151
|
Bao K, Wang M, Liu L, Zhang D, Jin C, Zhang J, Shi L. Jinhong decoction protects sepsis-associated acute lung injury by reducing intestinal bacterial translocation and improving gut microbial homeostasis. Front Pharmacol 2023; 14:1079482. [PMID: 37081964 PMCID: PMC10110981 DOI: 10.3389/fphar.2023.1079482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Currently no specific treatments are available for sepsis and the associated syndromes including acute lung injury (ALI). Jinhong Decoction (JHD) is a traditional Chinese prescription, and it has been applied clinically as an efficient and safe treatment for sepsis, but the underlying mechanism remains unknown. The aim of the study was to explore the potential mechanisms of JHD ameliorating sepsis and concurrent ALI.Methods: The cecum ligation puncture (CLP)- induced murine sepsis model was established for determining the efficacy of JHD protecting CLP and ALI. The role of gut microbiota involved in the efficacy of JHD was evaluated by 16S rRNA sequencing and fecal microbiota transplantation (FMT). Translocation of intestinal Escherichia coli (E. coli) to lungs after CLP was verified by qPCR and in vivo-imaging. Intestinal permeability was analyzed by detecting FITC-dextran leakness. Junction proteins were evaluated by Western blotting and immunofluorescence.Results: JHD treatment remarkably increased survival rate of septic mice and alleviated sepsis-associated lung inflammation and injury. FMT suggested that the protective role for JHD was mediated through the regulation of gut microbiota. We further revealed that JHD administration partially restored the diversity and configuration of microbiome that was distorted by CLP operation. Of interest, the intestinal bacteria, E. coli particularly, was found to translocate into the lungs upon CLP via disrupting the intestinal mucosal barrier, leading to the inflammatory response and tissue damage in lungs. JHD impeded the migration and hence lung accumulation of intestinal E. coli, and thereby prevented severe ALI associated with sepsis. This effect is causatively related with the ability of JHD to restore intestinal barrier by up-regulating tight junctions.Conclusion: Our study unveils a mechanism whereby the migration of gut bacteria leads to sepsis-associated ALI, and we demonstrate the potential of JHD as an effective strategy to block this bacterial migration for treating sepsis and the associated immunopathology in the distal organs.
Collapse
Affiliation(s)
- Kaifan Bao
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Meiling Wang
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Li Liu
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Dongya Zhang
- Department of Medical Microbiology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Cuiyuan Jin
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Junfeng Zhang
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Liyun Shi
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, China
- *Correspondence: Liyun Shi,
| |
Collapse
|
152
|
Shen Y, Zhang L, Wu P, Huang Y, Xin S, Zhang Q, Zhao S, Sun H, Lei G, Zhang T, Han W, Wang Z, Jiang J, Yu X. Construction and evaluation of networks among multiple postoperative complications. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 232:107439. [PMID: 36870170 DOI: 10.1016/j.cmpb.2023.107439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/31/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND AND OBJECTIVE Postoperative complications confer an increased risk of reoperation, prolonged length of hospital stay, and increased mortality. Many studies have attempted to identify the complex associations among complications to preemptively interrupt their progression, but few studies have looked at complications as a whole to reveal and quantify their possible trajectories of progression. The main objective of this study was to construct and quantify the association network among multiple postoperative complications from a comprehensive perspective to elucidate the possible evolution trajectories. METHODS In this study, a Bayesian network model was proposed to analyze the associations among 15 complications. Prior evidence and score-based hill-climbing algorithms were used to build the structure. The severity of complications was graded according to their connection to death, with the association between them quantified using conditional probabilities. The data of surgical inpatients used in this study were collected from four regionally representative academic/teaching hospitals in a prospective cohort study in China. RESULTS In the network obtained, 15 nodes represented complications or death, and 35 arcs with arrows represented the directly dependent relationship between them. With three grades classified on that basis, the correlation coefficients of complications within grades increased with increased grade, ranging from -0.11 to -0.06, 0.16, and 0.21 to 0.4 in grade 1 to grade 3, respectively. Moreover, the probability of each complication in the network increased with the occurrence of any other complication, even mild complications. Most seriously, once cardiac arrest requiring cardiopulmonary resuscitation occurs, the probability of death will be up to 88.1%. CONCLUSIONS The present evolving network can facilitate the identification of strong associations among specific complications and provides a basis for the development of targeted measures to prevent further deterioration in high-risk patients.
Collapse
Affiliation(s)
- Yubing Shen
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, No.5, Dongdansantiao Street, Dong Cheng District, Beijing 100005, China
| | - Luwen Zhang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, No.5, Dongdansantiao Street, Dong Cheng District, Beijing 100005, China
| | - Peng Wu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, No.5, Dongdansantiao Street, Dong Cheng District, Beijing 100005, China
| | - Yuguang Huang
- Department of Anaesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shijie Xin
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Qiang Zhang
- Department of Neurosurgery, Qinghai Provincial People's Hospital, Xining, Qinghai Province, China
| | - Shengxiu Zhao
- Department of Nursing, Qinghai Provincial People's Hospital, Xining, Qinghai Province, China
| | - Hong Sun
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Han
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, No.5, Dongdansantiao Street, Dong Cheng District, Beijing 100005, China
| | - Zixing Wang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, No.5, Dongdansantiao Street, Dong Cheng District, Beijing 100005, China
| | - Jingmei Jiang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, No.5, Dongdansantiao Street, Dong Cheng District, Beijing 100005, China.
| | - Xiaochu Yu
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1, ShuaiFuYuan, WangFuJing, Dong Cheng District, Beijing 100730, China.
| |
Collapse
|
153
|
Wang J, Song Q, Yang S, Wang H, Meng S, Huang L, Li Q, Xu J, Xie J, Huang Y. Effects of hydrocortisone combined with vitamin C and vitamin B1 versus hydrocortisone alone on microcirculation in septic shock patients: A pilot study. Clin Hemorheol Microcirc 2023:CH221444. [PMID: 36911931 PMCID: PMC10357145 DOI: 10.3233/ch-221444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
OBJECTIVE To investigate the effects of hydrocortisone combined with vitamin C and vitamin B1 versus hydrocortisone on sublingual microcirculation in septic shock patients. METHODS This pilot study enrolled septic shock patients admitted to the ICU of a tertiary teaching hospital from February 2019 to January 2020. We randomly assigned the enrolled patients to the treatment group (hydrocortisone combined with vitamin C and vitamin B1 added to standard care) and the control group (hydrocortisone alone added to standard care) in a 1 : 1 ratio. The primary outcome was perfused small vascular density (sPVD) monitored by a sublingual microcirculation imaging system at 24 hours after treatment. RESULTS Twelve patients in the treatment group and ten in the control group completed the study. The baseline characteristics were comparable between the groups. No statistically significant difference was found in the sPVD between the groups at baseline. The sPVD in the treatment group was significantly higher than that in the control group at 4 hours after treatment (mean difference, 7.042; 95% CI, 2.227-11.857; P = 0.009) and 24 hours after treatment (mean difference, 7.075; 95% CI, 2.390-11.759; P = 0.008). CONCLUSIONS Compared with hydrocortisone, hydrocortisone combined with vitamin C and vitamin B1 significantly improves microcirculation in septic shock patients.
Collapse
Affiliation(s)
- Jinlong Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Qianwen Song
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shuhe Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Haofei Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shanshan Meng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lili Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Qing Li
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jingyuan Xu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jianfeng Xie
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yingzi Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
154
|
Fan X, Mai C, Zuo L, Huang J, Xie C, Jiang Z, Li R, Yao X, Fan X, Wu Q, Yan P, Liu L, Chen J, Xie Y, Leung ELH. Herbal formula BaWeiBaiDuSan alleviates polymicrobial sepsis-induced liver injury via increasing the gut microbiota Lactobacillus johnsonii and regulating macrophage anti-inflammatory activity in mice. Acta Pharm Sin B 2023; 13:1164-1179. [PMID: 36970196 PMCID: PMC10031256 DOI: 10.1016/j.apsb.2022.10.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/19/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Sepsis-induced liver injury (SILI) is an important cause of septicemia deaths. BaWeiBaiDuSan (BWBDS) was extracted from a formula of Panax ginseng C. A. Meyer, Lilium brownie F. E. Brown ex Miellez var. viridulum Baker, Polygonatum sibiricum Delar. ex Redoute, Lonicera japonica Thunb., Hippophae rhamnoides Linn., Amygdalus Communis Vas, Platycodon grandiflorus (Jacq.) A. DC., and Cortex Phelloderdri. Herein, we investigated whether the BWBDS treatment could reverse SILI by the mechanism of modulating gut microbiota. BWBDS protected mice against SILI, which was associated with promoting macrophage anti-inflammatory activity and enhancing intestinal integrity. BWBDS selectively promoted the growth of Lactobacillus johnsonii (L. johnsonii) in cecal ligation and puncture treated mice. Fecal microbiota transplantation treatment indicated that gut bacteria correlated with sepsis and was required for BWBDS anti-sepsis effects. Notably, L. johnsonii significantly reduced SILI by promoting macrophage anti-inflammatory activity, increasing interleukin-10+ M2 macrophage production and enhancing intestinal integrity. Furthermore, heat inactivation L. johnsonii (HI-L. johnsonii) treatment promoted macrophage anti-inflammatory activity and alleviated SILI. Our findings revealed BWBDS and gut microbiota L. johnsonii as novel prebiotic and probiotic that may be used to treat SILI. The potential underlying mechanism was at least in part, via L. johnsonii-dependent immune regulation and interleukin-10+ M2 macrophage production.
Collapse
Affiliation(s)
- Xiaoqing Fan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Chutian Mai
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Ling Zuo
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jumin Huang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Chun Xie
- Cancer Center, Faculty of Health Science; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
| | - Zebo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Runze Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Xiaojun Yao
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xingxing Fan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Qibiao Wu
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Peiyu Yan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Liang Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Jianxin Chen
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ying Xie
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
| |
Collapse
|
155
|
Peng K, Yang F, Qiu C, Yang Y, Lan C. Rosmarinic acid protects against lipopolysaccharide-induced cardiac dysfunction via activating Sirt1/PGC-1α pathway to alleviate mitochondrial impairment. Clin Exp Pharmacol Physiol 2023; 50:218-227. [PMID: 36350269 DOI: 10.1111/1440-1681.13734] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/17/2022] [Accepted: 11/02/2022] [Indexed: 11/10/2022]
Abstract
Sepsis-induced cardiomyopathy is a decisive factor that plays a critical role in the high mortality of septic patients in the critically ill. Mitochondrial dysfunction occurring during sepsis is a vital contributor to the pathogenesis of myocardial damage. Rosmarinic acid (RA), a natural poly-phenolic compound, has showed cardio-protective and mitochondrial protective effect. The present study was aimed to investigate the effect of RA on sepsis-induced cardiomyopathy. Adult mice were subjected to intraperitoneal injection of saline (control) or lipopolysaccharide (LPS, 5 mg/kg) to mimic sepsis-induced cardiomyopathy. Immediately after LPS challenge, vehicle or RA (100 mg/kg/day) was administrated via gavage. Cardiac function was examined with echocardiographic analyses 12 hours after LPS challenge and cumulative survival of mice was recorded for 8 days. Heart tissues were harvested 12 hours after LPS challenge to perform histological analyses and determine mitochondrial function. We found RA significantly improved cardiac function and survival of LPS-injected mice. Histologically, RA attenuated LPS-mediated cardiomyocyte damage, indicated by decreased cardiomyocyte apoptosis and improved myocardial swollen and disarrangement. Moreover, RA attenuated LPS-mediated myocardial mitochondrial dysfunction, indicated by improved mitochondrial ultrastructure, increased mitochondrial membrane potential (MMP), synthesis of adenosine triphosphate (ATP), markedly decreased reactive oxygen species (ROS) level and alleviated oxidative stress in heart tissues. RA treatment downregulated protein expression of Sirt1 and peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), and Sirt1 inhibition blocked protective effect of RA on LPS-induced myocardial damage and mitochondrial dysfunction. Collectively, RA attenuates LPS-induced cardiac dysfunction via activating Sirt1/PGC-1α pathway to alleviate mitochondrial impairment. It may be a promising cardio-protective drug to be used for septic patients.
Collapse
Affiliation(s)
- Ke Peng
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Fengyuan Yang
- Department of Nephrology, General Hospital of Western Theater Command, Chengdu, China
| | - Chenming Qiu
- Department of Burn and Plastic Surgery, General Hospital of Western Theater Command, Chengdu, China
| | - Yongjian Yang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China.,Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| | - Cong Lan
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China.,Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
156
|
Osa S, Enoki Y, Miyajima T, Akiyama M, Fujiwara Y, Taguchi K, Kim YG, Matsumoto K. SCIATIC DENERVATION-INDUCED SKELETAL MUSCLE ATROPHY IS ASSOCIATED WITH PERSISTENT INFLAMMATION AND INCREASED MORTALITY DURING SEPSIS. Shock 2023; 59:417-425. [PMID: 36427072 DOI: 10.1097/shk.0000000000002053] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT Background: Patients with underlying skeletal muscle atrophy are likely to develop aggravated sepsis. However, no study has experimentally verified the association between the prognosis of sepsis and muscle atrophy, and the mechanism of aggravation of sepsis under muscle atrophy remains unclear. In this study, we investigated the effect of skeletal muscle atrophy induced by sciatic denervation (DN), an experimental muscle atrophy model, on sepsis prognosis. Methods: Skeletal muscle atrophy was induced by DN of the sciatic nerve in C57BL/6J male mice. Cecal ligation and puncture (CLP) was performed to induce sepsis. Results: The survival rates of the sham and DN groups 7 days after CLP were 63% and 35%, respectively, wherein an approximately 30% reduction was observed in the DN group ( P < 0.05, vs. sham-CLP). The DN group had a higher bacterial count in the blood 48 h after CLP ( P < 0.05, vs. sham-CLP). Notably, NOx (a metabolite of nitric oxide) concentrations in DN mice were higher than those in sham mice after CLP ( P < 0.05, vs. sham-CLP), whereas serum platelet levels were lower 48 h after CLP ( P < 0.05, vs. sham-CLP). In organ damage analysis, DN mice presented increased protein expression of the kidney injury molecule (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL), a kidney injury marker, after CLP (NGAL 48 h after CLP, P < 0.05, vs. sham-CLP; KIM-1 24 h after CLP, P < 0.01, vs. sham-CLP). Furthermore, nitro tyrosine levels in the kidneys of DN mice were higher 48 h after CLP compared with those in sham-CLP mice, indicating the accumulation of nitrative stress ( P < 0.05, vs. sham-CLP). Serum cytokine levels were increased in both groups after CLP, but decreased in the sham group 48 h after CLP and remained consistently higher in the DN group (tumor necrosis factor [TNF]-α: P < 0.05, sham-CLP vs. DN-CLP; interleukin (IL)-1β: P < 0.01, sham-CLP vs. DN-CLP; IL-6: P < 0.05, DN vs. DN-CLP; IL-10: P < 0.05, sham-CLP vs. DN-CLP). Conclusions: We verified that skeletal muscle atrophy induced by DN is associated with poor prognosis after CLP-induced sepsis. Importantly, mice with skeletal muscle atrophy presented worsening sepsis prognosis at late onset, including prolonged infection, persistent inflammation, and kidney damage accumulation, resulting in delayed recovery.
Collapse
Affiliation(s)
- Sumika Osa
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Shibakoen, Minato-ku, Tokyo, Japan
| | - Yuki Enoki
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Shibakoen, Minato-ku, Tokyo, Japan
| | - Taichi Miyajima
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Shibakoen, Minato-ku, Tokyo, Japan
| | - Masahiro Akiyama
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Honjo, Kumamoto Chuo-ku, Kumamoto, Japan
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Shibakoen, Minato-ku, Tokyo, Japan
| | - Yun-Gi Kim
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Shibakoen, Minato-ku, Tokyo, Japan
| |
Collapse
|
157
|
Zhang L, Meng W, Chen X, Wu L, Chen M, Zhou Z, Chen Y, Yuan L, Chen M, Chen J, Shui P. Multifunctional Nanoplatform for Mild Microwave-Enhanced Thermal, Antioxidative, and Chemotherapeutic Treatment of Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10341-10355. [PMID: 36790223 DOI: 10.1021/acsami.2c19198] [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: 06/18/2023]
Abstract
Rheumatoid arthritis (RA) is usually associated with excessive proliferation of M1-type proinflammatory macrophages, resulting in severe hypoxia and excess reactive oxygen species (ROS) in the joint cavity. Inhibiting M1-type proinflammatory macrophages and/or repolarizing them into M2 phenotype anti-inflammatory cells by alleviating hypoxia and scavenging ROS could be a promising strategy for RA treatment. In this work, a microwave-sensitive metal-organic framework of UiO-66-NH2 is constructed for coating a nanoenzyme of cerium oxide (CeO2) and loading with the drug celastrol (Cel) to give UiO-66-NH2/CeO2/Cel, which is ultimately wrapped with hyaluronic acid (HA) to form a nanocomposite UiO-66-NH2/CeO2/Cel@HA (UCCH). With the microwave-susceptible properties of UiO-66-NH2, the thermal effect of microwaves can eliminate the excessive proliferation of inflammatory cells. In addition, superoxide-like and catalase-like activities originating from CeO2 in UCCH are boosted to scavenge ROS and accelerate the decomposition of H2O2 to produce O2 under microwave irradiation. The nonthermal effect of microwaves could synergistically promote the repolarization of M1-type macrophages into the M2 phenotype. Accompanied by the release of the anti-RA chemotherapeutic drug Cel, UCCH can efficiently ameliorate RA in vitro and in vivo through microwave-enhanced multisynergistic effects. This strategy could inspire the design of other multisynergistic platforms enhanced by microwaves to exploit new treatment modalities in RA therapies.
Collapse
Affiliation(s)
- Lianying Zhang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wei Meng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaotong Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Libo Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Mingwa Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhaoxi Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongjian Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lixia Yuan
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ming Chen
- The People's Hospital of Gaozhou, Maoming 525200, China
| | - Jinxiang Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
158
|
Wu X, Guo LZ, Liu YH, Liu YC, Yang PL, Leung YS, Tai HC, Wang TD, Lin JCW, Lai CL, Chuang YH, Lin CH, Chou PT, Lai IR, Liu TM. Plasma riboflavin fluorescence as a diagnostic marker of mesenteric ischemia-reperfusion injury in rats. Thromb Res 2023; 223:146-154. [PMID: 36753876 DOI: 10.1016/j.thromres.2023.01.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Due to the delayed and vague symptoms, it is difficult to early diagnose mesenteric ischemia injuries in the dynamics of acute illness, leading to a 60-80 % mortality rate. Here, we found plasma fluorescence spectra can rapidly assess the severity of mesenteric ischemia injury in animal models. Ischemia-reperfusion damage of the intestine leads to multiple times increase in NADH, flavins, and porphyrin auto-fluorescence of blood. The fluorescence intensity ratio between blue-fluorophores and flavins can reflect the occurrence of shock. Using liquid chromatography and mass spectroscopy, we confirm that riboflavin is primarily responsible for the increased flavin fluorescence. Since humans absorb riboflavin from the intestine, its increase in plasma may indicate intestinal mucosa injury. Our work suggests a self-calibrated and reagent-free approach to identifying the emergence of fatal mesenteric ischemia in emergency departments or intensive care units.
Collapse
Affiliation(s)
- Xueqin Wu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Lun-Zhang Guo
- Department of Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Yi-Hung Liu
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Yu-Cheng Liu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Po-Lun Yang
- Department of Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Yun-Shiuan Leung
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Hwan-Ching Tai
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China.
| | - Tzung-Dau Wang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei 10002, Taiwan
| | - Jesse Chih-Wei Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chao-Lun Lai
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.
| | - Yueh-Hsun Chuang
- Department of Anesthesiology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Chih-Hsueh Lin
- Department of Nutrition, College of Medical and Health Care, Hungkuang University, Taichung City 433304, Taiwan
| | - Pi-Tai Chou
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - I-Rue Lai
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan; Department of Surgery, National Taiwan University Hospital, Taipei 100229, Taiwan.
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.
| |
Collapse
|
159
|
Jiang Y, Li Y, Zhang Y, Hu D, Zhang S, Wang C, Huang S, Zhang A, Jia Z, You R. NSC228155 alleviates septic cardiomyopathy via protecting mitochondria and inhibiting inflammation. Int Immunopharmacol 2023; 116:109847. [PMID: 36774857 DOI: 10.1016/j.intimp.2023.109847] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023]
Abstract
Septic cardiomyopathy is a lethal symptom of sepsis. Discovery of effective therapy that prevents cardiac injury in sepsis is critical in the clinical management of sepsis. NSC228155 is a novel compound with therapeutic potential on acute kidney injury by preventing apoptosis and protecting mitochondria. Whether NSC228155 protects against septic cardiomyopathy is unclear. In the present study, adult C57BL/6J mice were i.p injected with 5 mg/kg/day NSC228155 for 2 days before 10 mg/kg lipopolysaccharide (LPS) injection. Cardiac functional testing and sampling for serum and tissue were performed 12 and 24 h post LPS injection, respectively. NSC228155 significantly improved cardiac function examined by echocardiography, decreased the serum lactate dehydrogenase (LDH) and creatine kinase-MB, and pathologically alleviated cardiac injury in LPS mice. Accordingly, NSC228155 attenuated cardiomyocytes' mitochondrial damage as shown by decreased damaged mitochondrial ratio and activated signals for mitochondrial biogenesis, dynamics and mitophagy in LPS mice model. Metabolomics analysis demonstrated that NSC228155 corrected the metabolic disturbance involved in oxidative stress and energy metabolism, and decreased tissue injury metabolites in LPS-stimulated cardiac tissue. In the LPS-stimulated cardiac cell culture derived from human induced pluripotent stem cells, NSC228155 effectively restored the beating frequency, decreased LDH release, and protected mitochondria. NSC228155 also inhibited inflammation shown by decreased pro-inflammatory mediators in both serum and cardiac tissue in LPS model. Taken together, NSC228155 significantly improved cardiac function by directly preventing against cardiac cell injury and inhibiting inflammation in LPS model, hence may be a potential novel therapy against septic cardiomyopathy.
Collapse
Affiliation(s)
- Yuteng Jiang
- School of Medicine, Southeast University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yanwei Li
- School of Medicine, Southeast University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yiyuan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Dandan Hu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Shengnan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Chunli Wang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- School of Medicine, Southeast University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| | - Ran You
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
160
|
Miao H, Cui Z, Guo Z, Chen Q, Su W, Sun Y, Sun M, Ma X, Ding R. IDENTIFICATION OF SUBPHENOTYPES OF SEPSIS-ASSOCIATED LIVER DYSFUNCTION USING CLUSTER ANALYSIS. Shock 2023; 59:368-374. [PMID: 36562264 DOI: 10.1097/shk.0000000000002068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ABSTRACT Objectives: We attempted to identify and validate the subphenotypes of sepsis-associated liver dysfunction (SALD) using routine clinical information. Design: This article is a retrospective observational cohort study. Setting: We used the Medical Information Mart for Intensive Care IV database and the eICU Collaborative Research Database. Patients: We included adult patients (age ≥18 years) who developed SALD within the first 48 hours of intensive care unit (ICU) admission. We excluded patients who died or were discharged from the ICU within the first 48 hours of admission. Patients with abnormal liver function before ICU admission were also excluded. Measurements and Main Results: Patients in the MIMIC-IV 1.0 database served as a derivation cohort. Patients in the eICU database were used as validation cohort. We identified four subphenotypes of SALD (subphenotype α, β, γ, δ) using K-means cluster analysis in 5234 patients in derivation cohort. The baseline characteristics and clinical outcomes were compared between the phenotypes using one-way analysis of variance/Kruskal-Wallis test and the χ 2 test. Moreover, we used line charts to illustrate the trend of liver function parameters over 14 days after ICU admission. Subphenotype α (n = 1,055) was the most severe cluster, characterized by shock with multiple organ dysfunction (MODS) group. Subphenotype β (n = 1,179) had the highest median bilirubin level and the highest proportion of patients with underlying liver disease and coexisting coagulopathy (increased bilirubin group). Subphenotype γ (n = 1,661) was the cluster with the highest mean age and had the highest proportion of patients with chronic kidney disease (aged group). Subphenotype δ (n = 1,683) had the lowest 28-day and in-hospital mortality (mild group). The characteristics of clusters in the validation cohort were similar to those in the derivation cohort. In addition, we were surprised to find that GGT levels in subphenotype δ were significantly higher than in other subphenotypes, showing a different pattern from bilirubin. Conclusions: We identified four subphenotypes of SALD that presented with different clinical features and outcomes. These results can provide a valuable reference for understanding the clinical characteristics and associated outcomes to improve the management of patients with SALD in the ICU.
Collapse
Affiliation(s)
- He Miao
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhigang Cui
- School of Nursing, China Medical University, Shenyang, Liaoning Province, China
| | - Zhaotian Guo
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Qianhui Chen
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wantin Su
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yongqiang Sun
- Neusoft Corporation, Shenyang, Liaoning Province, China
| | - Mu Sun
- Neusoft Corporation, Shenyang, Liaoning Province, China
| | - Xiaochun Ma
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Renyu Ding
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
161
|
Yang T, Zhao S, Sun N, Zhao Y, Wang H, Zhang Y, Hou X, Tang Y, Gao X, Fan H. Network pharmacology and in vivo studies reveal the pharmacological effects and molecular mechanisms of Celastrol against acute hepatic injury induced by LPS. Int Immunopharmacol 2023; 117:109898. [PMID: 36827925 DOI: 10.1016/j.intimp.2023.109898] [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: 10/21/2022] [Revised: 01/28/2023] [Accepted: 02/12/2023] [Indexed: 02/24/2023]
Abstract
Sepsis is currently the main factor of death in the ICU, and the liver, as an important organ of immunity and stable metabolism, can be acutely damaged during sepsis, and the mortality rate of patients with sepsis complicated by acute liver injury is greatly increased. Celastrol (CEL) is derived from the root bark of Tripterygium wilfordii Hook.f.. As a traditional Chinese medicine, CEL has anti-inflammatory, anti-cancer, anti-oxidant, and other biological activities. Obtain CEL and AHI intersection targets via database and construct protein-protein interaction (PPI) network by STRING. GO functional enrichment and KEGG pathway analyses were performed by R studio. Targets were finally selected to perform molecular docking simulations with CEL. In vivo experiments based on the model of AHI were established by intraperitoneal injection of Lipopolysaccharide (LPS) 4 h, and pre-treated with CEL (0.5 mg/kg, 1 mg/kg, 1.5 mg/kg). The results are as follows: 273 genes with the intersection of CEL and AHI were obtained, and GO and KEGG enrichment analysis were used to design the mechanism of inflammation, apoptosis, and oxidative stress-related injury. By constructing the PPI network selected top 10 targets are: STAT3, RELA, MAPK1, MAPK3, TP53, AKT1, HSP90AA1, JUN, TNF, MAPK14, predicted CEL protection AHI design related pathways of MAPK and PI3K/AKT-related signal pathways. In vivo experiments, CEL inhibited the activation of MAPK and PI3K/AKT related pathways, reduced inflammatory response, apoptosis, and oxidative stress, and significantly improved LPS-induced AHI. In summary, this study predicted the mechanisms involved in the protective effect of CEL on AHI through network pharmacology. In vivo, CEL inhibited MAPK and PI3K/AKT-related signaling pathways, and reduced inflammatory response, apoptosis, and oxidative stress to protect LPS-induced AHI.
Collapse
Affiliation(s)
- Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Shuping Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Ning Sun
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Hui Wang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Yuntong Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Xiaoyu Hou
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Yulin Tang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Xiang Gao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China.
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China.
| |
Collapse
|
162
|
Lu Y, Shi Y, Wu Q, Sun X, Zhang WZ, Xu XL, Chen W. An Overview of Drug Delivery Nanosystems for Sepsis-Related Liver Injury Treatment. Int J Nanomedicine 2023; 18:765-779. [PMID: 36820059 PMCID: PMC9938667 DOI: 10.2147/ijn.s394802] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/29/2023] [Indexed: 02/16/2023] Open
Abstract
Sepsis, which is a systemic inflammatory response syndrome caused by infection, has high morbidity and mortality. Sepsis-related liver injury is one of the manifestations of sepsis-induced multiple organ syndrome. To date, an increasing number of studies have shown that the hepatic inflammatory response, oxidative stress, microcirculation coagulation dysfunction, and bacterial translocation play extremely vital roles in the occurrence and development of sepsis-related liver injury. In the clinic, sepsis-related liver injury is mainly treated by routine empirical methods on the basis of the primary disease. However, these therapies have some shortcomings, such as serious side effects, short duration of drug effects and lack of specificity. The emergence of drug delivery nanosystems can significantly improve drug bioavailability and reduce toxic side effects. In this paper, we reviewed drug delivery nanosystems designed for the treatment of sepsis-related liver injury according to their mechanisms (hepatic inflammation response, oxidative stress, coagulation dysfunction in the microcirculation, and bacterial translocation). Although much promising progress has been achieved, translation into clinical practice is still difficult. To this end, we also discussed the key issues currently facing this field, including immune system rejection and single treatment modalities. Finally, with the rigorous optimization of nanotechnology and the deepening of research, drug delivery nanosystems have great potential for the treatment of sepsis-related liver injury.
Collapse
Affiliation(s)
- Yi Lu
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yi Shi
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Qian Wu
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xin Sun
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Wei-Zhen Zhang
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China,Xiao-Ling Xu, Shulan International Medical College, Zhejiang Shuren University, 8 Shuren Street, Hangzhou, 310015, People’s Republic of China, Email
| | - Wei Chen
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China,Correspondence: Wei Chen, ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 725 South WanPing Road, Shanghai, 200032, People’s Republic of China, Tel +86-21-64385700-3522, Email
| |
Collapse
|
163
|
Ronco C, Chawla L, Husain-Syed F, Kellum JA. Rationale for sequential extracorporeal therapy (SET) in sepsis. Crit Care 2023; 27:50. [PMID: 36750878 PMCID: PMC9904264 DOI: 10.1186/s13054-023-04310-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/07/2023] [Indexed: 02/09/2023] Open
Abstract
Sepsis and septic shock remain drivers for morbidity and mortality in critical illness. The clinical picture of patients presenting with these syndromes evolves rapidly and may be characterised by: (a) microbial host invasion, (b) establishment of an infection focus, (c) opsonisation of bacterial products (e.g. lipopolysaccharide), (d) recognition of pathogens resulting in an immune response, (e) cellular and humoral effects of circulating pathogen and pathogen products, (f) immunodysregulation and endocrine effects of cytokines, (g) endothelial and organ damage, and (h) organ crosstalk and multiple organ dysfunction. Each step may be a potential target for a specific therapeutic approach. At various stages, extracorporeal therapies may target circulating molecules for removal. In sequence, we could consider: (a) pathogen removal from the circulation with affinity binders and cartridges (specific), (b) circulating endotoxin removal by haemoperfusion with polymyxin B adsorbers (specific), (c) cytokine removal by haemoperfusion with sorbent cartridges or adsorbing membranes (non-specific), (d) extracorporeal organ support with different techniques for respiratory and cardiac support (CO2 removal or extracorporeal membrane oxygenation), and renal support (haemofiltration, haemodialysis, or ultrafiltration). The sequence of events and the use of different techniques at different points for specific targets will likely require trials with endpoints other than mortality. Instead, the primary objectives should be to achieve the desired action by using extracorporeal therapy at a specific point.
Collapse
Affiliation(s)
- Claudio Ronco
- International Renal Research Institute of Vicenza, IRRIV Foundation, Department of Nephrology, Dialysis and Transplantation, St. Bortolo Hospital, aULSS8 Berica, Via Rodolfi, 37, 36100, Vicenza, Italy.
- Department of Medicine (DIMED), University of Padua, Via Giustiniani, 2, 35128, Padua, Italy.
| | - Lakhmir Chawla
- Department of Medicine, Veterans Affairs Medical Center, 3350 La Jolla Village Dr, San Diego, CA, 92161, USA
| | - Faeq Husain-Syed
- Department of Internal Medicine II, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Klinikstrasse 33, 35392 Giessen, Germany
- Division of Nephrology, University of Virginia School of Medicine, 1300 Jefferson Park Avenue, Charlottesville, VA, 22908, USA
| | - John A Kellum
- Center for Critical Care Nephrology, CRISMA, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
- Spectral Medical, 135 The West Mall, Unit 2, Toronto, M9C 1C2, Canada
| |
Collapse
|
164
|
Chen X, Zhu Y, Wei Y, Fan S, Xia L, Chen Q, Lu Y, Wu D, Liu X, Peng X. Glutamine alleviates intestinal injury in a murine burn sepsis model by maintaining intestinal intraepithelial lymphocyte homeostasis. Eur J Pharmacol 2023; 940:175480. [PMID: 36566008 DOI: 10.1016/j.ejphar.2022.175480] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Intestinal intraepithelial lymphocytes (IELs) play a sentinel role in the mucosal immune system because of their unique anatomical location in the epithelial layer. The disruption of IEL homeostasis is implicated in driving the intestinal injury of many typical inflammatory disorders, such as inflammatory bowel disease (IBD) and sepsis. Therefore, it is meaningful to alleviate intestinal injury by restoring IEL homeostasis in disease conditions. This study explores the effects of glutamine on intestinal IEL homeostasis in a murine model of burn sepsis. We report that glutamine inhibits inflammatory response and reduces injury in the small intestine of burn septic mice. This effect is attributed to the maintaining of IEL homeostasis by suppressing apoptosis and restoring the disrupted subpopulation balance induced by burn sepsis. Mechanistically, we show that glutamine does not affect the IL-15 dependent mechanisms that drive the maintenance and differentiation of IELs. Instead, glutamine sustains IEL homeostasis by upregulate aryl hydrocarbon receptor (AHR) and interleukin (IL)-22 transcription and expression. Consistently, the protective roles of glutamine in burn septic mice were repressed by further supplement with an AHR antagonist CH-223191. Collectively, our study reveals a new role of glutamine to maintain IEL homeostasis by activating the AHR signaling pathway, which in turn ameliorates intestinal injury in burn sepsis.
Collapse
Affiliation(s)
- Xiaoli Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yuanfeng Zhu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Lin Xia
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qian Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Dan Wu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
165
|
Suzuki Y, Kami D, Taya T, Sano A, Ogata T, Matoba S, Gojo S. ZLN005 improves the survival of polymicrobial sepsis by increasing the bacterial killing via inducing lysosomal acidification and biogenesis in phagocytes. Front Immunol 2023; 14:1089905. [PMID: 36820088 PMCID: PMC9938763 DOI: 10.3389/fimmu.2023.1089905] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/23/2023] [Indexed: 02/07/2023] Open
Abstract
Polymicrobial sepsis still has a high mortality rate despite the development of antimicrobial agents, elaborate strategies to protect major organs, and the investment of numerous medical resources. Mitochondrial dysfunction, which acts as the center of energy metabolism, is clearly the basis of pathogenesis. Drugs that act on PGC1α, the master regulator of mitochondrial biosynthesis, have shown useful effects in the treatment of sepsis; therefore, we investigated the efficacy of ZLN005, a PGC1α agonist, and found significant improvement in overall survival in an animal model. The mode of action of this effect was examined, and it was shown that the respiratory capacity of mitochondria was enhanced immediately after administration and that the function of TFEB, a transcriptional regulator that promotes lysosome biosynthesis and mutually enhances PGC1α, was enhanced, as was the physical contact between mitochondria and lysosomes. ZLN005 strongly supported immune defense in early sepsis by increasing lysosome volume and acidity and enhancing cargo degradation, resulting in a significant reduction in bacterial load. ZLN005 rapidly acted on two organelles, mitochondria and lysosomes, against sepsis and interactively linked the two to improve the pathogenesis. This is the first demonstration that acidification of lysosomes by a small molecule is a mechanism of action in the therapeutic strategy for sepsis, which will have a significant impact on future drug discovery.
Collapse
Affiliation(s)
- Yosuke Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihiko Taya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Arata Sano
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Pathology and Cell Regulation, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
166
|
Takahama M, Patil A, Johnson K, Cipurko D, Miki Y, Taketomi Y, Carbonetto P, Plaster M, Richey G, Pandey S, Cheronis K, Ueda T, Gruenbaum A, Dudek SM, Stephens M, Murakami M, Chevrier N. Organism-Wide Analysis of Sepsis Reveals Mechanisms of Systemic Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526342. [PMID: 36778287 PMCID: PMC9915512 DOI: 10.1101/2023.01.30.526342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sepsis is a systemic response to infection with life-threatening consequences. Our understanding of the impact of sepsis across organs of the body is rudimentary. Here, using mouse models of sepsis, we generate a dynamic, organism-wide map of the pathogenesis of the disease, revealing the spatiotemporal patterns of the effects of sepsis across tissues. These data revealed two interorgan mechanisms key in sepsis. First, we discover a simplifying principle in the systemic behavior of the cytokine network during sepsis, whereby a hierarchical cytokine circuit arising from the pairwise effects of TNF plus IL-18, IFN-γ, or IL-1β explains half of all the cellular effects of sepsis on 195 cell types across 9 organs. Second, we find that the secreted phospholipase PLA2G5 mediates hemolysis in blood, contributing to organ failure during sepsis. These results provide fundamental insights to help build a unifying mechanistic framework for the pathophysiological effects of sepsis on the body.
Collapse
|
167
|
PCSK9 Promotes Endothelial Dysfunction During Sepsis Via the TLR4/MyD88/NF-κB and NLRP3 Pathways. Inflammation 2023; 46:115-128. [PMID: 35930089 DOI: 10.1007/s10753-022-01715-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 11/05/2022]
Abstract
Endothelial dysfunction often accompanies sepsis. We aimed to explore the role of PCSK9 in septic endothelial dysfunction. Sepsis was induced by lipopolysaccharide (LPS) treatment of human umbilical vein endothelial cells (HUVECs) in vitro and cecal ligation and puncture (CLP) surgery in mice in vivo. Evolocumab (EVC) and Pep 2-8, PCSK9 inhibitors, were subsequently used to determine the role of PCSK9 in sepsis-induced endothelial dysfunction in vitro and in vivo, respectively. In addition, the TLR4 agonist, Kdo2-Lipid A ammonium (KLA), was used to determine the related mechanism. Protein expression of eNOS, VE-cadherin, PCSK9, TLR4, MyD88, p-p65, p65, NLRP3, ASC, and caspase-1 p20 in mice aortas and HUVECs was measured by western blotting, while mRNA expression of TNFα, IL-1β, and IL-18 was determined by qRT-PCR. The level of inflammatory cytokines of mouse aortas was visualized by immunohistochemistry. Vasodilation of the aorta was detected by vascular reactivity experiments. The 96-h survival rate after CLP was assessed. Our findings showed that the expression of eNOS and VE-cadherin decreased, and PCSK9 expression increased, in septic HUVECs or mice. Inhibition of PCSK9 increased eNOS and VE-cadherin expression. Activation of the TLR4/MyD88/NF-κB and NLRP3 pathways may be responsible for PCSK9-induced endothelial dysfunction in sepsis. Vascular reactivity tests and survival studies showed that inhibition of PCSK9 could prevent the decrease in endothelium-dependent vasodilation function and improve the survival rates of septic mice. In summary, our results suggested that increased PCSK9 expression during sepsis activated the TLR4/MyD88/NF-κB and NLRP3 pathways to induce inflammation, which resulted in vascular endothelial dysfunction and decreased survival rates. Thus, inhibition of PCSK9 may be a potential clinical therapeutic target to improve vascular endothelial function in sepsis.
Collapse
|
168
|
Xu M, Shao Y, Lin K, Liu Y, Lin Y, Lin Y, Yang R, Liu L, Yin M, Liao S, Jiang S, He J. Genetic Arg-304-His substitution in GRK5 protects against sepsis progression by alleviating NF-κB-mediated inflammation. Int Immunopharmacol 2023; 115:109629. [PMID: 36584571 DOI: 10.1016/j.intimp.2022.109629] [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: 10/19/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Previous studies have demonstrated that G protein-coupled receptor kinase 5 (GRK5) exerts a pivotal regulatory effect on the inflammation associated with sepsis. The present study aimed to investigate the clinical association of GRK5 genetic variants with sepsis and to further explore the underlying genetic mechanisms involved in regulating sepsis-induced inflammatory responses and the pathogenesis of sepsis. METHODS This case-control study enrolled 1081 septic patients and 1147 matched controls for genotyping of GRK5 rs2230349 and rs2230345 polymorphisms. The effect of these genetic variants on GRK5-mediated inflammatory responses was analyzed in peripheral blood mononuclear cells (PBMCs) and THP-1 macrophages. A clinically relevant polymicrobial sepsis model was established by subjecting wild-type (WT) and GRK5-knockout mice to cecal ligation and puncture (CLP) to evaluate the role of GRK5 in sepsis. RESULTS We identified significant differences in the genotype/allele distribution of rs2230349 G > A, but not rs2230345, between the sepsis subtype and septic shock subgroups (GA + AA vs. GG genotype, OR = 0.698, 95% CI = 0.547-0.893, P = 0.004; A vs. G allele, OR = 0.753, 95% CI = 0.620-0.919, P = 0.005) and between the survivor and nonsurvivor subgroups (GA + AA vs. GG genotype, OR = 0.702, 95% CI = 0.531-0.929, P = 0.015; A vs. G allele, OR = 0.753, 95% CI = 0.298-0.949, P = 0.017). PBMCs carrying the sepsis-associated protective A allele produced significantly lower levels of TNF-α and IL-1β upon LPS stimulation. The results from the in vitro experiment showed that the Arg-304-His substitution caused by the rs2230349 G-to-A mutation in GRK5 significantly decreased the LPS-induced production of several proinflammatory cytokines, such as TNF-α, IL-6, IL-1β and MCP-1, via the IκB-α/NF-κB signaling pathway in THP-1 macrophages. Furthermore, GRK5-knockout mice exhibited a significant decrease in IκB-α phosphorylation/degradation, the p-p65/p65 ratio, the p-p50/p50 ratio, p65 nuclear translocation and downstream cytokine (TNF-α, IL-6, IL-1β and VCAM-1) production compared to WT mice after CLP surgery. A significant improvement in 7-day survival rate in GRK5-KO septic mice was observed in the presence of antibiotics. CONCLUSIONS The Arg-304-His substitution caused by the rs2230349 G-to-A mutation in GRK5 might disrupt GRK5 function and alleviate IKB-α/NF-κB-mediated inflammatory responses, which ultimately conferred a genetic protective effect against susceptibility to sepsis progression and mortality. These results may, to some extent, explain the heterogeneity of the clinical prognoses of septic patients and provide novel opportunities for individualized approaches for sepsis treatment.
Collapse
Affiliation(s)
- Mingwei Xu
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, Guangdong, PR China.
| | - Yiming Shao
- The Intensive Care Unit, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, PR China; The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China.
| | - Kaisheng Lin
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, Guangdong, PR China.
| | - Yuchun Liu
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, Guangdong, PR China.
| | - Yao Lin
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, Guangdong, PR China.
| | - Yingying Lin
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, Guangdong, PR China.
| | - Ruoxuan Yang
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, Guangdong, PR China.
| | - Lizhen Liu
- The Clinical Medical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China.
| | - Mingkang Yin
- The Clinical Medical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China.
| | - Shuanglin Liao
- The Intensive Care Unit, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, PR China.
| | - Shaoru Jiang
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, Guangdong, PR China.
| | - Junbing He
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, Guangdong, PR China.
| |
Collapse
|
169
|
Impacts of Curcumin Treatment on Experimental Sepsis: A Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2252213. [PMID: 36756300 PMCID: PMC9902115 DOI: 10.1155/2023/2252213] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 02/03/2023]
Abstract
Background and Aims Sepsis is defined as a life-threatening organ dysfunction due to a dysregulated host immune response to an infection. Curcumin is a yellow polyphenol derived from the rhizome of Curcuma longa with anti-inflammatory and antioxidant properties scientifically proven, a condition that allowed its use as a tool in the treatment of sepsis. Thus, the purpose of this article was to systematically review the evidence on the impact of curcumin's anti-inflammatory effect on experimental sepsis. Methods For this, the PubMed, MEDLINE, EMBASE, Scopus, Web of Science, and LILACS databases were used, and the research was not limited to a specific publication period. Only original articles in English using in vivo experimental models (rats or mice) of sepsis induction performed by administration of lipopolysaccharide (LPS) or cecal ligation and perforation surgery (CLP) were included in the study. Studies using curcumin in dry extract or with a high degree of purity were included. At initial screening, 546 articles were selected, and of these, 223 were eligible for primary evaluation. Finally, 12 articles with full text met all inclusion criteria. Our results showed that curcumin may inhibit sepsis-induced complications such as brain, heart, liver, lungs, and kidney damage. Curcumin can inhibit inflammatory factors, prevent oxidative stress, and regulate immune responses in sepsis. Additionally, curcumin increased significantly the survival rates after experimental sepsis in several studies. The modulation of the immune response and mortality by curcumin reinforces its protective effect on sepsis and indicates a potential therapeutic tool for the treatment of sepsis.
Collapse
|
170
|
Luo M, He Q. Development of a prognostic nomogram for sepsis associated-acute respiratory failure patients on 30-day mortality in intensive care units: a retrospective cohort study. BMC Pulm Med 2023; 23:43. [PMID: 36717800 PMCID: PMC9885567 DOI: 10.1186/s12890-022-02302-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/23/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Acute respiratory failure is a type of sepsis complicated by severe organ failure. We have developed a new nomogram for predicting the 30-day risk of death in patients through a retrospective study. METHOD Data was collected and extracted from MIMICIV, with 768 eligible cases randomly assigned to the primary cohort (540) and the validation cohort (228). The final six factors were included by Cox regression analysis to create the Nomogram, the accuracy of the Nomogram was assessed using the C-index and calibration curve, and finally, the clinical usefulness of the Nomogram was evaluated using DCA in. RESULTS Multivariate Cox regression analysis showed that age, DBP, lactate, PaO2, platelet, mechanical ventilation were independent factors for 30-day mortality of SA-ARF. The nomogram established based on the six factors. The C-index of nomogram in the primary cohort is 0.731 (95% CI 0.657-0.724) and 0.722 (95%CI 0.622-0.759) in the validation cohort. Besides, the decision curve analysis (DCA) confirmed the clinical usefulness of the nomogram. CONCLUSION The study developed and validated a risk prediction model for SA-ARF patients that can help clinicians reasonably determine disease risk and further confirm its clinical utility using internal validation.
Collapse
Affiliation(s)
- Mengdi Luo
- grid.263901.f0000 0004 1791 7667Southwest Jiaotong University of Medicine/Southwest Jiaotong University Affiliated Chengdu Third People’s Hospital, Chengdu, 610031 Sichuan China
| | - Qing He
- grid.263901.f0000 0004 1791 7667Southwest Jiaotong University of Medicine/Southwest Jiaotong University Affiliated Chengdu Third People’s Hospital, Chengdu, 610031 Sichuan China
| |
Collapse
|
171
|
Han YC, Shen ZJ, Wang YN, Xiang RL, Xie HZ. LncRNA-mRNA expression profile and functional network of vascular dysfunction in septic rats. Eur J Med Res 2023; 28:11. [PMID: 36611198 PMCID: PMC9824925 DOI: 10.1186/s40001-022-00961-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND We used microarrays to analyse the changes in long non-coding RNAs (lncRNAs) and mRNAs in aorta tissue in model rats with lipopolysaccharide-induced sepsis and determined the lncRNA-mRNA and lncRNA-miRNA-mRNA functional networks. METHODS Wistar rats were intraperitoneally injected with lipopolysaccharide, and the lncRNA and mRNA expression profiles in the aorta were evaluated using microarrays. The functions of the differentially expressed mRNAs were analysed using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses. We then constructed coding/non-coding co-expression and competing endogenous RNA networks to study the mechanisms related to sepsis in rats. RESULTS We identified 503 differentially expressed lncRNAs and 2479 differentially expressed mRNAs in the model rats with lipopolysaccharide-induced sepsis. Mitochondrial fission process 1 (MTFP1) was the most significantly down-regulated mRNA. Bioinformatics analysis showed that the significantly down-regulated mRNAs in the sepsis models were in pathways related to mitochondrial structure, function, and energy metabolism. Coding/non-coding co-expression and competing endogenous RNA analyses were conducted using 12 validated lncRNAs in combination with all mRNAs. The coding/non-coding co-expression analysis showed that the 12 validated lncRNAs were mainly regulatory factors for abnormal energy metabolism, including mitochondrial structure damage and aberrant mitochondrial dynamics. The competing endogenous RNA analysis revealed that the potential functions of these 12 lncRNAs might be related to the inflammatory response. CONCLUSION We determined the differentially expressed lncRNAs and mRNAs in the aorta of septic rats using microarrays. Further studies on these lncRNAs will help elucidate the mechanism of sepsis at the genetic level and may identify potential therapeutic targets.
Collapse
Affiliation(s)
- Ye-Chen Han
- grid.413106.10000 0000 9889 6335Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730 China
| | - Zhu-Jun Shen
- grid.413106.10000 0000 9889 6335Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730 China
| | - Yi-Ning Wang
- grid.413106.10000 0000 9889 6335Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730 China
| | - Ruo-Lan Xiang
- grid.11135.370000 0001 2256 9319Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, 100191 China
| | - Hong-Zhi Xie
- grid.413106.10000 0000 9889 6335Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730 China
| |
Collapse
|
172
|
Zhang K, Wang Y, Chen S, Mao J, Jin Y, Ye H, Zhang Y, Liu X, Gong C, Cheng X, Huang X, Hoeft A, Chen Q, Li X, Fang X. TREM2 hi resident macrophages protect the septic heart by maintaining cardiomyocyte homeostasis. Nat Metab 2023; 5:129-146. [PMID: 36635449 PMCID: PMC9886554 DOI: 10.1038/s42255-022-00715-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 11/22/2022] [Indexed: 01/14/2023]
Abstract
Sepsis-induced cardiomyopathy (SICM) is common in septic patients with a high mortality and is characterized by an abnormal immune response. Owing to cellular heterogeneity, understanding the roles of immune cell subsets in SICM has been challenging. Here we identify a unique subpopulation of cardiac-resident macrophages termed CD163+RETNLA+ (Mac1), which undergoes self-renewal during sepsis and can be targeted to prevent SICM. By combining single-cell RNA sequencing with fate mapping in a mouse model of sepsis, we demonstrate that the Mac1 subpopulation has distinct transcriptomic signatures enriched in endocytosis and displays high expression of TREM2 (TREM2hi). TREM2hi Mac1 cells actively scavenge cardiomyocyte-ejected dysfunctional mitochondria. Trem2 deficiency in macrophages impairs the self-renewal capability of the Mac1 subpopulation and consequently results in defective elimination of damaged mitochondria, excessive inflammatory response in cardiac tissue, exacerbated cardiac dysfunction and decreased survival. Notably, intrapericardial administration of TREM2hi Mac1 cells prevents SICM. Our findings suggest that the modulation of TREM2hi Mac1 cells could serve as a therapeutic strategy for SICM.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Wang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shiyu Chen
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiali Mao
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Jin
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Ye
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Zhang
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiwang Liu
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenchen Gong
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuejun Cheng
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoli Huang
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Andreas Hoeft
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Bonn, Bonn, Germany
| | - Qixing Chen
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xuekun Li
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Xiangming Fang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
173
|
Liu T, Zhang C, Ying J, Wang Y, Yan G, Zhou Y, Lu G. Inhibition of the intracellular domain of Notch1 results in vascular endothelial cell dysfunction in sepsis. Front Immunol 2023; 14:1134556. [PMID: 37205094 PMCID: PMC10185824 DOI: 10.3389/fimmu.2023.1134556] [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: 12/30/2022] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Background Notch signaling is critical for regulating the function of vascular endothelial cells (ECs). However, the effect of the intracellular domain of Notch1 (NICD) on EC injury in sepsis remains unclear. Methods We established a cell model of vascular endothelial dysfunction and induced sepsis in a mouse model via lipopolysaccharide (LPS) injection and cecal ligation and puncture (CLP). Endothelial barrier function and expression of endothelial-related proteins were determined using CCK-8, permeability, flow cytometry, immunoblot, and immunoprecipitation assays. The effect of NICD inhibition or activation on endothelial barrier function was evaluated in vitro. Melatonin was used for NICD activation in sepsis mice. The survival rate, Evans blue dye of organs, vessel relaxation assay, immunohistochemistry, ELISA, immunoblot were used to explore the specific role of melatonin for sepsis induced vascular dysfunction in vivo. Results We found that LPS, interleukin 6, and serum collected from septic children could inhibit the expression of NICD and its downstream regulator Hes1, which impaired endothelial barrier function and led to EC apoptosis through the AKT pathway. Mechanistically, LPS decreased the stability of NICD by inhibiting the expression of a deubiquitylating enzyme, ubiquitin-specific proteases 8 (USP8). Melatonin, however, upregulated USP8 expression, thus maintaining the stability of NICD and Notch signaling, which ultimately reduced EC injury in our sepsis model and elevated the survival rate of septic mice. Conclusions We found a previously uncharacterized role of Notch1 in mediating vascular permeability during sepsis, and we showed that inhibition of NICD resulted in vascular EC dysfunction in sepsis, which was reversed by melatonin. Thus, the Notch1 signaling pathway is a potential target for the treatment of sepsis.
Collapse
Affiliation(s)
- Tingyan Liu
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Caiyan Zhang
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Jiayun Ying
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Yaodong Wang
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Gangfeng Yan
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Yufeng Zhou
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
- *Correspondence: Yufeng Zhou, ; Guoping Lu,
| | - Guoping Lu
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
- *Correspondence: Yufeng Zhou, ; Guoping Lu,
| |
Collapse
|
174
|
Dou W, Qi F, Li Y, Wei F, Hu Q, Yao Z, Wang J, Zhang L, Tang Z. Charge-biased nanofibrous membranes with uniform charge distribution and hemocompatibility for enhanced selective adsorption of endotoxin from plasma. J Memb Sci 2023. [DOI: 10.1016/j.memsci.2022.121134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
175
|
Leng Y, Li Y, Wang J, Deng P, Wang W, Wu J, Wang W, Weng C. Sepsis as an independent risk factor in atrial fibrillation and cardioembolic stroke. Front Endocrinol (Lausanne) 2023; 14:1056274. [PMID: 36793274 PMCID: PMC9922695 DOI: 10.3389/fendo.2023.1056274] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/05/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Electrolyte balance is an important factor to sustain the homeostasis of human body environment and in sepsis pathogenesis. Many current cohort-based studies have already revealed that electrolyte disorder may intensify sepsis and induce stroke. However, the corresponding randomized controlled trials did not show that electrolyte disorder in sepsis has a harmful effect on stroke. OBJECTIVES The aim of this study was to examine the association of genetically sepsis-derived electrolyte disorder with stroke risk using meta-analysis and Mendelian randomization. RESULTS In four studies (182,980 patients), electrolyte disorders were compared with stroke incidence in patients with sepsis. The pooled odds ratio (OR) of stroke is 1.79 [95% confidence interval (CI): 1.23-3.06; p < 0.05], which shows a significant association between electrolyte disorder and stroke in sepsis patients. Furthermore, in order to evaluate the causal association between stroke risk and sepsis-derived electrolyte disorder, a two-sample Mendelian randomization (MR) study was conducted. The genetic variants extracted from a genome-wide association study (GWAS) of exposure data that are strongly associated with frequently used sepsis were used as instrumental variables (IVs). Based on the IVs' corresponding effect estimates, we estimated overall stroke risk, cardioembolic stroke risk, and stroke induced by large/small vessels from a GWAS meta-analysis with 10,307 cases and 19,326 controls. As a final step to verify the preliminary MR results, we performed sensitivity analysis using multiple types of Mendelian randomization analysis. CONCLUSION Our study revealed the association between electrolyte disorder and stroke in sepsis patients, and the correlation between genetic susceptibility to sepsis and increased risk of cardioembolic stroke, hinting that cardiogenic diseases and accompanying electrolyte disorder interference in due course could help sepsis patients get more benefits in stroke prevention.
Collapse
Affiliation(s)
- Yiming Leng
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yalan Li
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Peizhi Deng
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jingjing Wu
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wenjuan Wang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Wenjuan Wang, ; Chunyan Weng,
| | - Chunyan Weng
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Wenjuan Wang, ; Chunyan Weng,
| |
Collapse
|
176
|
Balance Cell Apoptosis and Pyroptosis of Caspase-3-Activating Chemotherapy for Better Antitumor Therapy. Cancers (Basel) 2022; 15:cancers15010026. [PMID: 36612023 PMCID: PMC9817729 DOI: 10.3390/cancers15010026] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy is a standard treatment modality in clinic that exerts an antitumor effect via the activation of the caspase-3 pathway, inducing cell death. While a number of chemotherapeutic drugs have been developed to combat various types of tumors, severe side effects have been their common limitation, due to the nonspecific drug biodistribution, bringing significant pain to cancer patients. Recently, scientists found that, besides apoptosis, chemotherapy could also cause cell pyroptosis, both of which have great influence on the therapeutic index. For example, cell apoptosis is, generally, regarded as the main mechanism of killing tumor cells, while cell pyroptosis in tumors promotes treatment efficacy, but in normal tissue results in toxicity. Therefore, significant research efforts have been paid to exploring the rational modulation mode of cell death induced by chemotherapy. This critical review aims to summarize recent progress in the field, focusing on how to balance cell apoptosis and pyroptosis for better tumor chemotherapy. We first reviewed the mechanisms of chemotherapy-induced cell apoptosis and pyroptosis, in which the activated caspase-3 is the key signaling molecule for regulating both types of cell deaths. Then, we systematically discussed the rationale and methods of switching apoptosis to pyroptosis for enhanced antitumor efficacy, as well as the blockage of pyroptosis to decrease side effects. To balance cell pyroptosis in tumor and normal tissues, the level of GSDME expression and tumor-targeting drug delivery are two important factors. Finally, we proposed potential future research directions, which may provide guidance for researchers in the field.
Collapse
|
177
|
Zhang R, Liu H, Dai D, Ding X, Wang D, Wang Y, Shi X, Zhang S, Duan X, Wang H, Luo Y, Liu S, Han B, Zhang X, Fang Y, Yang J, Xu W, Sun T. Adjunctive sepsis therapy with aminophylline (STAP): a randomized controlled trial. Chin Med J (Engl) 2022; 135:2843-2850. [PMID: 36728571 PMCID: PMC9944697 DOI: 10.1097/cm9.0000000000002282] [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: 07/16/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Sepsis is a serious disease caused by infection. Aminophylline has anti-asthma and anti-inflammatory effects. We aimed to explore the safety and effect of aminophylline in sepsis. METHODS We conducted a clinical randomized controlled trial involving 100 patients diagnosed with sepsis within 48 h after intensive care unit (ICU) admission in two sites. All patients were randomized in a 1:1 ratio to receive standard therapy with or without aminophylline. The primary clinical outcome was all-cause mortality at 28 days. RESULTS From September 27, 2018 to February 12, 2020, we screened 277 septic patients and eventually enrolled 100 patients, with 50 assigned to the aminophylline group and 50 to the usual-care group. At 28 days, 7 of 50 patients (14.0%) in the aminophylline group had died, compared with 16 of 50 (32.0%) in the usual-care group ( P = 0.032). Cox regression showed that the aminophylline group had a lower hazard of death (hazard ratio = 0.312, 95% confidence interval: 0.129-0.753). Compared with the usual-care group, patients in the aminophylline group had a longer survival time ( P = 0.039 by the log-rank test). The effects of aminophylline on vasopressor dose, oxygenation index, and sequential organ failure assessment score were time-dependent with treatment. There were no significant differences in total hospitalization days, ICU hospitalization days, and rates of serious adverse events (all P > 0.05). No adverse events were observed in the trial. CONCLUSIONS Aminophylline as an adjunct therapy could significantly reduce the risk of death and prolong the survival time of patients with sepsis. TRIAL REGISTRATION ChiCTR.org.cn, ChiCTR1800019173.
Collapse
Affiliation(s)
- Ruifang Zhang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Huan Liu
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Dongmei Dai
- Department of Intensive Care Unit, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xianfei Ding
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Dong Wang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Yan Wang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Xuexiu Shi
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Shuguang Zhang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Xiaoguang Duan
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Haixu Wang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Yonggang Luo
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Shaohua Liu
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Bing Han
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Xiaojuan Zhang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Yu Fang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Jing Yang
- Precision Medicine Monitoring Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wangbin Xu
- Department of Intensive Care Unit, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Tongwen Sun
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| |
Collapse
|
178
|
Fibroblast growth factor 5 overexpression ameliorated lipopolysaccharide-induced apoptosis of hepatocytes through regulation of the phosphoinositide-3-kinase/protein kinase B pathway. Chin Med J (Engl) 2022; 135:2859-2868. [PMID: 36728504 PMCID: PMC9943982 DOI: 10.1097/cm9.0000000000002540] [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: 08/19/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Sepsis is a systemic inflammatory syndrome induced by several infectious agents. Multiple organs are affected by sepsis, including the liver, which plays an important role in metabolism and immune homeostasis. Fibroblast growth factors (FGFs) participate in several biological processes, although the role of FGF5 in sepsis is unclear. METHODS In this study, lipopolysaccharide (LPS) was administrated to mice to establish a sepsis-induced liver injury. A similar in vitro study was conducted using L-02 hepatocytes. Western blot and immunohistochemistry staining were performed to evaluate the FGF5 expression level in liver tissues and cells. Inflammatory cell infiltrations, cleaved-caspase-3 expressions, reactive oxygen species and levels of inflammatory cytokines were detected by immunofluorescence, dihydroethidium staining, and reverse transcription quantitative polymerase chain reaction analysis, respectively. Flow cytometry was used to detect the apoptosis level of cells. In addition, ribonucleic acid (RNA)-sequencing was applied to explore the possible mechanism by which FGF5 exerted effects. RESULTS LPS administration caused FGF5 down-regulation in the mouse liver as well as in L-02 hepatocytes. Additionally, with FGF5 overexpression, liver injury and the level of hepatocyte apoptosis were ameliorated. Further, RNA sequencing performed in hepatocytes revealed the phosphoinositide-3-kinase/protein kinase B (PI3K/AKT) pathway as a possible pathway regulated by FGF5 . This was supported using an inhibitor of the PI3K/AKT pathway, which abrogated the protective effect of FGF5 in LPS-induced hepatocyte injury. CONCLUSION The anti-apoptotic effect of FGF5 on hepatocytes suffering from LPS has been demonstrated and was dependent on the activation of the PI3K/AKT signaling pathway.
Collapse
|
179
|
Atreya MR, Cvijanovich NZ, Fitzgerald JC, Weiss SL, Bigham MT, Jain PN, Schwarz AJ, Lutfi R, Nowak J, Allen GL, Thomas NJ, Grunwell JR, Baines T, Quasney M, Haileselassie B, Lindsell CJ, Alder MN, Wong HR. Integrated PERSEVERE and endothelial biomarker risk model predicts death and persistent MODS in pediatric septic shock: a secondary analysis of a prospective observational study. Crit Care 2022; 26:210. [PMID: 35818064 PMCID: PMC9275255 DOI: 10.1186/s13054-022-04070-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/21/2022] [Indexed: 11/12/2022] Open
Abstract
Background Multiple organ dysfunction syndrome (MODS) is a critical driver of sepsis morbidity and mortality in children. Early identification of those at risk of death and persistent organ dysfunctions is necessary to enrich patients for future trials of sepsis therapeutics. Here, we sought to integrate endothelial and PERSEVERE biomarkers to estimate the composite risk of death or organ dysfunctions on day 7 of septic shock. Methods We measured endothelial dysfunction markers from day 1 serum among those with existing PERSEVERE data. TreeNet® classification model was derived incorporating 22 clinical and biological variables to estimate risk. Based on relative variable importance, a simplified 6-biomarker model was developed thereafter. Results Among 502 patients, 49 patients died before day 7 and 124 patients had persistence of MODS on day 7 of septic shock. Area under the receiver operator characteristic curve (AUROC) for the newly derived PERSEVEREnce model to predict death or day 7 MODS was 0.93 (0.91–0.95) with a summary AUROC of 0.80 (0.76–0.84) upon tenfold cross-validation. The simplified model, based on IL-8, HSP70, ICAM-1, Angpt2/Tie2, Angpt2/Angpt1, and Thrombomodulin, performed similarly. Interaction between variables—ICAM-1 with IL-8 and Thrombomodulin with Angpt2/Angpt1—contributed to the models’ predictive capabilities. Model performance varied when estimating risk of individual organ dysfunctions with AUROCS ranging from 0.91 to 0.97 and 0.68 to 0.89 in training and test sets, respectively. Conclusions The newly derived PERSEVEREnce biomarker model reliably estimates risk of death or persistent organ dysfunctions on day 7 of septic shock. If validated, this tool can be used for prognostic enrichment in future pediatric trials of sepsis therapeutics. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13054-022-04070-5.
Collapse
|
180
|
Li J, Lu Y, Lin G. Blocking cGAS/STING signaling protects against sepsis-associated acute liver injury. Int Immunopharmacol 2022; 113:109276. [DOI: 10.1016/j.intimp.2022.109276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 11/05/2022]
|
181
|
Li Y, Sun P, Chang K, Yang M, Deng N, Chen S, Su B. Effect of Continuous Renal Replacement Therapy with the oXiris Hemofilter on Critically Ill Patients: A Narrative Review. J Clin Med 2022; 11:jcm11226719. [PMID: 36431196 PMCID: PMC9695587 DOI: 10.3390/jcm11226719] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Critically ill patients with sepsis and severe COVID-19 are commonly characterized by a dysregulated immune response and an acute kidney injury. Continuous renal replacement therapy (CRRT) is now proposed as a promising adjuvant therapy to treat these critically ill patients by removing cytokines, pathogen-associated molecular patterns, and damage-associated molecular patterns from the blood. Although multiple hemofilters, including high-cutoff membranes, the oXiris hemofilter, the CytoSorb hemoadsorption device, and the Toraymyxin hemoperfusion cartridge, have been used in current clinical practice, the use of the oXiris hemofilter in critically ill patients is of particular interest because it is the only kind of hemofilter that can provide renal replacement therapy, remove endotoxins, and adsorb cytokines simultaneously. During the past five years, a growing body of literature has shown that CRRT with the oXiris hemofilter can improve hemodynamics and organ function and can decrease cytokines and endotoxins in both septic and COVID-19 patients. Here, we performed a narrative review to describe the development history of the oXiris hemofilter and to discuss the therapeutic effect of oXiris-CRRT on critically ill patients by searching the PubMed, Web of Science, and clinicaltrials.gov databases for articles published from inception to 8 September 2022 (updated on 1 November) with an English language restriction. We also summarized the current knowledge on anticoagulation techniques and safety concerns when delivering oXiris-CRRT sessions.
Collapse
Affiliation(s)
- Yupei Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peiyan Sun
- West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Kaixi Chang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mei Yang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Nephrology, The First People’s Hospital of Shuangliu District, Chengdu 610200, China
| | - Ningyue Deng
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shanshan Chen
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Baihai Su
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
- West China School of Medicine, Sichuan University, Chengdu 610041, China
- Med-X Center for Materials, Sichuan University, Chengdu 610041, China
- Med+ Biomaterial Institute of West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: ; Tel.: +86-028-85423341
| |
Collapse
|
182
|
Cano-Gamez E, Burnham KL, Goh C, Allcock A, Malick ZH, Overend L, Kwok A, Smith DA, Peters-Sengers H, Antcliffe D. An immune dysfunction score for stratification of patients with acute infection based on whole-blood gene expression. Sci Transl Med 2022; 14:eabq4433. [PMID: 36322631 PMCID: PMC7613832 DOI: 10.1126/scitranslmed.abq4433] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Dysregulated host responses to infection can lead to organ dysfunction and sepsis, causing millions of global deaths each year. To alleviate this burden, improved prognostication and biomarkers of response are urgently needed. We investigated the use of whole-blood transcriptomics for stratification of patients with severe infection by integrating data from 3149 samples from patients with sepsis due to community-acquired pneumonia or fecal peritonitis admitted to intensive care and healthy individuals into a gene expression reference map. We used this map to derive a quantitative sepsis response signature (SRSq) score reflective of immune dysfunction and predictive of clinical outcomes, which can be estimated using a 7- or 12-gene signature. Last, we built a machine learning framework, SepstratifieR, to deploy SRSq in adult and pediatric bacterial and viral sepsis, H1N1 influenza, and COVID-19, demonstrating clinically relevant stratification across diseases and revealing some of the physiological alterations linking immune dysregulation to mortality. Our method enables early identification of individuals with dysfunctional immune profiles, bringing us closer to precision medicine in infection.
Collapse
Affiliation(s)
- Eddie Cano-Gamez
- Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK,Wellcome Sanger Institute, Wellcome Genome Campus; Cambridge, CB10 1SA, UK
| | - Katie L Burnham
- Wellcome Sanger Institute, Wellcome Genome Campus; Cambridge, CB10 1SA, UK
| | - Cyndi Goh
- Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK,The Jenner Institute, University of Oxford; Oxford, OX3 7DQ, UK
| | - Alice Allcock
- Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Zunaira H. Malick
- Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Lauren Overend
- Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - Andrew Kwok
- Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK
| | - David A. Smith
- Wellcome Centre for Human Genetics, University of Oxford; Oxford, OX3 7BN, UK,Chinese Academy of Medical Science Oxford Institute, University of Oxford; Oxford, OX3 7BN, UK
| | - Hessel Peters-Sengers
- Centre for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam; 1100 DD Amsterdam Southeast, Netherlands,Department of Epidemiology and Data Science, Amsterdam Public Health, Amsterdam University Medical Centers, University of Amsterdam, 1100 DD Amsterdam Southeast, Netherlands,The Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, 1100 DD Amsterdam Southeast, Netherlands
| | - David Antcliffe
- Division of Anaesthesia, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London; London, SW7 2AZ, UK
| |
Collapse
|
183
|
Mora-Obando D, Lomonte B, Pla D, Guerrero-Vargas JA, Ayerbe-González S, Gutiérrez JM, Sasa M, Calvete JJ. Half a century of research on Bothrops asper venom variation: Biological and biomedical implications. Toxicon 2022; 221:106983. [DOI: 10.1016/j.toxicon.2022.106983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
|
184
|
Guo J, Wang R, Min F. Ginsenoside Rg1 ameliorates sepsis-induced acute kidney injury by inhibiting ferroptosis in renal tubular epithelial cells. J Leukoc Biol 2022; 112:1065-1077. [PMID: 35774015 DOI: 10.1002/jlb.1a0422-211r] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/27/2022] [Indexed: 12/24/2022] Open
Abstract
Acute kidney injury (AKI) represents a prevailing complication of sepsis, and its onset involves ferroptosis. Ginsenoside Rg1 exerts a positive effect on kidney diseases. This study explored the action of ginsenoside Rg1 in sepsis-induced AKI (SI-AKI) by regulating ferroptosis in renal tubular epithelial cells (TECs). Sepsis rat models were established using cecal ligation and puncture (CLP) and cell models were established by treating human renal TECs (HK-2) with LPS to induce ferroptosis. Serum creatinine (SCr) and blood urea nitrogen (BUN) and urine KIM1 contents in rats were determined by ELISA kits. Kidney tissues were subjected to immunohistochemical and H&E stainings. Iron concentration, malondialdehyde (MDA), glutathione (GSH), and ferroptosis-related protein (ferritin light chain [FTL], ferritin heavy chain [FTH], GSH peroxidase 4 [GPX4], and Ferroptosis suppressor protein 1 [FSP1]) levels in kidney tissues and HK-2 cells were measured using ELISA kits and Western blotting. HK-2 cell viability was detected by cell counting kit-8, and cell death was observed via propidium iodide staining. Reactive oxygen species accumulation in cells was detected using C11 BODIPY 581/591 as a molecular probe. In CLP rats, ginsenoside Rg1 reduced SCr, BUN, KIM1, and NGAL levels, thus palliating SI-AKI. Additionally, ginsenoside Rg1 decreased iron content, FTL, FTH, and MDA levels, and elevated GPX4, FSP1, and GSH levels, thereby inhibiting lipid peroxidation and ferroptosis. Moreover, FSP1 knockdown annulled the inhibition of ginsenoside Rg1 on ferroptosis. In vitro experiments, ginsenoside Rg1 raised HK-2 cell viability and lowered iron accumulation and lipid peroxidation during ferroptosis, and its antiferroptosis activity was dependent on FSP1. Ginsenoside Rg1 alleviates SI-AKI, possibly resulting from inhibition of ferroptosis in renal TECs through FSP1.
Collapse
Affiliation(s)
- Jun Guo
- Department of Critical Care Medicine, Union Jiangbei Hospital, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Rong Wang
- Department of Critical Care Medicine, Union Jiangbei Hospital, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Fei Min
- Department of Critical Care Medicine, Union Jiangbei Hospital, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
185
|
Labiner HE, Sas KM, Baur JA, Sims CA. Sirtuin 1 deletion increases inflammation and mortality in sepsis. J Trauma Acute Care Surg 2022; 93:672-678. [PMID: 35857031 PMCID: PMC10673225 DOI: 10.1097/ta.0000000000003751] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Sepsis is a hyperinflammatory response to infection that can lead to multiorgan failure and eventually death. Often, the onset of multiorgan failure is heralded by renal dysfunction. Sirtuin 1 (SIRT1) promotes cellular stress resilience by inhibiting inflammation and promoting mitochondrial function. We hypothesize that SIRT1 plays an important role in limiting the inflammatory responses that drive organ failure in sepsis, predominantly via expression in myeloid cells. METHODS We performed cecal ligation and puncture (CLP) on whole body SIRT1 knockout (S1KO) and myeloid cell-specific S1KO (S1KO-LysMCre) mice on a C57BL/6J background. Serum interleukin (IL)-6 was quantified by enzyme-linked immunosorbent assay. Renal mitochondrial complex activity was measured using Oxygraph-2k (Oroboros Instruments, Innsbruck, Austria). Blood urea nitrogen (BUN) was measured from serum. Survival was monitored for up to 5 days. RESULTS Following CLP, S1KO mice had decreased renal mitochondrial complex I-dependent respiratory capacity (241.7 vs. 418.3 mmolO2/mg/min, p = 0.018) and renal mitochondrial complex II-dependent respiratory capacity (932.3 vs. 1,178.4, p = 0.027), as well as reduced rates of fatty acid oxidation (187.3 vs. 250.3, p = 0.022). Sirtuin 1 knockout mice also had increased BUN (48.0 mg/dL vs. 16.0 mg/dL, p = 0.049). Interleukin-6 levels were elevated in S1KO mice (96.5 ng/mL vs. 45.6 ng/mL, p = 0.028) and S1KO-LysMCre mice (35.8 ng/mL vs. 24.5 ng/mL, p = 0.033) compared with controls 12 hours after surgery. Five-day survival in S1KO (33.3% vs. 83.3%, p = 0.025) and S1KO-LysMCre (60% vs. 100%, p = 0.049) mice was decreased compared with controls. CONCLUSION Sirtuin 1 deletion increases systemic inflammation in sepsis. Renal mitochondrial dysfunction, kidney injury, and mortality following CLP were all exacerbated by SIRT1 deletion. Similar effects on inflammation and survival were seen following myeloid cell-specific SIRT1 deletion, indicating that SIRT1 activity in myeloid cells may be a significant contributor for the protective effects of SIRT1 in sepsis.
Collapse
Affiliation(s)
- Hanna E. Labiner
- Division of Trauma, Critical Care, and Burn at The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, 43210
| | - Kelli M. Sas
- Division of Trauma, Critical Care, and Burn at The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, 43210
| | - Joseph A. Baur
- Institute for Diabetes, Obesity and Metabolism and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Carrie A. Sims
- Division of Trauma, Critical Care, and Burn at The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, 43210
| |
Collapse
|
186
|
Ye Z, Gao M, Ge C, Lin W, Zhang L, Zou Y, Peng Q. Association between albumin infusion and septic patients with coronary heart disease: A retrospective study based on medical information mart for intensive care III database. Front Cardiovasc Med 2022; 9:982969. [PMID: 36337861 PMCID: PMC9626508 DOI: 10.3389/fcvm.2022.982969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Coronary heart disease (CHD) is a common comorbidity in intensive care unit (ICU) patients, particularly in the elderly. This particular population may have worse conditions during sepsis, and it presents an overwhelming challenge for clinical practice. Previous studies suggested that patients with CHD have an increased risk of cardiovascular events, and low albumin concentration worsens the prognosis of patients with stable CHD. Hypoalbuminemia in patients with sepsis is common due to nutritional disorders, excessive consumption, and leakage. Albumin is a fluid often used for resuscitation in patients with sepsis. However, albumin infusion in patients with sepsis and CHD has rarely been studied. The effects and safety of albumin infusion in patients with sepsis and CHD remain unclear. Therefore, we collected medical information from Mimic-III (Mimic-III) and compared the all-cause mortality and cardiovascular mortality at 28- or 90-day between the albumin and non-albumin groups in septic patients with CHD. A total of 2,027 patients with sepsis and CHD were included in our study, with 405 in the albumin group and 1,622 in the non-albumin group. After propensity score matching (PSM), 350 pairs were included in our study. Improved survival benefits were found in the albumin group at the 28-day all-cause mortality compared with the non-albumin group (hazard ratio [HR], 0.54; 95% CI: 0.38–0.78; p = 0.0009). However, no difference was detected in the 90-day survival benefits (HR, 0.80, 95% CI: 0.60–1.06, p = 0.1207). Albumin infusion did not reverse cardiovascular mortality neither at 28th day nor at 90th day (cardiovascular mortality: 28 days, HR, 0.52, 95% CI: 0.23–1.19, p = 0.1218; 90 days, HR, 0.66, 95% CI: 0.33–1.33, p = 0.2420).
Collapse
Affiliation(s)
- Zhiwen Ye
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ming Gao
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China
| | - Chenglong Ge
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wenrui Lin
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yu Zou,
| | - Qianyi Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Qianyi Peng,
| |
Collapse
|
187
|
Xia G, Shi H, Su Y, Han B, Shen C, Gao S, Chen Z, Xu C. Photoactivated adenylyl cyclases attenuate sepsis-induced cardiomyopathy by suppressing macrophage-mediated inflammation. Front Immunol 2022; 13:1008702. [PMID: 36330522 PMCID: PMC9624221 DOI: 10.3389/fimmu.2022.1008702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/30/2022] [Indexed: 11/18/2022] Open
Abstract
Sepsis-induced myocardiopathy, characterized by innate immune cells infiltration and proinflammatory cytokines release, may lead to perfusion failure or even life-threatening cardiogenic shock. Macrophages-mediated inflammation has been shown to contribute to sepsis-induced myocardiopathy. In the current study, we introduced two photoactivated adenylyl cyclases (PACs), Beggiatoa sp. PAC (bPAC) and Beggiatoa sp. IS2 PAC (biPAC) into macrophages by transfection to detect the effects of light-induced regulation of macrophage pro-inflammatory response and LPS-induced sepsis-induced myocardiopathy. By this method, we uncovered that blue light-induced bPAC or biPAC activation considerably inhibited the production of pro-inflammatory cytokines IL-1 and TNF-α, both at mRNA and protein levels. Further, we assembled a GelMA-Macrophages-LED system, which consists of GelMA—a type of light crosslink hydrogel, gene modulated macrophages and wireless LED device, to allow light to regulate cardiac inflammation in situ with murine models of LPS-induced sepsis. Our results showed significant inhibition of leukocytes infiltration, especially macrophages and neutrophils, suppression of pro-inflammatory cytokines release, and alleviation of sepsis-induced cardiac dysfunction. Thus, our study may represent an emerging means to treat sepsis-induced myocardiopathy and other cardiovascular diseases by photo-activated regulating macrophage function.
Collapse
Affiliation(s)
- Guofang Xia
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyu Shi
- Wusong Central Hospital, Shanghai, China
| | - Yuanyuan Su
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beibei Han
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiqiang Gao
- Institute of Physiology, Department of Neurophysiology, Julius-Maximilians-University of Wuerzburg, Wuerzburg, Germany
| | - Zhong Chen
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Congfeng Xu, ; Zhong Chen,
| | - Congfeng Xu
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Congfeng Xu, ; Zhong Chen,
| |
Collapse
|
188
|
Wiedermann CJ. Antithrombin as Therapeutic Intervention against Sepsis-Induced Coagulopathy and Disseminated Intravascular Coagulation: Lessons Learned from COVID-19-Associated Coagulopathy. Int J Mol Sci 2022; 23:ijms232012474. [PMID: 36293332 PMCID: PMC9604230 DOI: 10.3390/ijms232012474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 12/04/2022] Open
Abstract
Recent research has contributed significantly to our understanding of the pathogenesis of acute disseminated intravascular coagulation. COVID-19 can be considered as a new underlying condition of disseminated intravascular coagulation. In this narrative review, current evidence is presented regarding biomarker differences between sepsis-induced and COVID-19-associated coagulopathies, supporting the importance of acquired antithrombin deficiency in the early differential diagnosis of septic coagulopathy and its potential impact on treatment with endogenous anticoagulants. Establishing new scoring systems for septic coagulopathy in combination with endogenous anticoagulant biomarker activities may allow for the identification of those in the heterogeneous population of sepsis patients who are more likely to benefit from targeted specific treatment interventions.
Collapse
Affiliation(s)
- Christian J. Wiedermann
- Institute of General Practice, Claudiana—College of Health Professions, 39100 Bolzano, Italy;
- Department of Public Health, Medical Decision Making and HTA, University of Health Sciences, Medical Informatics and Technology—Tyrol, 6060 Hall in Tyrol, Austria
| |
Collapse
|
189
|
Visfatin Amplifies Cardiac Inflammation and Aggravates Cardiac Injury via the NF-κB p65 Signaling Pathway in LPS-Treated Mice. Mediators Inflamm 2022; 2022:3306559. [PMID: 36262545 PMCID: PMC9576419 DOI: 10.1155/2022/3306559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/09/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Background Visfatin is an adipocytokine that has been demonstrated to be involved in cardiovascular diseases. This study aims at determining the role of visfatin in sepsis-induced cardiac injury and identify its possible mechanisms. Methods Dynamic changes in visfatin expression in mice with lipopolysaccharide- (LPS-) induced septicemia were measured. Additionally, mice were pretreated with visfatin and further administered LPS to observe the effects of visfatin on cardiac injury. Finally, septic mice were also pretreated with JSH-23 to investigate whether visfatin regulates cardiac injury via the NF-κB p65 pathway. Results Visfatin expression levels in both the heart and serum were increased in LPS-treated mice and peaked at 6 hours, and visfatin was derived from cardiac macrophages. In septic mice, pretreatment with visfatin reduced the survival rate, worsened cardiac dysfunction, and increased the expression of cardiac injury markers, including creatine kinase myocardial bound (CK-MB) and lactate dehydrogenase (LDH). Treatment with visfatin also increased the infiltration of CD3+ cells and F4/80+ cells, amplified the cardiac inflammatory response, and elevated myocardial cell apoptosis. Treatment with JSH-23 reversed the effects of visfatin in septic mice. Conclusions This study showed that visfatin amplifies the cardiac inflammatory response and aggravates cardiac injury through the p65 signaling pathway. Visfatin may be a clinical target for preventing cardiac injury in sepsis.
Collapse
|
190
|
Wang Q, Deng J, Sun J, Zhang H, Liu D, Gao C, Qiu J, Liu W, Qu G, Wen D, Du J, Zhang A, Zeng L, Jiang J. PDGFR kinase inhibitor protects against septic death via regulation of BTLA. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1917-1928. [PMID: 35918604 PMCID: PMC9345782 DOI: 10.1007/s11427-021-2136-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Sepsis, defined as life-threatening organ failure caused by a dysregulated host response to severe infection, is a major cause of death among intensive care unit patients. Therapies targeting on immunomodulatory is a new research field in sepsis treatment. B- and T-lymphocyte attenuator (BTLA) is an inhibitory costimulatory factor molecule of B and T lymphocytes. Studies have shown that elevated expression of BTLA in lymphocytes can reduce mortality in sepsis, but its regulatory compounds and the underlying mechanism remains to be elucidated. Here, we show that treatment with CP-673451 significantly decreases mortality of septic mouse. CP-673451 is a PDGFR kinase inhibitor which can promote the expression of BTLA, inhibit the release of chemokines such as CXCL13, and reduce first the chemotaxis of B cells to the peripheral blood and vital organs. CP-673451 also inhibits both the release of cytokines and chemokines such as IL-1β, IL-6, IL-10, TNF-α, CCL1, CCL2 and CCL7 and reduces both the chemotactic ability of T cells. This suggests that CP-673451 may prevent septic death by inhibiting lymphocyte chemotaxis and alleviating "cytokine storm". In conclusion, our study provides a new therapeutic target and an effective compound for sepsis treatment.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Jin Deng
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Jianhui Sun
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Huacai Zhang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Di Liu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Chu Gao
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Jinchao Qiu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Wenyi Liu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Guoxin Qu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Dalin Wen
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Juan Du
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Anqiang Zhang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - Ling Zeng
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China.
| | - Jianxin Jiang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
191
|
Xiang Z, Chen X, Zhou X, Qin Y, Zhao X, Wang Y, Li Q, Huang B. Development and application of a novel aldehyde nanoparticle-based amplified luminescent proximity homogeneous assay for rapid quantitation of pancreatic stone protein. Clin Chim Acta 2022; 535:120-130. [PMID: 36030885 DOI: 10.1016/j.cca.2022.08.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/04/2022] [Accepted: 08/20/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Timely diagnosis of bacterial infections is important to prevent sepsis. Classical infection biomarkers have some flaws, and common detection methods are time-consuming. Thus, we aimed to establish an efficient detection method that precisely detects pancreatic stone protein (PSP) in human plasma for the timely diagnosis of bacterial infections. METHODS Based on the novel amplified luminescent proximity homogeneous assay (AlphaLISA) method, donor and acceptor beads modified with aldehyde groups were directly coupled to the anti-PSP antibodies. PSP was quickly detected by a double-antibody sandwich method. Plasma samples from healthy individuals, bacterially infected patients, and acute-phase response patients were tested. RESULTS The detection time of the developed method is only 5 min. The results of PSP-AlphaLISA and time-resolved fluorescence were consistent (ρ = 0.9722). The plasma PSP levels of patients with bacterial infection were significantly higher than those of acute-phase response patients and healthy individuals (P < 0.05). PSP levels in patients with bacterial infection with sepsis were significantly higher than those in patients with bacterial infection without sepsis (P < 0.05). CONCLUSIONS The PSP-AlphaLISA exhibited excellent performance and may be applied to the differential diagnosis between bacterial infection and sepsis in patients without interference from patients with acute-phase response.
Collapse
Affiliation(s)
- Zhongyi Xiang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xindong Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiumei Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xueqin Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qian Li
- Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
| |
Collapse
|
192
|
Xiang Z, Zhang Z, Chen X, Zhang L, Zhou X, Qin Y, Zhao X, Wang Y, Huang B, Tang H. Development and application of amplified luminescent proximity homogeneous assay for quantitation of heparin-binding protein. Anal Biochem 2022; 657:114906. [PMID: 36152874 DOI: 10.1016/j.ab.2022.114906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022]
Abstract
A fast and highly sensitive amplified luminescent proximity homogeneous assay (AlphaLISA) method was developed for quantitation of plasma heparin-binding protein levels. In this study, a method directly coupling donor and acceptor beads modified with aldehyde groups to anti-HBP antibodies was proposed, which can effectively simplify the steps and shorten the reaction time to achieve faster detection. Therefore, the developed method required only 15 min of reaction time to generate results. Compared with the approved commercial kit, the developed method had a wider linear range (2.78-500 ng/mL). The excellent linear range means that the method can better exploit the value of HBP in clinical applications. Meanwhile, results of amplified luminescent proximity homogeneous assay and fluorescence dry quantitative immunoassay had good correlation and consistency (ρ = 0.9181). Moreover, the plasma HBP concentrations of patients with bacterial infection were significantly higher than those of healthy individuals (P < 0.0001), indicating the potential applicability of the proposed method for predicting the incidence of bacterial infections. Importantly, the newly developed method is expected to serve as an alternative to the traditional assay method and provides a completely new platform for other biomarkers that require rapid detection.
Collapse
Affiliation(s)
- Zhongyi Xiang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zhibin Zhang
- Clinical Laboratory, Wuxi Hospital of Traditional Chinese Medicine, Wuxi, China
| | - Xindong Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Lele Zhang
- Clinical Laboratory Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Xiumei Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xueqin Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
| | - Huqiang Tang
- Clinical Laboratory Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, China.
| |
Collapse
|
193
|
Mück A, Herter T. [69/m-Recurrent diverticulitis of the pelvic colon, tachypnea and altered mental status : Preparation course anesthesiological intensive care medicine: case 9]. DIE ANAESTHESIOLOGIE 2022; 71:48-55. [PMID: 35925178 DOI: 10.1007/s00101-022-01146-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Alexander Mück
- Klinik für Anästhesiologie und operative Intensivmedizin, Universitätsklinikum Augsburg, Stenglinstraße 2, 86156, Augsburg, Deutschland.
| | - Tobias Herter
- Klinik für Anästhesie, Intensiv- & Notfallmedizin, St. Josefskrankenhaus Freiburg, Freiburg, Deutschland
| |
Collapse
|
194
|
Sorrentino JT, Golden GJ, Morris C, Painter CD, Nizet V, Campos AR, Smith JW, Karlsson C, Malmström J, Lewis NE, Esko JD, Gómez Toledo A. Vascular Proteome Responses Precede Organ Dysfunction in a Murine Model of Staphylococcus aureus Bacteremia. mSystems 2022; 7:e0039522. [PMID: 35913192 PMCID: PMC9426442 DOI: 10.1128/msystems.00395-22] [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: 04/26/2022] [Accepted: 07/16/2022] [Indexed: 12/24/2022] Open
Abstract
Vascular dysfunction and organ failure are two distinct, albeit highly interconnected, clinical outcomes linked to morbidity and mortality in human sepsis. The mechanisms driving vascular and parenchymal damage are dynamic and display significant molecular cross talk between organs and tissues. Therefore, assessing their individual contribution to disease progression is technically challenging. Here, we hypothesize that dysregulated vascular responses predispose the organism to organ failure. To address this hypothesis, we have evaluated four major organs in a murine model of Staphylococcus aureus sepsis by combining in vivo labeling of the endothelial cell surface proteome, data-independent acquisition (DIA) mass spectrometry, and an integrative computational pipeline. The data reveal, with unprecedented depth and throughput, that a septic insult evokes organ-specific proteome responses that are highly compartmentalized, synchronously coordinated, and significantly correlated with the progression of the disease. These responses include abundant vascular shedding, dysregulation of the intrinsic pathway of coagulation, compartmentalization of the acute phase response, and abundant upregulation of glycocalyx components. Vascular cell surface proteome changes were also found to precede bacterial invasion and leukocyte infiltration into the organs, as well as to precede changes in various well-established cellular and biochemical correlates of systemic coagulopathy and tissue dysfunction. Importantly, our data suggest a potential role for the vascular proteome as a determinant of the susceptibility of the organs to undergo failure during sepsis. IMPORTANCE Sepsis is a life-threatening response to infection that results in immune dysregulation, vascular dysfunction, and organ failure. New methods are needed for the identification of diagnostic and therapeutic targets. Here, we took a systems-wide approach using data-independent acquisition (DIA) mass spectrometry to track the progression of bacterial sepsis in the vasculature leading to organ failure. Using a murine model of S. aureus sepsis, we were able to quantify thousands of proteins across the plasma and parenchymal and vascular compartments of multiple organs in a time-resolved fashion. We showcase the profound proteome remodeling triggered by sepsis over time and across these compartments. Importantly, many vascular proteome alterations precede changes in traditional correlates of organ dysfunction, opening a molecular window for the discovery of early markers of sepsis progression.
Collapse
Affiliation(s)
- James T. Sorrentino
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Gregory J. Golden
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Claire Morris
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Chelsea D. Painter
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - Alexandre Rosa Campos
- The Cancer Center and The Inflammatory and Infectious Disease Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Jeffrey W. Smith
- The Cancer Center and The Inflammatory and Infectious Disease Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Christofer Karlsson
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, BMC, Lund, Sweden
| | - Johan Malmström
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, BMC, Lund, Sweden
| | - Nathan E. Lewis
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- National Biologics Facility, Technical University of Denmark, Krogens-Lyngby, Denmark
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Alejandro Gómez Toledo
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, BMC, Lund, Sweden
| |
Collapse
|
195
|
Matouk AI, El-Daly M, Habib HA, Senousy S, Naguib Abdel Hafez SM, Kasem AW, Almalki WH, Alzahrani A, Alshehri A, Ahmed ASF. Protective effects of menthol against sepsis-induced hepatic injury: Role of mediators of hepatic inflammation, apoptosis, and regeneration. Front Pharmacol 2022; 13:952337. [PMID: 36120368 PMCID: PMC9476320 DOI: 10.3389/fphar.2022.952337] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Liver dysfunction in sepsis is a major complication that amplifies multiple organ failure and increases the risk of death. Inflammation and oxidative stress are the main mediators in the pathophysiology of sepsis. Therefore, we investigated the role of menthol, a natural antioxidant, against sepsis-induced liver injury in female Wistar rats. Sepsis was induced by cecal ligation and puncture (CLP). Menthol (100 mg/kg) was given intragastric 2 h after CLP. Blood samples and liver tissues were collected 24 h after surgery. Menthol significantly (p < 0.05) attenuated the sepsis-induced elevation in serum liver enzymes and improved the hepatic histopathological changes. Menthol treatment significantly (p < 0.05) decreased hepatic levels of tumor necrosis factor-alpha, malondialdehyde, total nitrite, and cleaved caspase-3. It restored the hepatic levels of superoxide dismutase and reduced glutathione. Additionally, menthol significantly (p < 0.05) increased hepatic levels of B-cell lymphoma 2 (Bcl-2); an anti-apoptotic factor, and proliferating cell nuclear antigen (PCNA), a biomarker of regeneration and survival. Our results showed the therapeutic potential of menthol against liver injury induced by sepsis.
Collapse
Affiliation(s)
- Asmaa I. Matouk
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Mahmoud El-Daly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Heba A. Habib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Shaymaa Senousy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | | | - AlShaimaa W. Kasem
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Minya, Egypt
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdulaziz Alzahrani
- Department of Pharmacology and Toxicology, College of Clinical Pharmacy, AlBaha University, Al Bahah, Saudi Arabia
| | - Ahmed Alshehri
- Department of Pharmacology and Toxicology, College of Clinical Pharmacy, AlBaha University, Al Bahah, Saudi Arabia
| | - Al-Shaimaa F. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
- *Correspondence: Al-Shaimaa F. Ahmed,
| |
Collapse
|
196
|
Zhang B, Yin S, Guo C, Gao Z, Li T, Lee W, Shen J, Yang X. Fragments of bombinakinin M exist in lipopolysaccharide-stimulated skin secretions of Bombina maxima and show lipopolysaccharide-neutralizing activity. J Immunol Methods 2022; 509:113343. [PMID: 36029800 DOI: 10.1016/j.jim.2022.113343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
Lipopolysaccharide (LPS) is a major pathogen-associated pattern molecule that can initiate lethal sepsis. Bioactive peptides in amphibian skin secretions, especially antimicrobial peptides, are essential components of the host immune system and help fight the microbial invasion. In this study, two peptides: peptide 1 (KINRKGPRPPG) and peptide 2 (INRKGPRPPG) were isolated, from skin secretions of the Chinese red belly frog (Bombina maxima). After stimulation with LPS, peptide 1 showed direct LPS-binding activity, low cytotoxicity, immunoregulatory functions in vitro, and neutralizing LPS effects in animal models. Thus, natural peptide 1 exhibits potential as an ideal candidate against LPS.
Collapse
Affiliation(s)
- Baiyu Zhang
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Saige Yin
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Caifen Guo
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Zhenhua Gao
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Tonghai Li
- Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Wenhui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms, Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Jihong Shen
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, China.
| |
Collapse
|
197
|
Xu F, Zhang L, Huang T, Han D, Yang R, Zheng S, Feng A, Huang L, Yin H, Lyu J. Effects of growth trajectory of shock index within 24 h on the prognosis of patients with sepsis. Front Med (Lausanne) 2022; 9:898424. [PMID: 36072946 PMCID: PMC9441919 DOI: 10.3389/fmed.2022.898424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/02/2022] [Indexed: 01/09/2023] Open
Abstract
BackgroundSepsis is a serious disease with high clinical morbidity and mortality. Despite the tremendous advances in medicine and nursing, treatment of sepsis remains a huge challenge. Our purpose was to explore the effects of shock index (SI) trajectory changes on the prognosis of patients within 24 h after the diagnosis of sepsis.MethodsThis study was based on Medical Information Mart for Intensive Care IV (MIMIC- IV). The effects of SI on the prognosis of patients with sepsis were investigated using C-index and restricted cubic spline (RCS). The trajectory of SI in 24 h after sepsis diagnosis was classified by latent growth mixture modeling (LGMM). Cox proportional hazard model, double robust analysis, and subgroup analysis were conducted to investigate the influence of SI trajectory on in-hospital death and secondary outcomes.ResultsA total of 19,869 patients were eventually enrolled in this study. C-index showed that SI had a prognostic value independent of Sequential Organ Failure Assessment for patients with sepsis. Moreover, the results of RCS showed that SI was a prognostic risk factor. LGMM divided SI trajectory into seven classes, and patients with sepsis in different classes had notable differences in prognosis. Compared with the SI continuously at a low level of 0.6, the SI continued to be at a level higher than 1.0, and the patients in the class whose initial SI was at a high level of 1.2 and then declined had a worse prognosis. Furthermore, the trajectory of SI had a higher prognostic value than the initial SI.ConclusionBoth initial SI and trajectory of SI were found to be independent factors that affect the prognosis of patients with sepsis. Therefore, in clinical treatment, we should closely monitor the basic vital signs of patients and arrive at appropriate clinical decisions on basis of their change trajectory.
Collapse
Affiliation(s)
- Fengshuo Xu
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Nosocomial Infection Management, Luoyang Orthopedic-Traumatological Hospital, Orthopedics Hospital of Henan Province, Zhengzhou, China
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Luming Zhang
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tao Huang
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Didi Han
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Rui Yang
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Shuai Zheng
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Aozi Feng
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liying Huang
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Haiyan Yin
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Haiyan Yin,
| | - Jun Lyu
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, China
- *Correspondence: Jun Lyu,
| |
Collapse
|
198
|
Liu R, Luo X, Li J, Lei Y, Zeng F, Huang X, Lan Y, Yang F. Melatonin: A window into the organ-protective effects of sepsis. Biomed Pharmacother 2022; 154:113556. [PMID: 35994818 DOI: 10.1016/j.biopha.2022.113556] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/22/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022] Open
Abstract
Sepsis is an uncontrolled host response to infection. In some cases, it progresses to multi-organ insufficiency, leading to septic shock and increased risk of mortality. Various organ support strategies are currently applied clinically, but they are still inadequate in terms of reducing mortality. Melatonin is a hormone that regulates sleep and wakefulness, and it is associated with a reduced risk of death in patients with sepsis. Evidence suggests that melatonin may help protect organ function from sepsis-related damage. Here, we review information related to the role of melatonin in protecting organ function during sepsis and explore its potential clinical applications, with the aim of providing an effective therapeutic strategy for treating sepsis-induced organ insufficiency.
Collapse
Affiliation(s)
- Rongan Liu
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiu Luo
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiajia Li
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Lei
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fan Zeng
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaobo Huang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunping Lan
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Fuxun Yang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
199
|
Jia N, Li G, Wang X, Cao Q, Chen W, Wang C, Chen L, Ma X, Zhang X, Tao Y, Zang J, Mo X, Hu J. Staphylococcal superantigen-like protein 10 induces necroptosis through TNFR1 activation of RIPK3-dependent signal pathways. Commun Biol 2022; 5:813. [PMID: 35962126 PMCID: PMC9374677 DOI: 10.1038/s42003-022-03752-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/22/2022] [Indexed: 11/11/2022] Open
Abstract
Staphylococcal aureus (S. aureus) infection can lead to a wide range of diseases such as sepsis and pneumonia. Staphylococcal superantigen-like (SSL) proteins, expressed by all known S. aureus strains, are shown to be involved in immune evasion during S. aureus infection. Here, we show that SSL10, an SSL family protein, exhibits potent cytotoxicity against human cells (HEK293T and HUVEC) by inducing necroptosis upon binding to its receptor TNFR1 on the cell membrane. After binding, two distinct signaling pathways are activated downstream of TNFR1 in a RIPK3-dependent manner, i.e., the RIPK1-RIPK3-MLKL and RIPK3-CaMKII-mitochondrial permeability transition pore (mPTP) pathways. Knockout of ssl10 in S. aureus significantly reduces cytotoxicity of the culture supernatants of S. aureus, indicating that SSL10 is involved in extracellular cytotoxicity during infection. We determined the crystal structure of SSL10 at 1.9 Å resolution and identified a positively charged surface of SSL10 responsible for TNFR1 binding and cytotoxic activity. This study thus provides the description of cytotoxicity through induction of necroptosis by the SSL10 protein, and a potential target for clinical treatment of S. aureus-associated diseases. The Staphylococcal superantigen like protein 10 induces necroptosis in human cells through binding to TNFR1 by both the N- and C-terminal domains and activating RIPK1-RIPK3-MLKL and RIPK3-CaMKII-mitochondrial permeability transition pore pathways.
Collapse
Affiliation(s)
- Nan Jia
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, China.,The Laboratory of Pediatric Infectious Diseases, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Guo Li
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Xing Wang
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Qing Cao
- Department of Infectious Diseases, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Wanbiao Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Chengliang Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Ling Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Xiaoling Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Xuan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Yue Tao
- The Laboratory of Pediatric Infectious Diseases, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jianye Zang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, China.
| | - Xi Mo
- The Laboratory of Pediatric Infectious Diseases, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jinfeng Hu
- The Laboratory of Pediatric Infectious Diseases, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China. .,Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
200
|
Ultraporous Polyquaternium-Carboxylated Chitosan Composite Hydrogel Spheres with Anticoagulant, Antibacterial, and Rapid Endotoxin Removal Profiles for Sepsis Treatment. Biomacromolecules 2022; 23:3728-3742. [PMID: 35926229 DOI: 10.1021/acs.biomac.2c00583] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hemoperfusion is an important method to remove endotoxins and save the lives of patients with sepsis. However, the current adsorbents for hemoperfusion have disadvantages of insufficient endotoxin adsorption capacity, poor blood compatibility, and so on. Herein, we proposed a novel emulsion templating (ET) method to prepare ultraporous and double-network carboxylated chitosan (CCS)-poly(diallyl dimethylammonium chloride) (PDDA) hydrogel spheres (ET-CCSPD), bearing both negative and positive charges. CCS was introduced to balance the strong positive charges of PDDA to improve hemocompatibility, and emulsion templates endowed the adsorbent with an ultraporous structure for enhanced adsorption efficacy. The ET-CCSPDs neither damaged blood cells nor activated complement responses. In addition, the activated partial thromboplastin time (APTT) was prolonged to 8.5 times, which was beneficial for reducing the injection of anticoagulant in patients. The ET-CCSPDs had excellent scavenging performance against bacteria and endotoxin, with removal ratios of 96.7% for E. coli and 99.8% for S. aureus, respectively, and the static removal ratio of endotoxin in plasma was as high as 99.1% (C0 = 5.50 EU/mL, critical illness level). An adsorption cartridge filled with the ET-CCSPDs could remove 84.7% of endotoxin within 1 h (C0 = 100 EU/mL in PBS). Interestingly, the ET-CCSPDs had a good inhibitory effect on the cytokines produced by endotoxin-mediated septic blood. By developing the ET method to prepare ultraporous and double-network adsorbents, the problems of low adsorption efficiency and poor blood compatibility of traditional endotoxin adsorbents have been solved, thus opening a new route to fabricate absorbents for blood purification.
Collapse
|