1
|
Zhu CL, Wang Y, Ren SC, Yu CM, Sun XY, Liu ZL, Li QQ, Guo DZ, Chen Y, You J, Wang JF. THE DELIVERY OF PD-L1 SIRNA BY NEUTROPHIL-TARGETED LIPID NANOPARTICLES EFFECTIVELY AMELIORATES SEPSIS. Shock 2024; 62:707-715. [PMID: 39158541 DOI: 10.1097/shk.0000000000002450] [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: 08/20/2024]
Abstract
ABSTRACT Background: Sepsis, a complex and life-threatening disease, poses a significant global burden affecting over 48 million individuals. Recently, it has been reported that programmed death-ligand 1 (PD-L1) expressed on neutrophils is involved in both inflammatory organ dysfunction and immunoparalysis in sepsis. However, there is a dearth of strategies to specifically target PD-L1 in neutrophils in vivo . Methods: We successfully developed two lipid nanoparticles (LNPs) specifically targeting neutrophils by delivering PD-L1 siRNA via neutrophil-specific antibodies and polypeptides. In vivo and in vitro experiments were performed to detect lipid nanoparticles into neutrophils. A mouse cecal ligation and puncture model was used to detect neutrophil migration, neutrophil extracellular traps level, and organ damage. Result: The PD-L1 siRNA-loaded LNPs that target neutrophils suppressed inflammation, reduced the release of neutrophil extracellular traps, and inhibited T-lymphocyte apoptosis. This approach could help maintain homeostasis of both the immune and inflammatory responses during sepsis. Furthermore, the PD-L1 siRNA-loaded LNPs targeting neutrophils have the potential to ameliorate the multiorgan damage and lethality resulting from cecal ligation and puncture. Conclusions: Taken together, our data identify a previously unknown drug delivery strategy targeting neutrophils, which represents a novel, safe, and effective approach to sepsis therapy.
Collapse
Affiliation(s)
- Cheng-Long Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Yi Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Shi-Chun Ren
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | | | | | | | | | - De-Zhi Guo
- The Battalion 3 of Cadet Brigade, School of Basic Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Yu Chen
- The Battalion 5 of Cadet Brigade, School of Basic Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Jia You
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Jia-Feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Wu X, Zhao X, Li F, Wang Y, Ou Y, Zhang H, Li X, Wu X, Wang L, Li M, Zhang Y, Liu J, Xing M, Liu H, Tan Y, Wang Y, Xie Y, Zhang H, Luo Y, Li H, Wang J, Sun L, Li Y, Zhang H. MLKL-mediated endothelial necroptosis drives vascular damage and mortality in systemic inflammatory response syndrome. Cell Mol Immunol 2024; 21:1309-1321. [PMID: 39349742 PMCID: PMC11527879 DOI: 10.1038/s41423-024-01217-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/11/2024] [Indexed: 11/02/2024] Open
Abstract
The hypersecretion of cytokines triggers life-threatening systemic inflammatory response syndrome (SIRS), leading to multiple organ dysfunction syndrome (MODS) and mortality. Although both coagulopathy and necroptosis have been identified as important factors in the pathogenesis of SIRS, the specific cell types that undergo necroptosis and the interrelationships between coagulopathy and necroptosis remain unclear. In this study, we utilized visualization analysis via intravital microscopy to demonstrate that both anticoagulant heparin and nonanticoagulant heparin (NAH) pretreatment protect mice against TNF-α-induced mortality in SIRS. Moreover, the deletion of Mlkl or Ripk3 resulted in decreased coagulation and reduced mortality in TNF-α-induced SIRS. These findings suggest that necroptosis plays a key role upstream of coagulation in SIRS-related mortality. Furthermore, using a genetic lineage tracing mouse model (Tie2-Cre;Rosa26-tdT), we tracked endothelial cells (ECs) and verified that EC necroptosis is responsible for the vascular damage observed in TNF-α-treated mice. Importantly, Mlkl deletion in vascular ECs in mice had a similar protective effect against lethal SIRS by blocking EC necroptosis to protect the integrity of the endothelium. Collectively, our findings demonstrated that RIPK3-MLKL-dependent necroptosis disrupted vascular integrity, resulting in coagulopathy and multiorgan failure, eventually leading to mortality in SIRS patients. These results highlight the importance of targeting vascular EC necroptosis for the development of effective treatments for SIRS patients.
Collapse
Affiliation(s)
- Xiaoxia Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xiaoming Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Fang Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yang Wang
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yangjing Ou
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Haiwei Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xiaoming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xuanhui Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Lingxia Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Ming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yue Zhang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jianling Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Mingyan Xing
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Han Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yongchang Tan
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yangyang Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yangyang Xie
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Hanwen Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hong Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Jing Wang
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Liming Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China
| | - Yu Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
3
|
Kim J, Kim JH, Kim Y, Lee J, Lee HJ, Koh SJ, Im JP, Kim JS. iRhom2 deficiency reduces sepsis-induced mortality associated with the attenuation of lung macrophages in mice. Histochem Cell Biol 2024; 162:415-428. [PMID: 39134731 PMCID: PMC11393161 DOI: 10.1007/s00418-024-02318-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/13/2024]
Abstract
Sepsis has a high mortality rate and leads to multi-organ failure, including lung injury. Inactive rhomboid protease family protein (iRhom2) has been identified as accountable for the release of TNF-α, a crucial mediator in the development of sepsis. This study aimed to evaluate the role of iRhom2 in sepsis and sepsis-induced acute lung injury (ALI). TNF-α and IL-6 secretion in vitro by peritoneal macrophages from wild-type (WT) and iRhom2 knoukout (KO) mice was assessed by enzyme-linked immunosorbent assay. Cecal ligation and puncture (CLP)-induced murine sepsis model was used for in vivo experiments. To evaluate the role of iRhom2 deficiency on survival during sepsis, both WT and iRhom2 KO mice were monitored for 8 consecutive days following the CLP. For histologic and biochemical examination, the mice were killed 18 h after CLP. iRhom2 deficiency improved the survival of mice after CLP. iRhom2 deficiency decreased CD68+ macrophage infiltration in lung tissues. Multiplex immunohistochemistry revealed that the proportion of Ki-67+ CD68+ macrophages was significantly lower in iRhom2 KO mice than that in WT mice after CLP. Moreover, CLP-induced release of TNF-α and IL-6 in the serum were significantly inhibited by iRhom2 deficiency. iRhom2 deficiency reduced NF-kB p65 and IκBα phosphorylation after CLP. iRhom2 deficiency reduces sepsis-related mortality associated with attenuated macrophage infiltration and proliferation in early lung injury. iRhom2 may play a pivotal role in the pathogenesis of sepsis and early stage of sepsis-induced ALI. Thus, iRhom2 may be a potential therapeutic target for the management of sepsis and sepsis-induced ALI.
Collapse
Affiliation(s)
- Jihye Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, 101 Daehak-ro Jongno-gu, Seoul, 03080, South Korea
- Center for Health Promotion and Optimal Aging, Seoul National University Hospital, Seoul, South Korea
| | - Jee Hyun Kim
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | - Younghoon Kim
- Department of Pathology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jooyoung Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, 101 Daehak-ro Jongno-gu, Seoul, 03080, South Korea
| | - Hyun Jung Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, 101 Daehak-ro Jongno-gu, Seoul, 03080, South Korea
| | - Seong-Joon Koh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, 101 Daehak-ro Jongno-gu, Seoul, 03080, South Korea
| | - Jong Pil Im
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, 101 Daehak-ro Jongno-gu, Seoul, 03080, South Korea
| | - Joo Sung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, 101 Daehak-ro Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
4
|
Gao M, Zha Y, Sheng N, Cao Y, Yao W, Bao B, Shan M, Cheng F, Yu S, Zhang Y, Geng T, Liu S, Yan H, Chen P, Zhang J, Zhang L. Integrated transcriptomics and lipidomics reveals protective effect in vascular endothelial barrier of a polysaccharide from Typhae Pollen. Int J Biol Macromol 2024; 282:136817. [PMID: 39490477 DOI: 10.1016/j.ijbiomac.2024.136817] [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: 06/11/2024] [Revised: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Endothelial cell dysfunction caused by inflammation and even vascular leakage are important manifestations of blood stasis syndrome (BSS). Reversible regulation of vascular integrity to cure BSS has attracted considerable interest. Herein, a novel acidic polysaccharide (TPP-4) was purified and characterized from Typhae Pollen, a typical traditional Chinese medicine for treating BSS, especially for bleeding caused by blood stasis. A series of structural characterization methods, including spectroscopic methods (FT-IR and UV), chromatographic methods (HPGPC, HPAEC-PDA and GC-MS) and NMR, have been used to reveal the fine structure of TPP-4. TPP-4 was a homogeneous heteropolysaccharide comprised with RG-I backbone. TPP-4 showed fantastic activities in vascular integrity regulation both in vitro (HUVECs) and in vivo (zebrafish). Transcriptomics revealed that SOX7 and lipid metabolism were the potential targets. Lipidomics showed that TPP-4 could regulate lipid metabolism disorders caused by vascular inflammation, particularly affecting LPE levels. The above regulatory effects were furtherly demonstrated to be related with VEGFA/PI3K/mTOR signaling pathway through various molecular biological experiments.
Collapse
Affiliation(s)
- Mingliang Gao
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yuling Zha
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Nian Sheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yudan Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weifeng Yao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Beihua Bao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Mingqiu Shan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Fangfang Cheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Sheng Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yi Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ting Geng
- Nanjing University of Chinese Medicine Hanlin College, Taizhou 225300, China.
| | - Shengjin Liu
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hui Yan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Peidong Chen
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Juanjuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China.
| | - Li Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
5
|
Baillie JK, Angus D, Burnham K, Calandra T, Calfee C, Gutteridge A, Hacohen N, Khatri P, Langley R, Ma'ayan A, Marshall J, Maslove D, Prescott HC, Rowan K, Scicluna BP, Seymour C, Shankar-Hari M, Shapiro N, Joost Wiersinga W, Singer M, Randolph AG. Causal inference can lead us to modifiable mechanisms and informative archetypes in sepsis. Intensive Care Med 2024:10.1007/s00134-024-07665-4. [PMID: 39432104 PMCID: PMC7616750 DOI: 10.1007/s00134-024-07665-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024]
Abstract
Medical progress is reflected in the advance from broad clinical syndromes to mechanistically coherent diagnoses. By this metric, research in sepsis is far behind other areas of medicine-the word itself conflates multiple different disease mechanisms, whilst excluding noninfectious syndromes (e.g., trauma, pancreatitis) with similar pathogenesis. New technologies, both for deep phenotyping and data analysis, offer the capability to define biological states with extreme depth. Progress is limited by a fundamental problem: observed groupings of patients lacking shared causal mechanisms are very poor predictors of response to treatment. Here, we discuss concrete steps to identify groups of patients reflecting archetypes of disease with shared underlying mechanisms of pathogenesis. Recent evidence demonstrates the role of causal inference from host genetics and randomised clinical trials to inform stratification analyses. Genetic studies can directly illuminate drug targets, but in addition they create a reservoir of statistical power that can be divided many times among potential patient subgroups to test for mechanistic coherence, accelerating discovery of modifiable mechanisms for testing in trials. Novel approaches, such as subgroup identification in-flight in clinical trials, will improve efficiency. Within the next decade, we expect ongoing large-scale collaborative projects to discover and test therapeutically relevant sepsis archetypes.
Collapse
Affiliation(s)
- J Kenneth Baillie
- Baillie Gifford Pandemic Science Hub, Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, UK.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK.
- Intensive Care Unit, Royal Infirmary of Edinburgh, Edinburgh, UK.
- International Sepsis Forum, Murphy, NC, USA.
| | - Derek Angus
- International Sepsis Forum, Murphy, NC, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA
- UPMC Health System, Pittsburgh, PA, USA
| | | | - Thierry Calandra
- International Sepsis Forum, Murphy, NC, USA
- Service of Immunology and Allergy, Department of Medicine, Lausanne, Switzerland
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Carolyn Calfee
- International Sepsis Forum, Murphy, NC, USA
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Departments of Medicine and Anesthesia, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Palo Alto, CA, USA
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Raymond Langley
- College of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Avi Ma'ayan
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Marshall
- International Sepsis Forum, Murphy, NC, USA
- Unity Health Toronto, Toronto, ON, Canada
| | - David Maslove
- Department of Critical Care Medicine, Queen's University, Kingston, ON, Canada
| | - Hallie C Prescott
- International Sepsis Forum, Murphy, NC, USA
- University of Michigan, Ann Arbor, MI, USA
| | - Kathy Rowan
- International Sepsis Forum, Murphy, NC, USA
- Intensive Care National Audit & Research Centre, London, UK
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei hospital, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences bldg., University of Malta, Msida, Malta
| | - Christopher Seymour
- International Sepsis Forum, Murphy, NC, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Manu Shankar-Hari
- Intensive Care Unit, Royal Infirmary of Edinburgh, Edinburgh, UK
- International Sepsis Forum, Murphy, NC, USA
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Nathan Shapiro
- International Sepsis Forum, Murphy, NC, USA
- Harvard University, Boston, USA
| | - W Joost Wiersinga
- International Sepsis Forum, Murphy, NC, USA
- Division of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mervyn Singer
- International Sepsis Forum, Murphy, NC, USA
- University College London, London, UK
| | - Adrienne G Randolph
- International Sepsis Forum, Murphy, NC, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
- Departments of Anaesthesia and Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Shen L, Tao C, Zhu K, Cai L, Yang S, Jin J, Ren Y, Xiao Y, Zhang Y, Lai D, Tou J. Key platelet genes play important roles in predicting the prognosis of sepsis. Sci Rep 2024; 14:23530. [PMID: 39384856 PMCID: PMC11464784 DOI: 10.1038/s41598-024-74052-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/23/2024] [Indexed: 10/11/2024] Open
Abstract
Sepsis is a life-threatening organ malfunction induced by an imbalanced immunological reaction to infection in the host. Many studies have utilized traditional RNA sequencing (RNA-seq) data to identify important biological targets to predict sepsis prognosis. However, alterations in core cells and functional status cannot be effectively detected in sepsis patients. The goal of this study was to identify key cells through single-cell RNA-seq (scRNA-seq), and combine bulk RNA-seq data and multiple algorithm analysis to construct a stable prognostic model for sepsis. The scRNA-seq and bulk RNA-seq data from sepsis patients were collected from the Gene Expression Omnibus (GEO) database. The R package "Seurat" was used to process the scRNA-seq data. Cell communication was investigated using the R package "CellChat". The pseudo-time of the cells was calculated using the R package "monocle". The R package "limma" was used to identify differentially expressed genes (DEGs) between the sepsis group and the control group. Weighted gene correlation network analysis (WGCNA) was used to identify critical modules. Eight kinds of machine learning and 90 algorithm combinations were used to construct the prognostic model for sepsis. Quantitative real-time PCR (qRT‒PCR) was performed to determine the expression of key genes in the cecal ligation and puncture (CLP)-induced sepsis mouse model. The immunological status and related properties of DEGs were then investigated in the high- and low-risk groups delineated by the model. By combining the scRNA-seq data from nine samples, 13 clusters and 9 cell types were identified. CellChat analysis revealed that the number and strength of interactions between platelets and a variety of cells increased. We identified key platelet genes from the scRNA-seq data and combined these genes and the results of differential analysis and WGCNA of the bulk RNA-seq data. After univariate Cox regression analysis, we calculated the Cindex of the model constructed by the combination of 90 algorithms, and we finally determined the "CoxBoost + Lasso" combination. Multivariate Cox regression was used to construct the final prognostic model. The qRT-PCR results revealed significant differences in five key prognostic genes between the CLP and sham groups. The data was classified into high- and low-risk groups based on the model score. The high-risk group had a poorer survival rate and less immune infiltration. We identified the importance of platelets in sepsis patients through scRNA-seq, and established prognostic models with key genes that were identified via scRNA-seq combined with bulk RNA-seq analysis. The results of this model were closely associated with patient survival rates and immunological status and this model is useful for the prognostic management of sepsis.
Collapse
Affiliation(s)
- Leiting Shen
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chang Tao
- Department of Urology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Kun Zhu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Linghao Cai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Sisi Yang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jingyi Jin
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yichao Ren
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yi Xiao
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuebai Zhang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Jinfa Tou
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
7
|
Geng X, Xia X, Liang Z, Li S, Yue Z, Zhang H, Guo L, Ma S, Jiang S, Lian X, Zhou J, Sung LA, Wang X, Yao W. Tropomodulin1 exacerbates inflammatory response in macrophages by negatively regulating LPS-induced TLR4 endocytosis. Cell Mol Life Sci 2024; 81:402. [PMID: 39276234 PMCID: PMC11401823 DOI: 10.1007/s00018-024-05424-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/24/2024] [Accepted: 08/17/2024] [Indexed: 09/16/2024]
Abstract
The excessive inflammation caused by the prolonged activation of Toll-like receptor 4 (TLR4) and its downstream signaling pathways leads to sepsis. CD14-mediated endocytosis of TLR4 is the key step to control the amount of TLR4 on cell membrane and the activity of downstream pathways. The actin cytoskeleton is necessary for receptor-mediated endocytosis, but its role in TLR4 endocytosis remains elusive. Here we show that Tropomodulin 1 (Tmod1), an actin capping protein, inhibited lipopolysaccharide (LPS)-induced TLR4 endocytosis and intracellular trafficking in macrophages. Thus it resulted in increased surface TLR4 and the upregulation of myeloid differentiation factor 88 (MyD88)-dependent pathway and the downregulation of TIR domain-containing adaptor-inducing interferon-β (TRIF)-dependent pathway, leading to the enhanced secretion of inflammatory cytokines, such as TNF-α and IL-6, and the reduced secretion of cytokines, such as IFN-β. Macrophages deficient with Tmod1 relieved the inflammatory response in LPS-induced acute lung injury mouse model. Mechanistically, Tmod1 negatively regulated LPS-induced TLR4 endocytosis and inflammatory response through modulating the activity of CD14/Syk/PLCγ2/IP3/Ca2+ signaling pathway, the reorganization of actin cytoskeleton, and the membrane tension. Therefore, Tmod1 is a key regulator of inflammatory response and immune functions in macrophages and may be a potential target for the treatment of excessive inflammation and sepsis.
Collapse
Affiliation(s)
- Xueyu Geng
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University Health Center, Beijing, 100191, China
| | - Xue Xia
- Nanjing Institute of Measurement and Testing Technology, Nanjing, 210049, Jiangsu Province, China
| | - Zhenhui Liang
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University Health Center, Beijing, 100191, China
| | - Shuo Li
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zejun Yue
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University Health Center, Beijing, 100191, China
| | - Huan Zhang
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lina Guo
- Department of Rehabilitation Medicine, Caoxian People's Hospital, Heze, 274400, Shandong Province, China
| | - Shan Ma
- Chengde Medical College, Chengde, 067000, Hebei Province, China
| | - Siyu Jiang
- Chengde Medical College, Chengde, 067000, Hebei Province, China
| | - Xiang Lian
- Department of Emergency, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Jing Zhou
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lanping Amy Sung
- Department of Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Xifu Wang
- Department of Emergency, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| | - Weijuan Yao
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University Health Center, Beijing, 100191, China.
| |
Collapse
|
8
|
Behal ML, Flannery AH, Miano TA. The times are changing: A primer on novel clinical trial designs and endpoints in critical care research. Am J Health Syst Pharm 2024; 81:890-902. [PMID: 38742701 PMCID: PMC11383190 DOI: 10.1093/ajhp/zxae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Michael L Behal
- Department of Pharmacy, University of Tennessee Medical Center, Knoxville, TN, USA
| | - Alexander H Flannery
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - Todd A Miano
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, and Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
Gu Q, Huang P, Yang Q, Meng X, Zhao M. A nomogram to predict 28-day mortality in patients with sepsis combined coronary artery disease: retrospective study based on the MIMIC-III database. Front Med (Lausanne) 2024; 11:1433809. [PMID: 39296895 PMCID: PMC11408215 DOI: 10.3389/fmed.2024.1433809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Object Establish a clinical prognosis model of coronary heart disease (CHD) to predict 28-day mortality in patients with sepsis. Method The data were collected retrospectively from septic patients with a previous history of coronary heart disease (CHD) from the Medical Information Mart for Intensive Care (MIMIC)-III database. The included patients were randomly divided into the training cohorts and validation cohorts. The variables were selected using the backward stepwise selection method of Cox regression, and a nomogram was subsequently constructed. The nomogram was compared to the Sequential Organ Failure Assessment (SOFA) model using the C-index, area under the receiver operating characteristic curve (AUC) over time, Net reclassification index (NRI), Integrated discrimination improvement index (IDI), calibration map, and decision curve analysis (DCA). Result A total of 800 patients were included in the study. We developed a nomogram based on age, diastolic blood pressure (DBP), pH, lactate, red blood cell distribution width (RDW), anion gap, valvular heart disease, peripheral vascular disease, and acute kidney injury (AKI) stage. The nomogram was evaluated using C-index, AUC, NRI, IDI, calibration plot, and DCA. Our findings revealed that this nomogram outperformed the SOFA score in predicting 28-day mortality in sepsis patients.
Collapse
Affiliation(s)
- Quankuan Gu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Ping Huang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Qiuyue Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Xianglin Meng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
10
|
Yang Z, Gao Y, Zhao L, Lv X, Du Y. Molecular mechanisms of Sepsis attacking the immune system and solid organs. Front Med (Lausanne) 2024; 11:1429370. [PMID: 39267971 PMCID: PMC11390691 DOI: 10.3389/fmed.2024.1429370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Remarkable progress has been achieved in sepsis treatment in recent times, the mortality rate of sepsis has experienced a gradual decline as a result of the prompt administration of antibiotics, fluid resuscitation, and the implementation of various therapies aimed at supporting multiple organ functions. However, there is still significant mortality and room for improvement. The mortality rate for septic patients, 22.5%, is still unacceptably high, accounting for 19.7% of all global deaths. Therefore, it is crucial to thoroughly comprehend the pathogenesis of sepsis in order to enhance clinical diagnosis and treatment methods. Here, we summarized classic mechanisms of sepsis progression, activation of signal pathways, mitochondrial quality control, imbalance of pro-and anti- inflammation response, diseminated intravascular coagulation (DIC), cell death, presented the latest research findings for each mechanism and identify potential therapeutic targets within each mechanism.
Collapse
Affiliation(s)
- Zhaoyun Yang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yan Gao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xuejiao Lv
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yanwei Du
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
11
|
Baljinnyam T, Fukuda S, Niimi Y, Prough D, Enkhbaatar P. Combined treatment with vitamin C, hydrocortisone and thiamine does not attenuate morbidity and mortality of septic sheep. Lab Anim Res 2024; 40:27. [PMID: 39135077 PMCID: PMC11318330 DOI: 10.1186/s42826-024-00213-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/01/2024] [Accepted: 07/14/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Sepsis is associated with a highest mortality rate in the ICU. Present study tests the efficacy of combined therapy with vitamin C, hydrocortisone and thiamine (combined therapy) in the ovine model of sepsis induced by Pseudomonas aeruginosa. In this study, sepsis was induced in sheep by instillation of Pseudomonas aeruginosa (1 × 1011 CFU) into the lungs via bronchoscope, under anesthesia. Nine hours after injury, intravenous infusion of vitamin C (0.75 g every 6 h), hydrocortisone (25 mg every 6 h), and thiamine (100 mg every 12 h) or saline was given to the treatment and control groups. Cardiopulmonary variables were recorded. RESULTS The survival rate was 16.7% in control and 33.3% in treatment groups. In the control group, mean arterial pressure dropped from 93.6 ± 8.6 to 75.5 ± 9.7 mmHg by 9 h, which was not affected by the combined therapy. Pulmonary dysfunction was not attenuated by the combined therapy either. The combined therapy had no effect on increased extravascular lung water content and fluid effusion into thoracic cavity. The bacterial number in the bronchoalveolar lavage fluid was significantly increased in the treatment group than the control group. The blood bacterial number remained comparable between groups. CONCLUSIONS Combined vitamin C, hydrocortisone, and thiamine did not attenuate severity of ovine sepsis.
Collapse
Affiliation(s)
- Tuvshintugs Baljinnyam
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA.
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA.
| | - Satoshi Fukuda
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Yosuke Niimi
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Donald Prough
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
12
|
Robey RC, Logue C, Caird CA, Hansel J, Hellyer TP, Simpson J, Dark P, Mathioudakis AG, Felton T. Immunomodulatory drugs in sepsis: a systematic review and meta-analysis. Anaesthesia 2024; 79:869-879. [PMID: 38523060 DOI: 10.1111/anae.16263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 03/26/2024]
Abstract
Dysregulation of the host immune response has a central role in the pathophysiology of sepsis. There has been much interest in immunomodulatory drugs as potential therapeutic adjuncts in sepsis. We conducted a systematic review and meta-analysis of randomised controlled trials evaluating the safety and clinical effectiveness of immunomodulatory drugs as adjuncts to standard care in the treatment of adults with sepsis. Our primary outcomes were serious adverse events and all-cause mortality. Fifty-six unique, eligible randomised controlled trials were identified, assessing a range of interventions including cytokine inhibitors; anti-inflammatories; immune cell stimulators; platelet pathway inhibitors; and complement inhibitors. At 1-month follow-up, the use of cytokine inhibitors was associated with a decreased risk of serious adverse events, based on 11 studies involving 7138 patients (RR (95%CI) 0.95 (0.90-1.00), I2 = 0%). The only immunomodulatory drugs associated with an increased risk of serious adverse events were toll-like receptor 4 antagonists (RR (95%CI) 1.18 (1.04-1.34), I2 = 0% (two trials, 567 patients)). Based on 18 randomised controlled trials, involving 11,075 patients, cytokine inhibitors reduced 1-month mortality (RR (95%CI) 0.88 (0.78-0.98), I2 = 57%). Mortality reduction was also shown in the subgroup of 13 randomised controlled trials that evaluated anti-tumour necrosis factor α interventions (RR (95%CI) 0.93 (0.87-0.99), I2 = 0%). Anti-inflammatory drugs had the largest apparent effect on mortality at 2 months at any dose (two trials, 228 patients, RR (95%CI) 0.64 (0.51-0.80), I2 = 0%) and at 3 months at any dose (three trials involving 277 patients, RR (95%CI) 0.67 (0.55-0.81), I2 = 0%). These data indicate that, except for toll-like receptor 4 antagonists, there is no evidence of safety concerns for the use of immunomodulatory drugs in sepsis, and they may show some short-term mortality benefit for selected drugs.
Collapse
Affiliation(s)
- R C Robey
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - C Logue
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - C A Caird
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - J Hansel
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - T P Hellyer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - J Simpson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - P Dark
- University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Critical Care Unit, Northern Care Alliance NHS Foundation Trust, Salford Care Organisation, Salford, UK
| | - A G Mathioudakis
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - T Felton
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
13
|
Soltan Dallal MM, Siavashi M, Karimaei S, Siavashi V, Abdi M, Yaseri M, Razavi SA, Bakhtiari R. The effect of thyme essential oil and endothelial progenitor stem cells on lipopolysaccharide-induced sepsis in C57BL/6 mice. Biotechnol Appl Biochem 2024; 71:835-848. [PMID: 38515313 DOI: 10.1002/bab.2580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 03/02/2024] [Indexed: 03/23/2024]
Abstract
Sepsis is a potentially fatal syndrome related to severe systemic inflammation developed by infection. Despite different antimicrobial therapies, morbidity and mortality rates remain high. Herbs along with cell therapy have been introduced as a promising option to improve the symptoms of sepsis. The present study aimed to evaluate the therapeutic effect of simultaneous administration of thyme essential oil (TEO) and endothelial progenitor stem cells (EPCs) on lipopolysaccharide (LPS)-induced sepsis in C57BL/6 mice. Sepsis was induced in C57Bl/6J mice by intraperitoneal injection of LPS, followed 2 h later by an intravenous injection of EPCs or oral administration of TEO or simultaneous administration of TEO and EPCs. After 10 days, the complete blood cell, renal and liver factors, serum levels of inflammatory cytokines, and angiogenic factors were measured. Simultaneous treatment with EPCs and TEO significantly increased the survival of mice with sepsis and modulated the inflammatory response by reducing the serum levels of pro-inflammatory cytokines. Moreover, this treatment significantly reduced the level of white blood cells and neutrophils and increased the number of red blood cells, the percentage of hematocrit, and hemoglobin. The combination of TEO with EPCs decreased organ injuries and was assessed by lower levels of the liver enzymes alanine aminotransferase and aspartate aminotransferase compared to the sepsis group. Administration of EPCs and TEO also significantly improved angiogenic factors, lung function, and toll-like receptor 4 expression. EPCs in combination with TEO increase survival in the LPS-induced sepsis mice model by acting on several targets. Thus, the combination of TEO with EPCs can be a feasible approach for the future clinical treatment and control of sepsis.
Collapse
Affiliation(s)
- Mohammad Mehdi Soltan Dallal
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Siavashi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Karimaei
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Siavashi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Milad Abdi
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Yaseri
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Alireza Razavi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ronak Bakhtiari
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
La Via L, Sangiorgio G, Stefani S, Marino A, Nunnari G, Cocuzza S, La Mantia I, Cacopardo B, Stracquadanio S, Spampinato S, Lavalle S, Maniaci A. The Global Burden of Sepsis and Septic Shock. EPIDEMIOLOGIA 2024; 5:456-478. [PMID: 39189251 PMCID: PMC11348270 DOI: 10.3390/epidemiologia5030032] [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: 06/13/2024] [Revised: 07/07/2024] [Accepted: 07/18/2024] [Indexed: 08/28/2024] Open
Abstract
A dysregulated host response to infection causes organ dysfunction in sepsis and septic shock, two potentially fatal diseases. They continue to be major worldwide health burdens with high rates of morbidity and mortality despite advancements in medical care. The goal of this thorough review was to present a thorough summary of the current body of knowledge about the prevalence of sepsis and septic shock worldwide. Using widely used computerized databases, a comprehensive search of the literature was carried out, and relevant studies were chosen in accordance with predetermined inclusion and exclusion criteria. A narrative technique was used to synthesize the data that were retrieved. The review's conclusions show how widely different locations and nations differ in terms of sepsis and septic shock's incidence, prevalence, and fatality rates. Compared to high-income countries (HICs), low- and middle-income countries (LMICs) are disproportionately burdened more heavily. We talk about risk factors, comorbidities, and difficulties in clinical management and diagnosis in a range of healthcare settings. The review highlights the need for more research, enhanced awareness, and context-specific interventions in order to successfully address the global burden of sepsis and septic shock.
Collapse
Affiliation(s)
- Luigi La Via
- Department of Anaesthesia and Intensive Care, University Hospital Policlinico “G. Rodolico-San Marco”, 24046 Catania, Italy
| | - Giuseppe Sangiorgio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (G.S.); (S.S.); (S.S.)
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (G.S.); (S.S.); (S.S.)
| | - Andrea Marino
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (A.M.); (G.N.); (B.C.); (S.S.)
| | - Giuseppe Nunnari
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (A.M.); (G.N.); (B.C.); (S.S.)
| | - Salvatore Cocuzza
- Department of Medical, Surgical Sciences and Advanced Technologies “GF Ingrassia” ENT Section, University of Catania, 95123 Catania, Italy; (S.C.); (I.L.M.)
| | - Ignazio La Mantia
- Department of Medical, Surgical Sciences and Advanced Technologies “GF Ingrassia” ENT Section, University of Catania, 95123 Catania, Italy; (S.C.); (I.L.M.)
| | - Bruno Cacopardo
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (A.M.); (G.N.); (B.C.); (S.S.)
| | - Stefano Stracquadanio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (G.S.); (S.S.); (S.S.)
| | - Serena Spampinato
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (A.M.); (G.N.); (B.C.); (S.S.)
| | - Salvatore Lavalle
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (S.L.); (A.M.)
| | - Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (S.L.); (A.M.)
| |
Collapse
|
15
|
Atreya MR, Huang M, Moore AR, Zheng H, Hasin-Brumshtein Y, Fitzgerald JC, Weiss SL, Cvijanovich NZ, Bigham MT, Jain PN, Schwarz AJ, Lutfi R, Nowak J, Thomas NJ, Quasney M, Dahmer MK, Baines T, Haileselassie B, Lautz AJ, Stanski NL, Standage SW, Kaplan JM, Zingarelli B, Sahay R, Zhang B, Sweeney TE, Khatri P, Sanchez-Pinto LN, Kamaleswaran R. Identification and transcriptomic assessment of latent profile pediatric septic shock phenotypes. Crit Care 2024; 28:246. [PMID: 39014377 PMCID: PMC11253460 DOI: 10.1186/s13054-024-05020-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Sepsis poses a grave threat, especially among children, but treatments are limited owing to heterogeneity among patients. We sought to test the clinical and biological relevance of pediatric septic shock subclasses identified using reproducible approaches. METHODS We performed latent profile analyses using clinical, laboratory, and biomarker data from a prospective multi-center pediatric septic shock observational cohort to derive phenotypes and trained a support vector machine model to assign phenotypes in an internal validation set. We established the clinical relevance of phenotypes and tested for their interaction with common sepsis treatments on patient outcomes. We conducted transcriptomic analyses to delineate phenotype-specific biology and inferred underlying cell subpopulations. Finally, we compared whether latent profile phenotypes overlapped with established gene-expression endotypes and compared survival among patients based on an integrated subclassification scheme. RESULTS Among 1071 pediatric septic shock patients requiring vasoactive support on day 1 included, we identified two phenotypes which we designated as Phenotype 1 (19.5%) and Phenotype 2 (80.5%). Membership in Phenotype 1 was associated with ~ fourfold adjusted odds of complicated course relative to Phenotype 2. Patients belonging to Phenotype 1 were characterized by relatively higher Angiopoietin-2/Tie-2 ratio, Angiopoietin-2, soluble thrombomodulin (sTM), interleukin 8 (IL-8), and intercellular adhesion molecule 1 (ICAM-1) and lower Tie-2 and Angiopoietin-1 concentrations compared to Phenotype 2. We did not identify significant interactions between phenotypes, common treatments, and clinical outcomes. Transcriptomic analysis revealed overexpression of genes implicated in the innate immune response and driven primarily by developing neutrophils among patients designated as Phenotype 1. There was no statistically significant overlap between established gene-expression endotypes, reflective of the host adaptive response, and the newly derived phenotypes, reflective of the host innate response including microvascular endothelial dysfunction. However, an integrated subclassification scheme demonstrated varying survival probabilities when comparing patient endophenotypes. CONCLUSIONS Our research underscores the reproducibility of latent profile analyses to identify pediatric septic shock phenotypes with high prognostic relevance. Pending validation, an integrated subclassification scheme, reflective of the different facets of the host response, holds promise to inform targeted intervention among those critically ill.
Collapse
Affiliation(s)
- Mihir R Atreya
- Division of Critical Care Medicine, MLC2005, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA.
| | - Min Huang
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew R Moore
- Stanford Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Hong Zheng
- Stanford Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | | | - Scott L Weiss
- Nemours Children's Health, Wilmington, DE, 19803, USA
| | | | | | - Parag N Jain
- Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Adam J Schwarz
- Children's Hospital of Orange County, Orange, CA, 92868, USA
| | - Riad Lutfi
- Riley Hospital for Children, Indianapolis, IN, 46202, USA
| | - Jeffrey Nowak
- Children's Hospital and Clinics of Minnesota, Minneapolis, MN, 55404, USA
| | - Neal J Thomas
- Penn State Hershey Children's Hospital, Hershey, PA, 17033, USA
| | - Michael Quasney
- C.S Mott Children's Hospital, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mary K Dahmer
- C.S Mott Children's Hospital, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Torrey Baines
- University of Florida Health Children's Hospital, Gainesville, FL, 32610, USA
| | | | - Andrew J Lautz
- Division of Critical Care Medicine, MLC2005, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Natalja L Stanski
- Division of Critical Care Medicine, MLC2005, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Stephen W Standage
- Division of Critical Care Medicine, MLC2005, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Jennifer M Kaplan
- Division of Critical Care Medicine, MLC2005, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Basilia Zingarelli
- Division of Critical Care Medicine, MLC2005, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Rashmi Sahay
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Bin Zhang
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | | | - Purvesh Khatri
- Stanford Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - L Nelson Sanchez-Pinto
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Health and Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Rishikesan Kamaleswaran
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
| |
Collapse
|
16
|
Bodinier M, Peronnet E, Llitjos JF, Kreitmann L, Brengel-Pesce K, Rimmelé T, Fleurie A, Textoris J, Venet F, Maucort-Boulch D, Monneret G. Integrated clustering of multiple immune marker trajectories reveals different immunotypes in severely injured patients. Crit Care 2024; 28:240. [PMID: 39010113 PMCID: PMC11247757 DOI: 10.1186/s13054-024-04990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND The immune response of critically ill patients, such as those with sepsis, severe trauma, or major surgery, is heterogeneous and dynamic, but its characterization and impact on outcomes are poorly understood. Until now, the primary challenge in advancing our understanding of the disease has been to concurrently address both multiparametric and temporal aspects. METHODS We used a clustering method to identify distinct groups of patients, based on various immune marker trajectories during the first week after admission to ICU. In 339 severely injured patients, we initially longitudinally clustered common biomarkers (both soluble and cellular parameters), whose variations are well-established during the immunosuppressive phase of sepsis. We then applied this multi-trajectory clustering using markers composed of whole blood immune-related mRNA. RESULTS We found that both sets of markers revealed two immunotypes, one of which was associated with worse outcomes, such as increased risk of hospital-acquired infection and mortality, and prolonged hospital stays. This immunotype showed signs of both hyperinflammation and immunosuppression, which persisted over time. CONCLUSION Our study suggest that the immune system of critically ill patients can be characterized by two distinct longitudinal immunotypes, one of which included patients with a persistently dysregulated and impaired immune response. This work confirms the relevance of such methodology to stratify patients and pave the way for further studies using markers indicative of potential immunomodulatory drug targets.
Collapse
Affiliation(s)
- Maxime Bodinier
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
| | - Estelle Peronnet
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
| | - Jean-François Llitjos
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
- Anesthesiology and Critical Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, 69003, Lyon, France
| | - Louis Kreitmann
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, W12 0NN, UK
| | - Karen Brengel-Pesce
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
| | - Thomas Rimmelé
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
- Anesthesiology and Critical Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, 69003, Lyon, France
| | - Aurore Fleurie
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
| | - Julien Textoris
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
- Anesthesiology and Critical Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, 69003, Lyon, France
| | - Fabienne Venet
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude, Bernard-Lyon 1, Lyon, France
| | - Delphine Maucort-Boulch
- Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- Équipe Biostatistique-Santé, Laboratoire de Biométrie Et Biologie Évolutive, CNRS UMR 5558, Villeurbanne, France
- Service de Biostatistique-Bioinformatique, Pôle Santé Publique, Hospices Civils de Lyon, Lyon, France
| | - Guillaume Monneret
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Immunology Laboratory and Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 place d'Arsonval, 69003, Lyon Cedex 03, France.
| |
Collapse
|
17
|
Burnham KL, Milind N, Lee W, Kwok AJ, Cano-Gamez K, Mi Y, Geoghegan CG, Zhang P, McKechnie S, Soranzo N, Hinds CJ, Knight JC, Davenport EE. eQTLs identify regulatory networks and drivers of variation in the individual response to sepsis. CELL GENOMICS 2024; 4:100587. [PMID: 38897207 PMCID: PMC11293594 DOI: 10.1016/j.xgen.2024.100587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
Sepsis is a clinical syndrome of life-threatening organ dysfunction caused by a dysregulated response to infection, for which disease heterogeneity is a major obstacle to developing targeted treatments. We have previously identified gene-expression-based patient subgroups (sepsis response signatures [SRS]) informative for outcome and underlying pathophysiology. Here, we aimed to investigate the role of genetic variation in determining the host transcriptomic response and to delineate regulatory networks underlying SRS. Using genotyping and RNA-sequencing data on 638 adult sepsis patients, we report 16,049 independent expression (eQTLs) and 32 co-expression module (modQTLs) quantitative trait loci in this disease context. We identified significant interactions between SRS and genotype for 1,578 SNP-gene pairs and combined transcription factor (TF) binding site information (SNP2TFBS) and predicted regulon activity (DoRothEA) to identify candidate upstream regulators. Overall, these approaches identified putative mechanistic links between host genetic variation, cell subtypes, and the individual transcriptomic response to infection.
Collapse
Affiliation(s)
- Katie L Burnham
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nikhil Milind
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK; University of Cambridge, Cambridge, UK
| | - Wanseon Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Andrew J Kwok
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kiki Cano-Gamez
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Yuxin Mi
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Ping Zhang
- Centre for Human Genetics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford, UK
| | | | - Nicole Soranzo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Charles J Hinds
- Centre for Translational Medicine & Therapeutics, William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Julian C Knight
- Centre for Human Genetics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford, UK.
| | | |
Collapse
|
18
|
Kim MJ, Choi EJ, Choi EJ. Evolving Paradigms in Sepsis Management: A Narrative Review. Cells 2024; 13:1172. [PMID: 39056754 PMCID: PMC11274781 DOI: 10.3390/cells13141172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Sepsis, a condition characterized by life-threatening organ dysfunction due to a dysregulated host response to infection, significantly impacts global health, with mortality rates varying widely across regions. Traditional therapeutic strategies that target hyperinflammation and immunosuppression have largely failed to improve outcomes, underscoring the need for innovative approaches. This review examines the development of therapeutic agents for sepsis, with a focus on clinical trials addressing hyperinflammation and immunosuppression. It highlights the frequent failures of these trials, explores the underlying reasons, and outlines current research efforts aimed at bridging the gap between theoretical advancements and clinical applications. Although personalized medicine and phenotypic categorization present promising directions, this review emphasizes the importance of understanding the complex pathogenesis of sepsis and developing targeted, effective therapies to enhance patient outcomes. By addressing the multifaceted nature of sepsis, future research can pave the way for more precise and individualized treatment strategies, ultimately improving the management and prognosis of sepsis patients.
Collapse
Affiliation(s)
- Min-Ji Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea;
| | - Eun-Joo Choi
- Department of Anesthesiology and Pain Medicine, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea;
| | - Eun-Jung Choi
- Department of Anatomy, School of Medicine, Daegu Catholic University, Duryugongwon-ro 17gil, Nam-gu, Daegu 42472, Republic of Korea
| |
Collapse
|
19
|
Lindell RB, Sayed S, Campos JS, Knight M, Mauracher AA, Hay CA, Conrey PE, Fitzgerald JC, Yehya N, Famularo ST, Arroyo T, Tustin R, Fazelinia H, Behrens EM, Teachey DT, Freeman AF, Bergerson JRE, Holland SM, Leiding JW, Weiss SL, Hall MW, Zuppa AF, Taylor DM, Feng R, Wherry EJ, Meyer NJ, Henrickson SE. Dysregulated STAT3 signaling and T cell immunometabolic dysfunction define a targetable, high mortality subphenotype of critically ill children. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.11.24308709. [PMID: 38946991 PMCID: PMC11213094 DOI: 10.1101/2024.06.11.24308709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Sepsis is the leading cause of death of hospitalized children worldwide. Despite the established link between immune dysregulation and mortality in pediatric sepsis, it remains unclear which host immune factors contribute causally to adverse sepsis outcomes. Identifying modifiable pathobiology is an essential first step to successful translation of biologic insights into precision therapeutics. We designed a prospective, longitudinal cohort study of 88 critically ill pediatric patients with multiple organ dysfunction syndrome (MODS), including patients with and without sepsis, to define subphenotypes associated with targetable mechanisms of immune dysregulation. We first assessed plasma proteomic profiles and identified shared features of immune dysregulation in MODS patients with and without sepsis. We then employed consensus clustering to define three subphenotypes based on protein expression at disease onset and identified a strong association between subphenotype and clinical outcome. We next identified differences in immune cell frequency and activation state by MODS subphenotype and determined the association between hyperinflammatory pathway activation and cellular immunophenotype. Using single cell transcriptomics, we demonstrated STAT3 hyperactivation in lymphocytes from the sickest MODS subgroup and then identified an association between STAT3 hyperactivation and T cell immunometabolic dysregulation. Finally, we compared proteomics findings between patients with MODS and patients with inborn errors of immunity that amplify cytokine signaling pathways to further assess the impact of STAT3 hyperactivation in the most severe patients with MODS. Overall, these results identify a potentially pathologic and targetable role for STAT3 hyperactivation in a subset of pediatric patients with MODS who have high severity of illness and poor prognosis.
Collapse
|
20
|
Mi Y, Burnham KL, Charles PD, Heilig R, Vendrell I, Whalley J, Torrance HD, Antcliffe DB, May SM, Neville MJ, Berridge G, Hutton P, Geoghegan CG, Radhakrishnan J, Nesvizhskii AI, Yu F, Davenport EE, McKechnie S, Davies R, O'Callaghan DJP, Patel P, Del Arroyo AG, Karpe F, Gordon AC, Ackland GL, Hinds CJ, Fischer R, Knight JC. High-throughput mass spectrometry maps the sepsis plasma proteome and differences in patient response. Sci Transl Med 2024; 16:eadh0185. [PMID: 38838133 DOI: 10.1126/scitranslmed.adh0185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
Sepsis, the dysregulated host response to infection causing life-threatening organ dysfunction, is a global health challenge requiring better understanding of pathophysiology and new therapeutic approaches. Here, we applied high-throughput tandem mass spectrometry to delineate the plasma proteome for sepsis and comparator groups (noninfected critical illness, postoperative inflammation, and healthy volunteers) involving 2612 samples (from 1611 patients) and 4553 liquid chromatography-mass spectrometry analyses acquired through a single batch of continuous measurements, with a throughput of 100 samples per day. We show how this scale of data can delineate proteins, pathways, and coexpression modules in sepsis and be integrated with paired leukocyte transcriptomic data (837 samples from n = 649 patients). We mapped the plasma proteomic landscape of the host response in sepsis, including changes over time, and identified features relating to etiology, clinical phenotypes (including organ failures), and severity. This work reveals subphenotypes informative for sepsis response state, disease processes, and outcome; identifies potential biomarkers; and advances opportunities for a precision medicine approach to sepsis.
Collapse
Affiliation(s)
- Yuxin Mi
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Katie L Burnham
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Philip D Charles
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Raphael Heilig
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Iolanda Vendrell
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
- Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Justin Whalley
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Hew D Torrance
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College, London SW7 2AZ, UK
| | - David B Antcliffe
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College, London SW7 2AZ, UK
- Department of Critical Care, Imperial College Healthcare NHS Trust, London W2 1NY, UK
| | - Shaun M May
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Matt J Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Georgina Berridge
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Paula Hutton
- Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7JX, UK
| | - Cyndi G Geoghegan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Jayachandran Radhakrishnan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | | | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emma E Davenport
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Stuart McKechnie
- Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7JX, UK
| | - Roger Davies
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College, London SW7 2AZ, UK
| | - David J P O'Callaghan
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College, London SW7 2AZ, UK
- Department of Critical Care, Imperial College Healthcare NHS Trust, London W2 1NY, UK
| | - Parind Patel
- Department of Critical Care, Imperial College Healthcare NHS Trust, London W2 1NY, UK
| | - Ana G Del Arroyo
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Anthony C Gordon
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College, London SW7 2AZ, UK
- Department of Critical Care, Imperial College Healthcare NHS Trust, London W2 1NY, UK
| | - Gareth L Ackland
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Charles J Hinds
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
- Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
- NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| |
Collapse
|
21
|
Gray CC, Armstead BE, Chung CS, Chen Y, Ayala A. VISTA nonredundantly regulates proliferation and CD69low γδ T cell accumulation in the intestine in murine sepsis. J Leukoc Biol 2024; 115:1005-1019. [PMID: 38035776 PMCID: PMC11135620 DOI: 10.1093/jleuko/qiad149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/21/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023] Open
Abstract
Sepsis is a dysregulated systemic immune response to infection i.e. responsible for ∼35% of in-hospital deaths at a significant fiscal healthcare cost. Our laboratory, among others, has demonstrated the efficacy of targeting negative checkpoint regulators (NCRs) to improve survival in a murine model of sepsis, cecal ligation and puncture (CLP). B7-CD28 superfamily member, V-domain immunoglobulin suppressor of T cell activation (VISTA), is an ideal candidate for strategic targeting in sepsis. VISTA is a 35 to 45 kDa type 1 transmembrane protein with unique biology that sets it apart from all other NCRs. We recently reported that VISTA-/- mice had a significant survival deficit post-CLP, which was rescued upon adoptive transfer of a VISTA-expressing pMSCV-mouse Foxp3-EF1α-GFP-T2A-puro stable Jurkat cell line (Jurkatfoxp3 T cells). Based on our prior study, we investigated the effector cell target of Jurkatfoxp3 T cells in VISTA-/- mice. γδ T cells are a powerful lymphoid subpopulation that require regulatory fine-tuning by regulatory T cells to prevent overt inflammation/pathology. In this study, we hypothesized that Jurkatfoxp3 T cells nonredundantly modulate the γδ T cell population post-CLP. We found that VISTA-/- mice have an increased accumulation of intestinal CD69low γδ T cells, which are not protective in murine sepsis. Adoptive transfer of Jurkatfoxp3 T cells decreased the intestinal γδ T cell population, suppressed proliferation, skewed remaining γδ T cells toward a CD69high phenotype, and increased soluble CD40L in VISTA-/- mice post-CLP. These results support a potential regulatory mechanism by which VISTA skews intestinal γδ T cell lineage representation in murine sepsis.
Collapse
MESH Headings
- Animals
- Sepsis/immunology
- Mice
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Cell Proliferation
- Humans
- Lectins, C-Type/metabolism
- Lectins, C-Type/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Mice, Knockout
- Mice, Inbred C57BL
- Intestines/immunology
- Intestines/pathology
- Jurkat Cells
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
Collapse
Affiliation(s)
- Chyna C Gray
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, United States
| | - Brandon E Armstead
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, United States
- Pathobiology Graduate Program, Brown University, Box G-B495, Providence, RI 02912, United States
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, United States
| | - Yaping Chen
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, United States
- Pathobiology Graduate Program, Brown University, Box G-B495, Providence, RI 02912, United States
| |
Collapse
|
22
|
Stevens J, Tezel O, Bonnefil V, Hapstack M, Atreya MR. Biological basis of critical illness subclasses: from the bedside to the bench and back again. Crit Care 2024; 28:186. [PMID: 38812006 PMCID: PMC11137966 DOI: 10.1186/s13054-024-04959-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
Critical illness syndromes including sepsis, acute respiratory distress syndrome, and acute kidney injury (AKI) are associated with high in-hospital mortality and long-term adverse health outcomes among survivors. Despite advancements in care, clinical and biological heterogeneity among patients continues to hamper identification of efficacious therapies. Precision medicine offers hope by identifying patient subclasses based on clinical, laboratory, biomarker and 'omic' data and potentially facilitating better alignment of interventions. Within the previous two decades, numerous studies have made strides in identifying gene-expression based endotypes and clinico-biomarker based phenotypes among critically ill patients associated with differential outcomes and responses to treatment. In this state-of-the-art review, we summarize the biological similarities and differences across the various subclassification schemes among critically ill patients. In addition, we highlight current translational gaps, the need for advanced scientific tools, human-relevant disease models, to gain a comprehensive understanding of the molecular mechanisms underlying critical illness subclasses.
Collapse
Affiliation(s)
- Joseph Stevens
- Division of Immunobiology, Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Oğuzhan Tezel
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Valentina Bonnefil
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Matthew Hapstack
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Mihir R Atreya
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA.
| |
Collapse
|
23
|
Antcliffe DB, Mi Y, Santhakumaran S, Burnham KL, Prevost AT, Ward JK, Marshall TJ, Bradley C, Al-Beidh F, Hutton P, McKechnie S, Davenport EE, Hinds CJ, O'Kane CM, McAuley DF, Shankar-Hari M, Gordon AC, Knight JC. Patient stratification using plasma cytokines and their regulators in sepsis: relationship to outcomes, treatment effect and leucocyte transcriptomic subphenotypes. Thorax 2024; 79:515-523. [PMID: 38471792 PMCID: PMC11137467 DOI: 10.1136/thorax-2023-220538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 02/21/2024] [Indexed: 03/14/2024]
Abstract
RATIONALE Heterogeneity of the host response within sepsis, acute respiratory distress syndrome (ARDS) and more widely critical illness, limits discovery and targeting of immunomodulatory therapies. Clustering approaches using clinical and circulating biomarkers have defined hyper-inflammatory and hypo-inflammatory subphenotypes in ARDS associated with differential treatment response. It is unknown if similar subphenotypes exist in sepsis populations where leucocyte transcriptomic-defined subphenotypes have been reported. OBJECTIVES We investigated whether inflammatory clusters based on cytokine protein abundance were seen in sepsis, and the relationships with previously described transcriptomic subphenotypes. METHODS Hierarchical cluster and latent class analysis were applied to an observational study (UK Genomic Advances in Sepsis (GAinS)) (n=124 patients) and two clinical trial datasets (VANISH, n=155 and LeoPARDS, n=484) in which the plasma protein abundance of 65, 21, 11 circulating cytokines, cytokine receptors and regulators were quantified. Clinical features, outcomes, response to trial treatments and assignment to transcriptomic subphenotypes were compared between inflammatory clusters. MEASUREMENTS AND MAIN RESULTS We identified two (UK GAinS, VANISH) or three (LeoPARDS) inflammatory clusters. A group with high levels of pro-inflammatory and anti-inflammatory cytokines was seen that was associated with worse organ dysfunction and survival. No interaction between inflammatory clusters and trial treatment response was found. We found variable overlap of inflammatory clusters and leucocyte transcriptomic subphenotypes. CONCLUSIONS These findings demonstrate that differences in response at the level of cytokine biology show clustering related to severity, but not treatment response, and may provide complementary information to transcriptomic sepsis subphenotypes. TRIAL REGISTRATION NUMBER ISRCTN20769191, ISRCTN12776039.
Collapse
Affiliation(s)
- David Benjamin Antcliffe
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Centre for Perioperative and Critical Care Research, Imperial College Healthcare NHS Trust, London, UK
| | - Yuxin Mi
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Shalini Santhakumaran
- Imperial Clinical Trials Unit, School of Public Health, Imperial College London, London, UK
| | - Katie L Burnham
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - A Toby Prevost
- Nightingale-Saunders Clinical Trials and Epidemiology Unit, King's College London, London, UK
| | - Josie K Ward
- Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Timothy J Marshall
- Department of Anaesthetics, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Central Clinical School Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Claire Bradley
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Farah Al-Beidh
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Paula Hutton
- Adult Intensive Care Unit, John Radcliffe Hospital, Oxford, UK
| | | | | | - Charles J Hinds
- William Harvey Research Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Cecilia M O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Daniel Francis McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
- Regional Intensive Care Unit, Royal Victoria Hospital, Belfast, UK
- Northern Ireland Clinical Trials Unit, Royal Hospitals, Belfast, UK
| | - Manu Shankar-Hari
- The Queen's Medical Research Institute, The University of Edinburgh College of Medicine and Veterinary Medicine, Edinburgh, UK
- Intensive Care Unit, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Anthony C Gordon
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Centre for Perioperative and Critical Care Research, Imperial College Healthcare NHS Trust, London, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Yin S, Peng Y, Lin Y, Wu H, Wang B, Wang X, Chen W, Liu T, Peng H, Li X, Xu J, Wang M. Bacterial heat shock protein: A new crosstalk between T lymphocyte and macrophage via JAK2/STAT1 pathway in bloodstream infection. Microbiol Res 2024; 282:127626. [PMID: 38330817 DOI: 10.1016/j.micres.2024.127626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024]
Abstract
Bloodstream infection (BSI) refers to the infection of blood by pathogens. Severe immune response to BSI can lead to sepsis, a systemic infection leading to multiple organ dysfunction, coupled with drug resistance, mortality, and limited clinical treatment options. This work aims to further investigate the new interplay between bacterial exocrine regulatory protein and host immune cells in the context of highly drug-resistant malignant BSI. Whether interfering with related regulatory signaling pathways can reverse the inflammatory disorder of immune cells. In-depth analysis of single-cell sequencing results in Septic patients for potential immunodeficiency factors. Analysis of key proteins enriched by host cells and key pathways using proteomics. Cell models and animal models validate the pathological effects of DnaK on T cells, MAITs, macrophages, and osteoclasts. The blood of patients was analyzed for the immunosuppression of T cells and MAITs. We identified that S. maltophilia-DnaK was enriched in immunodeficient T cells. The activation of the JAK2/STAT1 axis initiated the exhaustion of T cells. Septic patients with Gram-negative bacterial infections exhibited deficiencies in MAITs, which correspond to IFN-γ. Cellular and animal experiments confirmed that DnaK could facilitate MAIT depletion and M1 polarization of macrophages. Additionally, Fludarabine mitigated M1 polarization of blood, liver, and spleen in mice. Interestingly, DnaK also repressed osteoclastogenesis of macrophages stimulated by RANKL. S.maltophilia-DnaK prompts the activation of the JAK2/STAT1 axis in T cells and the M1 polarization of macrophages. Targeting the DnaK's crosstalk can be a potentially effective approach for treating the inflammatory disorder in the broad-spectrum drug-resistant BSI.
Collapse
Affiliation(s)
- Sheng Yin
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Yizhi Peng
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Laboratory Medicine, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan 410031, China
| | - YingRui Lin
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Hongzheng Wu
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Bingqi Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaofan Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Wanxin Chen
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Tianyao Liu
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Huanqie Peng
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xianping Li
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Min Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
25
|
Catling FJR, Nagendran M, Festor P, Bien Z, Harris S, Faisal AA, Gordon AC, Komorowski M. Can Machine Learning Personalize Cardiovascular Therapy in Sepsis? Crit Care Explor 2024; 6:e1087. [PMID: 38709088 DOI: 10.1097/cce.0000000000001087] [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: 05/07/2024] Open
Abstract
Large randomized trials in sepsis have generally failed to find effective novel treatments. This is increasingly attributed to patient heterogeneity, including heterogeneous cardiovascular changes in septic shock. We discuss the potential for machine learning systems to personalize cardiovascular resuscitation in sepsis. While the literature is replete with proofs of concept, the technological readiness of current systems is low, with a paucity of clinical trials and proven patient benefit. Systems may be vulnerable to confounding and poor generalization to new patient populations or contemporary patterns of care. Typical electronic health records do not capture rich enough data, at sufficient temporal resolution, to produce systems that make actionable treatment suggestions. To resolve these issues, we recommend a simultaneous focus on technical challenges and removing barriers to translation. This will involve improving data quality, adopting causally grounded models, prioritizing safety assessment and integration into healthcare workflows, conducting randomized clinical trials and aligning with regulatory requirements.
Collapse
Affiliation(s)
- Finneas J R Catling
- Institute of Healthcare Engineering, University College London, London, United Kingdom
- Division of Anaesthetics, Pain Medicine, and Intensive Care, Imperial College London, London, United Kingdom
| | - Myura Nagendran
- Division of Anaesthetics, Pain Medicine, and Intensive Care, Imperial College London, London, United Kingdom
- UKRI Centre for Doctoral Training in AI for Healthcare, Imperial College London, London, United Kingdom
| | - Paul Festor
- UKRI Centre for Doctoral Training in AI for Healthcare, Imperial College London, London, United Kingdom
- Department of Computing, Imperial College London, London, United Kingdom
| | - Zuzanna Bien
- School of Life Course & Population Sciences, King's College London, United Kingdom
| | - Steve Harris
- Department of Critical Care, University College London Hospital, London, United Kingdom
- Institute of Health Informatics, University College London, London, United Kingdom
| | - A Aldo Faisal
- UKRI Centre for Doctoral Training in AI for Healthcare, Imperial College London, London, United Kingdom
- Department of Computing, Imperial College London, London, United Kingdom
- Institute of Artificial and Human Intelligence, Universität Bayreuth, Bayreuth, Germany
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Anthony C Gordon
- Division of Anaesthetics, Pain Medicine, and Intensive Care, Imperial College London, London, United Kingdom
| | - Matthieu Komorowski
- Division of Anaesthetics, Pain Medicine, and Intensive Care, Imperial College London, London, United Kingdom
| |
Collapse
|
26
|
Qiao X, Yin J, Zheng Z, Li L, Feng X. Endothelial cell dynamics in sepsis-induced acute lung injury and acute respiratory distress syndrome: pathogenesis and therapeutic implications. Cell Commun Signal 2024; 22:241. [PMID: 38664775 PMCID: PMC11046830 DOI: 10.1186/s12964-024-01620-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis, a prevalent critical condition in clinics, continues to be the leading cause of death from infections and a global healthcare issue. Among the organs susceptible to the harmful effects of sepsis, the lungs are notably the most frequently affected. Consequently, patients with sepsis are predisposed to developing acute lung injury (ALI), and in severe cases, acute respiratory distress syndrome (ARDS). Nevertheless, the precise mechanisms associated with the onset of ALI/ARDS remain elusive. In recent years, there has been a growing emphasis on the role of endothelial cells (ECs), a cell type integral to lung barrier function, and their interactions with various stromal cells in sepsis-induced ALI/ARDS. In this comprehensive review, we summarize the involvement of endothelial cells and their intricate interplay with immune cells and stromal cells, including pulmonary epithelial cells and fibroblasts, in the pathogenesis of sepsis-induced ALI/ARDS, with particular emphasis placed on discussing the several pivotal pathways implicated in this process. Furthermore, we discuss the potential therapeutic interventions for modulating the functions of endothelial cells, their interactions with immune cells and stromal cells, and relevant pathways associated with ALI/ARDS to present a potential therapeutic strategy for managing sepsis and sepsis-induced ALI/ARDS.
Collapse
Affiliation(s)
- Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Liangge Li
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
27
|
Khan N, Kumar V, Li P, Schlapbach LJ, Boyd AW, Coulthard MG, Woodruff TM. Inhibiting Eph/ephrin signaling reduces vascular leak and endothelial cell dysfunction in mice with sepsis. Sci Transl Med 2024; 16:eadg5768. [PMID: 38657024 DOI: 10.1126/scitranslmed.adg5768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
Sepsis is a life-threatening disease caused by a dysregulated host response to infection, resulting in 11 million deaths globally each year. Vascular endothelial cell dysfunction results in the loss of endothelial barrier integrity, which contributes to sepsis-induced multiple organ failure and mortality. Erythropoietin-producing hepatocellular carcinoma (Eph) receptors and their ephrin ligands play a key role in vascular endothelial barrier disruption but are currently not a therapeutic target in sepsis. Using a cecal ligation and puncture (CLP) mouse model of sepsis, we showed that prophylactic or therapeutic treatment of mice with EphA4-Fc, a decoy receptor and pan-ephrin inhibitor, resulted in improved survival and a reduction in vascular leak, lung injury, and endothelial cell dysfunction. EphA2-/- mice also exhibited reduced mortality and pathology after CLP compared with wild-type mice. Proteomics of plasma samples from mice with sepsis after CLP revealed dysregulation of a number of Eph/ephrins, including EphA2/ephrin A1. Administration of EphA4-Fc to cultured human endothelial cells pretreated with TNF-α or ephrin-A1 prevented loss of endothelial junction proteins, specifically VE-cadherin, with maintenance of endothelial barrier integrity. In children admitted to hospital with fever and suspected infection, we observed that changes in EphA2/ephrin A1 in serum samples correlated with endothelial and organ dysfunction. Targeting Eph/ephrin signaling may be a potential therapeutic strategy to reduce sepsis-induced endothelial dysfunction and mortality.
Collapse
Affiliation(s)
- Nemat Khan
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| | - Pengcheng Li
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Luregn J Schlapbach
- Children's Intensive Care Research Program, Child Health Research Centre, University of Queensland, Brisbane, QLD 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Department of Intensive Care and Neonatology, and Children's Research Center, University Children's Hospital Zürich, University of Zürich, 8032 Zürich, Switzerland
| | - Andrew W Boyd
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia
| | - Mark G Coulthard
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
28
|
Thomas-Rüddel D, Bauer M, Moita LF, Helbig C, Schlattmann P, Ehler J, Rahmel T, Meybohm P, Gründling M, Schenk H, Köcher T, Brunkhorst FM, Gräler M, Heger AJ, Weis S. Epirubicin for the Treatment of Sepsis and Septic Shock (EPOS-1): study protocol for a randomised, placebo-controlled phase IIa dose-escalation trial. BMJ Open 2024; 14:e075158. [PMID: 38653508 PMCID: PMC11043739 DOI: 10.1136/bmjopen-2023-075158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION Sepsis remains the major cause of death among hospitalised patients in intensive care. While targeting sepsis-causing pathogens with source control or antimicrobials has had a dramatic impact on morbidity and mortality of sepsis patients, this strategy remains insufficient for about one-third of the affected individuals who succumb. Pharmacological targeting of mechanisms that reduce sepsis-defining organ dysfunction may be beneficial. When given at low doses, the anthracycline epirubicin promotes tissue damage control and lessens the severity of sepsis independently of the host-pathogen load by conferring disease tolerance to infection. Since epirubicin at higher doses can be myelotoxic, a first dose-response trial is necessary to assess the potential harm of this drug in this new indication. METHODS AND ANALYSIS Epirubicin for the Treatment of Sepsis and Septic Shock-1 is a randomised, double-blind, placebo-controlled phase 2 dose-escalation phase IIa clinical trial to assess the safety of epirubicin as an adjunctive in patients with sepsis. The primary endpoint is the 14-day myelotoxicity. Secondary and explorative outcomes include 30-day and 90-day mortality, organ dysfunction, pharmacokinetic/pharmacodynamic (PK/PD) and cytokine release. Patients will be randomised in three consecutive phases. For each study phase, patients are randomised to one of the two study arms (epirubicin or placebo) in a 4:1 ratio. Approximately 45 patients will be recruited. Patients in the epirubicin group will receive a single dose of epirubicin (3.75, 7.5 or 15 mg/m2 depending on the study phase. After each study phase, a data and safety monitoring board will recommend continuation or premature stopping of the trial. The primary analyses for each dose level will report the proportion of myelotoxicity together with a 95% CI. A potential dose-toxicity association will be analysed using a logistic regression model with dose as a covariate. All further analyses will be descriptive. ETHICS AND DISSEMINATION The protocol is approved by the German Federal Institute for Drugs and Medical Devices. The results will be submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT05033808.
Collapse
Affiliation(s)
- Daniel Thomas-Rüddel
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | | | - Christiane Helbig
- Center for Clinical Studies, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Peter Schlattmann
- Institute of Medical Statistics, Computer Sciences, and Data Science, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Johannes Ehler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Tim Rahmel
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Patrick Meybohm
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Matthias Gründling
- Department of Anesthesiology, Greifswald, University Hospital of Greifswald, Greifswald, Germany
| | - Heiko Schenk
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Thomas Köcher
- Vienna BioCenter Core Facilities GmbH, Wien, Austria
| | - Frank M Brunkhorst
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Markus Gräler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Ann-Julika Heger
- Center for Clinical Studies, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
- Leibniz Institute for Infection Biology and Natural Products Research, Hans-Knöll Institute - HKI, Jena, Germany
| |
Collapse
|
29
|
Tang G, Luo Y, Song H, Liu W, Huang Y, Wang X, Zou S, Sun Z, Hou H, Wang F. The immune landscape of sepsis and using immune clusters for identifying sepsis endotypes. Front Immunol 2024; 15:1287415. [PMID: 38707899 PMCID: PMC11066285 DOI: 10.3389/fimmu.2024.1287415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
Background The dysregulated immune response to sepsis still remains unclear. Stratification of sepsis patients into endotypes based on immune indicators is important for the future development of personalized therapies. We aimed to evaluate the immune landscape of sepsis and the use of immune clusters for identifying sepsis endotypes. Methods The indicators involved in innate, cellular, and humoral immune cells, inhibitory immune cells, and cytokines were simultaneously assessed in 90 sepsis patients and 40 healthy controls. Unsupervised k-means cluster analysis of immune indicator data were used to identify patient clusters, and a random forest approach was used to build a prediction model for classifying sepsis endotypes. Results We depicted that the impairment of innate and adaptive immunity accompanying increased inflammation was the most prominent feature in patients with sepsis. However, using immune indicators for distinguishing sepsis from bacteremia was difficult, most likely due to the considerable heterogeneity in sepsis patients. Cluster analysis of sepsis patients identified three immune clusters with different survival rates. Cluster 1 (36.7%) could be distinguished from the other clusters as being an "effector-type" cluster, whereas cluster 2 (34.4%) was a "potential-type" cluster, and cluster 3 (28.9%) was a "dysregulation-type" cluster, which showed the lowest survival rate. In addition, we established a prediction model based on immune indicator data, which accurately classified sepsis patients into three immune endotypes. Conclusion We depicted the immune landscape of patients with sepsis and identified three distinct immune endotypes with different survival rates. Cluster membership could be predicted with a model based on immune data.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hongyan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Chen KL, Chou RH, Chang CC, Kuo CS, Wei JH, Huang PH, Lin SJ. The high-density lipoprotein cholesterol (HDL-C)-concentration-dependent association between anti-inflammatory capacity and sepsis: A single-center cross-sectional study. PLoS One 2024; 19:e0296863. [PMID: 38603717 PMCID: PMC11008828 DOI: 10.1371/journal.pone.0296863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/20/2023] [Indexed: 04/13/2024] Open
Abstract
INTRODUCTION Known to have pleiotropic functions, high-density lipoprotein (HDL) helps to regulate systemic inflammation during sepsis. As preserving HDL-C level is a promising therapeutic strategy for sepsis, the interaction between HDL and sepsis worth further investigation. This study aimed to determine the impact of sepsis on HDL's anti-inflammatory capacity and explore its correlations with disease severity and laboratory parameters. METHODS AND MATERIALS We enrolled 80 septic subjects admitted to the intensive care unit and 50 controls admitted for scheduled coronary angiography in this cross-sectional study. We used apolipoprotein-B depleted (apoB-depleted) plasma to measure the anti-inflammatory capacity of HDL-C. ApoB-depleted plasma's anti-inflammatory capacity is defined as its ability to suppress tumor necrosis factor-α-induced vascular cell adhesion molecule-1 (VCAM-1) expression in human umbilical-vein endothelial cells. A subgroup analysis was conducted to investigate in septic subjects according to disease severity. RESULTS ApoB-depleted plasma's anti-inflammatory capacity was reduced in septic subjects relative to controls (VCAM-1 mRNA fold change: 50.1% vs. 35.5%; p < 0.0001). The impairment was more pronounced in septic subjects with than in those without septic shock (55.8% vs. 45.3%, p = 0.0022). Both associations were rendered non-significant with the adjustment for the HDL-C level. In sepsis patients, VCAM-1 mRNA fold change correlated with the SOFA score (Spearman's r = 0.231, p = 0.039), lactate level (r = 0.297, p = 0.0074), HDL-C level (r = -0.370, p = 0.0007), and inflammatory markers (C-reactive protein level: r = 0.441, p <0.0001; white blood cell: r = 0.353, p = 0.0013). CONCLUSION ApoB-depleted plasma's anti-inflammatory capacity is reduced in sepsis patients and this association depends of HDL-C concentration. In sepsis patients, this capacity correlates with disease severity and inflammatory markers. These findings explain the prognostic role of the HDL-C level in sepsis and indirectly support the rationale for targeting HDL-C as sepsis treatment.
Collapse
Affiliation(s)
- Kai-Lee Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ruey-Hsing Chou
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Chin Chang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Sung Kuo
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jih-Hua Wei
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shing-Jong Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Healthcare and Services Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Heart Center, Cheng-Hsin General Hospital, Taipei, Taiwan
| |
Collapse
|
31
|
Merk D, Cox FF, Jakobs P, Prömel S, Altschmied J, Haendeler J. Dose-Dependent Effects of Lipopolysaccharide on the Endothelium-Sepsis versus Metabolic Endotoxemia-Induced Cellular Senescence. Antioxidants (Basel) 2024; 13:443. [PMID: 38671891 PMCID: PMC11047739 DOI: 10.3390/antiox13040443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The endothelium, the innermost cell layer of blood vessels, is not only a physical barrier between the bloodstream and the surrounding tissues but has also essential functions in vascular homeostasis. Therefore, it is not surprising that endothelial dysfunction is associated with most cardiovascular diseases. The functionality of the endothelium is compromised by endotoxemia, the presence of bacterial endotoxins in the bloodstream with the main endotoxin lipopolysaccharide (LPS). Therefore, this review will focus on the effects of LPS on the endothelium. Depending on the LPS concentration, the outcomes are either sepsis or, at lower concentrations, so-called low-dose or metabolic endotoxemia. Sepsis, a life-threatening condition evoked by hyperactivation of the immune response, includes breakdown of the endothelial barrier resulting in failure of multiple organs. A deeper understanding of the underlying mechanisms in the endothelium might help pave the way to new therapeutic options in sepsis treatment to prevent endothelial leakage and fatal septic shock. Low-dose endotoxemia or metabolic endotoxemia results in chronic inflammation leading to endothelial cell senescence, which entails endothelial dysfunction and thus plays a critical role in cardiovascular diseases. The identification of compounds counteracting senescence induction in endothelial cells might therefore help in delaying the onset or progression of age-related pathologies. Interestingly, two natural plant-derived substances, caffeine and curcumin, have shown potential in preventing endothelial cell senescence.
Collapse
Affiliation(s)
- Dennis Merk
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
| | - Fiona Frederike Cox
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
- Medical Faculty, Institute for Translational Pharmacology, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Philipp Jakobs
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
| | - Simone Prömel
- Department of Biology, Institute of Cell Biology, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Joachim Altschmied
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
- Medical Faculty, Cardiovascular Research Institute Düsseldorf, CARID, University Hospital and Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Judith Haendeler
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
- Medical Faculty, Cardiovascular Research Institute Düsseldorf, CARID, University Hospital and Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
32
|
Kumar NR, Balraj TA, Shivashankar KK, Jayaram TC, Prashant A. Inflammaging in Multidrug-Resistant Sepsis of Geriatric ICU Patients and Healthcare Challenges. Geriatrics (Basel) 2024; 9:45. [PMID: 38667512 PMCID: PMC11049875 DOI: 10.3390/geriatrics9020045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/08/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Multidrug-resistant sepsis (MDR) is a pressing concern in intensive care unit (ICU) settings, specifically among geriatric patients who experience age-related immune system changes and comorbidities. The aim of this review is to explore the clinical impact of MDR sepsis in geriatric ICU patients and shed light on healthcare challenges associated with its management. We conducted a comprehensive literature search using the National Center for Biotechnology Information (NCBI) and Google Scholar search engines. Our search incorporated keywords such as "multidrug-resistant sepsis" OR "MDR sepsis", "geriatric ICU patients" OR "elderly ICU patients", and "complications", "healthcare burdens", "diagnostic challenges", and "healthcare challenges" associated with MDR sepsis in "ICU patients" and "geriatric/elderly ICU patients". This review explores the specific risk factors contributing to MDR sepsis, the complexities of diagnostic challenges, and the healthcare burden faced by elderly ICU patients. Notably, the elderly population bears a higher burden of MDR sepsis (57.5%), influenced by various factors, including comorbidities, immunosuppression, age-related immune changes, and resource-limited ICU settings. Furthermore, sepsis imposes a significant economic burden on healthcare systems, with annual costs exceeding $27 billion in the USA. These findings underscore the urgency of addressing MDR sepsis in geriatric ICU patients and the need for tailored interventions to improve outcomes and reduce healthcare costs.
Collapse
Affiliation(s)
- Nishitha R. Kumar
- Department of Biochemistry, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysuru 570015, India; (N.R.K.); (K.K.S.)
| | - Tejashree A. Balraj
- Department of Microbiology, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysuru 570015, India;
| | - Kusuma K. Shivashankar
- Department of Biochemistry, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysuru 570015, India; (N.R.K.); (K.K.S.)
| | - Tejaswini C. Jayaram
- Department of Geriatrics, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysuru 570015, India;
| | - Akila Prashant
- Department of Biochemistry, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysuru 570015, India; (N.R.K.); (K.K.S.)
- Department of Medical Genetics, JSS Medical College and Hospital, JSS Academy of Higher Education & Research, Mysuru 570015, India
| |
Collapse
|
33
|
Wang Z, Guo Z, Wang X, Chen F, Wang Z, Wang Z. ASSESSING THE CAUSAL RELATIONSHIP BETWEEN SEPSIS AND AUTOIMMUNE: A MENDELIAN RANDOMIZATION STUDY. Shock 2024; 61:564-569. [PMID: 37856654 DOI: 10.1097/shk.0000000000002246] [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: 10/21/2023]
Abstract
ABSTRACT Objective : Numerous epidemiological studies have identified a potential link between sepsis and a variety of autoimmune disorders. The primary objective of this study is to delve deeper into this connection, investigating the potential causal relationship between sepsis and autoimmune disorders through the application of Mendelian randomization (MR). Methods : To assess the potential genetic impact on sepsis risk relating to susceptibility toward immune-related outcomes, we used summary data from the largest European genome-wide association studies (GWAS) on these conditions using a two-sample MR framework. Single nucleotide polymorphisms-which had strong associations with the nine traits-were extracted from the GWAS and examined their effects in an extensive European sepsis GWAS (486,484 cases and 474,841 controls). We used inverse-variance weighted MR, weighted median, and MR Egger for analyses, supplementing these with sensitivity analyses and assessing level pleiotropy using MR methodologies. We also executed a reverse MR analysis to test sepsis' causal effects on the designated autoimmune traits. Results : With primary sclerosing cholangitis being the exception, our MR analysis suggests that susceptibility toward most autoimmune diseases does not affect sepsis risks. The reverse MR analysis did not validate any influence of sepsis susceptibility over other autoimmune diseases. Our primary inverse-variance weighted MR analysis outcomes found general confirmation through our sensitivity MR examinations. Variance in the exposures, as dictated by the single nucleotide polymorphism sets used as MR instruments, ranged between 4.88 × 10 -5 to 0.005. Conclusion : Our MR research, centered on a European population, does not validate a correlation between susceptibility to the majority of autoimmune disorders and sepsis risk. Associations discerned in epidemiological studies may owe partly to shared biological or environmental confounders. The risk susceptibility for primary sclerosing cholangitis does relate to sepsis risk, opening doors for personalized precision treatments in the future.
Collapse
Affiliation(s)
- Ziyi Wang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
34
|
Shankar-Hari M, Calandra T, Soares MP, Bauer M, Wiersinga WJ, Prescott HC, Knight JC, Baillie KJ, Bos LDJ, Derde LPG, Finfer S, Hotchkiss RS, Marshall J, Openshaw PJM, Seymour CW, Venet F, Vincent JL, Le Tourneau C, Maitland-van der Zee AH, McInnes IB, van der Poll T. Reframing sepsis immunobiology for translation: towards informative subtyping and targeted immunomodulatory therapies. THE LANCET. RESPIRATORY MEDICINE 2024; 12:323-336. [PMID: 38408467 PMCID: PMC11025021 DOI: 10.1016/s2213-2600(23)00468-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 02/28/2024]
Abstract
Sepsis is a common and deadly condition. Within the current model of sepsis immunobiology, the framing of dysregulated host immune responses into proinflammatory and immunosuppressive responses for the testing of novel treatments has not resulted in successful immunomodulatory therapies. Thus, the recent focus has been to parse observable heterogeneity into subtypes of sepsis to enable personalised immunomodulation. In this Personal View, we highlight that many fundamental immunological concepts such as resistance, disease tolerance, resilience, resolution, and repair are not incorporated into the current sepsis immunobiology model. The focus for addressing heterogeneity in sepsis should be broadened beyond subtyping to encompass the identification of deterministic molecular networks or dominant mechanisms. We explicitly reframe the dysregulated host immune responses in sepsis as altered homoeostasis with pathological disruption of immune-driven resistance, disease tolerance, resilience, and resolution mechanisms. Our proposal highlights opportunities to identify novel treatment targets and could enable successful immunomodulation in the future.
Collapse
Affiliation(s)
- Manu Shankar-Hari
- Institute for Regeneration and Repair, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK.
| | - Thierry Calandra
- Service of Immunology and Allergy, Center of Human Immunology Lausanne, Department of Medicine and Department of Laboratory Medicine and Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | | | - Michael Bauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine and Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Hallie C Prescott
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Julian C Knight
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kenneth J Baillie
- Institute for Regeneration and Repair, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Lieuwe D J Bos
- Department of Intensive Care, Academic Medical Center, Amsterdam, Netherlands
| | - Lennie P G Derde
- Intensive Care Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Simon Finfer
- Critical Care Division, The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | - Richard S Hotchkiss
- Department of Anesthesiology and Critical Care Medicine, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - John Marshall
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON, Canada
| | | | - Christopher W Seymour
- Department of Critical Care Medicine, The Clinical Research, Investigation, and Systems Modeling of Acute illness (CRISMA) Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fabienne Venet
- Immunology Laboratory, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | | | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, Paris, France
| | - Anke H Maitland-van der Zee
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Iain B McInnes
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine and Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
35
|
Zhou BW, Zhang WJ, Zhang FL, Yang X, Ding YQ, Yao ZW, Yan ZZ, Zhao BC, Chen XD, Li C, Liu KX. Propofol improves survival in a murine model of sepsis via inhibiting Rab5a-mediated intracellular trafficking of TLR4. J Transl Med 2024; 22:316. [PMID: 38549133 PMCID: PMC10976826 DOI: 10.1186/s12967-024-05107-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Propofol is a widely used anesthetic and sedative, which has been reported to exert an anti-inflammatory effect. TLR4 plays a critical role in coordinating the immuno-inflammatory response during sepsis. Whether propofol can act as an immunomodulator through regulating TLR4 is still unclear. Given its potential as a sepsis therapy, we investigated the mechanisms underlying the immunomodulatory activity of propofol. METHODS The effects of propofol on TLR4 and Rab5a (a master regulator involved in intracellular trafficking of immune factors) were investigated in macrophage (from Rab5a-/- and WT mice) following treatment with lipopolysaccharide (LPS) or cecal ligation and puncture (CLP) in vitro and in vivo, and peripheral blood monocyte from sepsis patients and healthy volunteers. RESULTS We showed that propofol reduced membrane TLR4 expression on macrophages in vitro and in vivo. Rab5a participated in TLR4 intracellular trafficking and both Rab5a expression and the interaction between Rab5a and TLR4 were inhibited by propofol. We also showed Rab5a upregulation in peripheral blood monocytes of septic patients, accompanied by increased TLR4 expression on the cell surface. Propofol downregulated the expression of Rab5a and TLR4 in these cells. CONCLUSIONS We demonstrated that Rab5a regulates intracellular trafficking of TLR4 and that propofol reduces membrane TLR4 expression on macrophages by targeting Rab5a. Our study not only reveals a novel mechanism for the immunomodulatory effect of propofol but also indicates that Rab5a may be a potential therapeutic target against sepsis.
Collapse
Affiliation(s)
- Bo-Wei Zhou
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wen-Juan Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Fang-Ling Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiao Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yu-Qi Ding
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zhi-Wen Yao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zheng-Zheng Yan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Bing-Cheng Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiao-Dong Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
36
|
Sahoo DK, Wong D, Patani A, Paital B, Yadav VK, Patel A, Jergens AE. Exploring the role of antioxidants in sepsis-associated oxidative stress: a comprehensive review. Front Cell Infect Microbiol 2024; 14:1348713. [PMID: 38510969 PMCID: PMC10952105 DOI: 10.3389/fcimb.2024.1348713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
Sepsis is a potentially fatal condition characterized by organ dysfunction caused by an imbalanced immune response to infection. Although an increased inflammatory response significantly contributes to the pathogenesis of sepsis, several molecular mechanisms underlying the progression of sepsis are associated with increased cellular reactive oxygen species (ROS) generation and exhausted antioxidant pathways. This review article provides a comprehensive overview of the involvement of ROS in the pathophysiology of sepsis and the potential application of antioxidants with antimicrobial properties as an adjunct to primary therapies (fluid and antibiotic therapies) against sepsis. This article delves into the advantages and disadvantages associated with the utilization of antioxidants in the therapeutic approach to sepsis, which has been explored in a variety of animal models and clinical trials. While the application of antioxidants has been suggested as a potential therapy to suppress the immune response in cases where an intensified inflammatory reaction occurs, the use of multiple antioxidant agents can be beneficial as they can act additively or synergistically on different pathways, thereby enhancing the antioxidant defense. Furthermore, the utilization of immunoadjuvant therapy, specifically in septic patients displaying immunosuppressive tendencies, represents a promising advancement in sepsis therapy.
Collapse
Affiliation(s)
- Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - David Wong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Anil Patani
- Department of Biotechnology, Smt. S. S. Patel Nootan Science and Commerce College, Sankalchand Patel University, Gujarat, India
| | - Biswaranjan Paital
- Redox Regulation Laboratory, Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Gujarat, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Gujarat, India
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
37
|
Torrance HD, Zhang P, Longbottom ER, Mi Y, Whalley JP, Allcock A, Kwok AJ, Cano-Gamez E, Geoghegan CG, Burnham KL, Antcliffe DB, Davenport EE, Pearse RM, O’Dwyer MJ, Hinds CJ, Knight JC, Gordon AC. A Transcriptomic Approach to Understand Patient Susceptibility to Pneumonia After Abdominal Surgery. Ann Surg 2024; 279:510-520. [PMID: 37497667 PMCID: PMC10829899 DOI: 10.1097/sla.0000000000006050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
OBJECTIVE To describe immune pathways and gene networks altered following major abdominal surgery and to identify transcriptomic patterns associated with postoperative pneumonia. BACKGROUND Nosocomial infections are a major healthcare challenge, developing in over 20% of patients aged 45 or over undergoing major abdominal surgery, with postoperative pneumonia associated with an almost 5-fold increase in 30-day mortality. METHODS From a prospective consecutive cohort (n=150) undergoing major abdominal surgery, whole-blood RNA was collected preoperatively and at 3 time-points postoperatively (2-6, 24, and 48 h). Twelve patients diagnosed with postoperative pneumonia and 27 matched patients remaining infection-free were identified for analysis with RNA-sequencing. RESULTS Compared to preoperative sampling, 3639 genes were upregulated and 5043 downregulated at 2 to 6 hours. Pathway analysis demonstrated innate-immune activation with neutrophil degranulation and Toll-like-receptor signaling upregulation alongside adaptive-immune suppression. Cell-type deconvolution of preoperative RNA-sequencing revealed elevated S100A8/9-high neutrophils alongside reduced naïve CD4 T-cells in those later developing pneumonia. Preoperatively, a gene-signature characteristic of neutrophil degranulation was associated with postoperative pneumonia acquisition ( P =0.00092). A previously reported Sepsis Response Signature (SRSq) score, reflecting neutrophil dysfunction and a more dysregulated host response, at 48 hours postoperatively, differed between patients subsequently developing pneumonia and those remaining infection-free ( P =0.045). Analysis of the novel neutrophil gene-signature and SRSq scores in independent major abdominal surgery and polytrauma cohorts indicated good predictive performance in identifying patients suffering later infection. CONCLUSIONS Major abdominal surgery acutely upregulates innate-immune pathways while simultaneously suppressing adaptive-immune pathways. This is more prominent in patients developing postoperative pneumonia. Preoperative transcriptomic signatures characteristic of neutrophil degranulation and postoperative SRSq scores may be useful predictors of subsequent pneumonia risk.
Collapse
Affiliation(s)
- Hew D. Torrance
- Division of Anaesthetics, Pain Medicine & Intensive Care Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, London. UK
| | - Ping Zhang
- Wellcome Centre for Human Genetics, University of Oxford, Oxford. UK
- Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford, UK
| | - E. Rebecca Longbottom
- Centre for Translational Medicine & Therapeutics, William Harvey Institute, Faculty of Medicine & Dentistry at Queen Mary University of London, London. UK
| | - Yuxin Mi
- Wellcome Centre for Human Genetics, University of Oxford, Oxford. UK
| | - Justin P. Whalley
- Wellcome Centre for Human Genetics, University of Oxford, Oxford. UK
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL
| | - Alice Allcock
- Wellcome Centre for Human Genetics, University of Oxford, Oxford. UK
| | - Andrew J. Kwok
- Wellcome Centre for Human Genetics, University of Oxford, Oxford. UK
| | - Eddie Cano-Gamez
- Wellcome Centre for Human Genetics, University of Oxford, Oxford. UK
| | | | - Katie L. Burnham
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - David B. Antcliffe
- Division of Anaesthetics, Pain Medicine & Intensive Care Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, London. UK
| | - Emma E. Davenport
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Rupert M. Pearse
- Centre for Translational Medicine & Therapeutics, William Harvey Institute, Faculty of Medicine & Dentistry at Queen Mary University of London, London. UK
| | - Michael J. O’Dwyer
- Department of Anaesthesia and Critical Care, St Vincent’s University Hospital, Dublin. Ireland
| | - Charles J. Hinds
- Centre for Translational Medicine & Therapeutics, William Harvey Institute, Faculty of Medicine & Dentistry at Queen Mary University of London, London. UK
| | - Julian C. Knight
- Wellcome Centre for Human Genetics, University of Oxford, Oxford. UK
- Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford, UK
| | - Anthony C. Gordon
- Division of Anaesthetics, Pain Medicine & Intensive Care Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, London. UK
| |
Collapse
|
38
|
Kannan SK, Kim CY, Heidarian M, Berton RR, Jensen IJ, Griffith TS, Badovinac VP. Mouse Models of Sepsis. Curr Protoc 2024; 4:e997. [PMID: 38439603 PMCID: PMC10917121 DOI: 10.1002/cpz1.997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Human sepsis is a complex disease that manifests with a diverse range of phenotypes and inherent variability among individuals, making it hard to develop a comprehensive animal model. Despite this difficulty, numerous models have been developed that capture many key aspects of human sepsis. The robustness of these models is vital for conducting pre-clinical studies to test and develop potential therapeutics. In this article, we describe four different models of murine sepsis that can be used to address different scientific questions relevant to the pathology and immune response during and after a septic event. Basic Protocol 1 details a non-synchronous cecal ligation and puncture (CLP) model of sepsis, where mice are subjected to polymicrobial exposure through surgery at different time points within 2 weeks. This variation in sepsis onset establishes each mouse at a different state of inflammation and cytokine levels that mimics the variability observed in humans when they present in the clinic. This model is ideal for studying the long-term impact of sepsis on the host. Basic Protocol 2 is also a type of polymicrobial sepsis, where injection of a specific amount of cecal slurry from a donor mouse into the peritoneum of recipient mice establishes immediate inflammation and sepsis without any need for surgery. Basic Protocol 3 describes infecting mice with a defined gram-positive or -negative bacterial strain to model a subset of sepsis observed in humans infected with a single pathogen. Basic Protocol 4 describes administering LPS to induce sterile endotoxemia. This form of sepsis is observed in humans exposed to bacterial toxins from the environment. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Non-synchronous cecal ligation and puncture Basic Protocol 2: Cecal slurry model of murine sepsis Basic Protocol 3: Monomicrobial model of murine sepsis Basic Protocol 4: LPS model of murine sepsis.
Collapse
Affiliation(s)
- Shravan-Kumar Kannan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA
| | - Caleb Y. Kim
- Microbiology, Immunology, and Cancer Biology Program, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Roger R. Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA
| | - Isaac J. Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Thomas S. Griffith
- Microbiology, Immunology, and Cancer Biology Program, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Urology, University of Minnesota, Minneapolis, Minnesota, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota, USA
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, USA
- Microbiology, Immunology, and Cancer Biology Program, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
39
|
Carlton EF, Perry-Eaddy MA, Prescott HC. Context and Implications of the New Pediatric Sepsis Criteria. JAMA 2024; 331:646-649. [PMID: 38245890 DOI: 10.1001/jama.2023.27979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Affiliation(s)
- Erin F Carlton
- Department of Pediatrics, University of Michigan, Ann Arbor
- Susan B. Meister Child Health Evaluation and Research, Ann Arbor, Michigan
| | - Mallory A Perry-Eaddy
- University of Connecticut School of Nursing, Storrs
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington
- Pediatric Intensive Care Unit, Connecticut Children's, Hartford
| | - Hallie C Prescott
- Department of Internal Medicine, University of Michigan, Ann Arbor
- VA Center for Clinical Management Research, Ann Arbor, Michigan
| |
Collapse
|
40
|
Takahama M, Patil A, Richey G, Cipurko D, Johnson K, Carbonetto P, Plaster M, Pandey S, Cheronis K, Ueda T, Gruenbaum A, Kawamoto T, Stephens M, Chevrier N. A pairwise cytokine code explains the organism-wide response to sepsis. Nat Immunol 2024; 25:226-239. [PMID: 38191855 PMCID: PMC10834370 DOI: 10.1038/s41590-023-01722-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024]
Abstract
Sepsis is a systemic response to infection with life-threatening consequences. Our understanding of the molecular and cellular impact of sepsis across organs remains rudimentary. Here, we characterize the pathogenesis of sepsis by measuring dynamic changes in gene expression across organs. To pinpoint molecules controlling organ states in sepsis, we compare the effects of sepsis on organ gene expression to those of 6 singles and 15 pairs of recombinant cytokines. Strikingly, we find that the pairwise effects of tumor necrosis factor plus interleukin (IL)-18, interferon-gamma or IL-1β suffice to mirror the impact of sepsis across tissues. Mechanistically, we map the cellular effects of sepsis and cytokines by computing changes in the abundance of 195 cell types across 9 organs, which we validate by whole-mouse spatial profiling. Our work decodes the cytokine cacophony in sepsis into a pairwise cytokine message capturing the gene, cell and tissue responses of the host to the disease.
Collapse
Affiliation(s)
- Michihiro Takahama
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | - Gabriella Richey
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Denis Cipurko
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Katherine Johnson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Peter Carbonetto
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Research Computing Center, University of Chicago, Chicago, IL, USA
| | - Madison Plaster
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Surya Pandey
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Katerina Cheronis
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Tatsuki Ueda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Adam Gruenbaum
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | | | - Matthew Stephens
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
41
|
Padovani CM, Yin K. Immunosuppression in Sepsis: Biomarkers and Specialized Pro-Resolving Mediators. Biomedicines 2024; 12:175. [PMID: 38255280 PMCID: PMC10813323 DOI: 10.3390/biomedicines12010175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Severe infection can lead to sepsis. In sepsis, the host mounts an inappropriately large inflammatory response in an attempt to clear the invading pathogen. This sustained high level of inflammation may cause tissue injury and organ failure. Later in sepsis, a paradoxical immunosuppression occurs, where the host is unable to clear the preexisting infection and is susceptible to secondary infections. A major issue with sepsis treatment is that it is difficult for physicians to ascertain which stage of sepsis the patient is in. Sepsis treatment will depend on the patient's immune status across the spectrum of the disease, and these immune statuses are nearly polar opposites in the early and late stages of sepsis. Furthermore, there is no approved treatment that can resolve inflammation without contributing to immunosuppression within the host. Here, we review the major mechanisms of sepsis-induced immunosuppression and the biomarkers of the immunosuppressive phase of sepsis. We focused on reviewing three main mechanisms of immunosuppression in sepsis. These are lymphocyte apoptosis, monocyte/macrophage exhaustion, and increased migration of myeloid-derived suppressor cells (MDSCs). The biomarkers of septic immunosuppression that we discuss include increased MDSC production/migration and IL-10 levels, decreased lymphocyte counts and HLA-DR expression, and increased GPR18 expression. We also review the literature on the use of specialized pro-resolving mediators (SPMs) in different models of infection and/or sepsis, as these compounds have been reported to resolve inflammation without being immunosuppressive. To obtain the necessary information, we searched the PubMed database using the keywords sepsis, lymphocyte apoptosis, macrophage exhaustion, MDSCs, biomarkers, and SPMs.
Collapse
Affiliation(s)
- Cristina M. Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, NJ 08084, USA;
| | | |
Collapse
|
42
|
Liang P, Wu Y, Qu S, Younis M, Wang W, Wu Z, Huang X. Exploring the biomarkers and potential therapeutic drugs for sepsis via integrated bioinformatic analysis. BMC Infect Dis 2024; 24:32. [PMID: 38166628 PMCID: PMC10763157 DOI: 10.1186/s12879-023-08883-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Sepsis is a life-threatening condition caused by an excessive inflammatory response to an infection, associated with high mortality. However, the regulatory mechanism of sepsis remains unclear. RESULTS In this study, bioinformatics analysis revealed the novel key biomarkers associated with sepsis and potential regulators. Three public datasets (GSE28750, GSE57065 and GSE95233) were employed to recognize the differentially expressed genes (DEGs). Taking the intersection of DEGs from these three datasets, GO and KEGG pathway enrichment analysis revealed 537 shared DEGs and their biological functions and pathways. These genes were mainly enriched in T cell activation, differentiation, lymphocyte differentiation, mononuclear cell differentiation, and regulation of T cell activation based on GO analysis. Further, pathway enrichment analysis revealed that these DEGs were significantly enriched in Th1, Th2 and Th17 cell differentiation. Additionally, five hub immune-related genes (CD3E, HLA-DRA, IL2RB, ITK and LAT) were identified from the protein-protein interaction network, and sepsis patients with higher expression of hub genes had a better prognosis. Besides, 14 drugs targeting these five hub related genes were revealed on the basis of the DrugBank database, which proved advantageous for treating immune-related diseases. CONCLUSIONS These results strengthen the new understanding of sepsis development and provide a fresh perspective into discriminating the candidate biomarkers for predicting sepsis as well as identifying new drugs for treating sepsis.
Collapse
Affiliation(s)
- Pingping Liang
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong Province, Zhuhai, 519000, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong Province, Zhuhai, 519000, China
| | - Siying Qu
- Department of Clinical Laboratory, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, The Second People's Hospital of Zhuhai, Guangdong Province, Zhuhai, 519020, China
| | - Muhammad Younis
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong Province, Zhuhai, 519000, China
| | - Wei Wang
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China
| | - Zhilong Wu
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China.
| | - Xi Huang
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China.
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong Province, Zhuhai, 519000, China.
| |
Collapse
|
43
|
Kim YT, Huh JW, Choi YH, Yoon HK, Nguyen TT, Chun E, Jeong G, Park S, Ahn S, Lee WK, Noh YW, Lee KS, Ahn HS, Lee C, Lee SM, Kim KS, Suh GJ, Jeon K, Kim S, Jin M. Highly secreted tryptophanyl tRNA synthetase 1 as a potential theranostic target for hypercytokinemic severe sepsis. EMBO Mol Med 2024; 16:40-63. [PMID: 38177528 PMCID: PMC10883277 DOI: 10.1038/s44321-023-00004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 01/06/2024] Open
Abstract
Despite intensive clinical and scientific efforts, the mortality rate of sepsis remains high due to the lack of precise biomarkers for patient stratification and therapeutic guidance. Secreted human tryptophanyl-tRNA synthetase 1 (WARS1), an endogenous ligand for Toll-like receptor (TLR) 2 and TLR4 against infection, activates the genes that signify the hyperinflammatory sepsis phenotype. High plasma WARS1 levels stratified the early death of critically ill patients with sepsis, along with elevated levels of cytokines, chemokines, and lactate, as well as increased numbers of absolute neutrophils and monocytes, and higher Sequential Organ Failure Assessment (SOFA) scores. These symptoms were recapitulated in severely ill septic mice with hypercytokinemia. Further, injection of WARS1 into mildly septic mice worsened morbidity and mortality. We created an anti-human WARS1-neutralizing antibody that suppresses proinflammatory cytokine expression in marmosets with endotoxemia. Administration of this antibody into severe septic mice attenuated cytokine storm, organ failure, and early mortality. With antibiotics, the antibody almost completely prevented fatalities. These data imply that blood-circulating WARS1-guided anti-WARS1 therapy may provide a novel theranostic strategy for life-threatening systemic hyperinflammatory sepsis.
Collapse
Affiliation(s)
- Yoon Tae Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Jin Won Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yun Hui Choi
- R&D Center, MirimGENE, Incheon, Republic of Korea
| | | | | | - Eunho Chun
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Geunyeol Jeong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Sunyoung Park
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Sungwoo Ahn
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Won-Kyu Lee
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Young-Woock Noh
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Kyoung Sun Lee
- Non-Clinical Evaluation Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Hee-Sung Ahn
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Cheolju Lee
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Sang Min Lee
- Department of Internal Medicine, Gil Medical Center, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Kyung Su Kim
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Gil Joon Suh
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyeongman Jeon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, Institute for Artificial Intelligence and Biomedical Research, The interdisciplinary graduate program in integrative biotechnology, College of Pharmacy & College of Medicine, Gangnam Severance Hospital, Yonsei University, Incheon, Republic of Korea
| | - Mirim Jin
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
- R&D Center, MirimGENE, Incheon, Republic of Korea.
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.
- Department of Microbiology, College of Medicine, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
44
|
Giamarellos-Bourboulis EJ, Aschenbrenner AC, Bauer M, Bock C, Calandra T, Gat-Viks I, Kyriazopoulou E, Lupse M, Monneret G, Pickkers P, Schultze JL, van der Poll T, van de Veerdonk FL, Vlaar APJ, Weis S, Wiersinga WJ, Netea MG. The pathophysiology of sepsis and precision-medicine-based immunotherapy. Nat Immunol 2024; 25:19-28. [PMID: 38168953 DOI: 10.1038/s41590-023-01660-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/21/2023] [Indexed: 01/05/2024]
Abstract
Sepsis remains a major cause of morbidity and mortality in both low- and high-income countries. Antibiotic therapy and supportive care have significantly improved survival following sepsis in the twentieth century, but further progress has been challenging. Immunotherapy trials for sepsis, mainly aimed at suppressing the immune response, from the 1990s and 2000s, have largely failed, in part owing to unresolved patient heterogeneity in the underlying immune disbalance. The past decade has brought the promise to break this blockade through technological developments based on omics-based technologies and systems medicine that can provide a much larger data space to describe in greater detail the immune endotypes in sepsis. Patient stratification opens new avenues towards precision medicine approaches that aim to apply immunotherapies to sepsis, on the basis of precise biomarkers and molecular mechanisms defining specific immune endotypes. This approach has the potential to lead to the establishment of immunotherapy as a successful pillar in the treatment of sepsis for future generations.
Collapse
Affiliation(s)
- Evangelos J Giamarellos-Bourboulis
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Anna C Aschenbrenner
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Institute of Artificial Intelligence, Center for Medical Data Science, Vienna, Austria
| | - Thierry Calandra
- Service of Immunology and Allergy and Center of Human Immunology Lausanne, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Irit Gat-Viks
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Evdoxia Kyriazopoulou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Mihaela Lupse
- Infectious Diseases Hospital, University of Medicine and Pharmacy 'Iuliu Hatieganu', Cluj-Napoca, Romania
| | - Guillaume Monneret
- Joint Research Unit HCL-bioMérieux, EA 7426 'Pathophysiology of Injury-Induced Immunosuppression' (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Immunology Laboratory, Edouard Herriot Hospital - Hospices Civils de Lyon, Lyon, France
| | - Peter Pickkers
- Department of Intensive Care, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joachim L Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and the University of Bonn, Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Tom van der Poll
- Division of Infectious Diseases, Amsterdam University Medical Centers (Amsterdam UMC), Center for Experimental and Molecular Medicine (CEMM), University of Amsterdam, Amsterdam, The Netherlands
| | - Frank L van de Veerdonk
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander P J Vlaar
- Department of Intensive Care and Laboratory of Experimental Intensive Care and Anesthesiology L.E.C.A. Amsterdam Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Sebastian Weis
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute-HKI, Jena, Germany
| | - W Joost Wiersinga
- Division of Infectious Diseases, Amsterdam University Medical Centers (Amsterdam UMC), Center for Experimental and Molecular Medicine (CEMM), University of Amsterdam, Amsterdam, The Netherlands
| | - Mihai G Netea
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
45
|
Truong N, Cottingham AL, Dharmaraj S, Shaw JR, Lasola JJM, Goodis CC, Fletcher S, Pearson RM. Multimodal nanoparticle-containing modified suberoylanilide hydroxamic acid polymer conjugates to mitigate immune dysfunction in severe inflammation. Bioeng Transl Med 2024; 9:e10611. [PMID: 38193117 PMCID: PMC10771562 DOI: 10.1002/btm2.10611] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/04/2023] [Accepted: 10/01/2023] [Indexed: 01/10/2024] Open
Abstract
Excessive immune activation and immunosuppression are opposing factors that contribute to the dysregulated innate and adaptive immune responses seen in severe inflammation and sepsis. Here, a novel analog of the histone deacetylase inhibitor (HDACi), suberoylanilide hydroxamic acid (SAHA-OH), was incorporated into immunomodulatory poly(lactic acid)-based nanoparticles (iNP-SAHA) by employing a prodrug approach through the covalent modification of poly(lactic-co-glycolic acid) (PLGA) with SAHA-OH. iNP-SAHA formulation allowed for controlled incorporation and delivery of SAHA-OH from iNP-SAHA and treatment led to multimodal biological responses including significant reductions in proinflammatory cytokine secretions and gene expression, while increasing the survival of primary macrophages under lipopolysaccharide (LPS) challenge. Using a lethal LPS-induced endotoxemia mouse model of sepsis, iNP-SAHA administration improved the survival of mice in a dose-dependent manner and tended to improve survival at the lowest doses compared to iNP control. Further, iNP-SAHA reduced the levels of plasma proinflammatory cytokines and chemokines associated with sepsis more significantly than iNP and similarly improved inflammation-induced spleen and liver toxicity as iNP, supporting its potential polypharmacological activity. Collectively, iNP-SAHA offers a potential drug delivery approach to modulate the multifaceted inflammatory responses observed in diseases such as sepsis.
Collapse
Affiliation(s)
- Nhu Truong
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Andrea L. Cottingham
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Shruti Dharmaraj
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Jacob R. Shaw
- Department of Microbiology and ImmunologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | | | - Christopher C. Goodis
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Steven Fletcher
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Ryan M. Pearson
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
- Department of Microbiology and ImmunologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
46
|
Ji A, Trumbauer AC, Noffsinger VP, Meredith LW, Dong B, Wang Q, Guo L, Li X, De Beer FC, Webb NR, Tannock LR, Starr ME, Waters CM, Shridas P. Deficiency of Acute-Phase Serum Amyloid A Exacerbates Sepsis-Induced Mortality and Lung Injury in Mice. Int J Mol Sci 2023; 24:17501. [PMID: 38139330 PMCID: PMC10744229 DOI: 10.3390/ijms242417501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Serum amyloid A (SAA) is a family of proteins, the plasma levels of which may increase >1000-fold in acute inflammatory states. We investigated the role of SAA in sepsis using mice deficient in all three acute-phase SAA isoforms (SAA-TKO). SAA deficiency significantly increased mortality rates in the three experimental sepsis mouse models: cecal ligation and puncture (CLP), cecal slurry (CS) injection, and lipopolysaccharide (LPS) treatments. SAA-TKO mice had exacerbated lung pathology compared to wild-type (WT) mice after CLP. A bulk RNA sequencing performed on lung tissues excised 24 h after CLP indicated significant enrichment in the expression of genes associated with chemokine production, chemokine and cytokine-mediated signaling, neutrophil chemotaxis, and neutrophil migration in SAA-TKO compared to WT mice. Consistently, myeloperoxidase activity and neutrophil counts were significantly increased in the lungs of septic SAA-TKO mice compared to WT mice. The in vitro treatment of HL-60, neutrophil-like cells, with SAA or SAA bound to a high-density lipoprotein (SAA-HDL), significantly decreased cellular transmigration through laminin-coated membranes compared to untreated cells. Thus, SAA potentially prevents neutrophil transmigration into injured lungs, thus reducing exacerbated tissue injury and mortality. In conclusion, we demonstrate for the first time that endogenous SAA plays a protective role in sepsis, including ameliorating lung injury.
Collapse
Affiliation(s)
- Ailing Ji
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Andrea C. Trumbauer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Victoria P. Noffsinger
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Luke W. Meredith
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Brittany Dong
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
| | - Qian Wang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Ling Guo
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Xiangan Li
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Frederick C. De Beer
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Nancy R. Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Lisa R. Tannock
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Marlene E. Starr
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher M. Waters
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
| | - Preetha Shridas
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| |
Collapse
|
47
|
Atreya MR, Huang M, Moore AR, Zheng H, Hasin-Brumshtein Y, Fitzgerald JC, Weiss SL, Cvijanovich NZ, Bigham MT, Jain PN, Schwarz AJ, Lutfi R, Nowak J, Thomas NJ, Quasney M, Dahmer MK, Baines T, Haileselassie B, Lautz AJ, Stanski NL, Standage SW, Kaplan JM, Zingarelli B, Sweeney TE, Khatri P, Sanchez-Pinto LN, Kamaleswaran R. Derivation, validation, and transcriptomic assessment of pediatric septic shock phenotypes identified through latent profile analyses: Results from a prospective multi-center observational cohort. RESEARCH SQUARE 2023:rs.3.rs-3692289. [PMID: 38105983 PMCID: PMC10723552 DOI: 10.21203/rs.3.rs-3692289/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background Sepsis poses a grave threat, especially among children, but treatments are limited due to clinical and biological heterogeneity among patients. Thus, there is an urgent need for precise subclassification of patients to guide therapeutic interventions. Methods We used clinical, laboratory, and biomarker data from a prospective multi-center pediatric septic shock cohort to derive phenotypes using latent profile analyses. Thereafter, we trained a support vector machine model to assign phenotypes in a hold-out validation set. We tested interactions between phenotypes and common sepsis therapies on clinical outcomes and conducted transcriptomic analyses to better understand the phenotype-specific biology. Finally, we compared whether newly identified phenotypes overlapped with established gene-expression endotypes and tested the utility of an integrated subclassification scheme. Findings Among 1,071 patients included, we identified two phenotypes which we named 'inflamed' (19.5%) and an 'uninflamed' phenotype (80.5%). The 'inflamed' phenotype had an over 4-fold risk of 28-day mortality relative to those 'uninflamed'. Transcriptomic analysis revealed overexpression of genes implicated in the innate immune response and suggested an overabundance of developing neutrophils, pro-T/NK cells, and NK cells among those 'inflamed'. There was no significant overlap between endotypes and phenotypes. However, an integrated subclassification scheme demonstrated varying survival probabilities when comparing endophenotypes. Interpretation Our research underscores the reproducibility of latent profile analyses to identify clinical and biologically informative pediatric septic shock phenotypes with high prognostic relevance. Pending validation, an integrated subclassification scheme, reflective of the different facets of the host response, holds promise to inform targeted intervention among those critically ill.
Collapse
Affiliation(s)
- Mihir R Atreya
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Min Huang
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew R Moore
- Stanford Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA
| | - Hong Zheng
- Stanford Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Stanford, 94305, CA
| | | | | | - Scott L Weiss
- Nemours Children's Health, Wilmington, DE, 19803, USA
| | | | | | - Parag N Jain
- Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Adam J Schwarz
- Children's Hospital of Orange County, Orange, CA, 92868, USA
| | - Riad Lutfi
- Riley Hospital for Children, Indianapolis, IN, 46202, USA
| | - Jeffrey Nowak
- Children's Hospital and Clinics of Minnesota, Minneapolis, MN, 55404, USA
| | - Neal J Thomas
- Penn State Hershey Children's Hospital, Hershey, PA, 17033, USA
| | - Michael Quasney
- C.S Mott Children's Hospital, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mary K Dahmer
- C.S Mott Children's Hospital, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Torrey Baines
- University of Florida Health Shands Children's Hospital, Gainesville, FL, 32610, USA
| | | | - Andrew J Lautz
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Natalja L Stanski
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Stephen W Standage
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Jennifer M Kaplan
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | | | - Purvesh Khatri
- Stanford Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Stanford, 94305, CA
| | - L Nelson Sanchez-Pinto
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
- Department of Health and Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - Rishikesan Kamaleswaran
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, 30322, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, 30322, GA, USA
| |
Collapse
|
48
|
Nugent WH, Carr DA, Friedman J, Song BK. Novel transdermal curcumin therapeutic preserves endothelial barrier function in a high-dose LPS rat model. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:33-40. [PMID: 36656591 DOI: 10.1080/21691401.2022.2164584] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Sepsis is a devastating complication of infection and injury that, through widespread endothelial dysfunction, can cause perfusion deficits and multi-organ failure. To address the recognised need for therapeutics targetting the endothelial barrier, a topical formulation (CUR; VASCEPTOR™; Vascarta Inc, Summit, NJ) was developed to transdermally deliver bio-active concentrations of curcumin-an anti-inflammatory and nitric oxide promoter. Male, Sprague Dawley rats were treated daily with lipopolysaccharide (LPS, 10 mg/kg, IP) to induce endotoxemia, and topical applications of Vehicle Control (LPS + VC; N = 7) or Curcumin (LPS + CUR; N = 7). A third group received neither LPS nor treatment (No-LPS; N = 8). After 72 h, animals were surgically prepared for measurements of physiology and endothelial dysfunction in the exteriorised spinotrapezius muscle through the extravasation of 67 kDa TRITC-BSA (albumin) and 500 kDa FITC-dextran (dextran). At 72 h, LPS + VC saw weight loss, and increases to pulse pressure, lactate, pCO2, CXCL5 (vs No-LPS) and IL-6 (vs 0 h; p < 0.05). LPS + CUR was similar to No-LPS, but with hypotension. Phenylephrine response was increased in LPS + CUR. Regarding endothelial function, LPS + CUR albumin and dextran extravasation were significantly reduced versus LPS + VC suggesting that Curcumin mitigated endotoxemic endothelial dysfunction. The speculated mechanisms are nitric oxide modulation of the endothelium and/or an indirect anti-inflammatory effect.
Collapse
Affiliation(s)
| | | | - Joel Friedman
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA.,Vascarta, Inc, Summit, NJ, USA
| | - Bjorn K Song
- Song Biotechnologies LLC, Cockeysville, MD, USA.,Vascarta, Inc, Summit, NJ, USA
| |
Collapse
|
49
|
Jang SY, Kim SY, Song HA, Kim H, Chung KS, Lee JK, Lee KT. Protective effect of hydrangenol on lipopolysaccharide-induced endotoxemia by suppressing intestinal inflammation. Int Immunopharmacol 2023; 125:111083. [PMID: 37871380 DOI: 10.1016/j.intimp.2023.111083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023]
Abstract
Hydrangenol, a dihydroisocoumarin, isolated from the leaves of Hydrangea serrata, possesses anti-inflammatory, anti-obesity, and anti-photoaging activities. In this study, we investigated the protective effects of hydrangenol (HG) against lipopolysaccharide (LPS)-induced endotoxemia and elucidated the underlying molecular mechanisms of action in C57BL/6 mice. Oral administration of HG (20 or 40 mg/kg) significantly restored the survival rate and population of macrophages, T helper cells (CD3+/CD4+), and Th17 cells (CD3+/CD4+/CCR6+) in the spleens of mice with LPS-induced endotoxemia. HG suppressed the expression of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-1β, and Interferon (IFN)-γ and the mRNA and protein expressions of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) in the intestine and lung of LPS-treated mice. Molecular data showed that HG ameliorated the activation of nuclear factor kappa B (NF-κB) p65, signal transducers and activators of transcription 3 (STAT3), and c-Fos and c-Jun (AP-1 subunits) via the myeloid differentiation primary response 88 (MyD88) dependent toll-like receptor 4 (TLR4) signaling pathway in the LPS-treated mouse intestines. HG treatment caused the recovery of LPS-induced impaired tight junction (occludin and claudin-2) protein and mRNA expressions. Furthermore, HG improved LPS-induced gut dysbiosis in mice. Taken together, our results suggest that HG protects against LPS-induced endotoxemia by restoring immune cells and the capacity of the intestinal barrier, reducing intestinal inflammation, and improving the composition of the gut microbiota.
Collapse
Affiliation(s)
- Seo-Yun Jang
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea; Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Su-Yeon Kim
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea; Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
| | - Hyeon-A Song
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea; Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Hyeyun Kim
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea; Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
| | - Jong Kil Lee
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea; Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea.
| |
Collapse
|
50
|
Alfano DN, Miller MJ, Bubeck Wardenburg J. Endothelial ADAM10 utilization defines a molecular pathway of vascular injury in mice with bacterial sepsis. J Clin Invest 2023; 133:e168450. [PMID: 37788087 PMCID: PMC10688991 DOI: 10.1172/jci168450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023] Open
Abstract
The endothelium plays a critical role in the host response to infection and has been a focus of investigation in sepsis. While it is appreciated that intravascular thrombus formation, severe inflammation, and loss of endothelial integrity impair tissue oxygenation during sepsis, the precise molecular mechanisms that lead to endothelial injury remain poorly understood. We demonstrate here that endothelial ADAM10 was essential for the pathogenesis of Staphylococcus aureus sepsis, contributing to α-toxin-mediated (Hla-mediated) microvascular thrombus formation and lethality. As ADAM10 is essential for endothelial development and homeostasis, we examined whether other major human sepsis pathogens also rely on ADAM10-dependent pathways in pathogenesis. Mice harboring an endothelium-specific knockout of ADAM10 were protected against lethal Pseudomonas aeruginosa and Streptococcus pneumoniae sepsis, yet remained fully susceptible to group B streptococci and Candida albicans sepsis. These studies illustrate a previously unknown role for ADAM10 in sepsis-associated endothelial injury and suggest that understanding pathogen-specific divergent host pathways in sepsis may enable more precise targeting of disease.
Collapse
Affiliation(s)
| | - Mark J. Miller
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|