction with associated lncRNAs in transmitting downstream signals, holding promise in informing the development of novel therapeutic strategies aimed at ameliorating the debilitating effects of sepsis.
Collapse
Affiliation(s)
- Pingping Liang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Manman Zhu
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Xingzi Sun
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Li Wang
- Department of Obstetrics and Gynecology, Perinatal Medical Center, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Bin Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Siqi Ming
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Muhammad Younis
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Jianhua Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| |
Collapse
|
28
|
Yang H, Feng L, Jiang Z, Wu X, Zeng K. Amlexanox reduces new-onset atrial fibrillation risk in sepsis by downregulating S100A12: a Mendelian randomization study. Front Cardiovasc Med 2024; 11:1401314. [PMID: 39444551 PMCID: PMC11496243 DOI: 10.3389/fcvm.2024.1401314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Background Sepsis is characterized by high morbidity and mortality rates, alongside limited therapeutic efficacy. Atrial fibrillation (AF), the most common arrhythmia, has been closely linked to sepsis in prior research. However, the specific mechanisms through which sepsis leads to new-onset AF remain poorly understood. This study focuses on identifying critical genes that are dysregulated in the development of new-onset AF within the context of sepsis, with the goal of uncovering new potential targets for its diagnosis and prevention. Material and methods Our study began by applying Mendelian Randomization (MR) to assess the causal link between sepsis and AF. We then sourced sepsis and AF datasets from the Gene expression Omnibus (GEO) database. Using Weighted Gene Co-expression Network Analysis (WGCNA), we pinpointed key modules and genes associated with both sepsis and AF conditions. Protein-protein interaction (PPI) network was constructed. The Transcriptional Regulatory Relationships Unravelled by Sentence-based Text-mining (TRRUST) database helped build the transcription factor (TF) interaction network. Key genes were scrutinized through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) to delve into their roles in new-onset AF's pathophysiology during sepsis. We employed the CIBERSORT algorithm to evaluate immune infiltration and the association between key genes and immune cells. The Connectivity Map (CMap) database facilitated the prediction of potential small molecule compounds targeting key genes. To culminate, an acute sepsis mouse model was developed to validate the implicated mechanisms of key genes involved in new-onset AF during sepsis, and to assess the prophylactic effectiveness of identified drug candidates. Results MR revealed potential independent risk factors for new-onset AF in sepsis. S100A12 was identified as a core interaction gene with elevated levels in sepsis and AF, underscoring its diagnostic and predictive significance. S100A12, along with associated genes, was mainly linked to immune and inflammatory response signaling pathways, correlating with immune cell levels. Targeting S100A12 identifies five potential small molecule therapeutics: amlexanox, balsalazide, methandriol, olopatadine, and tiboloe. In animal studies, acute sepsis increased S100A12 expression in serum and atrial tissues, correlating positively with inflammatory markers (IL-1β, IL-6, TNF-α) and negatively with heart rate, indicating a predisposition to AF. Early amlexanox administration can reduced S100A12 expression, dampened inflammation, and lessened new-onset AF risk in sepsis. Conclusion This study demonstrates that sepsis may independently increase the risk of new-onset AF. We identified S100A12 as a key gene influencing the new-onset AF in sepsis through immune regulation, presenting considerable diagnostic and predictive value. Notably, amlexanox, by targeting S100A12 emerges as the most clinical relevant intervention for managing new-onset AF in sepsis patients.
Collapse
Affiliation(s)
- Hang Yang
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lin Feng
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhenjie Jiang
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaodan Wu
- Department of Anesthesiology, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Kai Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
29
|
Alves PT, de Souza AG, Bastos VAF, Miguel EL, Ramos ACS, Cameron LC, Goulart LR, Cunha TM. The Modulation of Septic Shock: A Proteomic Approach. Int J Mol Sci 2024; 25:10641. [PMID: 39408970 PMCID: PMC11476436 DOI: 10.3390/ijms251910641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Sepsis poses a significant challenge due its lethality, involving multiple organ dysfunction and impaired immune responses. Among several factors affecting sepsis, monocytes play a crucial role; however, their phenotype, proteomic profile, and function in septic shock remain unclear. Our aim was to fully characterize the subpopulations and proteomic profiles of monocytes seen in septic shock cases and discuss their possible impact on the disease. Peripheral blood monocyte subpopulations were phenotype based on CD14/CD16 expression by flow cytometry, and proteins were extracted from the monocytes of individuals with septic shock and healthy controls to identify changes in the global protein expression in these cells. Analysis using 2D-nanoUPLC-UDMSE identified 67 differentially expressed proteins in shock patients compared to controls, in which 44 were upregulated and 23 downregulated. These proteins are involved in monocyte reprogramming, immune dysfunction, severe hypotension, hypo-responsiveness to vasoconstrictors, vasodilation, endothelial dysfunction, vascular injury, and blood clotting, elucidating the disease severity and therapeutic challenges of septic shock. This study identified critical biological targets in monocytes that could serve as potential biomarkers for the diagnosis, prognosis, and treatment of septic shock, providing new insights into the pathophysiology of the disease.
Collapse
Affiliation(s)
- Patrícia Terra Alves
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
| | - Aline Gomes de Souza
- Department of Medical Imaging, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14040-900, SP, Brazil;
| | - Victor Alexandre F. Bastos
- Laboratory of Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Eduarda L. Miguel
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| | - Augusto César S. Ramos
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| | - L. C. Cameron
- Arthritis Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada;
- Lorraine Protein Biochemistry Group, Graduate Program in Neurology, Gaffrée e Guinle University Hospital, Rio de Janeiro 20270-004, RJ, Brazil
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
| | - Thúlio M. Cunha
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| |
Collapse
|
30
|
Catarina AV, Branchini G, Caceres RA, Fernandes RS, Costa BP, Machado KLDG, Becker T, Ferreira LF, Rigatto K, de Oliveira JR, Nunes FB. Fructose-1,6-bisphosphate reverses hypotensive effect caused by L-kynurenine in Wistar male rats. Physiol Rep 2024; 12:e70033. [PMID: 39396923 PMCID: PMC11471349 DOI: 10.14814/phy2.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 10/15/2024] Open
Abstract
Hypotension is one of the main characteristics of the systemic inflammation, basically caused by endothelial dysfunction. Studies have shown that the amino acid L-kynurenine (KYN) causes vasodilation in mammals, leading to hypotensive shock. In hypotensive shock, when activated by the KYN, the voltage-gated potassium channel encoded by the family KCNQ (Kv7) gene can cause vasodilation. Fructose-1,6-bisphosphate (FBP) it is being considered in studies an anti-inflammatory, antioxidant, immunomodulator, and a modulator of some ion channels (Ca2+, Na+, and K+). We analyzed the effects of KYN and FBP on mean blood pressure (MBP), systolic and diastolic (DBP) blood pressure, and heart rate variability (HRV) in Wistar rats. Results demonstrated that the administration of KYN significant decreased MBP, DBP, and increased HRV. Importantly, the FBP treatment reversed the KYN effects on MBP, DBP, and HRV. Molecular Docking Simulations suggested that KYN and FBP present a very close estimated free energy of binding and the same position into structure of KCNQ4. Our results did demonstrate that FBP blunted the decrease in BP, provoked by KYN. Results raise new hypotheses for future and studies in the treatment of hypotension resulting from inflammation.
Collapse
Affiliation(s)
- Anderson Velasque Catarina
- Graduate Program in Pathology—Laboratory of ComputationalMolecular, and Cellular Biophysics—Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| | - Gisele Branchini
- Graduate Program in Pathology—Laboratory of ComputationalMolecular, and Cellular Biophysics—Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| | - Rafael Andrade Caceres
- Department of PharmacosciencesUniversidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| | - Renata Streck Fernandes
- Graduate Program in Health Sciences—Laboratory of Translational Physiology—Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| | - Bruna Pasqualotto Costa
- Laboratory of Inflammation and Cellular Biophysics—Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS)Porto AlegreBrazil
| | | | - Tiago Becker
- Department of Mechanical EngineeringUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Luis Fernando Ferreira
- School of Electronics, Electrical Engineering and Computer SciencesQueen's University of BelfastBelfastUK
- Graduate Program in Medicine: Hepatology—Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| | - Katya Rigatto
- Graduate Program in Health Sciences—Laboratory of Translational Physiology—Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Inflammation and Cellular Biophysics—Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS)Porto AlegreBrazil
| | - Fernanda Bordignon Nunes
- Graduate Program in Pathology—Laboratory of ComputationalMolecular, and Cellular Biophysics—Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
- Laboratory of Inflammation and Cellular Biophysics—Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS)Porto AlegreBrazil
| |
Collapse
|
31
|
Fu ZP, Lee S, Wang RY, Wang YQ. Cronobacter sakazakii induced sepsis-associated arrhythmias through its outer membrane vesicles. iScience 2024; 27:110572. [PMID: 39228788 PMCID: PMC11369384 DOI: 10.1016/j.isci.2024.110572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/15/2024] [Accepted: 07/22/2024] [Indexed: 09/05/2024] Open
Abstract
Sepsis-induced arrhythmia, linked to sudden cardiac death, is associated with gut microbiota, though the exact relationship is unclear. This study aimed to elucidate the relationship between Cronobacter sakazakii (C. sakazakii) and arrhythmia. The relative abundance of C. sakazakii was increased in cecal ligation and puncture (CLP)-induced septic mice. Live C. sakazakii, supernatant, and outer membrane vesicles (OMVs) resulted in premature ventricular beat (PVB), sinus arrhythmia (SA), and increased arrhythmia and mortality in sepsis model through dysregulated ion channel proteins. Moreover, short-chain fatty acids (SCFAs) showed antibacterial effects in vitro. We confirmed sodium acetate (C2) and sodium butyrate (C4) protect from C. sakazakii-induced arrhythmia, and C2 and C4 protected from septic arrhythmia by activating free fatty acid receptor 2 and 3 (FFAR2 and FFAR3) in mice. These findings point to how C. sakazakii's OMVs trigger arrhythmia, and SCFAs may be a treatment for septic arrhythmia.
Collapse
Affiliation(s)
- Zhi-ping Fu
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Shuang Lee
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Rui-yao Wang
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Yu-qing Wang
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| |
Collapse
|
32
|
Borges A, Bento L. Organ crosstalk and dysfunction in sepsis. Ann Intensive Care 2024; 14:147. [PMID: 39298039 DOI: 10.1186/s13613-024-01377-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
Sepsis is a dysregulated immune response to an infection that leads to organ dysfunction. Sepsis-associated organ dysfunction involves multiple inflammatory mechanisms and complex metabolic reprogramming of cellular function. These mechanisms cooperate through multiple organs and systems according to a complex set of long-distance communications mediated by cellular pathways, solutes, and neurohormonal actions. In sepsis, the concept of organ crosstalk involves the dysregulation of one system, which triggers compensatory mechanisms in other systems that can induce further damage. Despite the abundance of studies published on organ crosstalk in the last decade, there is a need to formulate a more comprehensive framework involving all organs to create a more detailed picture of sepsis. In this paper, we review the literature published on organ crosstalk in the last 10 years and explore how these relationships affect the progression of organ failure in patients with septic shock. We explored these relationships in terms of the heart-kidney-lung, gut-microbiome-liver-brain, and adipose tissue-muscle-bone crosstalk in sepsis patients. A deep connection exists among these organs based on crosstalk. We also review how multiple therapeutic interventions administered in intensive care units, such as mechanical ventilation, antibiotics, anesthesia, nutrition, and proton pump inhibitors, affect these systems and must be carefully considered when managing septic patients. The progression to multiple organ dysfunction syndrome in sepsis patients is still one of the most frequent causes of death in critically ill patients. A better understanding and monitoring of the mechanics of organ crosstalk will enable the anticipation of organ damage and the development of individualized therapeutic strategies.
Collapse
Affiliation(s)
- André Borges
- Intensive Care Unit of Hospital de São José, Unidade de Urgência Médica, Rua José António Serrano, Lisbon, 1150-199, Portugal.
- NOVA Medical School, Campo dos Mártires da Pátria 130, Lisbon, 1169-056, Portugal.
| | - Luís Bento
- Intensive Care Unit of Hospital de São José, Unidade de Urgência Médica, Rua José António Serrano, Lisbon, 1150-199, Portugal
- NOVA Medical School, Campo dos Mártires da Pátria 130, Lisbon, 1169-056, Portugal
| |
Collapse
|
33
|
Arya R, Kumar R, Priyadarshi RN, Narayan R, Anand U. Vascular complications of liver abscess: A literature review. World J Meta-Anal 2024; 12:94519. [DOI: 10.13105/wjma.v12.i3.94519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/23/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Extensive vascular network and proximity to the gastrointestinal tract make the liver susceptible to abscess formation. While pyogenic liver abscesses account for the majority of liver abscesses in the Western world, amebic liver abscesses are more prevalent in tropical and developing nations. Most liver abscesses heal without complications. However, various vascular complications can occur in these patients, including compression of the inferior vena cava, thrombosis of the portal vein and/or hepatic veins, hepatic artery pseudoaneurysm, direct rupture into major vessels or the pericardium, and biliovascular fistula. These complications can present significant clinical challenges due to the potential for haemorrhage, ischemia, and systemic embolism, thereby increasing the risk of morbidity and mortality. Mechanical compression, flow stasis, inflammation, endothelial injury, and direct invasion are some of the proposed mechanisms that can cause vascular complications in the setting of a liver abscess. For the diagnosis, thorough assessment, and therapeutic planning of vascular complications, more sophisticated imaging techniques such as multidetector computed tomography angiography or magnetic resonance angiography may be necessary. Although most vascular complications resolve with abscess treatment alone, additional interventions may be required based on the nature, severity, and course of the complications. This article aims to provide a systematic update on the spectrum of vascular complications of liver abscesses, offering insights into their pathogenesis, diagnosis, and management strategies.
Collapse
Affiliation(s)
- Rahul Arya
- Department of Gastroenterology, All India Institute of Medical Sciences, Patna 801507, Bihar, India
| | - Ramesh Kumar
- Department of Gastroenterology, All India Institute of Medical Sciences, Patna 801507, Bihar, India
| | - Rajeev N Priyadarshi
- Department of Radiodiagnosis, All India Institute of Medical Sciences, Patna 801507, Bihar, India
| | - Ruchika Narayan
- Department of Radiodiagnosis, All India Institute of Medical Sciences, Patna 801507, Bihar, India
| | - Utpal Anand
- Department of Surgical Gastroenterology, All India Institute of Medical Sciences, Patna 801507, India
| |
Collapse
|
34
|
Refaie MMM, El-Hussieny M, Bayoumi AMA, Abdelraheem WM, Abdel-Hakeem EA, Shehata S. Sacubitril/valsartan alleviates sepsis-induced myocardial injury in rats via dual angiotensin receptor-neprilysin inhibition and modulation of inflammasome/caspase 1/IL1β pathway. Eur J Pharmacol 2024; 979:176834. [PMID: 39038638 DOI: 10.1016/j.ejphar.2024.176834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/30/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Sepsis is a life-threatening situation that ultimately affects cardiac function, leading to cardiomyopathy and myocardial injury as a result of uncontrolled response to infection.Till now, there is limited effective treatment to rescue those cases. Thus, novel therapeutic strategies should be identified to achieve better outcomes for septic patients. For the first time, we aimed to evaluate the effect of sacubitril/valsartan (Sac/Val) on sepsis-induced cardiac injury. Wistar male adult albino rats were randomly divided into four groups; Group I received the vehicle; Group II was given the vehicle plus 1 ml saline containing viable Escherichia coli (E. coli) (2.1 × 109 cfu) by intraperitoneal (i.p.) injection on the 1st and 2nd days; Group III received i.p. injection as group II plus oral administration of Sac/Val (30 mg/kg/day) and Nitro- ω-L-arginine (L-NNA) (25 mg/kg/day) for 7 days. Group IV was administered i.p. injection as group II plus oral administration of Sac/Val (30 mg/kg/day) for 7 days. Our data (n = 10) revealed successful induction of sepsis as it showed a significant increase in the measured cardiac enzymes, malondialdehyde (MDA), angiotensin II (Ang II), neprilysin, inflammasome, caspase 1, interleukin (IL)1β, and caspase 3 with cardiac histopathological changes, but there was a significant decrease in the antioxidants and blood pressure (BP). Co-administration of Sac/Val could obviously improve these changes. Interestingly, L-NNA given group showed a decrease in the cardioprotective effect of Sac/Val. Sac/Val could ameliorate sepsis induced cardiac damage via inhibition of Ang II and neprilysin with anti-inflammatory, anti-oxidant and anti-apoptotic properties.
Collapse
Affiliation(s)
| | - Maram El-Hussieny
- Department of Pathology, Faculty of Medicine, Minia University, 61511, El-Minia, Egypt.
| | - Asmaa M A Bayoumi
- Department of Biochemistry, Faculty of Pharmacy, Minia University, 61519, El-Minia, Egypt.
| | - Wedad M Abdelraheem
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Minia University, 61511, El-Minia, Egypt.
| | - Elshymaa A Abdel-Hakeem
- Department of Medical Physiology, Faculty of Medicine, Minia University, 61511, El-Minia, Egypt.
| | - Sayed Shehata
- Department of Cardiology, Faculty of Medicine, Minia University, 61511, El-Minia, Egypt.
| |
Collapse
|
35
|
Ziveri J, Le Guennec L, Dos Santos Souza I, Barnier JP, Walter SM, Diallo Y, Smail Y, Le Seac'h E, Bouzinba-Segard H, Faure C, Morand PC, Carel I, Perriere N, Schmitt T, Izac B, Letourneur F, Coureuil M, Rattei T, Nassif X, Bourdoulous S. Angiopoietin-like 4 protects against endothelial dysfunction during bacterial sepsis. Nat Microbiol 2024; 9:2434-2447. [PMID: 39103571 DOI: 10.1038/s41564-024-01760-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/12/2024] [Indexed: 08/07/2024]
Abstract
Loss of endothelial integrity and vascular leakage are central features of sepsis pathogenesis; however, no effective therapeutic mechanisms for preserving endothelial integrity are available. Here we show that, compared to dermal microvessels, brain microvessels resist infection by Neisseria meningitidis, a bacterial pathogen that causes sepsis and meningitis. By comparing the transcriptional responses to infection in dermal and brain endothelial cells, we identified angiopoietin-like 4 as a key factor produced by the brain endothelium that preserves blood-brain barrier integrity during bacterial sepsis. Conversely, angiopoietin-like 4 is produced at lower levels in the peripheral endothelium. Treatment with recombinant angiopoietin-like 4 reduced vascular leakage, organ failure and death in mouse models of lethal sepsis and N. meningitidis infection. Protection was conferred by a previously uncharacterized domain of angiopoietin-like 4, through binding to the heparan proteoglycan, syndecan-4. These findings reveal a potential strategy to prevent endothelial dysfunction and improve outcomes in patients with sepsis.
Collapse
Affiliation(s)
- Jason Ziveri
- Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | - Loïc Le Guennec
- Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | | | - Jean-Philipe Barnier
- Institut Necker Enfants Malades, Université Paris Cité, CNRS, Inserm, Paris, France
| | - Samuel M Walter
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Doctoral School in Microbiology and Environmental Science, University of Vienna, Vienna, Austria
| | - Youssouf Diallo
- Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | - Yasmine Smail
- Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | - Elodie Le Seac'h
- Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | | | - Camille Faure
- Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | | | - Irié Carel
- Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | | | | | - Brigitte Izac
- Institut Cochin, Université Paris Cité, CNRS, Inserm, Paris, France
| | | | - Mathieu Coureuil
- Institut Necker Enfants Malades, Université Paris Cité, CNRS, Inserm, Paris, France
| | - Thomas Rattei
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Doctoral School in Microbiology and Environmental Science, University of Vienna, Vienna, Austria
| | - Xavier Nassif
- Institut Necker Enfants Malades, Université Paris Cité, CNRS, Inserm, Paris, France
| | | |
Collapse
|
36
|
Shaheen N, Miao J, Li D, Xia B, Baoyinna B, Zhao Y, Zhao J. Indole-3-Acetic Acid Protects Against Lipopolysaccharide-induced Endothelial Cell Dysfunction and Lung Injury through the Activation of USP40. Am J Respir Cell Mol Biol 2024; 71:307-317. [PMID: 38761166 PMCID: PMC11376244 DOI: 10.1165/rcmb.2024-0159oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/18/2024] [Indexed: 05/20/2024] Open
Abstract
Lung microvascular endothelial cell (EC) dysfunction is the pathological hallmark of acute respiratory distress syndrome. Heat shock protein 90 (HSP90) is a key regulator in control of endothelial barrier disruption and inflammation. Our recent study has demonstrated that ubiquitin-specific peptidase 40 (USP40) preserves endothelial integrity by targeting HSP90β for its deubiquitination and inactivation. Indole-3-acetic acid (IAA), a plant hormone of the auxin class, can also be catabolized from dietary tryptophan by the intestinal microbiota. Accumulating evidence suggests that IAA reduces oxidative stress and inflammation and promotes intestinal barrier function. However, little is known about the role of IAA in endothelial cells and acute lung injury. In this study, we investigated the role of IAA in lung endothelial cell function in the context of acute lung injury. IAA exhibited EC barrier protection against LPS-induced reduction in transendothelial electrical resistance and inflammatory responses. The underlying mechanism of IAA on EC protective effects was investigated by examining the influence of IAA on degrees of HSP90 ubiquitination and USP40 activity. We identified that IAA, acting as a potential activator of USP40, reduces HSP90 ubiquitination, thereby protecting against LPS-induced inflammation in human lung microvascular endothelial cells as well as alleviating experimental lung injury. Furthermore, the EC protective effects of IAA against LPS-induced EC dysfunction and lung injury were abolished in USP40-deficient human lung microvascular endothelial cell and lungs of USP40 EC-specific knockout (USP40cdh5-ECKO) mice. Taken together, this study reveals that IAA protects against LPS-induced EC dysfunction and lung injury through the activation of USP40.
Collapse
Affiliation(s)
- Nargis Shaheen
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, and
| | - Jiaxing Miao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, and
| | - Donna Li
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, and
| | - Boyu Xia
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, and
| | - Boina Baoyinna
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, and
| | - Yutong Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, and
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Jing Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, and
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
37
|
Tang F, Zhao XL, Xu LY, Zhang JN, Ao H, Peng C. Endothelial dysfunction: Pathophysiology and therapeutic targets for sepsis-induced multiple organ dysfunction syndrome. Biomed Pharmacother 2024; 178:117180. [PMID: 39068853 DOI: 10.1016/j.biopha.2024.117180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Sepsis and septic shock are critical medical conditions characterized by a systemic inflammatory response to infection, significantly contributing to global mortality rates. The progression to multiple organ dysfunction syndrome (MODS) represents the most severe complication of sepsis and markedly increases clinical mortality. Central to the pathophysiology of sepsis, endothelial cells play a crucial role in regulating microcirculation and maintaining barrier integrity across various organs and tissues. Recent studies have underscored the pivotal role of endothelial function in the development of sepsis-induced MODS. This review aims to provide a comprehensive overview of the pathophysiology of sepsis-induced MODS, with a specific focus on endothelial dysfunction. It also compiles compelling evidence regarding potential small molecules that could attenuate sepsis and subsequent multi-organ damage by modulating endothelial function. Thus, this review serves as an essential resource for clinical practitioners involved in the diagnosing, managing, and providing intensive care for sepsis and associated multi-organ injuries, emphasizing the importance of targeting endothelial cells to enhance outcomes of the patients.
Collapse
Affiliation(s)
- Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao-Lan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li-Yue Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing-Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
38
|
Carrara M, Campitelli R, Guberti D, Monge Garcia MI, Ferrario M. The role of pulse wave analysis indexes for critically ill patients: a narrative review. Physiol Meas 2024; 45:08TR01. [PMID: 39094611 DOI: 10.1088/1361-6579/ad6acf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/02/2024] [Indexed: 08/04/2024]
Abstract
Objective.Arterial pulse wave analysis (PWA) is now established as a powerful tool to investigate the cardiovascular system, and several clinical studies have shown how PWA can provide valuable prognostic information over and beyond traditional cardiovascular risk factors. Typically these techniques are applied to chronic conditions, such as hypertension or aging, to monitor the slow structural changes of the vascular system which lead to important alterations of the arterial PW. However, their application to acute critical illness is not currently widespread, probably because of the high hemodynamic instability and acute dynamic alterations affecting the cardiovascular system of these patients.Approach.In this work we propose a review of the physiological and methodological basis of PWA, describing how it can be used to provide insights into arterial structure and function, cardiovascular biomechanical properties, and to derive information on wave propagation and reflection.Main results.The applicability of these techniques to acute critical illness, especially septic shock, is extensively discussed, highlighting the feasibility of their use in acute critical patients and their role in optimizing therapy administration and hemodynamic monitoring.Significance.The potential for the clinical use of these techniques lies in the ease of computation and availability of arterial blood pressure signals, as invasive arterial lines are commonly used in these patients. We hope that the concepts illustrated in the present review will soon be translated into clinical practice.
Collapse
Affiliation(s)
- Marta Carrara
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Riccardo Campitelli
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Diletta Guberti
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - M Ignacio Monge Garcia
- Intensive Care Department, Hospital Universitario SAS de Jerez, Jerez de la Frontera, Spain
| | - Manuela Ferrario
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
39
|
Huang X, Fei Y, Qiu X, Qian T, Shang Q, Cui J, Song Y, Sheng S, Xiao W, Yu Q, Wang T, Wang X. MiR-625-5p is a potential therapeutic target in sepsis by regulating CXCL16/CXCR6 axis and endothelial barrier. Int Immunopharmacol 2024; 137:112508. [PMID: 38889512 DOI: 10.1016/j.intimp.2024.112508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND MicroRNA plays an important role in the progression of sepsis. We found a significant increase of in miR-625-5p expression in the blood of patients with sepsis, and lipopolysaccharide (LPS)-stimulated EA.hy926 cells. To date, little is known about the specific biological function of miR-625-5p in sepsis. METHODS Changes in miR-625-5p expression were verified through quantitative real-time polymerase chain reaction in 45 patients with sepsis or septic shock and 30 healthy subjects. In vitro, EA.hy926 cells were treated with LPS. Transendothelial electrical resistance assay and FITC-dextran were used in evaluating endothelial barrier function. RESULTS Herein, patients with sepsis or septic shock had significantly higher miR-625-5p expression levels, chemokine (C-X-C motif) ligand 16 (CXCL16) levels, and glycocalyx components than the healthy controls, and miR-625-5p level was positively correlated with disease. Kaplan-Meier analysis demonstrated a strong association between miR-625-5p level and 28-day mortality. Furthermore, the miR-625-5p inhibitor significantly alleviated LPS-induced endothelial barrier injury in vitro. Then, miR-625-5p positively regulated CXCL16 and down-regulated miR-625-5p attenuated CXCL16 transcription and expression in EA.hy926 cells. CXCL16 knockout significantly alleviated vascular barrier dysfunction in the LPS-induced EA.hy926 cells. sCXCL16 treatment in EA.hy926 cells significantly increased endothelial hyperpermeability by disrupting endothelial glycocalyx, tight junction proteins, and adherens junction proteins through the modulation of C-X-C chemokine receptor type 6 (CXCR6). CONCLUSIONS Increase in miR-625-5p level may be an effective biomarker for predicting 28-day mortality in patients with sepsis/septic shock. miR-625-5p is a critical pathogenic factor for endothelial barrier dysfunction in LPS-induced EA.hy926 cells because it activates the CXCL16/CXCR6 axis.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yuxin Fei
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaoyu Qiu
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Tiantian Qian
- Department of Respiratory Medicine, Ji'nan Zhangqiu District People's Hospital, No. 1920 Mingshuihuiquan Road, Ji'nan, 250200, Shandong, China
| | - Quanmei Shang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jinfeng Cui
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yutong Song
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shurui Sheng
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wenhan Xiao
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Qilin Yu
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Tao Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Xiaozhi Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
40
|
Chiscano-Camón L, Ruiz-Sanmartin A, Bajaña I, Bastidas J, Lopez-Martinez R, Franco-Jarava C, Gonzalez JJ, Larrosa N, Riera J, Nuvials-Casals X, Ruiz-Rodríguez JC, Ferrer R. Current perspectives in the management of sepsis and septic shock. Front Med (Lausanne) 2024; 11:1431791. [PMID: 39211340 PMCID: PMC11358069 DOI: 10.3389/fmed.2024.1431791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Within patients with sepsis, there exists significant heterogeneity, and while all patients should receive conventional therapy, there are subgroups of patients who may benefit from specific therapies, often referred to as rescue therapies. Therefore, the identification of these specific patient subgroups is crucial and lays the groundwork for the application of precision medicine based on the development of targeted interventions. Over the years, efforts have been made to categorize sepsis into different subtypes based on clinical characteristics, biomarkers, or underlying mechanisms. For example, sepsis can be stratified into different phenotypes based on the predominant dysregulated host response. These phenotypes can range from hyperinflammatory states to immunosuppressive states and even mixed phenotypes. Each phenotype may require different therapeutic approaches to improve patient outcomes. Rescue strategies for septic shock may encompass various interventions, such as immunomodulatory therapies, extracorporeal support (e.g., ECMO), or therapies targeted at specific molecular or cellular pathways involved in the pathophysiology of sepsis. In recent years, there has been growing interest in precision medicine approaches to sepsis and phenotype identification. Precision medicine aims to tailor treatments to each individual patient based on their unique characteristics and disease mechanisms.
Collapse
Affiliation(s)
- Luis Chiscano-Camón
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Departament de Medicina, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Adolf Ruiz-Sanmartin
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ivan Bajaña
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Juliana Bastidas
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Rocio Lopez-Martinez
- Immunology Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Clara Franco-Jarava
- Immunology Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Juan José Gonzalez
- Microbiology Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Nieves Larrosa
- Microbiology Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Jordi Riera
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Xavier Nuvials-Casals
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Juan Carlos Ruiz-Rodríguez
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Departament de Medicina, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Ricard Ferrer
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Departament de Medicina, Universitat Autonoma de Barcelona, Barcelona, Spain
| |
Collapse
|
41
|
Ford VJ, Applefeld WN, Wang J, Sun J, Solomon SB, Sidenko S, Feng J, Sheffield C, Klein HG, Yu Z, Torabi‐Parizi P, Danner RL, Sachdev V, Solomon MA, Chen MY, Natanson C. Cardiac Magnetic Resonance Studies in a Large Animal Model That Simulates the Cardiac Abnormalities of Human Septic Shock. J Am Heart Assoc 2024; 13:e034026. [PMID: 39101510 PMCID: PMC11964030 DOI: 10.1161/jaha.123.034026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/30/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Septic shock is associated with increases in end-diastolic volume (EDV) and decreases in ejection fraction that reverse within 10 days. Nonsurvivors do not develop EDV increases. The mechanism is unknown. METHODS AND RESULTS Purpose-bred beagles (n=33) were randomized to receive intrabronchial Staphylococcus aureus or saline. Over 96 hours, cardiac magnetic resonance imaging and echocardiograms were performed. Tissue was obtained at 66 hours. From 0 to 96 hours after bacterial challenge, septic animals versus controls had significantly increased left ventricular wall edema (6%) and wall thinning with loss of mass (15%). On histology, the major finding was nonocclusive microvascular injury with edema in myocytes, the interstitium, and endothelial cells. Edema was associated with significant worsening of biventricular ejection fractions, ventricular-arterial coupling, and circumferential strain. Early during sepsis, (0-24 hours), the EDV decreased; significantly more in nonsurvivors (ie, greater diastolic dysfunction). From 24 to 48 hours, septic animals' biventricular chamber sizes increased; in survivors significantly greater than baseline and nonsurvivors, whose EDVs were not different from baseline. Preload, afterload, or heart rate differences did not explain these differential changes. CONCLUSIONS The cardiac dysfunction of sepsis is associated with wall edema. In nonsurvivors, at 0 to 24 hours, sepsis induces a more severe diastolic dysfunction, further decreasing chamber size. The loss of left ventricular mass with wall thinning in septic survivors may, in part, explain the EDV increases from 24 to 48 hours because of a potentially reparative process removing damaged wall tissue. Septic cardiomyopathy is most consistent with a nonocclusive microvascular injury resulting in edema causing reversible systolic and diastolic dysfunction with more severe diastolic dysfunction being associated with a decreased EDV and death.
Collapse
MESH Headings
- Animals
- Dogs
- Disease Models, Animal
- Shock, Septic/physiopathology
- Shock, Septic/complications
- Stroke Volume
- Magnetic Resonance Imaging
- Edema, Cardiac/physiopathology
- Edema, Cardiac/pathology
- Edema, Cardiac/diagnostic imaging
- Ventricular Function, Left
- Time Factors
- Humans
- Staphylococcal Infections/complications
- Staphylococcal Infections/physiopathology
- Echocardiography
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/etiology
- Male
Collapse
Affiliation(s)
- Verity J. Ford
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | - Willard N. Applefeld
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- Division of CardiologyDuke University Medical CenterDurhamNCUSA
| | - Jeffrey Wang
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- Emory UniversityAtlantaGAUSA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | - Steven B. Solomon
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | - Stanislav Sidenko
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Jing Feng
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | | | - Harvey G. Klein
- Department of Transfusion Medicine, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | - Zu‐Xi Yu
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | | | - Robert L. Danner
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Vandana Sachdev
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Michael A. Solomon
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Marcus Y. Chen
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
42
|
Salvail W, Salvail D, Chagnon F, Lesur O. Apelin-13 administration allows for norepinephrine sparing in a rat model of cecal ligation and puncture-induced septic shock. Intensive Care Med Exp 2024; 12:68. [PMID: 39103658 DOI: 10.1186/s40635-024-00650-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 07/21/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Infusion of exogenous catecholamines (i.e., norepinephrine [NE] and dobutamine) is a recommended treatment for septic shock with myocardial dysfunction. However, sustained catecholamine infusion is linked to cardiac toxicity and impaired responsiveness. Several pre-clinical and clinical studies have investigated the use of alternative vasopressors in the treatment of septic shock, with limited benefits and generally no effect on mortality. Apelin-13 (APL-13) is an endogenous positive inotrope and vasoactive peptide and has been demonstrated cardioprotective with vasomodulator and sparing life effects in animal models of septic shock. A primary objective of this study was to evaluate the NE-sparing effect of APL-13 infusion in an experimental sepsis-induced hypotension. METHODS For this goal, sepsis was induced by cecal ligation and puncture (CLP) in male rats and the arterial blood pressure (BP) monitored continuously via a carotid catheter. Monitoring, fluid resuscitation and experimental treatments were performed on conscious animals. Based on pilot assays, normal saline fluid resuscitation (2.5 mL/Kg/h) was initiated 3 h post-CLP and maintained up to the endpoint. Thus, titrated doses of NE, with or without fixed-doses of APL-13 or the apelin receptor antagonist F13A co-infusion were started when 20% decrease of systolic BP (SBP) from baseline was achieved, to restore SBP values ≥ 115 ± 1.5 mmHg (baseline average ± SEM). RESULTS A reduction in mean NE dose was observed with APL-13 but not F13A co-infusion at pre-determined treatment time of 4.5 ± 0.5 h (17.37 ± 1.74 µg/Kg/h [APL-13] vs. 25.64 ± 2.61 µg/Kg/h [Control NE] vs. 28.60 ± 4.79 µg/Kg/min [F13A], P = 0.0491). A 60% decrease in NE infusion rate over time was observed with APL-13 co-infusion, (p = 0.008 vs NE alone), while F13A co-infusion increased the NE infusion rate over time by 218% (p = 0.003 vs NE + APL-13). Associated improvements in cardiac function are likely mediated by (i) enhanced left ventricular end-diastolic volume (0.18 ± 0.02 mL [Control NE] vs. 0.30 ± 0.03 mL [APL-13], P = 0.0051), stroke volume (0.11 ± 0.01 mL [Control NE] vs. 0.21 ± 0.01 mL [APL-13], P < 0.001) and cardiac output (67.57 ± 8.63 mL/min [Control NE] vs. 112.20 ± 8.53 mL/min [APL-13], P = 0.0036), and (ii) a reduced effective arterial elastance (920.6 ± 81.4 mmHg/mL/min [Control NE] vs. 497.633.44 mmHg/mL/min. [APL-13], P = 0.0002). APL-13 administration was also associated with a decrease in lactate levels compared to animals only receiving NE (7.08 ± 0.40 [Control NE] vs. 4.78 ± 0.60 [APL-13], P < 0.01). CONCLUSION APL-13 exhibits NE-sparing benefits in the treatment of sepsis-induced shock, potentially reducing deleterious effects of prolonged exogenous catecholamine administration.
Collapse
Affiliation(s)
- William Salvail
- Centre de Recherche Clinique du CHU Sherbrooke (CRCHUS), CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- IPS Therapeutique Inc., Sherbrooke, QC, Canada
| | | | - Frédéric Chagnon
- Centre de Recherche Clinique du CHU Sherbrooke (CRCHUS), CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Olivier Lesur
- Centre de Recherche Clinique du CHU Sherbrooke (CRCHUS), CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Département de Soins Intensifs et Service de PneumologieCHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 3001 12th Avenue Nord, SherbrookeSherbrooke, QC, J1H 5N4, Canada.
- Département de Médecine, CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
43
|
Dhande D, Dhok A, Anjankar A, Nagpure S, Ganjare R. The Role of Mycobacterium indicus pranii in Sepsis Management: A Comprehensive Review of Clinical Outcomes and Therapeutic Potential. Cureus 2024; 16:e66772. [PMID: 39268263 PMCID: PMC11392011 DOI: 10.7759/cureus.66772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Sepsis is a critical condition characterized by a dysregulated immune response to infection, leading to systemic inflammation, multi-organ failure, and high mortality rates. Current treatments primarily involve antibiotics and supportive care, which address the infection and stabilize hemodynamics but do not directly modulate the inflammatory response. This limitation highlights the need for novel therapeutic approaches. This review aims to evaluate the role of Mycobacterium indicus pranii (MIP) in sepsis management, focusing on its clinical outcomes and therapeutic potential. By examining preclinical and clinical evidence, we seek to understand the efficacy, safety, and practical applications of MIP in treating sepsis. A comprehensive review of existing literature was conducted, including preclinical studies, clinical trials, and case reports involving MIP. The review synthesizes findings related to its mechanism of action, therapeutic efficacy, and safety profile. MIP has demonstrated significant immunomodulatory effects, including enhancing innate and adaptive immune responses and reducing excessive inflammation. Clinical trials have shown promising results, with MIP improving clinical outcomes and reducing sepsis-related complications. The agent's unique ability to modulate the cytokine storm associated with sepsis positions it as a potential adjunctive therapy. MIP offers a novel approach to managing sepsis by addressing immune dysregulation and inflammation. The evidence suggests that MIP could be a valuable adjunct to current treatments, improving patient outcomes and addressing some limitations of conventional therapies. Further research is needed to establish its role in clinical practice and to optimize treatment protocols.
Collapse
Affiliation(s)
- Devshree Dhande
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Archana Dhok
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ashish Anjankar
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | | | - Roshani Ganjare
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
44
|
Cherbi M, Merdji H, Labbé V, Bonnefoy E, Lamblin N, Roubille F, Levy B, Lim P, Khachab H, Schurtz G, Harbaoui B, Vanzetto G, Combaret N, Marchandot B, Lattuca B, Biendel-Picquet C, Leurent G, Gerbaud E, Puymirat E, Bonello L, Delmas C. Cardiogenic shock and infection: A lethal combination. Arch Cardiovasc Dis 2024; 117:470-479. [PMID: 39048471 DOI: 10.1016/j.acvd.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Cardiogenic shock and sepsis are severe haemodynamic states that are frequently present concomitantly, leading to substantial mortality. Despite its frequency and clinical significance, there is a striking lack of literature on the outcomes of combined sepsis and cardiogenic shock. METHODS FRENSHOCK was a prospective registry including 772 patients with cardiogenic shock from 49 centres. The primary endpoint was 1-month all-cause mortality. Secondary endpoints included heart transplantation, ventricular assistance device and all-cause death rate at 1year. RESULTS Among the 772 patients with cardiogenic shock included, 92 cases were triggered by sepsis (11.9%), displaying more frequent renal and hepatic acute injuries, with lower mean arterial pressure. Patients in the sepsis group required broader use of dobutamine (90.1% vs. 81.2%; P=0.16), norepinephrine (72.5% vs. 50.8%; P<0.01), renal replacement therapy (29.7% vs. 14%; P<0.01), non-invasive ventilation (36.3% vs. 24.4%; P=0.09) and invasive ventilation (52.7% vs. 35.9%; P=0.02). Sepsis-triggered cardiogenic shock resulted in higher 1-month (41.3% vs. 24.0%; adjusted hazard ratio: 1.94, 95% confidence interval: 1.36-2.76; P<0.01) and 1-year (62.0% vs. 42.9%; adjusted hazard ratio 1.75, 95% confidence interval 1.32-2.33; P<0.01) all-cause death rates. No significant difference was found at 1year for heart transplantation or ventricular assistance device (8.7% vs. 10.3%; adjusted odds ratio 0.72, 95% confidence interval 0.32-1.64; P=0.43). In patients with sepsis-triggered cardiogenic shock, neither the presence of a preexisting cardiomyopathy nor the co-occurrence of other cardiogenic shock triggers had any additional impact on death. CONCLUSIONS The association between sepsis and cardiogenic shock represents a common high-risk scenario, leading to higher short- and long-term death rates, regardless of the association with other cardiogenic shock triggers or the presence of preexisting cardiomyopathy.
Collapse
Affiliation(s)
- Miloud Cherbi
- Intensive Cardiac Care Unit, Rangueil University Hospital, 31059 Toulouse, France; Institute of Metabolic and Cardiovascular Diseases (I2MC), Inserm UMR-1048, 31432 Toulouse, France
| | - Hamid Merdji
- Medical Intensive Care Unit, CHU de Strasbourg, 67000 Strasbourg, France
| | - Vincent Labbé
- Cardiology Department, Hôpital Tenon, AP-HP, 75020 Paris, France
| | - Eric Bonnefoy
- Intensive Cardiac Care Unit, Lyon University Hospital, 69500 Bron, France
| | - Nicolas Lamblin
- Urgences et Soins Intensifs de Cardiologie, CHU de Lille, University of Lille, Inserm U1167, 59000 Lille, France
| | - François Roubille
- PhyMedExp, Université de Montpellier, Inserm, CNRS, Cardiology Department, CHU de Montpellier, 34295 Montpellier, France
| | - Bruno Levy
- CHRU Nancy, Réanimation Médicale Brabois, 54511 Vandœuvre-Lès-Nancy, France
| | - Pascal Lim
- Université Paris-Est Créteil, Inserm, IMRB, 94010 Créteil, France; Service de Cardiologie, Hôpital Universitaire Henri-Mondor, AP-HP, 94010 Créteil, France
| | - Hadi Khachab
- Intensive Cardiac Care Unit, Department of Cardiology, CH d'Aix-en-Provence, 13616 Aix-en-Provence, France
| | - Guillaume Schurtz
- PhyMedExp, Université de Montpellier, Inserm, CNRS, Cardiology Department, CHU de Montpellier, 34295 Montpellier, France
| | - Brahim Harbaoui
- Cardiology Department, Hôpital Croix-Rousse and Hôpital Lyon Sud, Hospices Civils de Lyon, 69004 Lyon, France; University of Lyon, CREATIS UMR 5220, Inserm U1044, INSA-15 Lyon, 69621 Villeurbanne, France
| | - Gerald Vanzetto
- Department of Cardiology, Hôpital de Grenoble, 38700 La Tronche, France
| | - Nicolas Combaret
- Department of Cardiology, CHU de Clermont-Ferrand, CNRS, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Benjamin Marchandot
- Université de Strasbourg, Pôle d'Activité Médico-Chirurgicale Cardiovasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, 67091 Strasbourg, France
| | - Benoit Lattuca
- Department of Cardiology, Nîmes University Hospital, Montpellier University, 30900 Nîmes, France
| | - Caroline Biendel-Picquet
- Intensive Cardiac Care Unit, Rangueil University Hospital, 31059 Toulouse, France; Institute of Metabolic and Cardiovascular Diseases (I2MC), Inserm UMR-1048, 31432 Toulouse, France
| | - Guillaume Leurent
- Department of Cardiology, CHU de Rennes, Inserm, LTSI UMR 1099, Université de Rennes 1, 35000 Rennes, France
| | - Edouard Gerbaud
- Intensive Cardiac Care Unit and Interventional Cardiology, Hôpital Cardiologique du Haut-Lévêque, 33604 Pessac, France; Bordeaux Cardio-Thoracic Research Centre, U1045, Bordeaux University, Hôpital Xavier-Arnozan, 33600 Pessac, France
| | - Etienne Puymirat
- Department of Cardiology, Hôpital Européen Georges-Pompidou, AP-HP, 75015 Paris, France; Université de Paris, 75006 Paris, France
| | - Laurent Bonello
- Aix-Marseille Université, 13385 Marseille, France; Intensive Care Unit, Department of Cardiology, Hôpital Nord, AP-HM, 13385 Marseille, France; Mediterranean Association for Research and Studies in Cardiology (MARS Cardio), 13015 Marseille, France
| | - Clément Delmas
- Intensive Cardiac Care Unit, Rangueil University Hospital, 31059 Toulouse, France; Institute of Metabolic and Cardiovascular Diseases (I2MC), Inserm UMR-1048, 31432 Toulouse, France.
| |
Collapse
|
45
|
Kappelmayer J, Debreceni IB, Fejes Z, Nagy B. Inflammation, Sepsis, and the Coagulation System. Hamostaseologie 2024; 44:268-276. [PMID: 38354835 DOI: 10.1055/a-2202-8544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Sepsis has been a major health problem for centuries and it is still the leading cause of hospital deaths. Several studies in the past decades have identified numerous biochemical abnormalities in severe cases, and many of these studies provide evidence of the perturbation of the hemostatic system. This can result in complications, such as disseminated intravascular coagulation that can lead to multiorgan failure. Nevertheless, large clinical studies have demonstrated that the simple approach of inhibiting the coagulation processes by any means fails to provide significant improvement in the survival of septic patients. A cause of this failure could be the fact that in sepsis the major clinical problems result not primarily from the presence of the infective agent or enhanced coagulation but from the complex dysregulated systemic host response to pathogens. If this overt reaction is not fully deciphered, appropriate interference is highly unlikely and any improvement by conventional therapeutic interventions would be limited. Cellular activation in sepsis can be targeted by novel approaches like inhibition of the heterotypic cellular interactions of blood cells by targeting surface receptors or posttranscriptional control of the hemostatic system by noncoding ribonucleic acid (RNA) molecules. Stable RNA molecules can affect the expression of several proteins. Thus, it can be anticipated that modulation of microRNA production would result in a multitude of effects that may be beneficial in septic cases. Here, we highlight some of the recent diagnostic possibilities and potential novel routes of the dysregulated host response.
Collapse
Affiliation(s)
- János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildikó Beke Debreceni
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
46
|
Ahmad D, Linares I, Pietropaoli A, Waugh RE, McGrath JL. Sided Stimulation of Endothelial Cells Modulates Neutrophil Trafficking in an In Vitro Sepsis Model. Adv Healthc Mater 2024; 13:e2304338. [PMID: 38547536 PMCID: PMC11338706 DOI: 10.1002/adhm.202304338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Indexed: 04/09/2024]
Abstract
While the role of dysregulated polymorphonuclear leukocyte (PMN) transmigration in septic mediated tissue damage is well documented, strategies to mitigate aberrant transmigration across endothelium have yet to yield viable therapeutics. Recently, microphysiological systems (MPS) have emerged as novel in vitro mimetics that facilitate the development of human models of disease. With this advancement, aspects of endothelial physiology that are difficult to assess with other models can be directly probed. In this study, the role of endothelial cell (EC) apicobasal polarity on leukocyte trafficking response is evaluated with the µSiM-MVM (microphysiological system enabled by a silicon membrane - microvascular mimetic). Here, ECs are stimulated either apically or basally with a cytokine cocktail to model a septic-like challenge before introducing healthy donor PMNs into the device. Basally oriented stimulation generated a stronger PMN transmigratory response versus apical stimulation. Importantly, healthy PMNs are unable to migrate towards a bacterial peptide chemoattractant when ECs are apically stimulated, which mimics the attenuated PMN chemotaxis seen in sepsis. Escalating the apical inflammatory stimulus by a factor of five is necessary to elicit high PMN transmigration levels across endothelium. These results demonstrate that EC apicobasal polarity modulates PMN transmigratory behavior and provides insight into the mechanisms underlying sepsis.
Collapse
Affiliation(s)
- Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Isabelle Linares
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Anthony Pietropaoli
- Department of Medicine, Pulmonary Diseases and Critical Care at the University of Rochester, Rochester, NY, 14627, USA
| | - Richard E Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| |
Collapse
|
47
|
Li X, Chen Y, Yuan Q, Zhou H, Lu L, Guo R. Neutrophil-to-lymphocyte ratio, monocyte-to-lymphocyte ratio, platelet-to-lymphocyte ratio associated with 28-day all-cause mortality in septic patients with coronary artery disease: a retrospective analysis of MIMIC-IV database. BMC Infect Dis 2024; 24:749. [PMID: 39075364 PMCID: PMC11288105 DOI: 10.1186/s12879-024-09516-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/17/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND High Neutrophil-to-Lymphocyte Ratio (NLR), Monocyte-to-Lymphocyte Ratio (MLR), Platelet-to-Lymphocyte Ratio (PLR) were associated with worse prognosis of patients with sepsis. In-hospital mortality has been reported to be higher in patients with coronary artery disease (CAD) and sepsis than those with sepsis alone. However, the relationship between NLR, MLR, PLR and mortality in septic patients with coronary artery disease (CAD) remains unclear. The study aimed to explore the association between NLR, MLR, PLR and 28-day all-cause mortality in septic patients with CAD. METHODS We performed an observational cohort study of septic patients with CAD from the Medical Information Mart for Intensive Care (MIMIC)-IV database between 2008 and 2019. The patients were categorized by three group (Q1: low levels, Q2: medium levels, Q3: high levels) based on tertiles of NLR, MLR, and PLR. The associations between NLR, MLR, PLR and 28-day all-cause mortality were examined using the Cox proportional hazards model. Subsequently, we applied receiver operating characteristic (ROC) analysis for predicting 28-day mortality in septic patients with CAD by combining NLR, MLR and PLR with the modified sequential organ failure assessment (mSOFA) scores. RESULTS Overall 1,175 septic patients with CAD were included in the study. Observed all-cause mortality rates in 28 days were 27.1%. Multivariate Cox proportional hazards regression analysis results showed that 28-day all-cause mortality of septic patients with CAD was significantly related to rising NLR levels (adjusted hazard ratio [aHR]: 1.02; 95% confidence interval [CI]: 1.01-1.02; P < 0.001), MLR levels (aHR: 1.29; 95%CI: 1.18-1.41; P < 0.001), and PLR levels (aHR: 1.0007; 95%CI: 1.0004-1.0011; P < 0.001). Meanwhile, the higher levels (Q3) group of NLR, MLR, and PLR also had a higher risk of 28-day all-cause mortality than the lower (Q1) group. The area under the ROC curve of NLR, MLR, PLR, and mSOFA score were 0.630 (95%CI 0.595-0.665), 0.611 (95%CI 0.576-0.646), 0.601 (95%CI 0.567-0.636) and 0.718 (95%CI 0.689-0.748), respectively. Combining NLR, MLR, and PLR with mSOFA scores may improve ability of predicting 28-day mortality (AUC: 0.737, 95%CI 0.709-0.766). CONCLUSION Higher levels of NLR, MLR and PLR were associated with 28-day all-cause mortality in septic patients with CAD. Further investigation will be needed to improve understanding of the pathophysiology of this relationship.
Collapse
Affiliation(s)
- Xicong Li
- Department of Cardiology, the 920th Hospital, Kunming Medical University, Kunming, 650032, Yunnan, China
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650032, Yunnan, China
| | - Yubiao Chen
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Qi Yuan
- Department of Cardiology, the 920th Hospital, Kunming Medical University, Kunming, 650032, Yunnan, China
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650032, Yunnan, China
| | - Hongya Zhou
- Department of Cardiology, the 920th Hospital, Kunming Medical University, Kunming, 650032, Yunnan, China
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650032, Yunnan, China
| | - Lifei Lu
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, China.
| | - Ruiwei Guo
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650032, Yunnan, China.
| |
Collapse
|
48
|
Kravitz MS, Kattouf N, Stewart IJ, Ginde AA, Schmidt EP, Shapiro NI. Plasma for prevention and treatment of glycocalyx degradation in trauma and sepsis. Crit Care 2024; 28:254. [PMID: 39033135 PMCID: PMC11265047 DOI: 10.1186/s13054-024-05026-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 07/06/2024] [Indexed: 07/23/2024] Open
Abstract
The endothelial glycocalyx, a gel-like layer that lines the luminal surface of blood vessels, is composed of proteoglycans, glycoproteins, and glycosaminoglycans. The endothelial glycocalyx plays an essential role in vascular homeostasis, and its degradation in trauma and sepsis can lead to microvascular dysfunction and organ injury. While there are no proven therapies for preventing or treating endothelial glycocalyx degradation, some initial literature suggests that plasma may have a therapeutic role in trauma and sepsis patients. Overall, the literature suggesting the use of plasma as a therapy for endothelial glycocalyx degradation is non-clinical basic science or exploratory. Plasma is an established therapy in the resuscitation of patients with hemorrhage for restoration of coagulation factors. However, plasma also contains other bioactive components, including sphingosine-1 phosphate, antithrombin, and adiponectin, which may protect and restore the endothelial glycocalyx, thereby helping to maintain or restore vascular homeostasis. This narrative review begins by describing the endothelial glycocalyx in health and disease: we discuss the overlapping disease mechanisms in trauma and sepsis that lead to its damage and introduce plasma transfusion as a potential therapy for prevention and treatment of endothelial glycocalyx degradation. Second, we review the literature on plasma as an exploratory therapy for endothelial glycocalyx degradation in trauma and sepsis. Third, we discuss the safety of plasma transfusion by reviewing the adverse events associated with plasma and other blood product transfusions, and we examine modern transfusion precautions that have enhanced the safety of plasma transfusion. We conclude that the literature proposes that plasma may have the potential to prevent and treat endothelial glycocalyx degradation in trauma and sepsis, indicating the need for further research.
Collapse
Affiliation(s)
- M S Kravitz
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - N Kattouf
- Department of Emergency Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - I J Stewart
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - A A Ginde
- Department of Emergency Medicine, University of Colorado School of Medicines, Aurora, CO, USA
| | - E P Schmidt
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - N I Shapiro
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
49
|
Tepebaşı MY, Aşcı H, Özmen Ö, Taner R, Temel EN, Garlı S. Cannabidiol ameliorates lipopolysaccharide-induced cardiovascular toxicity by its antioxidant and anti-inflammatory activity via regulating IL-6, Hif1α, STAT3, eNOS pathway. Mol Biol Rep 2024; 51:825. [PMID: 39023749 DOI: 10.1007/s11033-024-09772-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Systemic inflammation causes several organ damage by activating the intracellular signaling mechanisms. Heart and aorta tissues are the structures mostly affected by this situation. By examining underlying processes, this study sought to determine whether cannabidiol (CBD) may have protective effects against the cardiovascular damage brought on by lipopolysaccharide (LPS). MATERIALS AND METHODS A total of 32 female rats were randomly allocated to one of four groups: control, lipopolysaccharide (LPS) (5 mg/kg, i.p., single dose), LPS + CBD (5 mg/kg, i.p., single dose), and CBD groups. The rats were killed six hours after receiving LPS, and tissues from the heart and aorta were taken. Histopathological and immunohistochemical analyzes were performed. Oxidative stress was evaluated biochemically by spectrophotometric method. Expression levels of genes were studied by RT-qPCR method. RESULTS Histopathological analysis of the LPS group showed moderate hyperemia, hemorrhages, edema, inflammation, and myocardial cell damage. There was a slight to moderate increase in Cox-1, G-CSF, and IL-3 immunoexpressions, along with enhanced expressions of IL-6, Hif1α, and STAT3 genes, and decreased expressions of eNOS genes. Additionally, there were increased levels of TOS and decreased TAS levels observed biochemically. CBD treatment effectively reversed and improved all of these observed changes. CONCLUSIONS CBD protects the heart and aorta against systemic inflammation through its antioxidant and anti-inflammatory activity via regulating IL-6, Hif1α, STAT3, and eNOS intracellular pathways.
Collapse
Affiliation(s)
| | - Halil Aşcı
- Department of Medical Pharmacology, Faculty of Medical, Suleyman Demirel University, Isparta, Turkey
| | - Özlem Özmen
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Rümeysa Taner
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, Isparta, Turkey
| | - Esra Nurlu Temel
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medical, Suleyman Demirel University, Isparta, Turkey
| | - Simge Garlı
- Mehmet Akif Ersoy University Experimental Animal Production and Experimental Research Center, Burdur, Turkey
| |
Collapse
|
50
|
Srdić T, Đurašević S, Lakić I, Ružičić A, Vujović P, Jevđović T, Dakić T, Đorđević J, Tosti T, Glumac S, Todorović Z, Jasnić N. From Molecular Mechanisms to Clinical Therapy: Understanding Sepsis-Induced Multiple Organ Dysfunction. Int J Mol Sci 2024; 25:7770. [PMID: 39063011 PMCID: PMC11277140 DOI: 10.3390/ijms25147770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
Sepsis-induced multiple organ dysfunction arises from the highly complex pathophysiology encompassing the interplay of inflammation, oxidative stress, endothelial dysfunction, mitochondrial damage, cellular energy failure, and dysbiosis. Over the past decades, numerous studies have been dedicated to elucidating the underlying molecular mechanisms of sepsis in order to develop effective treatments. Current research underscores liver and cardiac dysfunction, along with acute lung and kidney injuries, as predominant causes of mortality in sepsis patients. This understanding of sepsis-induced organ failure unveils potential therapeutic targets for sepsis treatment. Various novel therapeutics, including melatonin, metformin, palmitoylethanolamide (PEA), certain herbal extracts, and gut microbiota modulators, have demonstrated efficacy in different sepsis models. In recent years, the research focus has shifted from anti-inflammatory and antioxidative agents to exploring the modulation of energy metabolism and gut microbiota in sepsis. These approaches have shown a significant impact in preventing multiple organ damage and mortality in various animal sepsis models but require further clinical investigation. The accumulation of this knowledge enriches our understanding of sepsis and is anticipated to facilitate the development of effective therapeutic strategies in the future.
Collapse
Affiliation(s)
- Tijana Srdić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Siniša Đurašević
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Iva Lakić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Aleksandra Ružičić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Predrag Vujović
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tanja Jevđović
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tamara Dakić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Jelena Đorđević
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tomislav Tosti
- Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sofija Glumac
- School of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (S.G.); (Z.T.)
| | - Zoran Todorović
- School of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (S.G.); (Z.T.)
| | - Nebojša Jasnić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| |
Collapse
|