1
|
Wang X, Zhou H. Impact of antiplatelet therapy on outcomes of sepsis: A systematic review and meta-analysis. PLoS One 2025; 20:e0322293. [PMID: 40299932 PMCID: PMC12040142 DOI: 10.1371/journal.pone.0322293] [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: 10/04/2024] [Accepted: 03/19/2025] [Indexed: 05/01/2025] Open
Abstract
OBJECTIVE Antiplatelet therapy has been studied for its potential benefits in various cardiovascular conditions, but its role in sepsis remains less clear. This review aims to systematically analyse the available evidence on the effects of antiplatelet therapy in sepsis to assess its potential benefits and risks. MATERIAL AND METHODS The studies published until 01st April 2024 from PubMed, Embase and Scopus databases were searched. Pooled effect sizes were reported as relative risks (RR) or weighted mean difference (WMD) with corresponding 95% confidence intervals (CI). Outcomes included mortality, length of intensive care unit (ICU) stay, hospital stay, and the risk of complications. The certainty of evidence was evaluated using GRADE. RESULTS Twenty-one studies were included. Antiplatelet therapy was associated with significantly lower risk of in-hospital mortality (RR 0.76, 95% CI: 0.67, 0.87), and mortality at one (RR 0.77, 95% CI: 0.66, 0.90) and three months (RR 0.77, 95% CI: 0.66, 0.90) follow up. Risk of complications was comparable in all patients (RR 1.01, 95% CI: 0.84, 1.21). ICU stay (in days) (WMD -0.23, 95% CI: -0.53, 0.07; N=7, I2=97.2%) and overall duration of hospital stay (in days) (WMD 0.63, 95% CI: -0.66, 1.92; N=6, I2=93.2%) was also statistically similar among patients who received and did not receive antiplatelet drugs. The certainty of evidence for the outcomes ranged from "low to very low". CONCLUSION Antiplatelet therapy appears safe and significantly lowers the risk of short-term mortality in septic patients. While antiplatelet therapy did not impact the duration of ICU or overall hospital stay, our findings underscore the potential of antiplatelet agents as a beneficial adjunctive therapy in sepsis management.
Collapse
Affiliation(s)
- Xufang Wang
- Department of Critical Care Medicine, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Huifei Zhou
- Department of Critical Care Medicine, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
2
|
Xu Y, Wang J, Yuan R, Qin Z, Long K, Gao P. Targeting the immuno-inflammatory-microbial network: a key strategy for sepsis treatment. Front Immunol 2025; 16:1575516. [PMID: 40297590 PMCID: PMC12034552 DOI: 10.3389/fimmu.2025.1575516] [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: 02/12/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Sepsis is a life-threatening condition caused by a dysregulated host response to infection, remaining a major global health challenge despite clinical advances. Therapeutic challenges arise from antibiotic misuse, incomplete understanding of its complex pathophysiology, and the unresolved interplay of immune dysregulation and microbiota disruption. Investigating microbial homeostasis in the shift from cytokine storm to immunosuppression may elucidate the interplay between microbial metabolites, immune dysfunction, and organ injury, providing a foundation for targeted therapies and drug development. Traditional Chinese Medicine (TCM) has demonstrated significant advantages in mitigating sepsis-associated cytokine storms and modulating gut microbiota homeostasis, offering a promising strategy for developing highly effective and less toxic targeted monomeric compounds. Elucidating the interactions within the immune-inflammation-microbiota network in sepsis paves the way for biomarker-driven personalized therapeutic approaches.
Collapse
Affiliation(s)
- Yue Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | | | | | | | - Peiyang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
Huang R, Lu B, Liu Y, Zhang W, Xu D, Yao Y, Zhu B, Li J, Xian S, Wu G, Xu L, Li L, Tong X, Sun H, Yan J, Chen Y, He H, Liu J, Xiao S, Ji S. Proper early weaning from ventilator assistance influences the overall survival of patients with severe burns: A case-control study. Respir Med 2025; 240:108023. [PMID: 40032161 DOI: 10.1016/j.rmed.2025.108023] [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: 10/10/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Ventilator assistance is crucial in the treatment of severely burned patients. However, there is no consensus on the optimal duration of ventilator assistance, and whether it has an impact on the short - and long-term prognosis of severely burned patients is still unclear. METHODS We incorporated 185 severely burned patients in our study. Kaplan-Meier analysis and Chi-square tests were applied to reveal the associations of days of ventilator assistance with patients' overall survival (OS) and other clinical variables. Multivariable Cox regression was applied, and the model was diagnosed by residual analysis, calibration curve analysis, decision curve analysis, and receiver operating characteristic curve analysis. 144 burned patients in Changhai Cohort were used for external validation. RESULTS Ventilator assistance >26 days was significantly associated with a shorter OS in severely burned patients (P-value <0.001). Besides, "days of ventilator assistance" was significantly correlated with the burn area (P-value <0.001), and the combination of inhalation injury (P-value <0.01), sepsis, DIC, AHF, AKI, ALF, and ARDS (P-value <0.001). The prognostic value of "days of ventilator assistance" was further validated in Changhai Cohort. CONCLUSION Prolonged ventilator assistance was associated with decreased OS in severely burned patients. Under the premise of the recovery of spontaneous respiratory function, early weaning from ventilator assistance might improve the prognosis of patients with severe burns. The findings on the optimal duration of ventilator assistance and its impact on survival in severely burned patients would interest healthcare professionals in respiratory and critical care medicine.
Collapse
Affiliation(s)
- Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Bingnan Lu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Yifan Liu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, People's Republic of China; BGI research, BGI-Hangzhou, 310012, Hangzhou, People's Republic of China
| | - Wei Zhang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Dayuan Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, People's Republic of China; BGI research, BGI-Hangzhou, 310012, Hangzhou, People's Republic of China
| | - Bolin Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China; BGI research, BGI-Hangzhou, 310012, Hangzhou, People's Republic of China
| | - Junqiang Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Guosheng Wu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Long Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Linhui Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Hanlin Sun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Jiale Yan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200126, People's Republic of China
| | - Yang Chen
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Heng He
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China
| | - Jun Liu
- Department of Anesthesiology, Shanghai Pulmonary Hospital affiliated to Tongji University, 507 Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China.
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical SciencesPeople's Republic of China.
| |
Collapse
|
4
|
Zhou Z, Xu D, Huang L, Cui Y, Chen H, Tang J. Farnesoid X Receptor Regulated Sepsis-Induced Abnormal Bile Acid Metabolism via the Fibroblast Growth Factor 15/Fibroblast Growth Factor Receptor 4 Pathway. Immun Inflamm Dis 2025; 13:e70155. [PMID: 40192065 PMCID: PMC11973727 DOI: 10.1002/iid3.70155] [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: 09/02/2024] [Revised: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE The study aims to investigate the mechanism of Farnesoid X receptor (FXR) activation in sepsis-induced abnormal bile acid metabolism and the metabolism status of each bile acid type. METHODS The sepsis mouse model was developed via lipopolysaccharide intraperitoneal injection and confirmed via hematoxylin and eosin (H&E) staining. FXR agonist activated the FXR/fibroblast growth factor (FGF)15/FGFR pathway via quantitative real-time polymerase chain reaction and Western blot. Consequently, metabolomics and bioinformatics analysis were conducted to identify the alterations in each kind of bile acid content following FXR agonist/inhibitor intervention. RESULTS The H&E staining indicated that FXR activation alleviates the liver injury of the sepsis mouse model. The increased FGF15 and FXFR expression levels and decreased CYP7A1 demonstrated FXR/FGF15/FGFR pathway activation following FXR agonist treatment. Furthermore, total bile acid, interleukin (IL)-6, and tumor necrosis factor-α concentrations were downregulated after FXR activation, whereas IL-10 concentration was upregulated, indicating the alleviated effect of FXR agonist in sepsis. Consequently, metabolomics and bioinformatics analysis determined that T-a-MCA were downregulated in both FXR agonist and inhibitor groups, whereas six bile acid types were altered in the control group. CONCLUSION FXR activation was crucial in alleviating sepsis-induced hepatic injury and cholestasis through the FGF15/FGFR signaling pathway, and FXR may act as a potential preventive and intervention target of sepsis.
Collapse
Affiliation(s)
- Ziyang Zhou
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Dan Xu
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Liou Huang
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Yuhui Cui
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Hui Chen
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life ScienceEast China Normal UniversityShanghaiChina
| | - Jianguo Tang
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| |
Collapse
|
5
|
Luo XT, Hu HR, Sun ZD, Zhang LH, Li Y. Multi-omics analysis reveals that low cathepsin S expression aggravates sepsis progression and worse prognosis via inducing monocyte polarization. Front Cell Infect Microbiol 2025; 15:1531125. [PMID: 40115073 PMCID: PMC11922721 DOI: 10.3389/fcimb.2025.1531125] [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: 11/19/2024] [Accepted: 02/12/2025] [Indexed: 03/23/2025] Open
Abstract
Background Monocytes represent a vital cellular subpopulation in the peripheral blood, crucial in the progression of sepsis. Nonetheless, the prognostic role and precise function of monocytes in sepsis are still inadequately understood. Methods Single-cell transcriptomic sequencing and bioinformatics analysis were performed on peripheral blood samples from septic patients to identify key molecules in cell subsets. Subsequently, the expression pattern of this molecule was validated through diverse biological experiments, encompassing quantitative RT-PCR, western blotting, and immunofluorescence. Finally, the functionality of this molecule was evaluated using its specific agonist. Results A total of 22 monocytes-related biomarkers were identified from single-cell and bulk RNA-seq analyses. Initially, LASSO analysis was performed to derive a prognostic signature composed of 4 key genes, including CD14, CTSS, CXCL8 and THBS1. Subsequently, mendelian randomization and survival analysis demonstrated that only CTSS showed crucially protective role in sepsis development and prognosis. Next, CTSS was confirmed to be lower expressed in peripheral monocytes of septic patients. Inflammatory markers (p < 0.05) and migration ability of LPS-activated monocytes were significantly reduced after CTSS agonist. In addition, CTSS agonist decreased the pulmonary tissue monocyte/macrophages infiltration in septic mice. Conclusion Monocyte marker CTSS represent a promising target for the diagnosis and prognosis evaluation of sepsis and plays a critical role in monocytes activation, tissue inflammatory response and macrophages infiltration. Thus, CTSS agonist probably serves as new drug for clinical protection against sepsis.
Collapse
Affiliation(s)
- Xiao-Ting Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hui-Rong Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhen-Dong Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Li-Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan Li
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
6
|
Xu T, Han J, Wang N, Huan Z, Yao H, Ge X. The protective role of kakkalide in sepsis-induced intestinal barrier dysfunction via inhibition of NF-κB pathway activation. J Clin Biochem Nutr 2025; 76:139-147. [PMID: 40151402 PMCID: PMC11936737 DOI: 10.3164/jcbn.24-182] [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: 10/20/2024] [Accepted: 11/16/2024] [Indexed: 03/29/2025] Open
Abstract
Sepsis, a systemic inflammatory response often triggered by infection, can lead to multi-organ failure, with the intestine being one of the most vulnerable organs. The nuclear factor kappa-B (NF-κB) pathway plays a crucial role in immune responses, inflammation, and cell survival, making it central to sepsis-induced intestinal damage. Kakkalide (KA), a bioactive compound known for its anti-inflammatory, cardiovascular, neuroprotective, and anti-diabetic properties, has potential therapeutic effects. However, its impact on sepsis-induced intestinal injury remains unclear. In this study, murine sepsis models were used both in vivo and in vitro to evaluate the protective effects of KA on intestinal histopathology, apoptosis, and inflammation. Results showed that KA significantly reduced intestinal damage and apoptosis, as evidenced by hematoxylin-eosin and TUNEL staining. KA also improved intestinal barrier integrity, as indicated by reduced diamine oxidase activity, d-lactic acid content, and fluorescein isothiocyanate intensity, along with increased expression of zonula occludens-1. Furthermore, KA alleviates inflammation by reducing the levels of tumor necrosis factor-α, interleukin-1β, prostaglandin E2, inducible nitric oxide synthase, and cyclooxygenase-2. Immunofluorescence and Western blot analysis revealed that KA inhibited the sepsis-induced phosphorylation of inhibitor-kappaBα and RelA (P65) and prevented P65's translocation to the nucleus. These findings were confirmed in lipopolysaccharide-induced Caco-2 cells, suggesting that KA protected the intestinal barrier during sepsis by suppressing the NF-κB pathway.
Collapse
Affiliation(s)
- Tongrong Xu
- Department of Critical Care Medicine, Changzhou No.2 People’s Hospital, Gehu Middle Road 68, Changzhou, Jiangsu, People’s Republic of China
| | - Jiahui Han
- Department of Emergency and Critical Care Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Liangxi Road 999, Wuxi, Jiangsu 214000, People’s Republic of China
| | - Nan Wang
- Department of Emergency and Critical Care Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Liangxi Road 999, Wuxi, Jiangsu 214000, People’s Republic of China
| | - Zhirong Huan
- Department of Emergency and Critical Care Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Liangxi Road 999, Wuxi, Jiangsu 214000, People’s Republic of China
| | - Hao Yao
- Department of Emergency and Critical Care Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Liangxi Road 999, Wuxi, Jiangsu 214000, People’s Republic of China
| | - Xin Ge
- Department of Emergency and Critical Care Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Liangxi Road 999, Wuxi, Jiangsu 214000, People’s Republic of China
- Orthopedic Institution of Wuxi City, Liangxi Road 999, Wuxi, Jiangsu 214000, People’s Republic of China
| |
Collapse
|
7
|
Li A, Liu P, Gan J, Fang W, Liu A. Phellodendrine Exerts Protective Effects on Intra-abdominal Sepsis by Inactivating AKT/NF-kB Signaling. Cell Biochem Biophys 2025:10.1007/s12013-024-01658-2. [PMID: 39953352 DOI: 10.1007/s12013-024-01658-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 02/17/2025]
Abstract
Acute kidney injury (AKI) and acute lung injury (ALI) are major complications of intra-abdominal sepsis, leading to increased mortality. Phellodendrine (PHE) is a characteristic and important active ingredient of Phellodendri Cortex, possessing multiple pharmacological properties. This study intends to explore the effect of PHE on intra-abdominal sepsis-induced AKI and ALI. An intra-abdominal infection-induced rat model of sepsis was established by fecal intraperitoneal injection, followed by the administration of PHE. ELISA was used to determine plasma levels of inflammatory cytokines. Hematoxylin-eosin, Periodic acid Schiff, and Masson trichrome staining were employed for histopathological analysis of rat kidney and lung tissues. Western blotting was used to estimate the AKT/NF-κB signaling-related protein levels. The results showed that PHE improved the survival rate of septic rats and reduced plasma levels of proinflammatory cytokines. PHE administration attenuated pathological lesions in the kidneys and lungs of septic rats. Mechanistically, PHE treatment blocked AKT/NF-κB signaling in septic rats' kidneys and lungs. In conclusion, PHE ameliorates intra-abdominal sepsis-induced kidney and lung injury possibly by inactivating AKT/NF-kB signaling.
Collapse
Affiliation(s)
- Ang Li
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng Liu
- Department of Emergency, Wuhan Fourth Hospital, Wuhan, China
| | - Jiaohong Gan
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijun Fang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Anjie Liu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
8
|
Song X, Liu C, Zhang M, Wei W, Yang L, Wang B, Huang Y, Song G, Wang F, Yang Y, Zhao Y, Zhang L, Fu P. The Efficacy and Safety of Continuous Veno-Venous Hemodiafiltration With High Cutoff Membrane Versus High Flux Membrane in Septic Acute Kidney Injury: A Randomized Controlled Study. Artif Organs 2025. [PMID: 39895488 DOI: 10.1111/aor.14963] [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: 09/06/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The application of high cutoff (HCO) membranes for continuous renal replacement therapy remains unclear in septic acute kidney injury (S-AKI) patients. METHODS S-AKI patients who received continuous veno-venous hemodiafiltration (CVVHDF) were randomly assigned to the experimental group (HCO membrane) and the control group (high flux membrane, HF membrane). Interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in serum and waste fluid were measured at 0, 2, 12, and 24 h after CVVHDF initiation and the 28-day mortality. RESULTS Eleven patients were randomized to the HCO group, and 9 patients in the HF group, with a mean age of 54.9 ± 3.2 years and 6 patients (30%) being female. After 24 h of treatment with CVVHDF, there were significant reductions in serum IL-6 and TNF-α concentrations in the HCO group (p = 0.001, 0.015) and HF group (p = 0.004, 0.031). The serum IL-6 reduction rate of the HCO group was significantly higher than that of the HF group (79.21% vs. 42.69%, p = 0.025), while serum TNF-α reduction rates were comparable between the 2 groups. There were no significant changes in serum albumin after 24 h using either HCO membrane (28.7 ± 1.7 g/L vs. 32.7 ± 1.6 g/L, p = 0.138) or HF membrane (29.6 ± 1.1 g/L vs. 32.6 ± 1.3 g/L, p = 0.055). The two groups had similar 24-h filter clotting rates and 28-day mortality. CONCLUSION While CVVHDF with the HCO membrane and HF membrane both achieved significant reductions in serum cytokine levels, the HCO membrane was associated with a greater reduction rate in IL-6 but not in TNF-α. No difference was observed in serum albumin, mortality, or filter clotting. TRIAL REGISTRATION Registry number: ChiCTR2000039725.
Collapse
Affiliation(s)
- Xiaowei Song
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
- Department of Nephrology, The Third People's Hospital of Chengdu, Chengdu, China
| | - Caihong Liu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Min Zhang
- Department of Nephrology, West China Hospital of Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Wei Wei
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Letian Yang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Bo Wang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yongxiu Huang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Guojiao Song
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Fang Wang
- Department of Nephrology, West China Hospital of Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yingying Yang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yuliang Zhao
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Ling Zhang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Ping Fu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Xi W, Wu W, Zhou L, Zhang Q, Yang S, Huang L, Lu Y, Wang J, Chi X, Kang Y. Multifunctional nanoparticles confers both multiple inflammatory mediators scavenging and macrophage polarization for sepsis therapy. Mater Today Bio 2025; 30:101421. [PMID: 39811612 PMCID: PMC11732566 DOI: 10.1016/j.mtbio.2024.101421] [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: 08/06/2024] [Revised: 11/29/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Sepsis is a serious and life-threatening condition, which can lead to organ failure and death clinically. Abnormally increased cell-free DNA (cfDNA) and inflammatory cytokines are involved in the development and progression of sepsis. Thus, cfDNA clearance and down-regulation of inflammatory factors are essential for the effective treatment of sepsis. Here we designed and constructed a polydopamine-based multifunctional nanoparticle for the treatment of sepsis. These nanoparticles (NPs) are composed of polydopamine (PDA) grafted with cationic polyethyleneimine (PEI). On the one hand, the NPs can utilize the electrostatic interaction to effectively adsorb cfDNA in blood, then effectively inhibiting the activation of toll like receptors (TLRs) and nuclear factor kappa B (NF-κB) pathways induced by cfDNA. On the other hand, the NPs have an immunomodulatory function, which can effectively convert pro-inflammatory macrophage (M1) into anti-inflammatory macrophage (M2), thus reduce the release of inflammatory cytokines and slow down the inflammatory storm of sepsis. In addition, the NPs possess good reactive oxygen species (ROS) scavenging ability. Briefly, the effective treatment of sepsis can be achieved by multiple strategies of effectively capturing the inflammatory triggering factor cfDNA, modulating the polarization of M1 macrophage to M2 macrophage and scavenging ROS, which has a promising clinical application.
Collapse
Affiliation(s)
- Wenjie Xi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Weijie Wu
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lili Zhou
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qi Zhang
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shushu Yang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lihong Huang
- Department of Orthopaedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, China
| | - Yijun Lu
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jing Wang
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinjin Chi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
10
|
Dabrowski W, Pfortmueller CA, Kotfis K, Jaroszynski A, Gagos M, Plotek W, Malbrain MLNG. Is there a place for natural agents with anti-inflammatory and antioxidative properties in critically ill patients? Potential usefulness of Xanthohumol. Pharmacol Ther 2025; 266:108766. [PMID: 39637948 DOI: 10.1016/j.pharmthera.2024.108766] [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: 09/16/2023] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Multi-organ dysfunction is a major issue in critically ill patients, where a significant inflammatory response appears to be the primary factor driving the degree of organ impairment, which correlates with the extent of organ injury. The management of inflammation requires a multidisciplinary approach, including antibiotics for infection control, circulatory and respiratory support, and correction of coagulation abnormalities. However, the use of anti-inflammatory treatments is typically restricted to a selected group of medications, with their effectiveness remaining the subject of extensive debate. Xanthohumol (Xn), a natural compound extracted from hops, possesses strong anti-inflammatory and antioxidative properties, with a mild anti-coagulation effect. Its biological activity is related to the inhibition of different inflammatory pathways, reduction in cytokine production and secretion, and an increase in antioxidative enzyme activity. This review examined the potential use of Xn as an adjuvant in the treatment of various pathologies in critically ill patients.
Collapse
Affiliation(s)
- Wojciech Dabrowski
- First Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Poland.
| | | | - Katarzyna Kotfis
- Department of Anaesthesiology, Intensive Care and Pain Management, Pomeranian Medical University of Szczecin, Poland
| | | | - Mariusz Gagos
- Department of Cell Biology, Maria Curie-Sklodowska University of Lublin, Poland
| | - Wlodzimierz Plotek
- First Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Poland
| | - Manu L N G Malbrain
- First Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Poland; Medical Data Management, Medaman, Geel, Belgium; International Fluid Academy, Lovenjoel, Belgium
| |
Collapse
|
11
|
Gu J, Wang DF, Lou JY. Identification of DNA damage repair-related genes in sepsis using bioinformatics and machine learning: An observational study. Medicine (Baltimore) 2025; 104:e41354. [PMID: 39889168 PMCID: PMC11789855 DOI: 10.1097/md.0000000000041354] [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: 04/09/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 02/02/2025] Open
Abstract
Sepsis is a life-threatening disease with a high mortality rate, for which the pathogenetic mechanism still unclear. DNA damage repair (DDR) is essential for maintaining genome integrity. This study aimed to explore the role of DDR-related genes in the development of sepsis and further investigated their molecular subtypes to enrich potential diagnostic biomarkers. Two Gene Expression Omnibus datasets (GSE65682 and GSE95233) were implemented to investigate the underlying role of DDR-related genes in sepsis. Three machine learning algorithms were utilized to identify the optimal feature genes. The diagnostic value of the selected genes was evaluated using the receiver operating characteristic curves. A nomogram was built to assess the diagnostic ability of the selected genes via "rms" package. Consensus clustering was subsequently performed to identify the molecular subtypes for sepsis. Furthermore, CIBERSORT was used to evaluate the immune cell infiltration of samples. Three different expressed DDR-related genes (GADD45A, HMGB2, and RPS27L) were identified as sepsis biomarkers. Receiver operating characteristic curves revealed that all 3 genes showed good diagnostic value. The nomogram including these 3 genes also exhibited good diagnostic efficiency. A notable difference in the immune microenvironment landscape was discovered between sepsis patients and healthy controls. Furthermore, all 3 genes were significantly associated with various immune cells. Our findings identify potential new diagnostic markers for sepsis that shed light on novel pathogenetic mechanism of sepsis and, therefore, may offer opportunities for potential intervention and treatment strategies.
Collapse
Affiliation(s)
- Jin Gu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dong-Fang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Ying Lou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Wang D, Huo R, Ye L. Identification of lethality-related m7G methylation modification patterns and the regulatory features of immune microenvironment in sepsis. Heliyon 2025; 11:e40870. [PMID: 39758389 PMCID: PMC11699318 DOI: 10.1016/j.heliyon.2024.e40870] [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: 02/04/2024] [Revised: 11/30/2024] [Accepted: 12/01/2024] [Indexed: 01/07/2025] Open
Abstract
Objectives N7-methylguanosine (m7G) modification is closely related to the occurrence of human diseases, but its roles in sepsis remain unclear. This study aimed to explore the patterns of lethality-related m7G regulatory factor-mediated RNA methylation modification and immune microenvironment regulatory features in sepsis. Methods Three sepsis-related datasets (E-MTAB-4421 and E-MTAB-4451 as training sets and GSE185263 as a validation set) were collected, and differentially expressed m7G-related genes were analyzed between survivors and non-survivors. Lethality-related m7G signature genes were then screened using machine learning methods, followed by the construction of a survival recognition model. Additionally, differences in immune cell distribution were determined and differentially expressed genes (DEGs) between different subtypes were analyzed. Weighted gene co-expression network analysis (WGCNA) was used to select important modules and related hub genes. Results In total, 10 differentially expressed m7G-related genes were identified between the survivors and non-survivors, and after further analysis, EIF4G3, EIF4E3, NSUN2, NUDT4, and GEMIN5 were identified as the optimal lethality-related m7G genes. A survival status diagnostic model was then constructed with a combined AUC of 0.678. Fifteen types of immune cells were significantly different between survivors and non-survivors. Sepsis samples were classified into two subtypes, with 22 types of immune cells showing significant differences. Subsequently, 1707 DEGs were identified between the two subtypes, which were significantly enriched in 91 GO terms and 16 KEGG pathways. Finally, the green module with |correlation| > 0.3 was found to be closely related to the subtypes and survival status; further, the top10 hub genes were obtained. Conclusion The constructed survival status diagnostic model based on the five lethality-related m7G signature genes may help predict the survival status of patients, and the 10 hub genes obtained may be potential therapeutic targets for sepsis.
Collapse
Affiliation(s)
- Dan Wang
- Department of Respiratory Medicine, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Xinghualing Area, 030000, Taiyuan, China
| | - Rujie Huo
- Department of Respiratory Medicine, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Xinghualing Area, 030000, Taiyuan, China
| | - Lu Ye
- Department of Respiratory Medicine, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Xinghualing Area, 030000, Taiyuan, China
| |
Collapse
|
13
|
Xu T, Song S, Zhu K, Yang Y, Wu C, Wang N, Lu S. Systemic inflammatory response index improves prognostic predictive value in intensive care unit patients with sepsis. Sci Rep 2025; 15:1908. [PMID: 39809872 PMCID: PMC11732978 DOI: 10.1038/s41598-024-81860-7] [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: 09/16/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Sepsis is a severe infectious disease with high mortality. However, the indicators used to evaluate its severity and prognosis are relatively complicated. The systemic inflammatory response index (SIRI), a new inflammatory indicator, has shown good predictive value in chronic infection, stroke, and cancer. The purpose of this study was to investigate the connection between sepsis and SIRI and evaluate its predictive usefulness. A total of 401 patients with sepsis were included in this study. Multiple linear regression and logistic regression analyses were performed to evaluate the relationship between SIRI and sepsis. The restricted cubic spline (RCS) method was employed to illustrate the dose-response relationship. The area under the curve (AUC) and decision curve analysis (DCA) were used to evaluate the prognostic value of SIRI. Multiple linear regression analysis revealed a significant positive correlation between SIRI and both blood cell count and Sequential Organ Failure Assessment (SOFA) score. Additionally, higher SIRI levels were significantly linked to a higher risk of sepsis worsening, according to logistic regression analysis. The RCS curve demonstrated that the risk of poor prognosis rose with increasing SIRI, particularly when SIRI exceeded 6.1. Furthermore, AUC and DCA results showed that SIRI had superior predictive value compared to traditional indicators. A higher SIRI is linked to a worse prognosis and more severe sepsis. SIRI may serve as a novel prognostic indicator in sepsis, though further clinical studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- Tuo Xu
- Xinxiang Central Hospital, Xinxiang, Henan, People's Republic of China
- The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
- Nantong University Hospital, Nantong, Jiangsu, People's Republic of China
| | - Shuaiwei Song
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, People's Republic of China
| | - Ke Zhu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yin Yang
- The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Chengyu Wu
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, People's Republic of China
| | - Naixue Wang
- Nantong Sixth People's Hospital, Nantong, Jiangsu, People's Republic of China
| | - Shu Lu
- Nantong University Hospital, Nantong, Jiangsu, People's Republic of China.
- Member of the Critical Care Medicine Branch of Jiangsu Physicians Association, Member of the Critical Care Ultrasound Group of Jiangsu Medical Association, Member of the Critical Care Medicine Branch of Nantong Medical Association, Member of Nantong Critical Care Medicine Quality Control Center, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226000, Jiangsu, People's Republic of China.
| |
Collapse
|
14
|
Ye C, Huang X, Tong Y, Chen Y, Zhao X, Xie W, Wang Y, Wang J, Zhang A, Mo Y. Overexpression of ALKBH5 alleviates LPS induced neuroinflammation via increasing NFKBIA. Int Immunopharmacol 2025; 144:113598. [PMID: 39571266 DOI: 10.1016/j.intimp.2024.113598] [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: 07/03/2024] [Revised: 10/13/2024] [Accepted: 11/06/2024] [Indexed: 12/15/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is a serious condition in which the immune system uncontrollably responds to infection, causing organ dysfunction. Neuroinflammation is one of the primary mechanisms underlying SAE. N6-methyladenosine (m6A) methylation is a common and reversible chemical modification of RNA molecules. Increasing evidence suggests that this modification plays a vital role in the inflammatory immune response. AlkB homolog 5 (ALKBH5) is an enzyme responsible for removing m6A modifications from RNA molecules and is known as a demethylase. However, the specific role of ALKBH5 in neuroinflammation remains unclear. To explore the role of ALKBH5 in neuroinflammation, researchers have used lipopolysaccharide (LPS) to induce inflammation in BV2 cells and mice. This study found that treatment of BV2 cells with LPS (1 μg/mL) significantly increased the total RNA m6A level and the ALKBH5 protein decreased significantly. Meanwhile, the NF-κB inflammatory signaling pathway was activated, leading to an obvious increase in IL-1β, IL-6, and TNF-α mRNA. The LPS-induced inflammatory response was alleviated when ALKBH5 was overexpressed in BV2 cells. This is due to a slower degradation rate of NFKBIA mRNA, an increase in NFKBIA protein levels, and inhibition of the NF-κB inflammatory signal pathway. When ALKBH5 was overexpressed in mice, as expected, there was an improvement in behavioral abnormalities induced by LPS. Compared to healthy volunteers, ALKBH5 mRNA levels were significantly decreased in peripheral blood mononuclear cells (PBMCs) from patients with sepsis and correlated with GCS and IL-6 levels. In summary, this study suggested that ALKBH5 is a potential therapeutic target for enhancing NFKBIA mRNA stability and alleviating neuroinflammation. Thus, ALKBH5 may provide new insights into the diagnosis and treatment of SAE.
Collapse
Affiliation(s)
- Changzhou Ye
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, China
| | | | - Yao Tong
- Wenzhou Medical University, China
| | | | - Xinyu Zhao
- Zhejiang Provincial People's Hospital, China
| | - Wenjing Xie
- The First People's Hospital of Yuhang District, Hangzhou, China
| | | | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Anqi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Yunchang Mo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, China
| |
Collapse
|
15
|
Sun XF, Luo WC, Huang SQ, Zheng YJ, Xiao L, Zhang ZW, Liu RH, Zhong ZW, Song JQ, Nan K, Qiu ZX, Zhong J, Miao CH. Immune-cell signatures of persistent inflammation, immunosuppression, and catabolism syndrome after sepsis. MED 2025:S2666-6340(24)00483-5. [PMID: 39824181 DOI: 10.1016/j.medj.2024.12.003] [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: 05/07/2024] [Revised: 10/13/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND Management of persistent inflammation, immunosuppression, and catabolism syndrome (PICS) after sepsis remains challenging for patients in the intensive care unit, experiencing poor quality of life and death. However, immune-cell signatures in patients with PICS after sepsis remain unclear. METHODS We determined immune-cell signatures of PICS after sepsis at single-cell resolution. Murine cecal ligation and puncture models of PICS were applied for validation. FINDINGS Immune functions of two enriched monocyte subpopulations, Mono1 and Mono4, were suppressed substantially in patients with sepsis and were partially restored in patients with PICS after sepsis and exhibited immunosuppressive and pro-apoptotic effects on B and CD8T cells. Patients with PICS and sepsis had reduced naive and memory B cells and proliferated plasma cells. Besides, naive and memory B cells in patients with PICS showed an active antigen processing and presentation gene signature compared to those with sepsis. PICS patients with better prognoses exhibited more active memory B cells and IGHA1-plasma cells. CD8TEMRA displayed signs of proliferation and immune dysfunction in the PICS-death group in contrast with the PICS-alive group. Megakaryocytes proliferation was more pronounced in patients with PICS and sepsis than in healthy controls, with notable changes in the anti-inflammatory and immunomodulatory effects observed in patients with PICS and verified in mice models. CONCLUSIONS Our study evaluated PICS after sepsis at the single-cell level, identifying the heterogeneity present within immune-cell subsets, facilitating the prediction of disease progression and the development of effective intervention. FUNDING This work was supported by the National Natural Science Foundation of China, Shanghai Municipal Health Commission "Yiyuan New Star" Youth Medical Talent Cultivating Program, and Shanghai Clinical Research Center for Anesthesiology.
Collapse
Affiliation(s)
- Xing-Feng Sun
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200438, China
| | - Wen-Chen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Shao-Qiang Huang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200438, China
| | - Yi-Jun Zheng
- Department of Critical Care and Pain Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Lei Xiao
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhong-Wei Zhang
- Department of Critical Care and Pain Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Rong-Hua Liu
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zi-Wen Zhong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Jie-Qiong Song
- Department of Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Ke Nan
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Zhi-Xin Qiu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Anesthesiology, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China.
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China.
| | - Chang-Hong Miao
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; Laboratory of Perioperative Stress and Protection, Shanghai 200032, China.
| |
Collapse
|
16
|
Li Y, Zhang X, Jiang G, Min X, Kong Q, Liu L, Wu J, Ding Z. Downregulation of HSPA12A protects heart against sepsis through suppressing mTOR-mediated inflammatory response in cardiomyocytes. Int Immunopharmacol 2025; 145:113721. [PMID: 39642573 DOI: 10.1016/j.intimp.2024.113721] [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: 09/25/2024] [Revised: 11/17/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE Over-activated immune response in hearts is the main pathological feature of septic cardiomyopathy, a fatal complication of sepsis with high mortality. Autophagy is capable to limit immune response by removing inflammatory mediators. Heat shock protein A12A (HSPA12A) encodes an atypical member of HSP70 family. This study aimed to investigate the role of HSPA12A in septic cardiomyopathy. METHODS Sepsis was induced by cecal ligation and puncture (CLP) for 6 h in mice in vivo or by LPS treatment for 24 h in primary cardiomyocytes in vitro. HSPA12A knockout (Hspa12a-/-) mice were generated by cre-loxp system. Echocardiography was performed to assess cardiac function. TUNEL and propidium iodide (PI) staining was used to indicate cardiomyocyte death. Inflammation-related factors were examined by qPCR and immunoblotting. Autophagy was evaluated by levels of LC3-II and p62. RESULTS Sepsis decreased HSPA12A expression in hearts and cardiomyocytes, while HSPA12A knockout in mice attenuated sepsis-induced cardiomyocyte death and cardiac dysfunction. Sepsis-induced activation of TLR4/MyD88/NF-κB-mediated inflammation was inhibited in hearts by HSPA12A knockout whereas was enhanced by HSPA12A overexpression in cardiomyocytes. Moreover, HSPA12A overexpression activated mTOR and inhibited autophagy in cardiomyocytes, while inhibition of mTOR by rapamycin diminished the HSPA12A-induced autophagy inhibition, inflammation activation, and cardiomyocyte death in septic cardiomyocytes. CONCLUSION Downregulation of HSPA12A inhibited mTOR to activated autophagy, thereby suppressed inflammatory response, and ultimately attenuated septic cardiomyopathy. Our findings identified HSPA12A as a driver for septic cardiomyopathy development, and strategies that inhibit HSPA12A in cardiomyocytes might be a potential therapeutic intervention.
Collapse
Affiliation(s)
- Yunfan Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaojin Zhang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Guohua Jiang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xinxu Min
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qiuyue Kong
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Li Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jun Wu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Zhengnian Ding
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
17
|
Wong D, Sahoo DK, Faivre C, Kopper J, Dersh K, Beachler T, Esser M. Oxidative stress in critically ill neonatal foals. J Vet Intern Med 2025; 39:e17297. [PMID: 39854109 PMCID: PMC11758150 DOI: 10.1111/jvim.17297] [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: 06/18/2024] [Accepted: 12/12/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Oxidative injury occurs in septic people, but the role of oxidative stress and antioxidants has rarely been evaluated in foals. OBJECTIVES/HYPOTHESIS To measure reactive oxygen species (ROS), biomarkers of oxidative injury, and antioxidants in neonatal foals. We hypothesized that ill foals would have higher blood concentrations of ROS and biomarkers of oxidative injury and lower concentrations of antioxidants compared to healthy foals. ANIMALS Seventy-two hospitalized and 21 healthy neonatal foals. METHODS Prospective cohort study. Reactive oxygen species (hydrogen peroxide [H2O2]), biomarkers of oxidative injury (malondialdehyde [MDA], protein carbonyl), and antioxidants (superoxide dismutase [SOD], catalase [CAT], glutathione, and glutathione reductase [GR] and peroxidase [GPx]) were measured from foals at admission. Measured variables were compared between healthy and ill foals using a 1-way ANOVA by Tukey's multiple comparisons test. RESULTS Ill foals (n = 51) had significantly higher mean concentrations of H2O2 (healthy 2.6 ± 1.4 nmol/mL, ill 6.8 ± 4.6 L nmol/mL; 95% CI), MDA (healthy 31.2 ± 14.4 nmol/mL, ill 114.3 ± 94.0 nmol/mL; 95% CI), and protein carbonyl (healthy 0.07 ± 0.01 nmol/mg protein, ill 0.12 ± 0.02 nmol/mg protein, 95% CI). Significant lower CAT (healthy 0.4 ± 0.3 mU/mg protein, ill 0.02 ± 0.02 mU/mg protein, 95% CI), glutathione (healthy 238.5 ± 101.9 μg/mL, ill 110.7 ± 37.8 μg/mL, 95% CI; P < .0001), GR (healthy 1.6 ± 1.8 mU/mg protein, ill 0.4 ± 0.5 mU/mg protein, 95% CI), and GPx (healthy 0.01 ± 0.003 mU/mg protein, ill 0.007 ± 0.002 mU/mg protein, 95% CI) were also noted. CONCLUSIONS AND CLINICAL IMPORTANCE Oxidative stress and lower antioxidant concentrations occur in ill and bacteremic neonatal foals. These variables should be considered during the treatment of ill foals.
Collapse
Affiliation(s)
- David Wong
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineIowa State UniversityAmesIowaUSA
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineIowa State UniversityAmesIowaUSA
| | - Cosette Faivre
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineIowa State UniversityAmesIowaUSA
| | - Jamie Kopper
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineIowa State UniversityAmesIowaUSA
| | - Katie Dersh
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineIowa State UniversityAmesIowaUSA
| | - Theresa Beachler
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineIowa State UniversityAmesIowaUSA
| | - Melissa Esser
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineIowa State UniversityAmesIowaUSA
| |
Collapse
|
18
|
Zhao J, Wu L, Zhang R, Yuan M, Huang J, Jia X, Mao X. Clostridium butyricum attenuates LPS-induced myocardial injury in septic mice by modulating CD4 + CD25 + FOXP3 + Treg. Immunobiology 2025; 230:152857. [PMID: 39642442 DOI: 10.1016/j.imbio.2024.152857] [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: 08/12/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
Sepsis-induced myocardial injury has become a major threat to patient health and safety. Intestinal microbiota imbalance plays a crucial role in sepsis regulation. Using 16srRNA technology, we explored how intestinal colonization of Clostridium butyricum over 28 days impacted mice with LPS-induced sepsis. Significant changes were noted in the gut microbiota of the mice, highlighting that C. butyricum can positively influence the immune state in septic myocardial injury models. The bacterium's ability to prevent intestinal mucosal damage and alleviate the immunosuppressive state during the later stages of sepsis by regulating CD4 + CD25 + FOXP3 + Treg cells is particularly noteworthy. This suggests a therapeutic role for C. butyricum in sepsis management by protecting against myocardial injury and improving immune regulation.
Collapse
Affiliation(s)
- Jinglin Zhao
- Department of Medical Laboratory, Kunming Children's Hospital, Children's Hospital Affiliated to Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Liuli Wu
- The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming 650500, China
| | - Rupan Zhang
- Yan'an Hospital of Kunming City, Kunming 650000, Yunnan Province, China
| | - Mei Yuan
- Department of Medical Laboratory, Kunming Children's Hospital, Children's Hospital Affiliated to Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Junchao Huang
- The First People's Hospital of Yunnan Province New Kun Hua Hospital, Kunming 650000, Yunnan Province, China
| | - Xiongfei Jia
- Department of Clinical laboratory,920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming 650000, Yunnan Province, China
| | - Xiaoqin Mao
- The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming 650500, China.
| |
Collapse
|
19
|
Zhang X, Ling C, Xiong Z, Gong T, Luo S, Liu X, Zhang L, Liao C, Lu Y, Huang X, Zhou W, Zhou S, Liu Y, Tang J. Desuccinylation of TBK1 by SIRT5 regulates inflammatory response of macrophages in sepsis. Cell Rep 2024; 43:115060. [PMID: 39673708 DOI: 10.1016/j.celrep.2024.115060] [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: 12/21/2023] [Revised: 08/19/2024] [Accepted: 11/21/2024] [Indexed: 12/16/2024] Open
Abstract
Tank-binding kinase 1 (TBK1) is a critical signal transducer in the nuclear factor κB (NF-κB) and interferon regulatory factor (IRF) pathways, essential for innate immunity. However, its negative regulation mechanisms remain unclear. This study demonstrates that TBK1 succinylation, regulated by desuccinylase SIRT5, inhibits lipopolysaccharide (LPS)/Toll-like receptor 4 (TLR4)-mediated NF-κB and IRF signaling activation. We identified three key succinylation sites on TBK1: K38, K154, and K692. In endotoxemia and sepsis models, reduced SIRT5 levels in macrophages increased TBK1 succinylation, inhibiting its binding to IRF3 and TRAF2 and suppressing the inflammatory response. In vivo, adoptive transfer of macrophages expressing the succinylation-resistant TBK1-2KR (K154/692R) mutant reversed the inflammatory cytokine suppression caused by SIRT5 deficiency, exacerbating sepsis-induced lung injury. These findings reveal a novel mechanism by which SIRT5 modulates TBK1 activity and macrophage-mediated inflammation during sepsis.
Collapse
Affiliation(s)
- Xuedi Zhang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China; Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No. 1333, Xinhu Road, Baoan District, Shenzhen, Guangdong 518110, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chunxiu Ling
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Ziying Xiong
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Ting Gong
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No. 1333, Xinhu Road, Baoan District, Shenzhen, Guangdong 518110, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shuhua Luo
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Xiaolei Liu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Lina Zhang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Chaoxiong Liao
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Yue Lu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Xiao Huang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Wending Zhou
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Shuangnan Zhou
- Senior Department of Infectious Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China.
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No. 1333, Xinhu Road, Baoan District, Shenzhen, Guangdong 518110, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Jing Tang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China.
| |
Collapse
|
20
|
Zou W, Sai L, Sai W, Song L, Wang G. Diagnostic and prognostic value of disulfidptosis-related genes in sepsis. INFECTIOUS MEDICINE 2024; 3:100143. [PMID: 39610785 PMCID: PMC11602581 DOI: 10.1016/j.imj.2024.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/17/2024] [Accepted: 08/26/2024] [Indexed: 11/30/2024]
Abstract
Background Sepsis is a disease associated with high morbidity and mortality rates, especially among the elderly and patients in intensive care units. Disulfidptosis, a newly identified form of cell death triggered by disulfide stress, is emerging as a significant factor in disease progression. This study aimed to explore the diagnostic and prognostic value of disulfidptosis-related genes in sepsis. Methods We obtained two datasets from the Gene Expression Omnibus (GEO) database to conduct our analysis. Functional enrichment analysis was performed to identify relevant biological pathways. A protein-protein interaction network was constructed to identify hub genes critical to sepsis. Additionally, we analyzed the immune infiltration status in sepsis patients. The diagnostic value of these hub genes for sepsis was evaluated using nomograms, receiver operating characteristic (ROC) curves, and calibration curves in both training and validation datasets. Finally, a miRNA-immune-related hub genes (miRNA-IHGs) regulatory network was developed to elucidate the synergistic interactions between miRNAs and their target genes. Results A total of 3,469 differentially expressed genes (DEGs) were identified, of which seven were related to disulfidptosis (DR-DEGs). Functional enrichment analysis showed that DR-DEGs were significantly enriched in pathways related to actin dynamics. Five hub genes (MYH10, ACTN4, MYH9, FLNA, and IQGAP1) were identified as central to these processes. The analysis of immune infiltration revealed significantly lower levels of 11 immune cell types, while macrophages and regulatory T cells were significantly elevated in sepsis patients. The area under the ROC curves (AUCs) of the IHGs risk prediction model were 0.917 and 0.894 for the training and validation sets, respectively. A miRNA-IHGs regulatory network, comprising 17 nodes and 27 edges, was constructed, with MYH9 being the most frequently regulated by miRNAs. Conclusion The pathophysiological process of sepsis appears to involve disulfidptosis, highlighting it as a potential new therapeutic targets for sepsis management.
Collapse
Affiliation(s)
- Wenlu Zou
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Lintao Sai
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Wen Sai
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Li Song
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Gang Wang
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
21
|
Liao Y, Zhang W, Zhou M, Zhu C, Zou Z. Ubiquitination in pyroptosis pathway: A potential therapeutic target for sepsis. Cytokine Growth Factor Rev 2024; 80:72-86. [PMID: 39294049 DOI: 10.1016/j.cytogfr.2024.09.001] [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: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Sepsis remains a significant clinical challenge, causing numerous deaths annually and representing a major global health burden. Pyroptosis, a unique form of programmed cell death characterized by cell lysis and the release of inflammatory mediators, is a crucial factor in the pathogenesis and progression of sepsis, septic shock, and organ dysfunction. Ubiquitination, a key post-translational modification influencing protein fate, has emerged as a promising target for managing various inflammatory conditions, including sepsis. This review integrates the current knowledge on sepsis, pyroptosis, and the ubiquitin system, focusing on the molecular mechanisms of ubiquitination within pyroptotic pathways activated during sepsis. By exploring how modulating ubiquitination can regulate pyroptosis and its associated inflammatory signaling pathways, this review provides insights into potential therapeutic strategies for sepsis, highlighting the need for further research into these complex molecular networks.
Collapse
Affiliation(s)
- Yan Liao
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Miao Zhou
- Department of Anesthesiology, the Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Chenglong Zhu
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| | - Zui Zou
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
22
|
Walachowski S, Garo L, Sharma A, Jayaraman A, Noon J, Reinhardt C, Bosmann M. Disruption of the C5a/C5aR1 axis confers protection against hookworm infection in lung. Front Immunol 2024; 15:1475165. [PMID: 39628481 PMCID: PMC11611822 DOI: 10.3389/fimmu.2024.1475165] [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: 08/03/2024] [Accepted: 10/23/2024] [Indexed: 12/06/2024] Open
Abstract
Hookworms are soil-transmitted parasitic nematodes that penetrate the host skin before migrating to the lungs. With an estimated 500-700 million people infected worldwide, hookworm infections are a neglected tropical disease and a significant cause of morbidity, particularly in children, pregnant women, and immunocompromised individuals. Although there is ample evidence that complement activation is pivotal to elicit a protective host immune response against invasive pathogens, its role in hookworm infection remains insufficiently explored. Here, we investigated the complement anaphylatoxin, C5a, during the early lung stage of infection with Nippostrongylus brasiliensis in C57BL/6J wild type and C5aR1-/- mice. Despite the previously reported ability of lung larvae to evade complement activation, C5a was detectable locally in lung tissue and bronchoalveolar lavages. Surprisingly, C5aR1 presence directly contributed to the pathogenicity of hookworm infection. The burden of viable parasites in the lungs was mitigated in C5aR1-/- mice, compared to C57BL/6J mice 48 hours post-infection. Additionally, C5aR1-/- mice showed significantly reduced lung injury, lower cytokine release, attenuated alveolar hemorrhage, and limited alveolar-capillary barrier disruption. Neutrophils were the most abundant and highest C5aR1-expressing cell type in the alveolar space after infection. Deficiency of C5aR1 reduced the influx of neutrophils, monocytes, and eosinophils to the pulmonary airways. RNA sequencing of alveolar neutrophils revealed C5aR1-dependent regulation of the novel nuclear protein, DEDD2. In conclusion, our findings highlight the impact of C5aR1 signaling in neutrophils during hookworm infection uncovering an unexpected downside of complement activation in parasitic infection.
Collapse
Affiliation(s)
- Sarah Walachowski
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Lucien Garo
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Arjun Sharma
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Mainz Research School of Translational Biomedicine (TransMed), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Archana Jayaraman
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jason Noon
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
23
|
Liu FJ, Wu J, Gong LJ, Yang HS, Chen H. Non-invasive vagus nerve stimulation in anti-inflammatory therapy: mechanistic insights and future perspectives. Front Neurosci 2024; 18:1490300. [PMID: 39605787 PMCID: PMC11599236 DOI: 10.3389/fnins.2024.1490300] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Non-invasive vagus nerve stimulation (VNS) represents a transformative approach for managing a broad spectrum of inflammatory and autoimmune conditions, including rheumatoid arthritis and inflammatory bowel disease. This comprehensive review delineates the mechanisms underlying VNS, emphasizing the cholinergic anti-inflammatory pathway, and explores interactions within the neuro-immune and vagus-gut axes based on both clinical outcomes and pre-clinical models. Clinical applications have confirmed the efficacy of VNS in managing specific autoimmune diseases, such as rheumatoid arthritis, and chronic inflammatory conditions like inflammatory bowel disease, showcasing the variability in stimulation parameters and patient responses. Concurrently, pre-clinical studies have provided insights into the potential of VNS in modulating cardiovascular and broader inflammatory responses, paving the way for its translational application in clinical settings. Innovations in non-invasive VNS technology and precision neuromodulation are enhancing its therapeutic potential, making it a viable option for patients who are unresponsive to conventional treatments. Nonetheless, the widespread adoption of this promising therapy is impeded by regulatory challenges, patient compliance issues, and the need for extensive studies on long-term efficacy and safety. Future research directions will focus on refining VNS technology, optimizing treatment parameters, and exploring synergistic effects with other therapeutic modalities, which could revolutionize the management of chronic inflammatory and autoimmune disorders.
Collapse
Affiliation(s)
- Fu-Jun Liu
- Neurology Medical Center II, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Jing Wu
- Department of Medical Imaging, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Li-Jun Gong
- Center of Surgical Anesthesia, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hong-Shuai Yang
- Central Operating Room, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Huan Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
Yamaga S, Murao A, Zhou M, Aziz M, Brenner M, Wang P. Radiation-induced eCIRP impairs macrophage bacterial phagocytosis. J Leukoc Biol 2024; 116:1072-1079. [PMID: 38920274 PMCID: PMC11531804 DOI: 10.1093/jleuko/qiae132] [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: 02/01/2024] [Revised: 04/29/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Macrophages are essential immune cells for host defense against bacterial pathogens after radiation injury. However, the role of macrophage phagocytosis in infection following radiation injury remains poorly examined. Extracellular cold-inducible RNA-binding protein is a damage-associated molecular pattern that dysregulates host immune system responses such as phagocytosis. We hypothesized that radiation-induced extracellular cold-inducible RNA-binding protein release impairs macrophage phagocytosis of bacteria. Adult healthy mice were exposed to 6.5 Gy total body irradiation. Primary peritoneal macrophages isolated from adult healthy mice were exposed to 6.5 Gy radiation. Extracellular cold-inducible RNA-binding protein-neutralizing monoclonal antibody was added to the cell culture prior to irradiation. Bacterial phagocytosis by peritoneal macrophages was assessed using pHrodo Green-labeled Escherichia coli 7 d after irradiation ex vivo and in vitro. Bacterial phagocytosis was also assessed after treatment with recombinant murine cold-inducible RNA-binding protein. Rac1 and ARP2 protein expression in cell lysates and extracellular cold-inducible RNA-binding protein levels in the peritoneal lavage were assessed by western blotting. Bacterial phagocytosis by peritoneal macrophages was significantly decreased after irradiation compared with controls ex vivo and in vitro. Rac1 and ARP2 expression in the peritoneal macrophages were downregulated after total body irradiation. Total body irradiation significantly increased extracellular cold-inducible RNA-binding protein levels in the peritoneal cavity. Recombinant murine cold-inducible RNA-binding protein significantly decreased bacterial phagocytosis in a dose-dependent manner. Extracellular cold-inducible RNA-binding protein monoclonal antibody restored bacterial phagocytosis by peritoneal macrophages after irradiation. Ionizing radiation exposure impairs bacterial phagocytosis by macrophages after irradiation. Neutralization of extracellular cold-inducible RNA-binding protein restores the phagocytic ability of macrophages after irradiation. Our findings elucidate a novel mechanism of immune dysfunction and provide a potential new therapeutic approach for limiting infection after radiation injury.
Collapse
Affiliation(s)
- Satoshi Yamaga
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
| | - Mian Zhou
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
| |
Collapse
|
25
|
Cheng H, Wang Z, Feng M, Tang Y, Zheng X, Zhang X, Lyu J. Predicting High-Flow Nasal Cannula Oxygen Therapy Failure in Patients With Acute Hypoxaemic Respiratory Failure Using Machine Learning: Model Development and External Validation. J Clin Nurs 2024. [PMID: 39468839 DOI: 10.1111/jocn.17518] [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: 05/06/2024] [Revised: 08/27/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
AIMS AND OBJECTIVES To develop and validate a prediction model for high-flow nasal cannula (HFNC) failure in patients with acute hypoxaemic respiratory failure (AHRF). BACKGROUND AHRF accounts for a major proportion of intensive care unit (ICU) admissions and is associated with high mortality. HFNC is a non-invasive respiratory support technique that can improve patient oxygenation. However, HFNC failure, defined as the need for escalation to invasive mechanical ventilation, can lead to delayed intubation, prolonged mechanical ventilation and increased risk of mortality. Timely and accurate prediction of HFNC failure has important clinical implications. Machine learning (ML) can improve clinical prediction. DESIGN Multicentre observational study. METHODS This study analysed 581 patients from an academic medical centre in Boston and 180 patients from Guangzhou, China treated with HFNC for AHRF. The Boston dataset was randomly divided into a training set (90%, n = 522) and an internal validation set (10%, n = 59), and the model was externally validated using the Guangzhou dataset (n = 180). A random forest (RF)-based feature selection method was used to identify predictive factors. Nine machine learning algorithms were selected to build the predictive model. The area under the receiver operating characteristic curve (AUC) and performance evaluation parameters were used to evaluate the models. RESULTS The final model included 38 features selected using the RF method, with additional input from clinical specialists. Models based on ensemble learning outperformed other models (internal validation AUC: 0.83; external validation AUC: 0.75). Important predictors of HFNC failure include Glasgow Coma Scale scores and Sequential Organ Failure Assessment scores, albumin levels measured during HFNC treatment, ROX index at ICU admission and sepsis. CONCLUSIONS This study developed an interpretable ML model that accurately predicts the risk of HFNC failure in patients with AHRF. RELEVANCE TO CLINICAL PRACTICE Clinicians and nurses can use ML models for early risk assessment and decision support in AHRF patients receiving HFNC. REPORTING METHOD TRIPOD checklist for prediction model studies was followed in this study. PATIENT OR PUBLIC CONTRIBUTION Patients were involved in the sample of the study.
Collapse
Affiliation(s)
- Hongtao Cheng
- School of Nursing, Jinan University, Guangzhou, Guangdong, China
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zichen Wang
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Mei Feng
- Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yonglan Tang
- School of Nursing, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoyu Zheng
- Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiaoshen Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
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
|
27
|
Machado-Junior PA, Dias MSS, de Souza ABF, Lopes LSE, Menezes TP, Talvani A, Brochard L, Bezerra FS. A short duration of mechanical ventilation alters redox status in the diaphragm and aggravates inflammation in septic mice. Respir Physiol Neurobiol 2024; 331:104361. [PMID: 39433197 DOI: 10.1016/j.resp.2024.104361] [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: 09/04/2024] [Revised: 10/07/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND Mechanical ventilation (MV) is a life support method used to treat patients with respiratory failure. High tidal volumes during MV can cause ventilator-induced lung injury (VILI), but also affect other organs, such as the diaphragm (Dia) causing ventilator-induced diaphragmatic dysfunction (VIDD). VIDD is often associated with a complicated course on MV. Sepsis can induce inflammation and oxidative stress, contributing to the impairment of the Dia and worsening of the prognosis. This study evaluated the additive or synergistic effects of a short course of mechanical ventilation on Dia in healthy and septic adult mice. METHODS 32 adult male C57BL/6 mice were randomly into four groups: Control (CG), non-ventilated animals instilled with saline solution (PBS1x); Lipopolysaccharide (LPS), non-ventilated animals instilled with PBS solution containing lipopolysaccharide; Mechanical Ventilation (MV) for 1 h, ventilated animals instilled with PBS solution; and Mechanical Ventilation and LPS (MV+LPS), ventilated animals instilled with PBS solution containing LPS. At the end of the experimental protocol, the animals were euthanized, then blood and diaphragm tissue samples were collected. RESULTS Evaluation of leukocyte/blood parameters and diaphragm muscle showed that MV, LPS and the combination of both were able to increase neutrophil count, creatine kinase, inflammatory mediators and oxidative stress in all groups compared to the control. MV and sepsis combined had additive effects on inflammation and lipid peroxidation. CONCLUSIONS A short course of Mechanical ventilation promotes inflammation and oxidative stress and, its combination with sepsis further increases local and systemic inflammation.
Collapse
Affiliation(s)
- Pedro Alves Machado-Junior
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Marcelo Santiago Soares Dias
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Leonardo Spinelli Estevão Lopes
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Tatiana Prata Menezes
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Laurent Brochard
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Qu H, Wu J, Pan Y, Abdulla A, Duan Z, Cheng W, Wang N, Chen H, Wang C, Yang J, Tang J, Yang C, Wu C, Xue X. Biomimetic Nanomodulator Regulates Oxidative and Inflammatory Stresses to Treat Sepsis-Associated Encephalopathy. ACS NANO 2024; 18:28228-28245. [PMID: 39367850 DOI: 10.1021/acsnano.4c08157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is a devastating complication of sepsis, affecting approximately 70% of patients with sepsis in intensive care units (ICU). Although the pathophysiological mechanisms remain elusive, sepsis is typically accompanied by systemic inflammatory response syndrome (SIRS) and hyper-oxidative conditions. Here, we introduce a biomimetic nanomodulator (mAOI NP) that specifically targets inflammation site and simultaneously regulates oxidative and inflammatory stresses. mAOI NPs are constructed using metal-coordinated polyphenolic antioxidants (tannic acid) and flavonoid quercetin, which are then coated with macrophage membrane to enhance pharmacokinetics and enable SAE targeting. In a cecal ligation and puncture (CLP)-induced severe sepsis model, mAOI NPs effectively mitigate oxidative stress by purging reactive oxygen species, repairing mitochondrial damage and activating the Nrf2/HO-1 signaling pathway; while polarizing M1 macrophages or microglia toward anti-inflammatory M2 subtype. mAOI NPs potently inhibit sepsis progress, prolong overall survival from 25 to 66% and enhance learning and memory capabilities in SAE mice. Further proteomics analysis reveals that mAOI NPs modulate neurodevelopment processes related to learning and memory formation while also exerting anti-inflammatory and antioxidative effects on brain tissue responses associated with SAE pathology. This study offers significant potential for improving patient outcomes and revolutionizing the treatment landscape for this devastating complication of sepsis.
Collapse
Affiliation(s)
- Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Aynur Abdulla
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiran Duan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ning Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiaojiao Yang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianguo Tang
- Department of Trauma-Emergency and Critical Care Medicine Center (TECCMC), Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Chunhui Yang
- Department of Trauma-Emergency and Critical Care Medicine Center (TECCMC), Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Chunrong Wu
- Department of Trauma-Emergency and Critical Care Medicine Center (TECCMC), Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
29
|
Joo HK, Kim S, Choi E, Jin H, Lee YR, Lee EO, Kim CS, Jeon BH. Heterozygous Apex1 deficiency exacerbates lipopolysaccharide-induced systemic inflammation in a murine model. Free Radic Biol Med 2024; 223:96-108. [PMID: 39067624 DOI: 10.1016/j.freeradbiomed.2024.07.033] [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: 03/15/2024] [Revised: 06/26/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
The biological role of apurinic/apyrimidinic endonuclease 1/redox factor-1 (Apex1) in modulating systemic inflammation remains unclear. This study aimed to assess the impact of Apex1 deficiency on systemic inflammation triggered by lipopolysaccharide (LPS) in a murine model. The methods involved transcriptomic analysis and assessments of inflammatory responses in age-matched 8-week-old Apex1+/- and wild-type Apex1+/+ mice, generated using the CRISPR/Cas9 system. Apex1+/- mice displayed no overt changes in body weight, however, Apex1 protein expressions in tissues were significantly reduced compared to wild-type mice. Furthermore, in Apex1+/- mice transcriptomic analysis showed that genes associated with antioxidant pathways were downregulated, and levels of superoxide production, 8-hydroxy-2'-deoxyguanosine (8-OHdG), and malondialdehyde (MDA) were increased. Moreover, hematological analysis showed increased neutrophil levels and a twofold increase in the count of splenic lymphocyte antigen 6 family member G+ (Ly6G+) neutrophils in the Apex1+/- mice compared to those in Apex1+/+ mice. Furthermore, following LPS treatment, the levels of cytokines and chemokines, including interleukin-1β, interleukin-10, tumor necrosis factor-α, and monocyte chemoattractant protein 1, increased in the Apex1+/- mice. The Kaplan-Meier curve showed a significant reduction in the survival rates of Apex1+/- mice treated with LPS compared to those of Apex1+/+ mice. The hepatic and lung injury scores and Ly6G+ neutrophil infiltration levels also increased in Apex1+/- mice after LPS treatment. These results showed that Apex1 deficiency exacerbated the LPS-induced tissue damage in the lung and liver. These findings illustrate that in vivo Apex1 deficiency exacerbates LPS-induced systemic inflammation, tissue damage, and mortality in a murine model, highlighting the crucial role of Apex1 in mitigating inflammatory responses and maintaining a holistic physiological equilibrium.
Collapse
Affiliation(s)
- Hee Kyoung Joo
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Physiology, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | - Sungmin Kim
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Physiology, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | - Eunju Choi
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Physiology, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | - Hao Jin
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Physiology, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | - Yu-Ran Lee
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Physiology, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | - Eun-Ok Lee
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Physiology, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | - Cuk-Seong Kim
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Physiology, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | - Byeong Hwa Jeon
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea; Department of Physiology, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea.
| |
Collapse
|
30
|
Jiang WX, Li HH. Circulating inflammatory cytokines and the risk of sepsis: a bidirectional mendelian randomization analysis. BMC Infect Dis 2024; 24:793. [PMID: 39112975 PMCID: PMC11304706 DOI: 10.1186/s12879-024-09689-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/30/2024] [Indexed: 08/11/2024] Open
Abstract
BACKGROUND Sepsis is a life-threatening condition that is characterized by multiorgan dysfunction and caused by dysregulated cytokine networks, which are closely associated with sepsis progression and outcomes. However, currently available treatment strategies that target cytokines have failed. Thus, this study aimed to investigate the interplay between genetically predicted circulating concentrations of cytokines and the outcomes of sepsis and to identify potential targets for sepsis treatment. METHODS Data related to 35 circulating cytokines in 31,112 individuals (including 11,643 patients with sepsis) were included in genome-wide association studies (GWASs) from the UK Biobank and FinnGen consortia. A bidirectional two-sample Mendelian randomization (MR) analysis was performed using single nucleotide polymorphisms (SNPs) to evaluate the causal effects of circulating cytokines on sepsis outcomes and other cytokines. RESULTS A total of 35 inflammatory cytokine genes were identified in the GWASs, and 11 cytokines, including Interleukin-1 receptor antagonist (IL-1ra), macrophage inflammatory protein 1 (MIP1α), IL-16, et al., were associated with sepsis outcome pairs according to the selection criteria of the cis-pQTL instrument. Multiple MR methods verified that genetically predicted high circulating levels of IL-1ra or MIP1α were negatively correlated with genetic susceptibility to risk of sepsis, including sepsis (28-day mortality), septicaemia, streptococcal and pneumonia-derived septicaemia (P ≤ 0.01). Furthermore, genetic susceptibility of sepsis outcomes except sepsis (28-day mortality) markedly associated with the circulating levels of five cytokines, including active plasminogen activator inhibitor (PAI), interleukin 7 (IL-7), tumour necrosis factor alpha (TNF-α), beta nerve growth factor (NGF-β), hepatic growth factor (HGF) (P < 0.05). Finally, we observed that the causal interaction network between MIP1α or IL-1ra and other cytokines (P < 0.05). CONCLUSIONS This comprehensive MR analysis provides insights into the potential causal mechanisms that link key cytokines, particularly MIP1α, with risk of sepsis, and the findings suggest that targeting MIP1α may be a potential strategy for preventing sepsis.
Collapse
Affiliation(s)
- Wen-Xi Jiang
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
31
|
Mun SJ, Cho E, Kim HK, Gil WJ, Yang CS. Enhancing acute inflammatory and sepsis treatment: superiority of membrane receptor blockade. Front Immunol 2024; 15:1424768. [PMID: 39081318 PMCID: PMC11286478 DOI: 10.3389/fimmu.2024.1424768] [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: 04/28/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Conditions such as acute pancreatitis, ulcerative colitis, delayed graft function and infections caused by a variety of microorganisms, including gram-positive and gram-negative organisms, increase the risk of sepsis and therefore mortality. Immune dysfunction is a characterization of sepsis, so timely and effective treatment strategies are needed. The conventional approaches, such as antibiotic-based treatments, face challenges such as antibiotic resistance, and cytokine-based treatments have shown limited efficacy. To address these limitations, a novel approach focusing on membrane receptors, the initiators of the inflammatory cascade, is proposed. Membrane receptors such as Toll-like receptors, interleukin-1 receptor, endothelial protein C receptor, μ-opioid receptor, triggering receptor expressed on myeloid cells 1, and G-protein coupled receptors play pivotal roles in the inflammatory response, offering opportunities for rapid regulation. Various membrane receptor blockade strategies have demonstrated efficacy in both preclinical and clinical studies. These membrane receptor blockades act as early stage inflammation modulators, providing faster responses compared to conventional therapies. Importantly, these blockers exhibit immunomodulatory capabilities without inducing complete immunosuppression. Finally, this review underscores the critical need for early intervention in acute inflammatory and infectious diseases, particularly those posing a risk of progressing to sepsis. And, exploring membrane receptor blockade as an adjunctive treatment for acute inflammatory and infectious diseases presents a promising avenue. These novel approaches, when combined with antibiotics, have the potential to enhance patient outcomes, particularly in conditions prone to sepsis, while minimizing risks associated with antibiotic resistance and immune suppression.
Collapse
Affiliation(s)
- Seok-Jun Mun
- Department of Bionano Engineering, Hanyang University, Seoul, Republic of Korea
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
| | - Euni Cho
- Department of Bionano Engineering, Hanyang University, Seoul, Republic of Korea
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
| | - Hyo Keun Kim
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
| | - Woo Jin Gil
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
| | - Chul-Su Yang
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Republic of Korea
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
- Department of Medicinal and Life Science, Hanyang University, Ansan, Republic of Korea
| |
Collapse
|
32
|
Li H, Ren Q, Hu Y, Guo F, Huang R, Lin L, Tan Z, Ma L, Fu P. SKLB023 protects against inflammation and apoptosis in sepsis-associated acute kidney injury via the inhibition of toll-like receptor 4 signaling. Int Immunopharmacol 2024; 139:112668. [PMID: 39008938 DOI: 10.1016/j.intimp.2024.112668] [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: 05/20/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is one of common critical illnesses with high morbidity and mortality. At present, effective therapeutic drugs for SA-AKI are remain lacking. SKLB023 is a synthetic small-molecule compound which exerts potent anti-inflammatory effects in our previous studies. Here, this study aimed to characterize the protective effect of SKLB023 on SA-AKI and explore its underlying mechanism. The SA-AKI experimental models have been established by cecum ligation/puncture (CLP) and lipopolysaccharide (LPS) injection in male C57BL/6J mice. SKLB023 was administered by gavage (50 or 25 mg/kg in CLP model and 50 mg/kg in LPS model) daily 3 days in advance and 30 min earlier on the day of modeling. Our results confirmed SKLB023 treatment could improve the survival of SA-AKI mice and ameliorate renal pathological injury, inflammation, and apoptosis in the two types of septic AKI mice. Mechanically, SKLB023 deceased the expression of TLR4 in LPS-triggered renal tubular epithelial cells, and inhibited the activation of downstream pathways including NF-κB and MAPK pathways. Our study suggested that SKLB023 is expected to be a potential drug for the prevention and treatment of septic AKI.
Collapse
Affiliation(s)
- Hui Li
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| | - Qian Ren
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| | - Yao Hu
- Department of Medicine Renal Division, Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu 610041, PR China
| | - Fan Guo
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| | - Rongshuang Huang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| | - Lin Lin
- West-district Outpatient Department, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| | - Zhouke Tan
- Department of Nephrology, Organ Transplant Center, Guizhou Province Key Laboratory of Cell Engineering, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, PR China.
| | - Liang Ma
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China.
| | - Ping Fu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, PR China
| |
Collapse
|
33
|
Kim D, Lee D, Lee J, Lee B, Ko SW. Association between the red cell distribution width and mortality in elderly patients with non-traumatic coma: An observational cohort study. Medicine (Baltimore) 2024; 103:e38773. [PMID: 38941367 PMCID: PMC11466147 DOI: 10.1097/md.0000000000038773] [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: 01/16/2024] [Accepted: 06/10/2024] [Indexed: 06/30/2024] Open
Abstract
Elevated red blood cell distribution width (RDW) can be associated with disease severity. However, studies on RDW for the prognosis of elderly patients with non-traumatic coma (NTC) are lacking. This study aims to examine the relationship between RDW and outcomes in elderly patients with NTC. This observational cohort study included elderly patients (aged ≥ 65 years) with NTC between January 2022 and December 2022. We measured RDW upon patient arrival at the emergency department (ED). We conducted a multivariable analysis using logistic regression of relevant covariates to predict in-hospital mortality. Survival curves based on 30-day mortality were designed using the Kaplan-Meier method. The primary outcome was in-hospital mortality, and the secondary outcome was 30-day mortality. A total of 689 patients were included in the study, and in-hospital mortality was 29.6% (n = 204). Our results found that the RDWs of non-survivors were significantly greater than those of survivors (14.6% vs 13.6%). Multivariable analysis showed that RDWs at ED arrival were independently associated with in-hospital mortality (odds ratio, 1.126; 95% confidence interval, 1.047-1.212; P < .001). The Kaplan-Meier curve indicated that the survival probability of patients with a low RDW was greater than those with a high RDW. Having a high RDW at ED arrival was associated with in-hospital mortality in elderly patients with NTC.
Collapse
Affiliation(s)
- Dongki Kim
- Department of Emergency Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Donghun Lee
- Department of Emergency Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Emergency Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jiho Lee
- Department of Emergency Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Byungkook Lee
- Department of Emergency Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Emergency Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Sang Won Ko
- Department of Emergency Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| |
Collapse
|
34
|
Chen S, Wang K, Fan Z, Zhou T, Li R, Zhang B, Chen J, Chi J, Wei K, Liu J, Liu Z, Ma J, Dong N, Liu J. Modulation of anti-cardiac fibrosis immune responses by changing M2 macrophages into M1 macrophages. Mol Med 2024; 30:88. [PMID: 38879491 PMCID: PMC11179216 DOI: 10.1186/s10020-024-00858-z] [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: 01/23/2024] [Accepted: 06/10/2024] [Indexed: 06/19/2024] Open
Abstract
BACKGROUND Macrophages play a crucial role in the development of cardiac fibrosis (CF). Although our previous studies have shown that glycogen metabolism plays an important role in macrophage inflammatory phenotype, the role and mechanism of modifying macrophage phenotype by regulating glycogen metabolism and thereby improving CF have not been reported. METHODS Here, we took glycogen synthetase kinase 3β (GSK3β) as the target and used its inhibitor NaW to enhance macrophage glycogen metabolism, transform M2 phenotype into anti-fibrotic M1 phenotype, inhibit fibroblast activation into myofibroblasts, and ultimately achieve the purpose of CF treatment. RESULTS NaW increases the pH of macrophage lysosome through transmembrane protein 175 (TMEM175) and caused the release of Ca2+ through the lysosomal Ca2+ channel mucolipin-2 (Mcoln2). At the same time, the released Ca2+ activates TFEB, which promotes glucose uptake by M2 and further enhances glycogen metabolism. NaW transforms the M2 phenotype into the anti-fibrotic M1 phenotype, inhibits fibroblasts from activating myofibroblasts, and ultimately achieves the purpose of treating CF. CONCLUSION Our data indicate the possibility of modifying macrophage phenotype by regulating macrophage glycogen metabolism, suggesting a potential macrophage-based immunotherapy against CF.
Collapse
Affiliation(s)
- Shiqi Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhengfeng Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tingwen Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rui Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bingxia Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jie Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiangyang Chi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Keke Wei
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Jincheng Liu
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Zongtao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Junwei Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
35
|
Liu K, Yang L, Wang P, Zhu J, Li F, Peng J, Huang K, Liang M. Myricanol attenuates sepsis-induced inflammatory responses by nuclear factor erythroid 2-related factor 2 signaling and nuclear factor kappa B/mitogen-activated protein kinase pathway via upregulating Sirtuin 1. Inflammopharmacology 2024; 32:1887-1901. [PMID: 38526770 DOI: 10.1007/s10787-024-01448-5] [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: 10/15/2023] [Accepted: 01/30/2024] [Indexed: 03/27/2024]
Abstract
Sepsis, a life-threatening condition characterized by dysregulated immune responses, remains a significant clinical challenge. Myricanol, a natural compound, plays a variety of roles in regulating lipid metabolism, anti-cancer, anti-neurodegeneration, and it could act as an Sirtuin 1 (SIRT1) activator. This study aimed to explore the therapeutic potential and underlying mechanism of myricanol in the lipopolysaccharide (LPS)-induced sepsis model. In vivo studies revealed that myricanol administration significantly improved the survival rate of LPS-treated mice, effectively mitigating LPS-induced inflammatory responses in lung tissue. Furthermore, in vitro studies demonstrated that myricanol treatment inhibited the expression of pro-inflammatory cytokines, attenuated signal pathway activation, and reduced oxidative stress in macrophages. In addition, we demonstrated that myricanol selectively enhances SIRT1 activation in LPS-stimulated macrophages, and all of the protective effect of myricanol were reversed through SIRT1 silencing. Remarkably, the beneficial effects of myricanol against LPS-induced sepsis were abolished in SIRT1 myeloid-specific knockout mice, underpinning the critical role of SIRT1 in mediating myricanol's therapeutic efficacy. In summary, this study provides significant evidence that myricanol acts as a potent SIRT1 activator, targeting inflammatory signal pathways and oxidative stress to suppress excessive inflammatory responses. Our findings highlight the potential of myricanol as a novel therapeutic agent for the treatment of LPS-induced sepsis.
Collapse
Affiliation(s)
- Kaiyuan Liu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liuye Yang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengchao Wang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingbo Zhu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengcen Li
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangtong Peng
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| |
Collapse
|
36
|
Chen X, Ma B, Yang Y, Zhang M, Xu F. Predicting the potentially exacerbation of severe viral pneumonia in hospital by MuLBSTA score joint CD4 + and CD8 +T cell counts: construction and verification of risk warning model. BMC Pulm Med 2024; 24:261. [PMID: 38811907 PMCID: PMC11137986 DOI: 10.1186/s12890-024-03073-y] [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: 12/13/2023] [Accepted: 05/22/2024] [Indexed: 05/31/2024] Open
Abstract
PURPOSE This study mainly focuses on the immune function and introduces CD4+, CD8+ T cells and their ratios based on the MuLBSTA score, a previous viral pneumonia mortality risk warning model, to construct an early warning model of severe viral pneumonia risk. METHODS A retrospective single-center observational study was operated from January 2021 to December 2022 at the People's Hospital of Liangjiang New Area, Chongqing, China. A total of 138 patients who met the criteria for viral pneumonia in hospital were selected and their data, including demographic data, comorbidities, laboratory results, CT scans, immunologic and pathogenic tests, treatment regimens, and clinical outcomes, were collected and statistically analyzed. RESULTS Forty-one patients (29.7%) developed severe or critical illness. A viral pneumonia severe risk warning model was successfully constructed, including eight parameters: age, bacterial coinfection, CD4+, CD4+/CD8+, multiple lung lobe infiltrations, smoking, hypertension, and hospital admission days. The risk score for severe illness in patients was set at 600 points. The model had good predictive performance (AUROC = 0.94397), better than the original MuLBSTA score (AUROC = 0.8241). CONCLUSION A warning system constructed based on immune function has a good warning effect on the risk of severe conversion in patients with viral pneumonia.
Collapse
Affiliation(s)
- Xi Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- Department of Critical Care Medicine, People's Hospital of Chongqing Liangjiang New Area, Chongqing, 401120, China
| | - Bei Ma
- Department of Critical Care Medicine, People's Hospital of Chongqing Liangjiang New Area, Chongqing, 401120, China
| | - Yu Yang
- Department of Critical Care Medicine, People's Hospital of Chongqing Liangjiang New Area, Chongqing, 401120, China
| | - Mu Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| | - Fang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
37
|
Wu Y, Wang L, Li Y, Cao Y, Wang M, Deng Z, Kang H. Immunotherapy in the context of sepsis-induced immunological dysregulation. Front Immunol 2024; 15:1391395. [PMID: 38835773 PMCID: PMC11148279 DOI: 10.3389/fimmu.2024.1391395] [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: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Sepsis is a clinical syndrome caused by uncontrollable immune dysregulation triggered by pathogen infection, characterized by high incidence, mortality rates, and disease burden. Current treatments primarily focus on symptomatic relief, lacking specific therapeutic interventions. The core mechanism of sepsis is believed to be an imbalance in the host's immune response, characterized by early excessive inflammation followed by late immune suppression, triggered by pathogen invasion. This suggests that we can develop immunotherapeutic treatment strategies by targeting and modulating the components and immunological functions of the host's innate and adaptive immune systems. Therefore, this paper reviews the mechanisms of immune dysregulation in sepsis and, based on this foundation, discusses the current state of immunotherapy applications in sepsis animal models and clinical trials.
Collapse
Affiliation(s)
- Yiqi Wu
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yun Li
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuan Cao
- Department of Emergency Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zihui Deng
- Department of Basic Medicine, Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Hongjun Kang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
38
|
Kim KM, Hwang NH, Hyun JS, Shin D. Recent Advances in IRAK1: Pharmacological and Therapeutic Aspects. Molecules 2024; 29:2226. [PMID: 38792088 PMCID: PMC11123835 DOI: 10.3390/molecules29102226] [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: 03/04/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Interleukin receptor-associated kinase (IRAK) proteins are pivotal in interleukin-1 and Toll-like receptor-mediated signaling pathways. They play essential roles in innate immunity and inflammation. This review analyzes and discusses the physiological functions of IRAK1 and its associated diseases. IRAK1 is involved in a wide range of diseases such as dry eye, which highlights its potential as a therapeutic target under various conditions. Various IRAK1 inhibitors, including Pacritinib and Rosoxacin, show therapeutic potential against malignancies and inflammatory diseases. The covalent IRAK1 inhibitor JH-X-119-01 shows promise in B-cell lymphomas, emphasizing the significance of covalent bonds in its activity. Additionally, the emergence of selective IRAK1 degraders, such as JNJ-101, provides a novel strategy by targeting the scaffolding function of IRAK1. Thus, the evolving landscape of IRAK1-targeted approaches provides promising avenues for increasingly safe and effective therapeutic interventions for various diseases.
Collapse
Affiliation(s)
| | | | - Ja-Shil Hyun
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon 21935, Republic of Korea
| | - Dongyun Shin
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon 21935, Republic of Korea
| |
Collapse
|
39
|
Zhang C, Wang S, Han Y, Zheng A, Liu G, Meng K, Yang P, Chen Z. Effects of Crude Extract of Glycyrrhiza Radix and Atractylodes macrocephala on Immune and Antioxidant Capacity of SPF White Leghorn Chickens in an Oxidative Stress Model. Antioxidants (Basel) 2024; 13:578. [PMID: 38790683 PMCID: PMC11118435 DOI: 10.3390/antiox13050578] [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: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
The natural edible characteristics of Chinese herbs have led more and more people to study them as an alternative product to antibiotics. In this study, crude extracts of Glycyrrhiza radix and Atractylodes macrocephala (abbreviated as GRAM) with glycyrrhizic acid content not less than 0.2 mg/g were selected to evaluate the effects of GRAM on the immune and antioxidant capacity of model animals. Thirty 21-day-old male Leghorn chickens were weighed and randomly assigned to one of three groups of ten animals each. The treatments comprised a control group (CON), in which saline was injected at day 31, day 33, and day 35, an LPS-treated group (LPS), in which LPS (0.5 mg/kg of BW) was injected at day 31, day 33, and day 35, and finally a GRAM and LPS-treated group, (G-L) in which a GRAM-treated diet (at GRAM 2 g/kg) was fed from day 21 to day 35 with LPS injection (0.5 mg/kg of BW) at day 31, day 33, and day 35. The results of diarrhea grade and serum antioxidant measurement showed that the LPS group had obvious diarrhea symptoms, serum ROS and MDA were significantly increased, and T-AOC was significantly decreased. The oxidative stress model of LPS was successfully established. The results of immune and antioxidant indexes showed that feeding GRAM significantly decreased levels of the pro-inflammatory factors TNF-α, IL-1β, and IL-6 (p < 0.05) and significantly increased levels of the anti-inflammatory factors IL-4 and IL-10 and levels of the antioxidant enzymes GSH-Px and CAT (p < 0.05). GRAM resisted the influence of LPS on ileum morphology, liver, and immune organs and maintained normal index values for ileum morphology, liver, and immune organs. In summary, this study confirmed the antidiarrheal effect of GRAM, which improved the immune and antioxidant capacity of model animals by regulating inflammatory cytokine levels and antioxidant enzyme activity in poultry.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Peilong Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China; (C.Z.); (S.W.); (Y.H.); (A.Z.); (G.L.); (K.M.)
| | - Zhimin Chen
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China; (C.Z.); (S.W.); (Y.H.); (A.Z.); (G.L.); (K.M.)
| |
Collapse
|
40
|
Girardis M, Coloretti I, Antonelli M, Berlot G, Busani S, Cortegiani A, De Pascale G, De Rosa FG, De Rosa S, Donadello K, Donati A, Forfori F, Giannella M, Grasselli G, Montrucchio G, Oliva A, Pasero D, Piazza O, Romagnoli S, Tascini C, Viaggi B, Tumbarello M, Viale P. Adjunctive immunotherapeutic agents in patients with sepsis and septic shock: a multidisciplinary consensus of 23. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2024; 4:28. [PMID: 38689337 PMCID: PMC11059820 DOI: 10.1186/s44158-024-00165-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND In the last decades, several adjunctive treatments have been proposed to reduce mortality in septic shock patients. Unfortunately, mortality due to sepsis and septic shock remains elevated and NO trials evaluating adjunctive therapies were able to demonstrate any clear benefit. In light of the lack of evidence and conflicting results from previous studies, in this multidisciplinary consensus, the authors considered the rational, recent investigations and potential clinical benefits of targeted adjunctive therapies. METHODS A panel of multidisciplinary experts defined clinical phenotypes, treatments and outcomes of greater interest in the field of adjunctive therapies for sepsis and septic shock. After an extensive systematic literature review, the appropriateness of each treatment for each clinical phenotype was determined using the modified RAND/UCLA appropriateness method. RESULTS The consensus identified two distinct clinical phenotypes: patients with overwhelming shock and patients with immune paralysis. Six different adjunctive treatments were considered the most frequently used and promising: (i) corticosteroids, (ii) blood purification, (iii) immunoglobulins, (iv) granulocyte/monocyte colony-stimulating factor and (v) specific immune therapy (i.e. interferon-gamma, IL7 and AntiPD1). Agreement was achieved in 70% of the 25 clinical questions. CONCLUSIONS Although clinical evidence is lacking, adjunctive therapies are often employed in the treatment of sepsis. To address this gap in knowledge, a panel of national experts has provided a structured consensus on the appropriate use of these treatments in clinical practice.
Collapse
Affiliation(s)
- Massimo Girardis
- Anesthesia and Intensive Care Medicine, Policlinico Di Modena, University of Modena and Reggio Emilia, Modena, Italy.
| | - Irene Coloretti
- Anesthesia and Intensive Care Medicine, Policlinico Di Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Antonelli
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Berlot
- Anesthesia and Intensive Care, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Stefano Busani
- Anesthesia and Intensive Care Medicine, Policlinico Di Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Cortegiani
- Department of Surgical, Oncological and Oral Science (Di.Chir.On.S.), University of Palermo, Palermo, Italy
- Department of Anaesthesia, Intensive Care and Emergency, Policlinico Paolo Giaccone, Palermo, Italy
| | - Gennaro De Pascale
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Silvia De Rosa
- Anesthesia and Intensive Care, Santa Chiara Regional Hospital, APSS, Trento, Italy
| | - Katia Donadello
- Department of Surgery, Dentistry, Ginaecology and Paediatrics, University of Verona, and Anesthesia and Intensive Care Unit B, University Hospital Integrated Trust of Verona, Verona, Italy
| | - Abele Donati
- Anesthesia and Intensive Care, Azienda Ospedaliero Universitaria Delle Marche, Ancona, Italy
| | - Francesco Forfori
- Anesthesia and Intensive Care, Anesthesia and Resuscitation Department, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Maddalena Giannella
- Department of Medical and Surgical Sciences Infectious Diseases Unit, IRCCS Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giacomo Grasselli
- Department of Anesthesia, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giorgia Montrucchio
- Department of Surgical Sciences, Departement of Anesthesia, Resuscitation and Emergency Torino, University of Turin, Turin, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Daniela Pasero
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Ornella Piazza
- University Hospital "San Giovanni Di Dio E Ruggi d'Aragona", Salerno, Italy
| | - Stefano Romagnoli
- Department of Health Science, Department of Anesthesia and Intensive Care, University of Florence, Careggi University Hospital, Florence, Italy
| | - Carlo Tascini
- Department of Medicine (DAME), Infectious Diseases Clinic, University of Udine, Udine, Italy
| | - Bruno Viaggi
- Anesthesia and Intensive Care, Careggi University Hospital, Florence, Italy
| | - Mario Tumbarello
- Infectious and Tropical Diseases Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences Infectious Diseases Unit, IRCCS Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
41
|
Li F, Qu H, Li Y, Liu J, Fu H. Establishment and assessment of mortality risk prediction model in patients with sepsis based on early-stage peripheral lymphocyte subsets. Aging (Albany NY) 2024; 16:7460-7473. [PMID: 38669099 PMCID: PMC11087126 DOI: 10.18632/aging.205772] [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: 11/24/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024]
Abstract
This study is aimed to explore the value of lymphocyte subsets in evaluating the severity and prognosis of sepsis. The counts of lymphocytes, CD3+ T cells, CD4+ T cells, CD8+ T cells, CD19+ B cells, and NK cells significantly decreased between day 1 and day 3 in both the survivor and the non-survivor groups. The peripheral lymphocyte subsets (PLS) at day 1 were not significantly different between the survivor and the non-survivor groups. However, at day 3, the counts of lymphocytes, CD3+ T cells, CD4+ T cells, and NK cells were remarkably lower in the non-survivor group. No significant differences in CD8+ T cells, or CD19+ B cells were observed. The PLS index was independently and significantly associated with the 28-day mortality risk in septic patients (OR: 3.08, 95% CI: 1.18-9.67). Based on these clinical parameters and the PLS index, we developed a nomograph for evaluating the individual mortality risk in sepsis. The area under the curve of prediction with the PLS index was significantly higher than that from the model with only clinical parameters (0.912 vs. 0.817). Our study suggests that the decline of PLS occurred in the early stage of sepsis. The new novel PLS index can be an independent predictor of 28-day mortality in septic patients. The prediction model based on clinical parameters and the PLS index has relatively high predicting ability.
Collapse
Affiliation(s)
- Fuzhu Li
- The First Affiliated Hospital, Department of Neurosurgical Intensive Care Unit, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, China
| | - Hongtao Qu
- The First Affiliated Hospital, Department of Neurosurgical Intensive Care Unit, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, China
| | - Yimin Li
- The First Affiliated Hospital, Department of Neurosurgical Intensive Care Unit, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, China
| | - Jie Liu
- Department of Emergency, Shenzhen United Family Hospital, Shenzhen, Guangdong 518048, China
| | - Hongyun Fu
- The Affiliated Nanhua Hospital, Department of Docimasiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China
| |
Collapse
|
42
|
Gong M, Qi S, Wu Z, Huang Y, Wu L, Wang X, He L, Lin L, Lin D. A novel therapeutic approach to modulate the inflammatory cascade: A timely exogenous local inflammatory response attenuates the sepsis-induced cytokine storm. Cytokine 2024; 176:156533. [PMID: 38340550 DOI: 10.1016/j.cyto.2024.156533] [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: 10/19/2023] [Revised: 01/03/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND The emergence of severe sepsis is contingent upon the occurrence of a cytokine storm (CS), a multifaceted process intricately entwined with the temporal dimension, thereby rendering the infection response remarkably intricate. Consequently, it becomes imperative to discern and accurately identify the optimal timing for interventions, predicated upon the dynamic timeline of inflammatory changes. Moreover, the administration of exogenous low-dose pro-inflammatory agents has exhibited the potential to impede the relentless progression of the inflammatory cascade. Hence, the present study aims to scrutinize the impact of exogenous Local Inflammatory Response (eLIR) on the body surface in the context of the inflammatory cascade during sepsis, within a temporal framework, with a particular emphasis on the point of exacerbation of inflammation. METHODS Rats were induced sterile sepsis by intraperitoneal injection of zymosan (ZY) at an appropriate dosage. The temporal progression of inflammatory changes and eLIR effects were described based on the trend of serum crucial inflammatory cytokines, tring to quest time-point of inflammatory aggravation in sepsis. Then, the varying degrees of surface inflammation caused by eLIR on this time point leading to the final effects on the inflammatory cascade response were explored. In addition, given the authentic pathological progression of sepsis, further observation was conducted on the impact of another intervention timing of eLIR on the inflammatory cascade. The survival rate was measured. Serum and organ related inflammatory cytokines were detected, and organ histopathology was investigated. RESULTS In present study, a dosage of 600 mg/kg ZY was found to be optimal for the sterile sepsis model. Initiating eLIR 6 h prior to ZY injection, the maximum effect point of eLIR could be precisely align with the inflammatory aggravation point of sterile sepsis. Initiating eLIR at this time, 3 sessions of eLIR were found to be more effective than 1 or 2 sessions in mitigating inflammatory responses during the initial stage of inflammation and the peak of inflammation. Notably, the findings also suggested that this intervention improve survival rate. In addition, the anti-inflammatory efficacy has been substantially diminished by the prompt initiation of 3 sessions of eLIR immediately after ZY injection at the onset of sepsis. Similarly, the current findings did not demonstrate a statistically significant enhancement in survival rates with eLIR at this time point. CONCLUSIONS Compared with the initial stage of inflammation, low-scale inflammation caused by a certain intensity of eLIR (3 sessions) on the body surface can more effectively pry the inflammation aggravation time-point, thereby shifting the pro-inflammatory to anti-inflammatory milieu, impeding the disproportionate cytokines release in inflammatory diseases, slowing down the inflammatory cascade, and improving the survival rate of sepsis.
Collapse
Affiliation(s)
- Meng Gong
- College of Acupuncture, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Shiyi Qi
- College of Acupuncture, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Zhiting Wu
- College of Acupuncture, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Ying Huang
- College of Acupuncture, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Lihua Wu
- Department of Otolaryngology, Fujian provincial hospital, Fuzhou, Fujian Province, China
| | - Xiangbin Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Lingling He
- College of Acupuncture, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Lili Lin
- College of Acupuncture, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China; Institute of Acupuncture and Meridian, Fujian Academy of Chinese Medical Sciences, Fuzhou, Fujian Province, China
| | - Dong Lin
- College of Acupuncture, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China; Institute of Acupuncture and Meridian, Fujian Academy of Chinese Medical Sciences, Fuzhou, Fujian Province, China.
| |
Collapse
|
43
|
Ratajczak MZ, Bujko K, Brzezniakiewicz-Janus K, Ratajczak J, Kucia M. Hematopoiesis Revolves Around the Primordial Evolutional Rhythm of Purinergic Signaling and Innate Immunity - A Journey to the Developmental Roots. Stem Cell Rev Rep 2024; 20:827-838. [PMID: 38363476 PMCID: PMC10984895 DOI: 10.1007/s12015-024-10692-9] [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] [Accepted: 02/04/2024] [Indexed: 02/17/2024]
Abstract
A cell's most significant existential task is to survive by ensuring proper metabolism, avoiding harmful stimuli, and adapting to changing environments. It explains why early evolutionary primordial signals and pathways remained active and regulate cell and tissue integrity. This requires energy supply and a balanced redox state. To meet these requirements, the universal intracellular energy transporter purine nucleotide-adenosine triphosphate (ATP) became an important signaling molecule and precursor of purinergic signaling after being released into extracellular space. Similarly, ancient proteins involved in intracellular metabolism gave rise to the third protein component (C3) of the complement cascade (ComC), a soluble arm of innate immunity. These pathways induce cytosol reactive oxygen (ROS) and reactive nitrogen species (RNS) that regulate the redox state of the cells. While low levels of ROS and RNS promote cell growth and differentiation, supra-physiological concentrations can lead to cell damage by pyroptosis. This balance explains the impact of purinergic signaling and innate immunity on cell metabolism, organogenesis, and tissue development. Subsequently, along with evolution, new regulatory cues emerge in the form of growth factors, cytokines, chemokines, and bioactive lipids. However, their expression is still modulated by both primordial signaling pathways. This review will focus on the data that purinergic signaling and innate immunity carry on their ancient developmental task in hematopoiesis and specification of hematopoietic stem/progenitor cells (HSPCs). Moreover, recent evidence shows both these regulatory pathways operate in a paracrine manner and inside HSPCs at the autocrine level.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland.
- Department of Hematology, University of Zielona Gora, Multi-Specialist Hospital Gorzow Wlkp., Gorzow Wielkopolski, Poland.
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA.
| | - Kamila Bujko
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine at Medical, University of Warsaw, Warsaw, Poland
| | | | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Magdalena Kucia
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine at Medical, University of Warsaw, Warsaw, Poland
| |
Collapse
|
44
|
Huang W, Wang L, Huang Z, Sun Z, Zheng B. Peroxiredoxin 3 has a crucial role in the macrophage polarization by regulating mitochondrial homeostasis. Respir Res 2024; 25:110. [PMID: 38431661 PMCID: PMC10909251 DOI: 10.1186/s12931-024-02739-9] [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: 10/15/2023] [Accepted: 02/19/2024] [Indexed: 03/05/2024] Open
Abstract
Acute lung injury (ALI) is one of the life-threatening complications of sepsis, and macrophage polarization plays a crucial role in the sepsis-associated ALI. However, the regulatory mechanisms of macrophage polarization in ALI and in the development of inflammation are largely unknown. In this study, we demonstrated that macrophage polarization occurs in sepsis-associated ALI and is accompanied by mitochondrial dysfunction and inflammation, and a decrease of PRDX3 promotes the initiation of macrophage polarization and mitochondrial dysfunction. Mechanistically, PRDX3 overexpression promotes M1 macrophages to differentiate into M2 macrophages, and enhances mitochondrial functional recovery after injury by reducing the level of glycolysis and increasing TCA cycle activity. In conclusion, we identified PRDX3 as a critical hub integrating oxidative stress, inflammation, and metabolic reprogramming in macrophage polarization. The findings illustrate an adaptive mechanism underlying the link between macrophage polarization and sepsis-associated ALI.
Collapse
Affiliation(s)
- Wenhui Huang
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lianfang Wang
- Department of Respiratory and Critical Care Medicine, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Guangxi, China
| | - Zhipeng Huang
- Dongguan Hospital of Integrated Chinese and Western Medicine, Dongguan, China
| | - Zhichao Sun
- The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine, Guangzhou, China
| | - Bojun Zheng
- Department of Critical Care Medicine, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
45
|
Bertozzi G, Ferrara M, Di Fazio A, Maiese A, Delogu G, Di Fazio N, Tortorella V, La Russa R, Fineschi V. Oxidative Stress in Sepsis: A Focus on Cardiac Pathology. Int J Mol Sci 2024; 25:2912. [PMID: 38474158 PMCID: PMC10931756 DOI: 10.3390/ijms25052912] [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: 02/09/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
This study aims to analyze post-mortem human cardiac specimens, to verify and evaluate the existence or extent of oxidative stress in subjects whose cause of death has been traced to sepsis, through immunohistological oxidative/nitrosative stress markers. Indeed, in the present study, i-NOS, NOX2, and nitrotyrosine markers were higher expressed in the septic death group when compared to the control group, associated with also a significant increase in 8-OHdG, highlighting the pivotal role of oxidative stress in septic etiopathogenesis. In particular, 70% of cardiomyocyte nuclei from septic death specimens showed positivity for 8-OHdG. Furthermore, intense and massive NOX2-positive myocyte immunoreaction was noticed in the septic group, as nitrotyrosine immunostaining intense reaction was found in the cardiac cells. These results demonstrated a correlation between oxidative and nitrosative stress imbalance and the pathophysiology of cardiac dysfunction documented in cases of sepsis. Therefore, subsequent studies will focus on the expression of oxidative stress markers in other organs and tissues, as well as on the involvement of the intracellular pattern of apoptosis, to better clarify the complex pathogenesis of multi-organ failure, leading to support the rationale for including therapies targeting redox abnormalities in the management of septic patients.
Collapse
Affiliation(s)
- Giuseppe Bertozzi
- SIC Medicina Legale, Via Potito Petrone, 85100 Potenza, Italy; (G.B.); (M.F.); (A.D.F.)
| | - Michela Ferrara
- SIC Medicina Legale, Via Potito Petrone, 85100 Potenza, Italy; (G.B.); (M.F.); (A.D.F.)
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy; (G.D.); (N.D.F.); (V.T.)
| | - Aldo Di Fazio
- SIC Medicina Legale, Via Potito Petrone, 85100 Potenza, Italy; (G.B.); (M.F.); (A.D.F.)
| | - Aniello Maiese
- Institute of Legal Medicine, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
| | - Giuseppe Delogu
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy; (G.D.); (N.D.F.); (V.T.)
| | - Nicola Di Fazio
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy; (G.D.); (N.D.F.); (V.T.)
| | - Vittoria Tortorella
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy; (G.D.); (N.D.F.); (V.T.)
| | - Raffaele La Russa
- Department of Clinical Medicine, Public Health, Life and Environment Science, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy; (G.D.); (N.D.F.); (V.T.)
| |
Collapse
|
46
|
Zhu X, Bai B, Ge X, Zheng B, Xiao Z, Tang Y, Fang L, Tang Y, Dai Y, Zhang B, Zhang Y. Costunolide attenuates LPS-induced inflammation and lung injury through inhibiting IKK/NF-κB signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1601-1610. [PMID: 37688623 DOI: 10.1007/s00210-023-02705-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
Inflammation is an important pathological process of many acute and chronic diseases, such as sepsis, arthritis, and cancer. Many factors can lead to an inflammatory state of the body, among which bacterial infection plays an important role. Bacterial infection often leads to sepsis, acute lung injury (ALI), or its more serious form of acute respiratory distress syndrome, which are the main fatal diseases in intensive care units. Costunolide has been reported to possess excellent anti-inflammatory activity; however, whether it can affect inflammation induced by gram-negative bacterial is still unclear. Lipopolysaccharide (LPS) stimulated mouse peritoneal macrophages (MPMs) to release proinflammatory cytokines was used as the cell model. The mouse model of sepsis and ALI was built through injecting intravenously and intratracheally of LPS. In the present study, costunolide inhibited LPS-induced inflammatory response through IKK/NF-κB signaling pathway in macrophages. In vivo, costunolide attenuated LPS-induced septic death in mice. Meanwhile, costunolide treatment alleviated LPS-induced lung injury and inflammation via inhibiting the infiltration of inflammatory cells and the expression of inflammatory cytokines. Taken together, these results demonstrated that costunolide could attenuate gram-negative bacterial induced inflammation and diseases and might be a potential candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xiaona Zhu
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China
| | - Bin Bai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiangting Ge
- Department of Pulmonary and Critical Care Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Bin Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zhongxiang Xiao
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China
| | - Yue Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Letong Fang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yelin Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yuanrong Dai
- Department of Pulmonary and Critical Care Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Bing Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China.
| | - Yali Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
47
|
Lai J, Liang J, Chen K, Guan B, Chen Z, Chen L, Fan J, Zhang Y, Li Q, Su J, Chen Q, Lin J. Carrimycin ameliorates lipopolysaccharide and cecal ligation and puncture-induced sepsis in mice. Chin J Nat Med 2024; 22:235-248. [PMID: 38553191 DOI: 10.1016/s1875-5364(24)60600-x] [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: 07/14/2023] [Indexed: 04/02/2024]
Abstract
Carrimycin (CA), sanctioned by China's National Medical Products Administration (NMPA) in 2019 for treating acute bronchitis and sinusitis, has recently been observed to exhibit multifaceted biological activities, encompassing anti-inflammatory, antiviral, and anti-tumor properties. Despite these applications, its efficacy in sepsis treatment remains unexplored. This study introduces a novel function of CA, demonstrating its capacity to mitigate sepsis induced by lipopolysaccharide (LPS) and cecal ligation and puncture (CLP) in mice models. Our research employed in vitro assays, real-time quantitative polymerase chain reaction (RT-qPCR), and RNA-seq analysis to establish that CA significantly reduces the levels of pro-inflammatory cytokines, namely tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6), in response to LPS stimulation. Additionally, Western blotting and immunofluorescence assays revealed that CA impedes Nuclear Factor Kappa B (NF-κB) activation in LPS-stimulated RAW264.7 cells. Complementing these findings, in vivo experiments demonstrated that CA effectively alleviates LPS- and CLP-triggered organ inflammation in C57BL/6 mice. Further insights were gained through 16S sequencing, highlighting CA's pivotal role in enhancing gut microbiota diversity and modulating metabolic pathways, particularly by augmenting the production of short-chain fatty acids in mice subjected to CLP. Notably, a comparative analysis revealed that CA's anti-inflammatory efficacy surpasses that of equivalent doses of aspirin (ASP) and TIENAM. Collectively, these findings suggest that CA exhibits significant therapeutic potential in sepsis treatment. This discovery provides a foundational theoretical basis for the clinical application of CA in sepsis management.
Collapse
Affiliation(s)
- Junzhong Lai
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jiadi Liang
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China; Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Kunsen Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Biyun Guan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Zhirong Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Linqin Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Jiqiang Fan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Qiumei Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China.
| | - Jizhen Lin
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China; The Department of Otolaryngology, Head & Neck Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
48
|
Jin S, Yin JB, Li W, Zang LL. Effect of neutrophil to lymphocyte ratio on prognosis of elderly patients with severe sepsis combined with diabetes mellitus. BMC Geriatr 2024; 24:211. [PMID: 38424501 PMCID: PMC10905898 DOI: 10.1186/s12877-024-04757-0] [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: 09/11/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND To investigate the predictive value of neutrophil-to-lymphocyte ratio (NLR) in the short-term prognosis of elderly patients with severe sepsis combined with diabetes mellitus (DM). METHODS The clinical data of 162 elderly patients with severe sepsis combined with DM from January 2018 to December 2022 were retrospectively collected. These patients were divided into a survival group (n = 104) and a death group (n = 58) according to 90-day prognosis. The number of neutrophils, lymphocytes, and NLR were compared. The optimal cut-off value for NLR to predict 90-day prognosis in elderly patients with severe sepsis combined with DM was determined using Receiver Operator Characteristic (ROC) curves, and the patients were divided into high and low NLR groups depending on the optimal cut-off value. The Kaplan-Meier method was used to plot the survival curves of the high and low NLR groups. Risk factors for the 90-day death in elderly patients with severe sepsis combined with DM were analyzed by a multivariate cox regression model. RESULTS There were no significant differences in gender, age, history of hypertension and hyperlipidemia, intensive care unit (ICU) stay, duration of mechanical ventilation, and oxygenation index between the survival group and death group (p > 0.05). However, acute physiological and chronic health evaluation II (APACHE II) scores, and sepsis-related organ failure assessment (SOFA) scores were significantly lower in the survival group compared with the death group (p < 0.05). In the survival group, neutrophils counts and NLR were much lower than those in the death group, while lymphocytes counts were much higher (p < 0.05). ROC curves showed that the optimal cut-off value for NLR to predict 90-day mortality in elderly patients with severe sepsis combined with DM was 3.482. Patients were divided into high NLR and low NLR groups based on whether NLR was ≥ 3.482. In terms of the log-rank test results, patients in the low NLR group had a significantly higher 90-day survival rate than those in the high NLR group (Logrank χ2 = 8.635, p = 0.003). The multivariate cox regression model showed that the length of ICU stay longer than 15 days and NLR ≥ 3.482 were independent risk factors for 90-day prognosis in elderly patients with severe sepsis combined with DM. CONCLUSION NLR ≥ 3.482 can be used to predict whether poor prognosis occurs in the short term after illness in elderly patients with severe sepsis combined with DM, and has good assessment value.
Collapse
Affiliation(s)
- Shan Jin
- Department of Neurology, Shandong Province 960th Hospital of the People's Liberation Army, 250031, Jinan, Shandong, China.
| | - Jun-Bin Yin
- Department of Neurology, Shandong Province 960th Hospital of the People's Liberation Army, 250031, Jinan, Shandong, China
| | - Wei Li
- Department of Neurology, Shandong Province 960th Hospital of the People's Liberation Army, 250031, Jinan, Shandong, China
| | - Li-Li Zang
- Department of Neurology, Shandong Province 960th Hospital of the People's Liberation Army, 250031, Jinan, Shandong, China
| |
Collapse
|
49
|
Ashayeripanah M, Vega-Ramos J, Fernandez-Ruiz D, Valikhani S, Lun ATL, White JT, Young LJ, Yaftiyan A, Zhan Y, Wakim L, Caminschi I, Lahoud MH, Lew AM, Shortman K, Smyth GK, Heath WR, Mintern JD, Roquilly A, Villadangos JA. Systemic inflammatory response syndrome triggered by blood-borne pathogens induces prolonged dendritic cell paralysis and immunosuppression. Cell Rep 2024; 43:113754. [PMID: 38354086 DOI: 10.1016/j.celrep.2024.113754] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 12/01/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Blood-borne pathogens can cause systemic inflammatory response syndrome (SIRS) followed by protracted, potentially lethal immunosuppression. The mechanisms responsible for impaired immunity post-SIRS remain unclear. We show that SIRS triggered by pathogen mimics or malaria infection leads to functional paralysis of conventional dendritic cells (cDCs). Paralysis affects several generations of cDCs and impairs immunity for 3-4 weeks. Paralyzed cDCs display distinct transcriptomic and phenotypic signatures and show impaired capacity to capture and present antigens in vivo. They also display altered cytokine production patterns upon stimulation. The paralysis program is not initiated in the bone marrow but during final cDC differentiation in peripheral tissues under the influence of local secondary signals that persist after resolution of SIRS. Vaccination with monoclonal antibodies that target cDC receptors or blockade of transforming growth factor β partially overcomes paralysis and immunosuppression. This work provides insights into the mechanisms of paralysis and describes strategies to restore immunocompetence post-SIRS.
Collapse
Affiliation(s)
- Mitra Ashayeripanah
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Javier Vega-Ramos
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia; School of Biomedical Sciences, Faculty of Medicine & Health and the UNSW RNA Institute, The University of New South Wales, Kensington, NSW 2052, Australia
| | - Shirin Valikhani
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Aaron T L Lun
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jason T White
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Louise J Young
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Atefeh Yaftiyan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Yifan Zhan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Linda Wakim
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Irina Caminschi
- Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Mireille H Lahoud
- Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Andrew M Lew
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ken Shortman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - William R Heath
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Justine D Mintern
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Antoine Roquilly
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia; Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000 Nantes, France; CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, 44000 Nantes, France.
| | - Jose A Villadangos
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3000, Australia; Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
50
|
Zeng J, Yang Z, Xu D, Song J, Liu Y, Qin J, Weng Z. NMI Functions as Immuno-regulatory Molecule in Sepsis by Regulating Multiple Signaling Pathways. Inflammation 2024; 47:60-73. [PMID: 37679586 DOI: 10.1007/s10753-023-01893-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/02/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023]
Abstract
Sepsis-induced tissue and organ damage is caused by an overactive inflammatory response, immune dysfunction, and coagulation dysfunction. Danger-associated molecular pattern (DAMP) molecules play a critical role in the excessive inflammation observed in sepsis. In our previous research, we identified NMI as a new type of DAMP molecule that promotes inflammation in sepsis by binding to toll-like receptor 4 (TLR4) on macrophage surfaces, activating the NF-κB pathway, and releasing pro-inflammatory cytokines. However, it is still unknown whether NMI plays a significant role in other pathways. Our analysis of bulk and single-cell transcriptome data from the GEO database revealed a significant increase in NMI expression in neutrophils and monocytes in sepsis patients. It is likely that NMI functions through multiple receptors in sepsis, including IFNAR1, IFNAR2, TNFR1, TLR3, TLR1, IL9R, IL10RB, and TLR4. Furthermore, the correlation between NMI expression and the activation of NF-κB, MAPK, and JAK pathways, as well as the up-regulation of their downstream pro-inflammatory factors, demonstrates that NMI may exacerbate the inflammatory response through these signaling pathways. Finally, we demonstrated that STAT1 phosphorylation was enhanced in RAW cells upon stimulation with NMI, supporting the activation of JAK signaling pathway by NMI. Collectively, these findings shed new light on the functional mechanism of NMI in sepsis.
Collapse
Affiliation(s)
- Jinhua Zeng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Zixin Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Dan Xu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Jierong Song
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Yingfang Liu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Jing Qin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Zhuangfeng Weng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|