1
|
Fang Y, Meng H, Wang J. Mechanisms of LPS-induced toxicity in endothelial cells and the protective role of geniposidic acid. Food Chem Toxicol 2025; 201:115488. [PMID: 40288513 DOI: 10.1016/j.fct.2025.115488] [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: 03/10/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Vascular inflammation and oxidative stress are critical pathogenic factors in cardiovascular diseases. Lipopolysaccharide (LPS)-induced endothelial cytotoxicity, driven by oxidative stress and inflammation, remains incompletely understood. This study highlights the molecular mechanisms underlying LPS toxicity, focusing on the ROS/JNK/NLRP3 signaling axis. LPS disrupts mitochondrial function, increases ROS accumulation, activates JNK phosphorylation, and induces NLRP3 inflammasome activation, culminating in pyroptosis through caspase-1-mediated GSDMD cleavage. Mechanistic studies with the JNK inhibitor SP600125 confirmed the critical role of the ROS/JNK/NLRP3 pathway in LPS-induced endothelial damage. Additionally, PGC-1α, a key regulator of mitochondrial homeostasis, was identified as a protective factor suppressed by LPS, exacerbating ROS overproduction and inflammasome activation. To validate these findings, geniposidic acid (GPA), a natural antioxidant and anti-inflammatory compound, was employed. GPA effectively reduced ROS levels, inhibited JNK activation, and suppressed pyroptosis, supporting its utility as a chemical tool to confirm the pivotal role of ROS/JNK/NLRP3 signaling. This study elucidates the intricate interplay between oxidative stress, mitochondrial dysfunction, and pyroptosis, providing a comprehensive framework for addressing inflammation-driven vascular damage.
Collapse
Affiliation(s)
- Yan Fang
- University of Science and Technology of China, Hefei, 230026, China; Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - He Meng
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jun Wang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
| |
Collapse
|
2
|
Lavarti R, Alvarez-Diaz T, Marti K, Kar P, Raju RP. The context-dependent effect of cellular senescence: From embryogenesis and wound healing to aging. Ageing Res Rev 2025; 109:102760. [PMID: 40318767 PMCID: PMC12145239 DOI: 10.1016/j.arr.2025.102760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Aging is characterized by a steady loss of physiological integrity, leading to impaired function and increased vulnerability to death. Cell senescence is a biological process that progresses with aging and is believed to be a key driver of age-related diseases. Senescence, a hallmark of aging, also demonstrates its beneficial physiological aspects as an anti-cancer, pro-regenerative, homeostatic, and developmental mechanism. A transitory response in which the senescent cells are quickly formed and cleared may promote tissue regeneration and organismal fitness. At the same time, senescence-related secretory phenotypes associated with extended senescence can have devastating effects. The fact that the interaction between senescent cells and their surroundings is very context-dependent may also help to explain this seemingly opposing pleiotropic function. Further, mitochondrial dysfunction is an often-unappreciated hallmark of cellular senescence and figures prominently in multiple feedback loops that induce and maintain the senescent phenotype. This review summarizes the mechanism of cellular senescence and the significance of acute senescence. We concisely introduced the context-dependent role of senescent cells and SASP, aspects of mitochondrial biology altered in the senescent cells, and their impact on the senescent phenotype. Finally, we conclude with recent therapeutic advancements targeting cellular senescence, focusing on acute injuries and age-associated diseases. Collectively, these insights provide a future roadmap for the role of senescence in organismal fitness and life span extension.
Collapse
Affiliation(s)
- Rupa Lavarti
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Tatiana Alvarez-Diaz
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kyarangelie Marti
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Parmita Kar
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States.
| |
Collapse
|
3
|
Chen K, Li Y, Zhang G, Zuo M, Bi H, Shi W, Cong B. A case report of fatal splenic rupture caused by multiple organ infection following foreign body ingestion in a detainee. Forensic Sci Res 2025; 10:owaf008. [PMID: 40256281 PMCID: PMC12007405 DOI: 10.1093/fsr/owaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/21/2025] [Indexed: 04/22/2025] Open
Abstract
Among forensic and clinical cases, infections caused by the ingestion of foreign bodies are common. In general, timely removal of the foreign body and appropriate treatment prevent serious consequences. We herein report a rare case of death due to massive bleeding caused by splenic rupture following foreign body ingestion. To our knowledge, no similar cases have been reported in the Chinese or international literature, making this case particularly noteworthy. In this instance, the decedent was in a detention centre for a criminal offence and swallowed a wire unnoticed. The wire remained in his stomach for >50 days, leading to a severe suppurative infection in the gastric tissue. This resulted in suppurative inflammation affecting multiple organs, including the liver, pancreas, and spleen. The condition ultimately led to the rupture of splenic vessels and the formation of a rare, massive haematoma beneath the splenic capsule. Based on medical records and histopathological findings, we infer that the wire had remained in the stomach for ~50 days, triggering severe suppurative infections in multiple organs. The spleen eventually ruptured, and the victim died of massive haemorrhage.
Collapse
Affiliation(s)
- Ke Chen
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Centre of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Department of Forensic Medicine, College of Medicine, Nantong University, Nantong, China
| | - Yingmin Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Centre of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Guozhong Zhang
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Centre of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Min Zuo
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Centre of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Haitao Bi
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Centre of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Weibo Shi
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Centre of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Centre of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
4
|
Al-Kadi A, Anter AF, Rofaeil RR, Sayed-Ahmed MM, Hafez SMNA, Ahmed ASF. Endothelin System Blockade Extenuates Sepsis-Induced Acute Heart and Kidney Injuries via Modulating ET-1/Klotho/p38-MAPK. Clin Exp Pharmacol Physiol 2025; 52:e70042. [PMID: 40228821 DOI: 10.1111/1440-1681.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/07/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Sepsis-induced organ failure is a major health problem, characterised by massive inflammatory and oxidative stress responses. Endothelin-1 (ET-1) is one of the peptides expressed during septicemia with proapoptotic, proinflammatory, and oxidant effects. ET-1 plays a role in heart and kidney injuries in sepsis. Accordingly, the current study was conducted to investigate, on a mechanistic basis, whether inhibition of ET-1 signalling either by blocking its receptors or inhibiting its formation attenuates sepsis-induced acute cardiorenal injuries. To analyse the role of ET-1 in sepsis, we used a cecal ligation and puncture (CLP) model of sepsis. The animals were divided into five groups: CLP non-treated group, CLP-treated groups with bosentan, ambrisentan, and phosphoramidon (30, 5, and 0.5 mg/kg, respectively), and sham-operated group. In addition to the same set of groups, survival analysis was assigned Survival rate, histopathological assessment, and cardiorenal functions were analysed. Oxidant and antioxidant activities, ET-1, IL-6, and lactate were measured. The expression of TNF-α, p38, Klotho, and caspase-3 was evaluated by immunohistochemistry. CLP caused acute cardiorenal damage, high mortality, upregulated levels of ET-1, IL-6, and lactate, as well as an imbalance in oxidant/antioxidant activities, elevated expression of TNF-α, p38, caspase-3 and reduced expression of klotho. Bosentan, ambrisentan, or phosphoramidon improved survival, reduced the levels of inflammatory and oxidative stress parameters, improved cardiorenal functions and structure, elevated the tissue contents of GSH and SOD, raised the expression of klotho protein, and reduced the cardiorenal expression of p38, TNF-α and caspase-3. Endothelin receptor antagonists (ERAs); bosentan and ambrisentan, or endothelin converting enzyme inhibitor (ECE-i) phosphoramidon, are promising agents against sepsis-induced organ damage. This was evident in their cardiorenal protective effects, up-regulation of klotho, suppression of inflammation, oxidation, apoptosis, and enhancement of the antioxidant status.
Collapse
Affiliation(s)
- Alaa Al-Kadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Aliaa F Anter
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Remon Roshdy Rofaeil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Deraya University, Minia, Egypt
- Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Mohamed M Sayed-Ahmed
- Pharmacology and Experimental Oncology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Sara Mohamed Naguib Abdel Hafez
- Histology and Cell Biology Department, Minia University, Faculty of Medicine, Minia University Faculty of Medicine, Minia, Egypt
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
5
|
Liu MW, Duan SX, Zhao XY, Wang QF, Yang SL, Ma N, Li X. Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review). Int J Mol Med 2025; 55:94. [PMID: 40242975 PMCID: PMC12045470 DOI: 10.3892/ijmm.2025.5535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Sepsis‑induced organ dysfunction syndrome (ODS) arises from a dysregulated response to infection, leading to multiple life‑threatening organ dysfunctions, and is a common complication in critically ill patients. Sepsis results in varying degrees of injury to the brain, lungs, kidneys and liver, culminating in immune dysfunction and multiple ODS (MODS). Current evidence indicates a direct correlation between the severity of organ injury and the prognosis of septic patients. Understanding the mechanisms of MODS in sepsis and developing effective management strategies are vital research areas. The protective effects of dexmedetomidine (DEX) on sepsis are well established, demonstrating its capacity to mitigate injuries to the brain, lungs, kidneys, liver and immune system. The present study reviews recent research progress on the role and mechanisms of action of DEX in the treatment of sepsis.
Collapse
Affiliation(s)
- Ming-Wei Liu
- Department of Emergency Medicine, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Shao-Xin Duan
- Department of Trauma, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Xue-Yan Zhao
- Department of Endocrinology, First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Qiong-Fen Wang
- Department of Gastroenterology, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Shan-Lan Yang
- Department of Oncology, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Ni Ma
- Department of Pharmacy, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| | - Xuan Li
- Department of Rehabilitation, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
| |
Collapse
|
6
|
Li C, Gao X, Liu Y, Yang B, Dai H, Zhao H, Li Y. The role of natural killer T cells in sepsis-associated acute kidney injury. Int Immunopharmacol 2025; 159:114953. [PMID: 40418883 DOI: 10.1016/j.intimp.2025.114953] [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: 01/31/2025] [Revised: 05/12/2025] [Accepted: 05/21/2025] [Indexed: 05/28/2025]
Abstract
The condition of sepsis, defined by the misregulation of the body's defensive mechanisms against infection, culminates in the potential for catastrophic organ damage and stands as a primary driver of mortality in Intensive Care Units (ICU) settings. Among patients in a critical condition, sepsis is a predominant factor in the development of acute kidney injury (AKI), and the death rate among those with both sepsis and AKI is considerably higher, underscoring the importance of addressing this health crisis. Sepsis-associated acute kidney injury (S-AKI) is a complex process involving inflammation, microcirculatory issues, and metabolic disorders. Among these, the inflammatory response has become a focal point of interest. Bridging the innate and adaptive immunity, natural killer T (NKT) cells can be rapidly activated in sepsis, contributing to sepsis-associated injury and downstream activation of inflammatory cells through the emission of Th1 or Th2 cytokines. They also contribute to S-AKI through the TNF-α/FasL and perforin pathways. Alpha-Galactosylceramide (α-GalCer), acting as a powerful activator for type I NKT (iNKT) cells, is able to regulate the secretory profile of iNKT cells, responding to the pro-inflammatory response and immunosuppressive profiles of sepsis. This review examines the part played by NKT cells in S-AKI and whether α-Galcer could function as a significant regulator in sepsis, based on studies of regression-related mechanisms.
Collapse
Affiliation(s)
- Cheng Li
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China; Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Xiaopo Gao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China; Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Yuan Liu
- Jiangxi Medical College, Nanchang University, Nanchang 330000, Jiangxi, China
| | - Bin Yang
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China; Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Hongkai Dai
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China; Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Hui Zhao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China; Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Yongshen Li
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China; Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China.
| |
Collapse
|
7
|
Fu H, Gao B, Zhou X, Hao Y, Liu C, Lan A, Tang J, Zhou F. DNA dioxygenase TET2 deficiency aggravates sepsis-induced acute lung injury by targeting ITGA10 via the PI3K/AKT signaling pathway. Cell Mol Biol Lett 2025; 30:60. [PMID: 40389853 PMCID: PMC12090539 DOI: 10.1186/s11658-025-00739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 05/06/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Sepsis-induced acute lung injury (ALI) is a clinical condition with high morbidity and mortality, and impaired endothelial function is the main pathological characteristic. As a member of DNA demethylases, ten-eleven translocation protein 2 (TET2) is involved in a variety of biological processes. However, the role of TET2 in endothelial dysfunction of sepsis-induced ALI remains unclear. METHODS We used cecal ligation and puncture (CLP) to establish a sepsis-induced acute lung injury mouse model and screened out Tet2 from TET family proteins. The results suggested that Tet2 was obviously declined. We used lipopolysaccharide (LPS) to stimulate human pulmonary microvascular endothelial cells (HPMECs) as an in vitro model, and we found the expression of TET2 was also decreased. Then we used small interfering RNAs and adenovirus to knockdown or overexpress TET2 to investigate the effect of TET2 on the function of HPMECs. The changes in sepsis-induced ALI symptoms were also analyzed in Tet2-deficient mice generated by adeno-associated virus 6 (AAV6). Next, RNA sequencing and KEGG analysis were used to find the TET2-regulated downstream target genes and signaling pathways under LPS stimulation. Finally, the rescue experiments were performed to analyze the role of target genes and signaling pathways modulated by TET2 in LPS-treated HPMECs. RESULTS TET2 and 5-hmC levels were significantly decreased in both in vitro and in vivo models of sepsis-induced ALI. TET2 knockdown exacerbated the dysfunction and apoptosis of HPMECs induced by LPS. Conversely, TET2 overexpression significantly alleviated these dysfunctions and reduced apoptosis. Meanwhile, the lung injury of Tet2-deficient mice was aggravated by increased inflammation and apoptosis. RNA sequencing and subsequent experiments showed that TET2 overexpression could increase the expression of Integrin α10 (ITGA10) by reducing the methylation level of ITGA10 promoter. This, in turn, activated the PI3K-AKT signaling pathway. Knocking down ITGA10 weakened the beneficial effects of TET2 overexpression in LPS-stimulated endothelial cells. CONCLUSIONS In our study, we demonstrated that TET2 deficiency aggravates endothelial cell dysfunction and promotes acute lung injury by targeting ITGA10 via the PI3K-AKT pathway. These findings indicate that TET2 may be a promising therapeutic target for treating sepsis-induced ALI.
Collapse
Affiliation(s)
- Hongxue Fu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bin Gao
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xin Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yingting Hao
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chang Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ailin Lan
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jingyi Tang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Fachun Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
8
|
Guo J, Sun J, Xiong M, Wang L, Han N, Wang T, He Z, Yuan C, Ma Y, Qi H, Hou Y, Jia Z. Pulmonary microvascular endothelial glycocalyx degradation as a key driver in COPD progression and its protection by Tongxinluo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156878. [PMID: 40424984 DOI: 10.1016/j.phymed.2025.156878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 05/15/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Chronic Obstructive Pulmonary Disease (COPD) is a major cause of morbidity and mortality worldwide. Pulmonary microvascular endothelial glycocalyx (PMEG) has been found to be significantly reduced in COPD, but the mechanism, cause, and effect of the reduction on COPD progression are inconclusive. OBJECTIVE This study aims to explore the mechanisms and consequences underlying PMEG degradation in COPD. Additionally, we strive to ascertain whether Tongxinluo (TXL)'s protective role in COPD is mediated through the preservation of PMEG integrity. METHODS A staged cigarette smoke (CS) exposure model was employed to investigate the timeline, trajectory, mechanisms, and causes of glycocalyx degradation, with in vitro validation. The in vivo glycocalyx degradation model was induced by intravenous injection of glycocalyx hydrolase along with CS exposure. The protective effect of TXL on glycocalyx integrity was examined in CS-exposed mice treated with TXL. RESULTS PMEG degradation occurs as early as 2 weeks after CS exposure and worsens as the disease advances. Multiple glycocalyx degrading enzyme upregulation at different time points collectively results in consistent glycocalyx component degradation. Mechanistically, CS or reactive oxygen species (ROS) exposure elevates pro-inflammatory cytokine secretion, leading to an increase in glycocalyx hydrolysis expression and subsequent PMEG degradation on the endothelial cell (EC) surface. PMEG degradation further promotes inflammatory cell infiltration and accelerates endothelial apoptosis, ultimately driving disease progression in COPD. TXL alleviates oxidative stress, reverses the upregulation of PMEG degrading enzyme, preserves PMEG integrity, reduces endothelial cell apoptosis, and mitigates COPD pathology. CONCLUSION In summary, this study provides groundbreaking insights into the role of PMEG degradation in COPD pathogenesis and introduces TXL as a novel therapeutic agent with the potential to preserve PMEG integrity and mitigate COPD progression. These findings significantly advance our understanding of COPD and offer innovative directions for future research and therapeutic development.
Collapse
Affiliation(s)
- Jing Guo
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China; The First Hospital of Hebei University of Chinese Medicine, Shijiazhuang 050011, Hebei, China
| | - Jiemeng Sun
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China; The First Hospital of Hebei University of Chinese Medicine, Shijiazhuang 050011, Hebei, China
| | - Mingyu Xiong
- Graduate School, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Le Wang
- Graduate School, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Ningxin Han
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, Hebei, China; Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050035, Hebei, China
| | - Tongxing Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, Hebei, China; Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050035, Hebei, China
| | - Zhuo He
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China
| | - Caiyun Yuan
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China
| | - Yan Ma
- Hebei Yiling Hospital, High-level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang 050091, Hebei, China
| | - Hui Qi
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, Hebei, China; Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050035, Hebei, China.
| | - Yunlong Hou
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, Hebei, China; Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050035, Hebei, China.
| | - Zhenhua Jia
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, Hebei, China; Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050035, Hebei, China; Hebei Yiling Hospital, High-level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang 050091, Hebei, China.
| |
Collapse
|
9
|
Wang Y, Zhang H, Miao C. Unraveling immunosenescence in sepsis: from cellular mechanisms to therapeutics. Cell Death Dis 2025; 16:393. [PMID: 40379629 DOI: 10.1038/s41419-025-07714-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/26/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
Sepsis is a life-threatening multiple organ dysfunction resulting from a dysregulated host response to infection, and patients with sepsis always exhibit a state of immune disorder characterized by both overwhelming inflammation and immunosuppression. The aging of immune system, namely "immunosenescence", has been reported to be correlated with high morbidity and mortality in elderly patients with sepsis. Initially, immunosenescence was considered as a range of age-related alterations in the immune system. However, increasing evidence has proven that persistent inflammation or even a short-term inflammatory challenge during sepsis could trigger accelerated aging of immune cells, which might further exacerbate inflammatory cytokine storm and promote the shift towards immunosuppression. Thus, premature immunosenescence is found in young sepsis individuals, which further aggravates immune disorders and induces the progression of sepsis. Furthermore, in old sepsis patients, the synergistic effects of both sepsis and aging may cause immunosenescence-associated alterations more significantly, resulting in more severe immune dysfunction and a worse prognosis. Therefore, it is necessary to explore the potential therapeutic strategies targeting immunosenescence during sepsis.
Collapse
Affiliation(s)
- Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Ji Y, Song H, Li L. Traditional Chinese medicine for sepsis: advancing from evidence to innovative drug discovery. Crit Care 2025; 29:193. [PMID: 40375087 PMCID: PMC12080179 DOI: 10.1186/s13054-025-05441-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/03/2025] [Indexed: 05/18/2025] Open
Abstract
The global health burden of sepsis is immense, characterized by significant loss of life and high healthcare costs. Traditional Chinese medicine (TCM), with its over two millennia of clinical practice in China, has gained attention as a potential adjunctive approach for sepsis. Here, we evaluated TCM applications in sepsis management, highlighting both potential benefits and methodological limitations of existing clinical evidence. Although various TCM preparations have been evaluated for sepsis treatment, the vast majority lack robust clinical evidence. Xuebijing Injection represents a rare example that has demonstrated efficacy in a large-scale, multicenter, randomized, double-blind, placebo-controlled trial. In contrast, the evidence supporting other preparations such as Shenfu and Shenmai Injections comes primarily from smaller, single-center studies with significant methodological limitations. There is a clear need for more high-quality, multicenter randomized controlled trials to rigorously evaluate these potentially beneficial but currently insufficiently validated TCM preparations. The pharmacological effects and underlying mechanisms of some bioactive compounds derived from TCM medications have been elucidated, shedding light on the potential of TCM-based anti-sepsis drug discovery. We underscore the importance of continued research to better integrate TCM with modern sepsis management, paving the way for the development of evidence-based TCM treatments for this challenging condition.
Collapse
Affiliation(s)
- Yun Ji
- Department of Surgical Intensive Care Unit, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, China.
| | - Hongyun Song
- Department of Rehabilitation in Traditional Chinese Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Libin Li
- Department of Surgical Intensive Care Unit, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
He J, Shao Z, Li Z, He Y, Zhang J, Zhong H, Li J, Liu Q, Shao Y. Mechanistic Insights into HOTAIR-Driven ADAM17/NF-Κb Activation and Endothelial Dysfunction in LPS-Challenged HUVECs. Immunol Invest 2025:1-27. [PMID: 40366882 DOI: 10.1080/08820139.2025.2503174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
INTRODUCTION HOX transcript antisense intergenic RNA (HOTAIR) has been implicated in inflammation and vascular pathology, but its role in regulation of ADAM17 and sepsis-induced endothelial injury remains unclear. METHODS LPS-treated human umbilical vein endothelial cells (HUVECs) modeled sepsis-induced endothelial injury, which were assessed via qRT-PCR, western blot and immunofluorescence. HOTAIR-knockout mice were treated with cecal ligation and perforation to establish sepsis model. RESULTS LPS-stimulation increased expression of HOTAIR and ADAM17 and decreased miR-326 levels in HUVECs. HOTAIR-knockdown by antisense oligonucleotides (ASOs) decreased ADAM17, TNF-α production and NF-κB activities; it also alleviated endothelial inflammation, VE-cadherin integrity damage, apoptosis and barrier dysfunction, while miR-326 inhibition reversed these effects. MiR-326 inhibited TNF-α/NF-κB via targeting ADAM17. Further experiments demonstrated recombinant TNF-α reversed the inhibitory effect of HOTAIR-ASOs on LPS-triggered TNF-α/NF-κB activation and downstream endothelial injury, which were further mitigated by NF-κB or p38 MAPK inhibitors. In-vivo experiments in HOTAIR-knockout mice confirmed the role of HOTAIR/miR-326/ADAM17 in regulating NF-κB and p38 MAPK inflammation, with improved lung injury and survival following sepsis. DISCUSSION The HOTAIR/miR-326/ADAM17 axis is a key regulator of inflammation, endothelial injury and barrier dysfunction during sepsis via modulation of TNF-α/NF-κB signaling, providing new insights into the mechanisms underlying endothelial injury in sepsis.
Collapse
Affiliation(s)
- Junbing He
- The Department of Emergency, The First Affiliated Hospital, Jinan University, Guangzhou, GD, China
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, GD, China
| | - Zixuan Shao
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| | - Zhuoji Li
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| | - Yufu He
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, GD, China
| | - Jingqi Zhang
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| | - Haotian Zhong
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| | - Jiekai Li
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, GD, China
| | - Qinghua Liu
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang Affiliated Hospital of Sun Yat-sen University, Jieyang, GD, China
- The Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GD, China
| | - Yiming Shao
- The Department of Emergency, The First Affiliated Hospital, Jinan University, Guangzhou, GD, China
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, GD, China
| |
Collapse
|
12
|
Li M, Zhang Y, Xiang K, Su Z, Li X, Song H, Wu X, Mo D, Ren M, Yang S. Ant Colony-Inspired Adaptive Peptide Nanoregulators Remodeling the Endothelial Barrier to Alleviate Inflammatory Responses. ACS NANO 2025; 19:16829-16849. [PMID: 40277092 DOI: 10.1021/acsnano.5c01730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Endothelial barrier disruption exacerbates inflammation and tissue injury, posing dual challenges of reconstructing tight junctions and precisely regulating the local microenvironment. Traditional multidrug therapies often struggle with rapid drug leakage due to barrier dysfunction and limited synergy between therapeutic agents. Here, a strategy is proposed inspired by the "ant colony collaboration", developing an "all-in-one" conformationally adaptive peptide nanoregulator (VJP NPs) through the intelligent integration of three functional peptides. VJP NPs strategically harness the overexpression of vascular cell adhesion protein 1 (VCAM-1), enabling selective targeting of the inflamed endothelium under the guidance of the VHPK peptide while accumulating within the inflammatory microenvironment. The nanoregulators disassemble in response to high ROS levels, efficiently scavenging excess ROS. Simultaneously, they release the PMX peptide, competitively binding to the complement receptor C5aR to regulate the complement cascade. Furthermore, they release the JIP peptide to restore the endothelial barrier, reducing immune cell infiltration. As demonstrated in a mouse model of acute lung injury (ALI), VJP NPs markedly promote pulmonary vascular endothelial barrier repair, effectively attenuating inflammatory responses and alleviating tissue injury. This peptide-based nanoplatform boosts peptide delivery efficiency via a nanoprodrug strategy and amplifies synergistic therapeutic effects, highlighting its potential in endothelial barrier restoration and inflammation modulation.
Collapse
Affiliation(s)
- Meng Li
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Yining Zhang
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Kai Xiang
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Zhikang Su
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Xinyi Li
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Haoyue Song
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Xianghao Wu
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Dingqiang Mo
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Mingxing Ren
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| | - Sheng Yang
- College of Stomatology, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biom-edical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing 401147, China
| |
Collapse
|
13
|
Cui J, Deng Y, Li X, Gao L, Li J, Li Z, Qu H, Chu Y, Gu Y, Meng M, Li R. Herbal-based Xuebijing injection ameliorated vascular endothelial dysfunction via inhibiting ACLY/MYB/RIG-I axis in sepsis-associated lung injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156573. [PMID: 40088739 DOI: 10.1016/j.phymed.2025.156573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Excessive endothelial pro-inflammatory response is an early hallmark of sepsis-induced acute lung injury (ALI). Xuebijing (XBJ), a traditional Chinese medicine, is widely used in clinical practice to treat sepsis. PURPOSE This study aims to investigate the molecular mechanisms underlying the beneficial effects of XBJ. METHODS Plasma samples from septic patients treated with or without XBJ were collected and analyzed. The mouse model of sepsis was established by intraperitoneal injection of LPS (10 mg/kg). XBJ (10 ml/kg) was administrated intraperitoneally twice before LPS challenge and one time after LPS challenge. The severity of lung injury and the levels of inflammation and coagulation were evaluated. In vitro, HUVEC were used to explore the mechanisms of XBJ and its compounds in regulating the ACLY/MYB/RIG-I axis. RESULTS XBJ significantly reduced the plasma levels of endothelial cell (EC) damage-related markers in septic patients. The in vivo and in vitro data demonstrated that XBJ alleviated LPS-induced lung injury and reduced the levels of inflammation and coagulation activation in ECs. XBJ inhibited the phosphorylation-dependent activation of ATP citrate lyase (ACLY), thereby suppressing the acetylation-dependent nuclear translocation of the transcription factor MYB. The expression of retinoic acid inducible gene I (RIG-I) was downregulated, leading to the inhibition of NF-κB signaling and EC pro-inflammatory and coagulation activation, which further alleviated sepsis-associated ALI. Moreover, XBJ compounds Quercetin, Ferulic Acid, Kaempferol and Paeoniflorin all showed inhibitory effects on the activation of the downstream MYB/RIG-I signaling by binding to ACLY protein. CONCLUSION Our study revealed a novel regulatory mechanism of XBJ in sepsis-induced EC dysfunction and ALI. The compounds in XBJ inhibited the activity of ACLY, thereby inhibiting the expression of RIG-I by reducing the acetylation of transcription factor MYB, leading to the alleviation of EC activation and lung injury induced by sepsis. Our findings provide a theoretical basis for the clinical application of XBJ and shedding light on novel therapeutic targets for treating sepsis.
Collapse
Affiliation(s)
- Jiayin Cui
- Laboratory for Marine Drugs and Bioproducts of Qingdao Marine Science and Technology Center, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, PR China; Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yunxin Deng
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Xuechuan Li
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Liya Gao
- Yantai Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256600, PR China
| | - Jinrong Li
- Yantai Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256600, PR China
| | - Zuxian Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yufeng Chu
- Neurocritical Care Unit, Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwu Road, Jinan, Shandong, 250012, PR China
| | - Yuchao Gu
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Mei Meng
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| |
Collapse
|
14
|
Cunha CMC, Abreu VHP, Estato V, Soares GMV, Moraes BPT, Oliveira GP, Silva JD, Silva PL, Immler R, Rocco PR, Sperandio M, Silva AR, Bozza PT, Castro-Faria-Neto HC, Gonçalves-de-Albuquerque CF. Bosutinib mitigates inflammation in experimental sepsis. Eur J Clin Invest 2025:e70055. [PMID: 40292988 DOI: 10.1111/eci.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Sepsis, a leading cause of death globally, lacks targeted and effective treatment. Its pathophysiology involves unbalanced inflammation, marked by a high release of inflammatory mediators, leukocyte recruitment, vascular changes and dysfunction of the nervous and respiratory systems. Src family tyrosine kinases (SFK) play a critical role in immune responses, and their inhibition can modulate excessive inflammation. This study investigates the potential of bosutinib, an SFK inhibitor, as a treatment for sepsis. METHODS Clinical signs, survival rates, systemic and neuronal inflammatory responses, cell recruitment, lung function and cerebral microcirculation were analysed in mice treated with bosutinib (3 mg/kg) or DMSO/saline followed by cecal ligation and puncture (CLP)-induced sepsis. RESULTS Bosutinib treatment reduced the severity of sepsis, improved survival rates and reduced the levels of pro-inflammatory cytokines and chemokines in peritoneal lavage, plasma and brain tissue. It also reduced cellular infiltration and bacterial growth at the infection site and protected lung function by reducing diffuse alveolar damage. Using intravital microscopy and laser speckle techniques, bosutinib improved capillary density and blood perfusion and reduced leukocyte recruitment and adhesion in the cerebral microcirculation of septic animals. CONCLUSIONS Bosutinib pretreatment attenuated dysregulated inflammatory responses and neurovascular changes in experimental sepsis.
Collapse
Affiliation(s)
- C M C Cunha
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - V H P Abreu
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - V Estato
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - G M V Soares
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - B P T Moraes
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - G P Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J D Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - P L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - R Immler
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - P R Rocco
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - M Sperandio
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - A R Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - P T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - H C Castro-Faria-Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - C F Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Yang JY, Li LL, Fu SZ. Association analysis of sepsis progression to sepsis-induced coagulopathy: a study based on the MIMIC-IV database. BMC Infect Dis 2025; 25:573. [PMID: 40259248 PMCID: PMC12013014 DOI: 10.1186/s12879-025-10972-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Sepsis-induced coagulopathy (SIC) is a severe complication of sepsis, characterized by poor prognosis and high mortality. However, the predictive factors for the development of SIC in sepsis patients remain to be determined. The aim of this study was to develop an easy-to-use and efficient nomogram for predicting the risk of sepsis patients developing SIC in the intensive care unit (ICU), based on common indicators and complications observed at admission. METHODS A total of 12, 455 sepsis patients from the MIMIC database were screened and randomly divided into training and validation cohorts. In the training cohort, LASSO regression was used for variable selection and regularization. The selected variables were then incorporated into a multivariable logistic regression model to construct the nomogram for predicting the risk of sepsis patients developing sepsis-induced coagulopathy (SIC). The model's predictive performance was evaluated using the area under the receiver operating characteristic curve (AUC), and its calibration was assessed through a calibration curve. Additionally, decision curve analysis (DCA) was performed to evaluate the clinical applicability of the model. External validation was conducted using data from the ICU database of Xingtai People's Hospital. RESULTS Among the 12, 455 sepsis patients, 5, 145 (41. 3%) developed SIC. The occurrence of SIC was significantly associated with the SOFA score, red blood cell count, red cell distribution width (RDW), white blood cell count, platelet count, INR, and lactate levels. Additionally, hypertension was identified as a potential protective factor. A nomogram was developed to predict the risk of SIC, which showed an AUC of 0. 81 (95% CI: 0. 79-0. 83) in the training set, 0. 83 (95% CI: 0. 82-0. 84) in the validation set, and 0. 79 (95% CI: 0. 74-0. 84) in the external validation. The calibration curve of the nomogram showed good consistency between the observed and predicted probabilities of SIC. CONCLUSIONS The novel nomogram demonstrates excellent predictive performance for the incidence of SIC in ICU patients with sepsis and holds promise for assisting clinicians in early identification and intervention of SIC. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- Jian-Yue Yang
- Department of Critical Care Medicine, Xingtai People's Hospital, Hebei, 054001, China
| | - Li-Li Li
- Department of Critical Care Medicine, Xingtai People's Hospital, Hebei, 054001, China
| | - Su-Zhen Fu
- Department of Critical Care Medicine, Xingtai People's Hospital, Hebei, 054001, China.
| |
Collapse
|
16
|
Wang Y, Zhang Z, Qu X, Zhou G. Role of the endothelial cell glycocalyx in sepsis-induced acute kidney injury. Front Med (Lausanne) 2025; 12:1535673. [PMID: 40255592 PMCID: PMC12006053 DOI: 10.3389/fmed.2025.1535673] [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: 11/27/2024] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Sepsis-induced acute kidney injury (S-AKI) is a common complication of sepsis. It occurs at high incidence and is associated with a high level of mortality in the intensive care unit (ICU). The pathophysiologic mechanisms underlying S-AKI are complex, and include renal vascular endothelial cell dysfunction. The endothelial glycocalyx (EG) is a polysaccharide/protein complex located on the cell membrane at the luminal surface of vascular endothelial cells that has anti-inflammatory, anti-thrombotic, and endothelial protective effects. Recent studies have shown that glycocalyx damage plays a causal role in S-AKI progression. In this review, we first describe the structure, location, and basic function of the EG. Second, we analyze the underlying mechanisms of EG degradation in sepsis and S-AKI. Finally, we provide a summary of the potential therapeutic strategies that target the EG.
Collapse
Affiliation(s)
- Yixun Wang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
- Yichang Sepsis Clinical Research Center, Yichang, Hubei, China
| | - Zhaohui Zhang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
- Yichang Sepsis Clinical Research Center, Yichang, Hubei, China
| | - Xingguang Qu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
- Yichang Sepsis Clinical Research Center, Yichang, Hubei, China
| | - Gaosheng Zhou
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
- Yichang Sepsis Clinical Research Center, Yichang, Hubei, China
| |
Collapse
|
17
|
Lee S, Wischmeyer PE, Mintz CD, Serbanescu MA. Recent Insights into the Evolving Role of the Gut Microbiome in Critical Care. Crit Care Clin 2025; 41:379-396. [PMID: 40021286 DOI: 10.1016/j.ccc.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
This review explores the evolving understanding of gut microbiota's role in critical illness, focusing on how acute illness and exposures in intensive care unit (ICU) environment negatively impact the gut microbiota and the implications of these changes on host responses in critically-ill patients. Focusing on recent findings from clinical and preclinical studies, we discuss the effects of inflammation, enteral nutrient deprivation, and antibiotics on gut microbial dynamics. This review aims to enhance comprehension of microbial dynamics in the ICU and their implications for clinical outcomes and therapeutic strategies.
Collapse
Affiliation(s)
- Seoho Lee
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Phipps 455 1800 Orleans Street, Baltimore, MD 21212, USA
| | - Paul E Wischmeyer
- Department of Anesthesiology, Duke University School of Medicine, 5692 HAFS Box 3094, 2301 Erwin Road, Durham, NC 27710, USA
| | - Cyrus D Mintz
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Phipps 455 1800 Orleans Street, Baltimore, MD 21212, USA
| | - Mara A Serbanescu
- Department of Anesthesiology, Duke University School of Medicine, 5692 HAFS Box 3094, 2301 Erwin Road, Durham, NC 27710, USA.
| |
Collapse
|
18
|
Wang D, Wang S, Liu J, Shi X, Xiong T, Li R, Wei W, Ji L, Huang Q, Gong X, Ai K. Nanomedicine Penetrating Blood-Pancreas Barrier for Effective Treatment of Acute Pancreatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413925. [PMID: 39950925 PMCID: PMC11967758 DOI: 10.1002/advs.202413925] [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: 10/29/2024] [Revised: 02/01/2025] [Indexed: 04/05/2025]
Abstract
Acute pancreatitis (AP) is a primary contributor to hospitalization and in-hospital mortality worldwide. Targeted elimination of mitochondrial reactive oxygen species (mtROS) within pancreatic acinar cells (PACs) represents an ideal strategy for treating AP. However, existing drugs fail to overcome the physiological barriers of the pancreas to effectively reach PACs mitochondria due to the trade-off between conventional positively charged mitochondrial-targeting groups and their inability to penetrate the blood-pancreas barrier (BPB). Here, a tungsten-based heteropolyacid nano-antioxidant (mTWNDs) is introduced, co-modified with tannic acid (TA) and melanin, enabling site-specific clearance of mtROS in PACs, offering a highly effective treatment for AP. TA exhibits a strong affinity for proline-rich type III collagen and the mitochondrial outer membrane protein TOM20. This unique property allows mTWNDs to traverse the damaged BPB-exposing type III collagen to reach PACs and subsequently penetrate mitochondria for targeted mtROS elimination. In cerulein-induced AP mice, mTWNDs reversed AP at 1/50th the dose of N-acetylcysteine, suppressing PACs apoptosis and inflammation by blocking the stimulator of the interferon genes pathway activation in macrophage. This study establishes a mitochondrial-targeting antioxidant nanomedicine strategy for AP treatment.
Collapse
Affiliation(s)
- Dan Wang
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Shuya Wang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Jinjin Liu
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Xiaojing Shi
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Tingli Xiong
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Ruishi Li
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Wei Wei
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Liandong Ji
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Qiong Huang
- Department of PharmacyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Xuejun Gong
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Kelong Ai
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of EducationXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
19
|
Turcato G, Filippi L, Zaboli A, Ferretto P, Milazzo D, Maggi M, Stefani F, Parodi M, Marchetti M, Wiedermann CJ. Relationship between fluid bolus administration and the prognostic role of serum albumin in patients with sepsis. Am J Med Sci 2025; 369:451-459. [PMID: 39638034 DOI: 10.1016/j.amjms.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Serum albumin plays a pivotal role in the exchange between interstitial and vascular compartments, and reduced levels of this biomarker appear to be associated with negative prognosis in septic patients. The correlation between the volume effect in sepsis therapy and the kinetics of serum albumin is unclear. AIM To investigate the relationship between serum albumin and fluid bolus in relation to its prognostic role in septic patients. METHODS A single-center prospective observational study conducted from September 2022 to February 2024. All patients with sepsis admitted from the Emergency Department to the Intermediate Medical Care Unit (IMCU) were considered. Post-fluid bolus serum albumin was obtained after fluid bolus. The albumin value was correlated with the volume effect of the fluid bolus, and multivariate models were performed to evaluate its potential independent effect on 30-day mortality. RESULTS 179 patients were enrolled. Pre-fluid bolus serum albumin was 2.55 g/dL (SD 0.51) with a multivariate OR for 30-day mortality of 1.170 (95 % CI 1.055-1.297, p = 0.003). After the fluid bolus, which resulted in a fluid balance percentage of +23.1 % (SD 7.1) and a mean Fractional Plasma Dilution of -0.48 (SD 0.18), albumin showed a mean decrease of -0.28 g/dL (SD 0.28) with a multivariate OR for 30-day mortality of 1.198 (95 % CI 1.065-1.348, p = 0.003). Post-fluid bolus albumin was negatively correlated with cumulative fluid balance and hemodilution. CONCLUSIONS The volume effect of fluid bolus is correlated with a decrease in serum albumin, and low albumin levels are associated with a high risk of mortality.
Collapse
Affiliation(s)
- Gianni Turcato
- Department of Internal Medicine, Intermediate Care Unit, Hospital Alto Vicentino (AULSS-7), Santorso, Italy; UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy.
| | - Lucia Filippi
- Department of Internal Medicine, Intermediate Care Unit, Hospital Alto Vicentino (AULSS-7), Santorso, Italy
| | - Arian Zaboli
- Innovation, Research and Teaching Service (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Bolzano, Italy
| | - Paolo Ferretto
- Department of Internal Medicine, Intermediate Care Unit, Hospital Alto Vicentino (AULSS-7), Santorso, Italy
| | - Daniela Milazzo
- Department of Internal Medicine, Intermediate Care Unit, Hospital Alto Vicentino (AULSS-7), Santorso, Italy
| | - Michael Maggi
- Department of Internal Medicine, Intermediate Care Unit, Hospital Alto Vicentino (AULSS-7), Santorso, Italy
| | - Francesca Stefani
- Department of Internal Medicine, Intermediate Care Unit, Hospital Alto Vicentino (AULSS-7), Santorso, Italy
| | - Marta Parodi
- Department of Internal Medicine, Intermediate Care Unit, Hospital Alto Vicentino (AULSS-7), Santorso, Italy
| | - Massimo Marchetti
- Department of Internal Medicine, Intermediate Care Unit, Hospital Alto Vicentino (AULSS-7), Santorso, Italy
| | | |
Collapse
|
20
|
Ge S, Wang XH, Fan J, Liu H, Xin Y, Li X, Yu Y, Yang YW, Gao H. A Dual-Pipeline Lactate Removal Strategy to Reverse Vascular Hyperpermeability for the Management of Lipopolysaccharide-Induced Sepsis. Adv Healthc Mater 2025; 14:e2403592. [PMID: 39887647 DOI: 10.1002/adhm.202403592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/22/2025] [Indexed: 02/01/2025]
Abstract
Sepsis is an underappreciated yet severe threat to human life, marked by organ dysfunction and high mortality resulting from disordered inflammatory responses to blood infection. Unfortunately, no specific drugs are available for effective sepsis treatment. As a pivotal biomarker for sepsis, lactate levels are closely related to vascular permeability and sepsis-associated mortality. Herein, a dual-pipeline lactate removal strategy is reported from circulating blood to ameliorate vascular permeability and lipopolysaccharide (LPS)-induced sepsis. This is achieved by formulating lactate oxidase (LOX)-encapsulated hollow manganese dioxide (HMnO2) nanohybrids (LOX@HMnO2-P[5]A) bearing pillar[5]arene (P[5]A) macrocycle with excellent host-guest properties. The highly biocompatible nanohybrids enable direct lactate consumption through LOX catalytic degradation and block lactate production by P[5]A-mediated LPS trapping, allowing for dual-pipeline lactate removal to maximize the reversal of lactate-mediated vascular hyperpermeability. Besides, HMnO2 cores decompose hydrogen peroxide produced from lactate oxidation into oxygen, further contributing to lactate consumption and mitigating the hypoxic inflammatory environment. In vivo investigations demonstrate that intravenous administration of LOX@HMnO2-P[5]A nanohybrids with extended blood circulation can effectively ameliorate endothelial barrier dysfunction, inflammatory responses, and multiple organ injury, ultimately improving survival outcomes in LPS-induced sepsis. Taken together, this dual-pipeline lactate removal strategy offers a promising approach for efficient sepsis treatment.
Collapse
Affiliation(s)
- Shuangfeng Ge
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China
| | - Xing-Huo Wang
- Institute for Sustainable Energy and Resources, College of Chemistry and Chemical Engineering, Qingdao University, 308 Ningxia Road, Qingdao, 266071, P. R. China
| | - Juntao Fan
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China
| | - Haofei Liu
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China
| | - Youtao Xin
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China
| | - Xiaohui Li
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China
| | - Yunjian Yu
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China
| | - Ying-Wei Yang
- College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
| | - Hui Gao
- State Key Laboratory of Separation Membranes and Membrane Processes & Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China
| |
Collapse
|
21
|
Hu B, Jiang J, Pan W, Chung CS, Gray C, Chen Y, Guo J, Ayala A. V-domain Ig Suppressor of T cell Activation Expression During Hemorrhage or Sepsis-Induced Acute Respiratory Distress Syndrome: Insights From a Mouse Model. J Surg Res 2025; 308:73-85. [PMID: 40086004 DOI: 10.1016/j.jss.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/15/2025] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Acute respiratory distress syndrome (ARDS) is a life-threatening pulmonary condition with significant mortality, largely due to a lack of therapeutic interventions grounded in its molecular pathophysiology. Immune checkpoint regulators, such as the V-domain Ig Suppressor of T cell Activation (VISTA), may provide novel immunotherapeutic strategies for ARDS by modulating the immune response, a concept extensively explored in cancer and autoimmune diseases. Investigating VISTA in the context of ARDS could unveil new therapeutic avenues. METHODS We used a mouse model of indirect ARDS by subjecting C57BL/6J mice to hemorrhage followed by cecal ligation and puncture. Systemic and localized inflammatory conditions were assessed using samples from blood, lung, and peritoneal fluid. Lung pathology was quantified by scoring hematoxylin and eosin-stained sections. Flow cytometry, enzyme-linked immunosorbent assay, and reverse transcription-polymerase chain reaction analyses concentrated on macrophages, neutrophils, endothelial cells, and epithelial cells to elucidate VISTA expression patterns. RESULTS Hemorrhage or cecal ligation and puncture-treated mice exhibited hallmark symptoms of indirect ARDS, including elevated levels of inflammatory cytokines and chemokines. Notably, VISTA expression was substantially upregulated on various cell types, including blood monocytes, lung macrophages, and both circulating and lung-infiltrating neutrophils, as well as on pulmonary epithelial cells and endothelial cells. CONCLUSIONS Our model replicates critical inflammatory and physiologic changes leading to ARDS, with the elevated expression of VISTA on immune and parenchymal cells suggesting its central involvement in lung injury. The findings propose VISTA as both a potential biomarker for lung damage and as a promising target for ARDS therapy.
Collapse
Affiliation(s)
- Baoji Hu
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China; Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China; Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island
| | - Jihong Jiang
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Pan
- Division of Infectious Diseases, Rhode Island Hospital/ The Warren Alpert Medical School at Brown University, Providence, Rhode Island
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island
| | - Chyna Gray
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island
| | - Yaping Chen
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island
| | - Jianrong Guo
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China; Department of Anesthesiology, Shanghai Gongli Hospital, Naval Military Medical University, Shanghai, China.
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital/ The Warren Alpert School at Medicine at Brown University, Providence, Rhode Island.
| |
Collapse
|
22
|
Bridges CA, Fu L, Yeow J, Huang X, Jackson M, Kuchel RP, Sterling JD, Baker SM, Lord MS. The interplay between endothelial glycocalyx maturity and both the toxicity and intracellular uptake of charged nanoparticles. Acta Biomater 2025; 196:293-306. [PMID: 40058617 DOI: 10.1016/j.actbio.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
Nanoparticles are widely studied for delivering treatments to target tissues, but few have reached clinical use. Most nanoparticles encounter blood vessels on their way to target tissues. The inner surface of these vessels is lined with endothelial cells covered by a glycocalyx, an extracellular matrix rich in anionic glycans. The role of the glycocalyx in nanoparticle interactions is not well understood. Here, we demonstrate that endothelial cells need extended culture times to synthesize a mature glycocalyx. Our research shows that branched polyethyleneimine functionalized gold nanoparticles bind to endothelial cells expressing either a developing or mature glycocalyx, with the interaction involving hyaluronan and heparan sulfate. These nanoparticles are subsequently internalized. Similar results were seen with poly(L-arginine). A mature glycocalyx protects cells by reducing the toxicity of these cationic nanoparticles. In contrast, lipoic acid-functionalized gold nanoparticles are internalized by cells with a developing glycocalyx, but not a mature one. Poly(L-glutamic acid) only interacts with cells when major glycans in the glycocalyx are degraded. These findings highlight the complex relationship between nanoparticle charge and structure, and their effects on toxicity, binding, and uptake by endothelial cells. This offers important insights for improving nanoparticle interactions with blood vessels in health and disease. STATEMENT OF SIGNIFICANCE: Endothelial cells lining blood vessels form a barrier through which nanoparticles must cross to reach target tissues. These cells are covered with a layer called the glycocalyx, which is rich in anionic glycans. However, the role of the glycocalyx in how nanoparticles interact with cells remains underexplored. Our research revealed that cells with a mature glycocalyx internalize cationic nanoparticles and experience reduced cytotoxicity. Conversely, a mature glycocalyx prevents anionic nanoparticles from entering cells. These results suggest that the structure of both the nanoparticles and the glycocalyx should be considered in future studies to improve the use of nanoparticles for medical applications.
Collapse
Affiliation(s)
- Claire A Bridges
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lu Fu
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jonathan Yeow
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xiaojing Huang
- Molecular Surface Interaction Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Miriam Jackson
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Rhiannon P Kuchel
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - James D Sterling
- College of Innovation, Entrepreneurship, and Economic Development, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | | | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
23
|
Liu J, Zeng X, Lin YQ, Peng YS, Liu L, Chen SH, Du YH. The Marine Compound Isaridin E Ameliorates Lipopolysaccharide-Induced Vascular Endothelial Inflammation via the Downregulation of the TLR4/NF-κB Signaling Pathway. Mar Drugs 2025; 23:145. [PMID: 40278266 PMCID: PMC12028784 DOI: 10.3390/md23040145] [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: 02/28/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Isaridin E, a cyclodepsipeptide derived from the marine fungus Beauveria felina (SYSU-MS7908), has been demonstrated to possess multiple biological properties. In this study, we employed both lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells (HUVECs) and a LPS-induced murine endotoxemia model to investigate its anti-inflammatory effects. Our results revealed that isaridin E suppressed the expression of pro-inflammatory cytokines and adhesion molecules in a concentration dependent manner, while also reducing monocyte adhesion to endothelial cells. Furthermore, this compound attenuated vascular hyperpermeability and inflammatory cell infiltration in the lungs, as well as preserving the integrity of the aortic and pulmonary tissues. At the molecular level, isaridin E was found to downregulate TLR4 expression, increase IκBα levels, and inhibit the LPS-induced phosphorylation and nuclear translocation of NF-κB p65. In conclusion, our findings indicate that isaridin E exerts robust anti-inflammatory effects in LPS-induced endotoxemia through the suppression of the TLR4/NF-κB signaling axis, positioning it as a promising therapeutic candidate for vascular inflammatory disorders.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Department of Pharmacy, The Second Clinical Medical College, Southern Medical University, Guangzhou 510280, China
| | - Xin Zeng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yu-Quan Lin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yu-Sheng Peng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Lan Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Sen-Hua Chen
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Yan-Hua Du
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
24
|
Fang J, Ding H, Huang J, Liu W, Hong T, Yang J, Wu Z, Li Z, Zhang S, Liu P, Fang Y, Wu J, Li X, Lin J. Mac-1 blockade impedes adhesion-dependent neutrophil extracellular trap formation and ameliorates lung injury in LPS-induced sepsis. Front Immunol 2025; 16:1548913. [PMID: 40226627 PMCID: PMC11985419 DOI: 10.3389/fimmu.2025.1548913] [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: 12/20/2024] [Accepted: 03/07/2025] [Indexed: 04/15/2025] Open
Abstract
Background Sepsis is a common critical condition that can lead to multiple organ injury. Sepsis-induced acute respiratory distress syndrome (ARDS) is frequently an important cause of poor prognosis and is associated with high mortality rates, despite existing therapeutic interventions. Neutrophil infiltration and extracellular traps (NET) are implicated in acute lung injury (ALI) and ARDS following sepsis. As circulating neutrophils infiltrate infected tissues, they come into direct contact with vascular endothelial cells (ECs). Although the ability of NETs to induce endothelial damage is well established, the specific role of direct EC-neutrophil interactions in NET formation and lung injury during sepsis is not fully understood. Methods In this study, NET formation was assessed when neutrophils were co-culture with ECs or separated from them and stimulated with phorbol 12-myristate 13-acetate (PMA), lipopolysaccharide (LPS), lipoteichoic acid (LTA), or septic plasma. Results We found that adhesion of neutrophils on ECs is critical in NET formation in response to LPS, LTA, or septic plasma in vitro. Blocking the macrophage-1 antigen (Mac-1) impeded NET formation, while inhibiting P-selectin glycoprotein ligand-1 (PSGL-1) or leukocyte function-associated antigen-1 (LFA-1) did not. This adhesion-dependent NET formation was reliant on the influx of extracellular calcium and peptidylarginine deiminase 4 (PAD4)-mediated citrullination of histone H3. However, Mac-1 blockade did not alter calcium influx. In a murine model of LPS-induced sepsis, Mac-1 blockade reduced NET release, lowered inflammatory cytokine levels, mitigated endothelial damage, and attenuated lung injury. Conclusion Our findings offer insights into the critical role of EC-neutrophil direct contact in NET formation during sepsis and propose Mac-1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jinhua Fang
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hongguang Ding
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiaqi Huang
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Wang Liu
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Tiantian Hong
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Junxian Yang
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhiwei Wu
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhuo Li
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shiying Zhang
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Peimin Liu
- Department of Nephrology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying Fang
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Jianhua Wu
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiangguo Lin
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
25
|
Zeng QB, Peng EL, Zhou Y, Lin QW, Zhong LC, He LP, Zhang NQ, Song JC. Explainable machine learning model for predicting septic shock in critically sepsis patients based on coagulation indexes: A multicenter cohort study. Chin J Traumatol 2025:S1008-1275(25)00032-X. [PMID: 40246624 DOI: 10.1016/j.cjtee.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/19/2024] [Accepted: 08/23/2024] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Septic shock is associated with high mortality and poor outcomes among sepsis patients with coagulopathy. Although traditional statistical methods or machine learning (ML) algorithms have been proposed to predict septic shock, these potential approaches have never been systematically compared. The present work aimed to develop and compare models to predict septic shock among patients with sepsis. METHODS It is a retrospective cohort study based on 484 patients with sepsis who were admitted to our intensive care units between May 2018 and November 2022. Patients from the 908th Hospital of Chinese PLA Logistical Support Force and Nanchang Hongdu Hospital of Traditional Chinese Medicine were respectively allocated to training (n=311) and validation (n=173) sets. All clinical and laboratory data of sepsis patients characterized by comprehensive coagulation indexes were collected. We developed 5 models based on ML algorithms and 1 model based on a traditional statistical method to predict septic shock in the training cohort. The performance of all models was assessed using the area under the receiver operating characteristic curve and calibration plots. Decision curve analysis was used to evaluate the net benefit of the models. The validation set was applied to verify the predictive accuracy of the models. This study also used SHapley Additive exPlanations method to assess variable importance and explain the prediction made by a ML algorithm. RESULTS Among all patients, 37.2% experienced septic shock. The characteristic curves of the 6 models ranged from 0.833 to 0.962 and 0.630 to 0.744 in the training and validation sets, respectively. The model with the best prediction performance was based on the support vector machine (SVM) algorithm, which was constructed by age, tissue plasminogen activator-inhibitor complex, prothrombin time, international normalized ratio, white blood cells, and platelet counts. The SVM model showed good calibration and discrimination and a greater net benefit in decision curve analysis. CONCLUSION The SVM algorithm may be superior to other ML and traditional statistical algorithms for predicting septic shock. Physicians can better understand the reliability of the predictive model by SHapley Additive exPlanations value analysis.
Collapse
Affiliation(s)
- Qing-Bo Zeng
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Intensive Care Unit, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, 330038, China
| | - En-Lan Peng
- Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Ye Zhou
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Qing-Wei Lin
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Lin-Cui Zhong
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Long-Ping He
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China
| | - Nian-Qing Zhang
- Intensive Care Unit, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, 330038, China
| | - Jing-Chun Song
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, 330002, China; Department of Critical Care Medicine, Changcheng Hospital Affiliated to Nanchang University, Nanchang, 330002, China.
| |
Collapse
|
26
|
Li Y, Ren S, Zhou S. Advances in sepsis research: Insights into signaling pathways, organ failure, and emerging intervention strategies. Exp Mol Pathol 2025; 142:104963. [PMID: 40139086 DOI: 10.1016/j.yexmp.2025.104963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Sepsis is a complex syndrome resulting from an aberrant host response to infection. A hallmark of sepsis is the failure of the immune system to restore balance, characterized by hyperinflammation or immunosuppression. However, the net effect of immune system imbalance and the clinical manifestations are highly heterogeneous among patients. In recent years, research interest has shifted from focusing on the pathogenicity of microorganisms to the molecular mechanisms of host responses which is also associated with biomarkers that can help early diagnose sepsis and guide treatment decisions. Despite significant advancements in medical science, sepsis remains a major challenge in healthcare, contributing to substantial morbidity and mortality worldwide. Further research is needed to improve our understanding of this condition and develop novel therapies to improve outcomes for patients with sepsis. This review explores the related signal pathways of sepsis and underscores recent advancements in understanding its mechanisms. Exploration of diverse biomarkers and the emerging concept of sepsis endotypes offer promising avenues for precision therapy in the future.
Collapse
Affiliation(s)
- Yehua Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, PR China.
| | - Siying Ren
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Shen'ao Zhou
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, CAS. Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
27
|
Fu Y, Gong T, Loughran PA, Li Y, Billiar TR, Liu Y, Wen Z, Fan J. Roles of TLR4 in macrophage immunity and macrophage-pulmonary vascular/lymphatic endothelial cell interactions in sepsis. Commun Biol 2025; 8:469. [PMID: 40119011 PMCID: PMC11928643 DOI: 10.1038/s42003-025-07921-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
In sepsis, acute lung injury (ALI) is a severe complication and a leading cause of death, involving complex mechanisms that include cellular and molecular interactions between immune and lung parenchymal cells. In recent decades, the role of Toll-like receptor 4 (TLR4) in mediating infection-induced inflammation has been extensively studied. However, how TLR4 facilitates interactions between innate immune cells and lung parenchymal cells in sepsis remains to be fully understood. This study aims to explore the role of TLR4 in regulating macrophage immunity and metabolism in greater depth. It also seeks to reveal how changes in these processes affect the interaction between macrophages and both pulmonary endothelial cells (ECs) and lymphatic endothelial cells (LECs). Using TLR4 knockout mice and the combined approaches of single-cell RNA sequencing and experimental validation, we demonstrate that in sepsis, TLR4-deficient macrophages upregulate Abca1, enhance cholesterol efflux, and reduce glycolysis, promoting M2 polarization and attenuating inflammation. These metabolic and phenotypic shifts significantly affect their interactions with pulmonary ECs and LECs. Mechanistically, we uncovered that TLR4 operates through multiple pathways in endothelial dysfunction: macrophage TLR4 mediates inflammatory damage to ECs/LECs, while endothelial TLR4 both directly sensitizes cells to lipopolysaccharide-induced injury and determines their susceptibility to macrophage-derived inflammatory signals. These findings reveal the complex role of TLR4 in orchestrating both immune-mediated and direct endothelial responses during sepsis-induced ALI, supporting that targeting TLR4 on multiple cell populations may present an effective therapeutic strategy.
Collapse
Affiliation(s)
- Yu Fu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Ting Gong
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, Shenzhen, 518110, China
| | - Patricia A Loughran
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Yuehua Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, Shenzhen, 518110, China
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
28
|
Zhang J, Cao Y, Shu W, Dong S, Sun Y, Ma X. Neutrophil-derived heparin-binding protein increases endothelial permeability in acute lung injury by promoting TRIM21 and the ubiquitination of P65. Cell Biol Toxicol 2025; 41:55. [PMID: 40045003 PMCID: PMC11882632 DOI: 10.1007/s10565-025-10005-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025]
Abstract
Acute lung injury (ALI), which poses a significant public health threat, is commonly caused by sepsis. ALI is associated with permeability and glycolysis changes in pulmonary microvascular endothelial cells. Our study demonstrates that heparin-binding protein (HBP), released from neutrophils during sepsis, exacerbates endothelial permeability and glycolysis, thereby triggering ALI. Through coimmunoprecipitation and mass spectrometry, TRIM21 was identified as a HBP interaction partner. Notably, HBP enhances the protein stability of TRIM21 by inhibiting K48 ubiquitination. TRIM21 binds to and promotes K63-linked ubiquitination of P65, facilitating its nuclear translocation. TRIM21 regulates HPMEC permeability and glycolysis in a manner dependent on P65 nuclear translocation. HBP stabilizes TRIM21 and enhances TRIM21 interactions with P65. Rescue experiments conducted in vivo and in vitro demonstrate that modulation of endothelial permeability and glycolysis by HBP is predominantly mediated through the TRIM21-P65 axis. Our results suggest that targeting the HBP/TRIM21/P65 axis is a novel therapeutic strategy to ameliorate ALI.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yong Cao
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Wenqi Shu
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Senxiao Dong
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yini Sun
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Xiaochun Ma
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, People's Republic of China.
| |
Collapse
|
29
|
Ma D, Wan X, Yang H, Yang L, Peng A, Yuan Q, Li Y, Xu S. Scrub Typhus Combined With Septic Shock Disseminated Intravascular Coagulation and Significant Hyperfibrinolysis: A Case Report and Review of the Literature. Case Rep Infect Dis 2025; 2025:1931423. [PMID: 40226813 PMCID: PMC11986193 DOI: 10.1155/crdi/1931423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction: Scrub typhus is an acute infectious disease caused by Orientia tsutsugamushi, whose pathophysiology is characterized by systemic small-vessel vasculitis. Its high misdiagnosis rate stems from its nonspecific clinical features. If not diagnosed and treated in time, patients may rapidly progress to multiorgan dysfunction syndrome (MODS) or even disseminated intravascular coagulation (DIC), posing a severe threat to life. Case Presentation: The patient was a 68-year-old male with "recurrent fever and dry cough for six days." He was admitted to the hospital with a diagnosis of scrub typhus. After admission, he developed severe acute respiratory distress syndrome (ARDS), MODS, septic shock, DIC with thrombocytopenia, hypofibrinogenemia, significant hyperfibrinolysis, and myocardial depression. The patient improved following treatment with doxycycline, moxifloxacin, renal replacement therapy, blood transfusion, antifibrinolysis, invasive mechanical ventilation, and other supportive therapies. The patient's coagulation profile in DIC caused by scrub typhus demonstrated significant hyperfibrinolysis, differing from that of garden-variety sepsis, and no similar cases were identified in a search of medical literature/databases. Conclusion: The fibrinolytic system in DIC caused by scrub typhus is excessively active, and antifibrinolytic therapy may benefit such patients. Further research on the distinct coagulation abnormalities in scrub typhus-associated DIC would be highly valuable compared to sepsis-associated DIC.
Collapse
Affiliation(s)
- Dewen Ma
- Intensive Care Unit, People's Hospital, Pu'er City, Yunnan, China
| | - Xiaohong Wan
- Intensive Care Unit, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Haihui Yang
- Department of Cardiology, People's Hospital, Pu'er City, Yunnan, China
| | - Liying Yang
- Intensive Care Unit, People's Hospital, Pu'er City, Yunnan, China
| | - Ankang Peng
- Department of Cardiology, People's Hospital, Pu'er City, Yunnan, China
| | - Quping Yuan
- Intensive Care Unit, People's Hospital, Pu'er City, Yunnan, China
| | - You Li
- Intensive Care Unit, People's Hospital, Pu'er City, Yunnan, China
| | - Shunhang Xu
- Department of Pediatrics, People's Hospital, Pu'er City, Yunnan, China
| |
Collapse
|
30
|
Chen X, Yang P. S100A4 participates in sepsis-induced endothelial cell inflammatory response and barrier damage by binding to BRD4. Clin Hemorheol Microcirc 2025; 89:302-311. [PMID: 40433870 DOI: 10.1177/13860291251313579] [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] [Indexed: 05/29/2025]
Abstract
BackgroundResearch has shown that S100A4 is upregulated in endothelial cells when exposed to serum from septic patients. This article aims to explore the role of endogenous S100A4 in lipopolysaccharide (LPS)-induced endothelial cells.MethodsA septic HUVECs injury model was established using LPS and transfected with siRNA-S100A4 or Ov-BRD4 plasmid. Targets of S100A4 were predicted using online databases, and immunoprecipitation (IP) was used to verify the binding of S100A4 and targets. Cell viability, levels of apoptosis, and the expression of apoptosis-related proteins were measured to assess cell injury. Transendothelial electrical resistance (TER) and the expression of tight junction proteins were measured to assess cell barrier function. Assessed the inflammatory response by measuring the levels of inflammatory factors, the adhesion of THP-1 monocytes, and the expression of adhesion molecules.ResultsDatabase prediction and IP verification indicated that S100A4 could bind to BRD4 in LPS-induced HUVECs, and the expression of S100A4 and BRD4 was increased in LPS-induced HUVECs. Interference with S100A4 significantly enhanced the cell viability and TER, reduced the apoptosis, TNFα, IL-1β, and IL-6 levels, and THP-1 adhesion number in LPS-treated HUVECs. Additionally, interference with S100A4 upregulated the expression of Bcl2, ZO-1, occludin, and claudin-4 proteins, and downregulated the expression of BRD4, Bax, cleaved caspase-3, ICAM-1, VCAM-1, and E-selectin proteins in LPS-induced HUVECs. However, overexpression of BRD4 significantly attenuated the protective effect of interfering with S100A4 on LPS-induced HUVECs.ConclusionS100A4 is involved in LPS-induced inflammatory response and barrier damage in HUVECs by binding to BRD4.
Collapse
Affiliation(s)
- Xiujing Chen
- Department of Infectious Diseases, Yueqing People's Hospital, Wenzhou City, Zhejiang, China
| | - Ping Yang
- Department of Critical Care Medicine, the second affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Cao X, Ma R, Wang Y, Huang Y, You K, Zhang L, Li H, Feng G, Chen T, Wang D, Sun K, Fang H, Shen X. Paeoniflorin protects the vascular endothelial barrier in mice with sepsis by activating RXRα signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156384. [PMID: 39826282 DOI: 10.1016/j.phymed.2025.156384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Sepsis is a life-threatening condition characterized by organ dysfunction resulting from the body's aberrant response to infection. A primary indicator of early sepsis is vascular leakage due to endothelial injury. The immunomodulatory effects of paeoniflorin are well established. However, its effect on vascular endothelial injury in sepsis remains to be verified. METHODS The sepsis model was established by cecal ligation and puncture (CLP), along with simultaneous administration of paeoniflorin. The therapeutic effectiveness of paeoniflorin was evaluated by assessing the survival rate, the bacterial load in blood and the histopathological lung tissue injury. The pulmonary vascular endothelial barrier integrity was assessed using immunofluorescence, western blot, Evans blue dye, and qPCR. Human umbilical vein endothelial cells (HUVECs) were used for in vitro validation and exploration of the underlying mechanisms. RESULTS The CLP mice exhibited significant damage to pulmonary tissue and breakdown of endothelial barrier. Administration of paeoniflorin markedly improved survival rates, mitigated lung injury, and preserved the integrity of the pulmonary vascular endothelial barrier in CLP mice which was confirmed by in vitro experiments. Pharmacological mechanism studies showed that the protective effects of paeoniflorin on the vascular endothelium was achieved through activation of RXRα signaling, which could be reversed by RXRα knockdown. CONCLUSION Our experiments demonstrates the protective effect of paeoniflorin on the vascular endothelial barrier through activation of the RXRα, thereby offering potential therapeutic options for sepsis treatment. We also identified RXRα as a novel transcription factor for VE-cadherin, providing a potential new intervention target for vascular endothelial barrier damage in sepsis.
Collapse
Affiliation(s)
- Xinyue Cao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Ruihua Ma
- Department of Anesthesiology, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China, 215125
| | - Yirui Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Yuran Huang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Keyuan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Lijie Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Haidong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Guize Feng
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Tongqing Chen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Dong Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Keyu Sun
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, PR China, 201199.
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, PR China, 200032; Department of Anesthesiology, Shanghai Geriatic Medical Center, Shanghai, PR China, 201104; Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, PR China, 201199.
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China.
| |
Collapse
|
32
|
Williams JG, Whitney JE, Weiss SL, Varisco BM, Yehya N, Atreya MR. Derivation and Validation of a Clinical and Endothelial Biomarker Risk Model to Predict Persistent Pediatric Sepsis-Associated Acute Respiratory Dysfunction. CHEST CRITICAL CARE 2025; 3:100120. [PMID: 40242498 PMCID: PMC12001826 DOI: 10.1016/j.chstcc.2024.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
BACKGROUND Sepsis-associated ARDS results in high morbidity and mortality in children. However, heterogeneity among patients makes identifying those at risk of persistent acute respiratory dysfunction challenging. Endothelial dysfunction is a key feature of ARDS pathophysiologic characteristics, contributing to lung injury in sepsis. Incorporating endothelial biomarkers into risk models may enhance prediction of those with persistent acute respiratory dysfunction. RESEARCH QUESTION Can clinical variables and endothelial biomarkers measured early in the course of sepsis predict risk of persistent acute respiratory dysfunction among critically ill children? STUDY DESIGN AND METHODS This was a multicenter derivation and single center test cohort study of prospectively enrolled children with sepsis. The derivation cohort was split into training and holdout validation sets. We trained TreeNet (Minitab, LLC) and classification and regression tree (CART) models using clinical and endothelial biomarkers measured on day 1 of septic shock to predict risk of sepsis-associated acute respiratory dysfunction (SA ARD) on day 3. The performance of the CART model was tested in the holdout validation data set and in the independent test cohort. RESULTS In the derivation (n = 625) and test (n = 162) cohorts, children with day 3 SA ARD showed increased mortality, length of mechanical ventilation, and PICU length of stay compared with those without. The TreeNet and CART models yielded comparable results. The variables included in the final CART model were presence of SA ARD on day 1, Pao2 to Fio2 ratio of < 250, soluble thrombomodulin, and vascular cell adhesion molecule 1 concentrations. This model showed an area under the receiver operating characteristic curve (AUC) of 0.88 in the training data set, sensitivity of 0.91 (95% CI, 0.86-0.94), specificity of 0.76 (95% CI, 0.68-0.82), and demonstrated reproducibility in validation data set and test cohort (AUC range, 0.78-0.83). INTERPRETATION We derived and validated predictive models incorporating clinical and endothelial biomarkers to identify pediatric patients with septic shock at high risk of persistent acute respiratory dysfunction. Pending prospective validation, such models may facilitate enrichment and targeted intervention in future clinical trials.
Collapse
Affiliation(s)
- James G Williams
- Division of Critical Care Medicine and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Jane E Whitney
- Division of Medical Critical Care, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Scott L Weiss
- Division of Critical Care Medicine, Nemour's Children's Hospital, Wilmington, DE
| | - Brian M Varisco
- Division of Critical Care Medicine and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Nadir Yehya
- Division of Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Mihir R Atreya
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center and Department of Pediatrics University of Cincinnati, College of Medicine, Cincinnati, OH
| |
Collapse
|
33
|
Wang Y, Wu J, Shao T, Su D, Ma X, Yu Z, Li N. PROGNOSTIC IMPLICATIONS OF CHANGES IN PLATELET TRAJECTORIES IN PATIENTS WITH SEPSIS: A RETROSPECTIVE ANALYSIS USING THE MEDICAL INFORMATION MART FOR INTENSIVE CARE IV DATABASE. Shock 2025; 63:371-378. [PMID: 39450919 DOI: 10.1097/shk.0000000000002493] [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: 10/26/2024]
Abstract
ABSTRACT Objective: Patients with sepsis often experience reductions or increases in platelet counts, but the implications of these temporal patterns on prognosis remain unclear. The aim of this study was to investigate the impact of changes in platelet trajectories on the clinical prognosis of sepsis. Methods: This study was a retrospective analysis using data from the Medical Information Mart for Intensive Care IV database. Patients with sepsis were identified from the database, and their platelet trajectories were categorized into four distinct models based on the changes in platelet counts over a period of 14 days after diagnosis of sepsis. The effect of these trajectories on patient prognosis was subsequently evaluated. Results: A total of 15,250 patients with sepsis were included to construct a model, and the following four distinct platelet count trajectories were identified: normal platelet levels (phenotype 1); persistently low platelet levels (phenotype 2); gradually increasing platelet levels exceeding the normal range (phenotype 3); and consistently significantly elevated platelet levels (phenotype 4). Statistically significant differences were found in the 28-day mortality, in-hospital mortality, and 90-day mortality among the four phenotypes. Multivariate regression analysis showed that compared to the group with normal platelet levels (phenotype 1), the group with persistently low platelet levels (phenotype 2) had higher in-hospital mortality (odds ratio [OR] = 1.34, 95% confidence interval [CI]: 1.16-1.54), 28-day mortality (OR = 1.69, 95% CI: 1.47-1.94), and 90-day mortality (OR = 1.50, 95% CI: 1.32-1.69). There was no difference in in-hospital mortality between phenotypes 3 and 4 compared to phenotype 1, although phenotype 4 showed an increase in 28-day mortality ( P < 0.05), and phenotype 3 showed a decreasing trend in 90-day mortality ( P < 0.05). The results of inverse probability weighting adjusted by regression were basically consistent with the above findings, except that there was no statistical difference in 28-day mortality between phenotype 4 and phenotype 1. In the subgroups based on age, weight, and antiplatelet drugs or therapies, there was an interaction between platelet levels and these factors. Conclusions: In patients with sepsis, a decrease in platelet count is associated with increased mortality, while a moderate increase in platelet count can reduce 90-day mortality. However, for patients with persistently elevated platelet counts, caution is advised when using antiplatelet drugs or therapies, as it may increase mortality.
Collapse
Affiliation(s)
- Yingxin Wang
- Department of Critical Care Medicine, Affiliated Hospital of Hebei University, BaoDing, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Fang S, Wang Y, Nan W, Feng Y, Su W, Wang Y, Jiang X. Unfractionated heparin may improve near-term survival in patients admitted to the ICU with sepsis attributed to pneumonia: an observational study using the MIMIC-IV database. Front Pharmacol 2025; 16:1518716. [PMID: 40083381 PMCID: PMC11903409 DOI: 10.3389/fphar.2025.1518716] [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: 10/28/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction Limited data are available on the use, duration, and dosage of anticoagulant therapy in patients with pneumonia-induced sepsis, and the survival benefits of heparin remain uncertain. This study aimed to assess whether heparin administration improves near-term survival in critically ill patients with pneumonia-induced sepsis and identify the optimal dosage and treatment duration. Methods This study utilized the Medical Information Mart for Intensive Care-IV (MIMIC-IV) database. The variance inflation factor was employed to exclude highly collinear variables. Propensity score matching (PSM), the Cox proportional hazards model, and Cox regression subgroup analysis were used to evaluate the outcomes of subcutaneous heparin prophylactic anticoagulation after intensive care unit (ICU) admission. The primary outcomes were 30-, 45-, and 60-d mortality rates. Secondary outcomes included ICU length of stay (LOS_ICU), hospital length of stay (LOS_Hospital), in-hospital mortality, and the incidence of gastrointestinal bleeding. Results We enrolled 1,586 adult patients with pneumonia-induced sepsis. After PSM, 1,176 patients remained (588 in the heparin group and 588 in the non-heparin group). The 45-d survival rate was significantly higher in the heparin-treated group than that in the non-heparin group (84.4% vs. 79.4%; HR: 0.75; 95% CI: 0.572-0.83; adjusted HR: 0.73, 95% CI: 0.563-0.964; P < 0.05). LOS_ICU and LOS_Hospital were significantly shorter in the heparin group (P < 0.001), with no significant difference in gastrointestinal bleeding incidence between the two groups. Cox proportional hazards models demonstrated that heparin dose and duration were strongly associated with 45-d survival. Subgroup analysis indicated a significant survival advantage in patients aged 18-60 years, without diabetes, chronic obstructive pulmonary disease, or stage 1 acute kidney injury, who received a daily heparin dose of 3 mL for more than 7 d. Conclusion Our study found that early administration of heparin, particularly in sufficient doses (Heparin Sodium 5,000 units/mL, 1 mL per dose, three times daily (TID)) for more than 7 d, was associated with reduced near-term mortality in critically ill patients with pneumonia-induced sepsis. These findings underscore the potential benefits of anticoagulant therapy in this high-risk patient population.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaodong Jiang
- The Second Hospital of Dalian Medical University, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
35
|
Li K, Feng J, Li M, Han L, Wu Y. Systematic Review of Interleukin-35 in Endothelial Dysfunction: A New Target for Therapeutic Intervention. Mediators Inflamm 2025; 2025:2003124. [PMID: 39974277 PMCID: PMC11839265 DOI: 10.1155/mi/2003124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial dysfunction is a significant factor in the pathogenesis of various diseases. In pathological states, endothelial cells (ECs) undergo activation, resulting in dysfunction characterized by the stimulation of inflammatory responses, oxidative stress, cell proliferation, blood coagulation, and vascular adhesions. Interleukin-35 (IL-35), a novel member of the IL-12 family, is primarily secreted by regulatory T cells (Tregs) and regulatory B cells (Bregs). The role of IL-35 in immunomodulation, antioxidative stress, resistance to apoptosis, control of EC activation, adhesion, and angiogenesis in ECs remains incompletely understood, as the specific mechanisms of IL-35 action and its regulation have yet to be fully elucidated. Therefore, this systematic review aims to comprehensively investigate the impact of IL-35 on ECs and their physiological roles in a range of conditions, including cardiovascular diseases, tumors, sepsis, and rheumatoid arthritis (RA), with the objective of elucidating the potential of IL-35 as a therapeutic target for these ailments.
Collapse
Affiliation(s)
- Kai Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Jie Feng
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| |
Collapse
|
36
|
Ku CJ, Yu X, Zhao QY, Grzegorski SJ, Daniel JG, Ferguson AC, Shavit JA. Loss of protein C vs protein S results in discrepant thrombotic phenotypes. Blood Adv 2025; 9:545-557. [PMID: 39657127 PMCID: PMC11821410 DOI: 10.1182/bloodadvances.2024013237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/25/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT Venous thrombosis is a leading cause of morbidity/mortality and associated with deficiencies of the anticoagulant protein C (PC; PROC) and its cofactor, protein S (PS; PROS1). Heterozygous mutations increase the risk of adult-onset thrombosis, whereas homozygous mutations result in pre/neonatal lethal thrombosis. Phenotypes of patients with PC and PS deficiency are generally considered clinically indistinguishable. Here, we generate proc (zebrafish PROC ortholog) and pros1 knockouts through genome editing in zebrafish and uncover partially discordant phenotypes. proc-/- mutants exhibited ∼70% lethality at 1 year of age, whereas pros1-/- survival was unaffected. Induced venous endothelial injury in both mutants revealed reduced occlusive thrombus formation. This is consistent with the consumptive coagulopathy of zebrafish antithrombin 3 knockouts, which also results in spontaneous venous thrombosis. However, proc and pros1 mutants revealed a discrepancy. Although both mutants demonstrated spontaneous thrombosis, proc-/- was localized to the cardiac and venous systems, whereas pros1-/- was intracardiac. Aside from coagulation, PC has been shown to have PS-independent roles in inflammation. proc mutants displayed altered inflammatory markers and defects in neutrophil migration independent of pros1. Transcriptomic analysis and gene knockdown identified novel proc genetic interactions with adgrf7, a G protein-coupled receptor (GPCR) not previously known to be involved in coagulation. In summary, our data reveal differences between PC- and PS-deficient thrombosis, with cardiovascular tissue-specific phenotypes and survival differences, suggesting the possibility of underlying clinical differences in affected patients. This model of complete proc-/- deficiency in an accessible organism will facilitate further in vivo study of these distinctions, as well as PS-dependent and -independent functions of PC.
Collapse
Affiliation(s)
- Chia-Jui Ku
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Xinge Yu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Queena Y. Zhao
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | | | | | | | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
37
|
Su E, Song X, Wei L, Xue J, Cheng X, Xie S, Jiang H, Liu M. Endothelial GSDMD underlies LPS-induced systemic vascular injury and lethality. JCI Insight 2025; 10:e182398. [PMID: 39927458 PMCID: PMC11948583 DOI: 10.1172/jci.insight.182398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/17/2024] [Indexed: 02/11/2025] Open
Abstract
Endothelial injury destroys endothelial barrier integrity, triggering organ dysfunction and ultimately resulting in sepsis-related death. Considerable attention has been focused on identifying effective targets for inhibiting damage to endothelial cells to treat endotoxemia-induced septic shock. Global gasdermin D (Gsdmd) deletion reportedly prevents death caused by endotoxemia. However, the role of endothelial GSDMD in endothelial injury and lethality in lipopolysaccharide-induced (LPS-induced) endotoxemia and the underlying regulatory mechanisms are unknown. Here, we show that LPS increases endothelial GSDMD level in aortas and lung microvessels. We demonstrated that endothelial Gsdmd deficiency, but not myeloid cell Gsdmd deletion, protects against endothelial injury and death in mice with endotoxemia or sepsis. In vivo experiments suggested that hepatocyte GSDMD mediated the release of high-mobility group box 1, which subsequently binds to the receptor for advanced glycation end products in endothelial cells to cause systemic vascular injury, ultimately resulting in acute lung injury and lethality in shock driven by endotoxemia or sepsis. Additionally, inhibiting endothelial GSDMD activation via a polypeptide inhibitor alleviated endothelial damage and improved survival in a mouse model of endotoxemia or sepsis. These data suggest that endothelial GSDMD is a viable pharmaceutical target for treating endotoxemia and endotoxemia-induced sepsis.
Collapse
Affiliation(s)
- Enyong Su
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyue Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Lili Wei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cardiology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai, China
| | - Junqiang Xue
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xuelin Cheng
- Department of Health Management Center, Zhongshan Hospital, and
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shiyao Xie
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong Jiang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ming Liu
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Health Management Center, Zhongshan Hospital, and
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Morin A, Missri L, Urbina T, Bonny V, Gasperment M, Bernier J, Baudel JL, Kattan E, Maury E, Joffre J, Ait-Oufella H. Relationship between skin microvascular blood flow and capillary refill time in critically ill patients. Crit Care 2025; 29:57. [PMID: 39905546 PMCID: PMC11792347 DOI: 10.1186/s13054-025-05285-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Capillary refill time (CRT) and skin blood flow (SBF) have been reported to be strong predictors of mortality in critically ill patients. However, the relationship between both parameters remains unclear. METHODS We conducted a prospective observational study in a tertiary teaching hospital. All patients older than 18 years admitted in the intensive care unit (ICU) with circulatory failure and a measurable CRT were included. We assessed index SBF by laser doppler flowmetry and CRT on the fingertip, at T0 (Within the first 48 h from admission) and T1 (4 to 6 h later). Correlation was computed using Spearman or Pearson's formula. RESULTS During a 2-month period, 50 patients were included, 54% were admitted for sepsis. At baseline median CRT was 2.0 [1.1-3.9] seconds and median SBF was 46 [20-184] PU. At baseline SBF strongly correlated with CRT (R2 = 0.89; p < 0.0001, curvilinear relationship), this correlation was maintained whether patients were septic or not (R2 = 0.94; p = 0.0013; R2 = 0.87; p < 0.0001, respectively), and whether they received norepinephrine or not (R2 = 0.97; p = 0.0035; R2 = 0.92; p < 0.0001, respectively). Between T0 and T1, changes in SBF also significantly correlated with changes in CRT (R2 = 0.34; p < 0.0001). SBF was related to tissue perfusion parameters such as arterial lactate level (p = 0.02), whilst no correlation was found with cardiac output. In addition, only survivors significantly improved their SBF between T0 and T1. SBF was a powerful predictor of day-28 mortality as the AUROC at T0 was 85% [95% IC [76-91]] and at T1 90% [95% IC [78-100]]. CONCLUSION We have shown that index CRT and SBF were correlated, providing evidence that CRT is a reliable marker of microvascular blood flow. Trial registration Comité de protection des personnes Ouest II N° 2023-A02046-39.
Collapse
Affiliation(s)
- Alexandra Morin
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
| | - Louai Missri
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
| | - Tomas Urbina
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
| | - Vincent Bonny
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
| | - Maxime Gasperment
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
| | - Juliette Bernier
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
| | - Jean-Luc Baudel
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
| | - Eduardo Kattan
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Eric Maury
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
| | - Jérémie Joffre
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France
- Centre de Recherche Saint-Antoine Inserm UMR-S 938, Sorbonne University, 75012, Paris, France
| | - Hafid Ait-Oufella
- Intensive Care Unit, Saint-Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France.
- Paris Cardiovascular Research Center, Inserm U970, University Paris-Cité, Paris, France.
| |
Collapse
|
39
|
Zhu L, Dong H, Li L, Liu X. The Mechanisms of Sepsis Induced Coagulation Dysfunction and Its Treatment. J Inflamm Res 2025; 18:1479-1495. [PMID: 39925935 PMCID: PMC11804232 DOI: 10.2147/jir.s504184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/22/2025] [Indexed: 02/11/2025] Open
Abstract
Sepsis is a critical condition characterized by organ dysfunction due to a dysregulated response to infection that poses significant global health challenges. Coagulation dysfunction is nearly ubiquitous among sepsis patients. Its mechanisms involve platelet activation, coagulation cascade activation, inflammatory reaction imbalances, immune dysregulation, mitochondrial damage, neuroendocrine network disruptions, and endoplasmic reticulum (ER) stress. These factors not only interact but also exacerbate one another, leading to severe organ dysfunction. This review illustrates the mechanisms of sepsis-induced coagulopathy, with a focus on tissue factor activation, endothelial glycocalyx damage, and the release of neutrophil extracellular traps (NETs), all of which are potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - He Dong
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Lin Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Xiaojie Liu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| |
Collapse
|
40
|
Liu H, Xu C, Hu Q, Wang Y. Sepsis-induced cardiomyopathy: understanding pathophysiology and clinical implications. Arch Toxicol 2025; 99:467-480. [PMID: 39601874 DOI: 10.1007/s00204-024-03916-x] [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: 07/23/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Sepsis is a life-threatening form of organ dysfunction resulting from a dysregulated response to infection. The complex pathogenesis of sepsis poses challenges because of the lack of reliable biomarkers for early identification and effective treatments. As sepsis progresses to severe forms, cardiac dysfunction becomes a major concern, often manifesting as ventricular dilation, a reduced ejection fraction, and a diminished contractile capacity, known as sepsis-induced cardiomyopathy (SIC). The absence of standardized diagnostic and treatment protocols for SIC leads to varied criteria being used across medical institutions and studies, resulting in significant outcome disparities. Despite the high prevalence of SIC, accurate statistical data are lacking. To understand how SIC affects sepsis prognosis, a thorough exploration of its pathophysiological mechanisms, including systemic factors and complex signalling within myocardial and immune cells, is required. Identifying the factors influencing SIC occurrence and progression is crucial and must be conducted within specific clinical contexts. In this review, the clinical manifestations, pathophysiological mechanisms, and treatment strategies for SIC are discussed, along with the clinical background. We aim to connect current practices with future research challenges, providing clear guidance for clinicians and researchers.
Collapse
Affiliation(s)
- Haoran Liu
- Emergency and Trauma College, Hainan Medical University, Haikou, People's Republic of China
| | - Chaoqun Xu
- School of Medicine, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, People's Republic of China
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Qin Hu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Yang Wang
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.
| |
Collapse
|
41
|
Zou F, Zou J, Du Q, Liu L, Li D, Zhao L, Tang M, Zuo L, Sun Z. XueBiJing injection improves the symptoms of sepsis-induced acute lung injury by mitigating oxidative stress and ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118732. [PMID: 39181287 DOI: 10.1016/j.jep.2024.118732] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE XBJ injection is approved by the China Food and Drug Administration for the adjunctive treatment of sepsis, and it is derived from the traditional Chinese medicine (TCM) prescription XuefuZhuyu Decoction. It consists of five Chinese herbal extracts: Carthamus tinctorius, Paeonia lactiflora, Salvia miltiorrhiza, Conioselinum anthriscoides 'Chuanxiong' and Angelica sinensis. AIM OF THE STUDY The purpose of this study was to explore the relationship between ferroptosis and acute septic lung injury, and to evaluate the improvement effect of XBJ injection on acute lung injury in sepsis. MATERIALS AND METHODS Acute lung injury was induced in rats by cecum ligation and puncture, and these rats were treated with XBJ injection. Oxidative stress and inflammation levels were assessed in serum and lung tissue, and tissue samples were collected for histological and protein analyses. To illustrate the mechanism of the improvement effect of XBJ on acute lung injury in sepsis, serum lipidomics was carried out to investigate whether XBJ prevents oxidative stress-induced lipid metabolism disorders. Furthermore, protein expression of ferroptosis-related genes was also examined. RESULTS XBJ was shown to be effective in alleviating sepsis-induced ALI. XBJ also improves sepsis-induced acute lung injury by reducing lipid peroxidation and inflammation and modulating ferroptosis pathways. Specifically, compared with the sham group, XBJ downregulated the levels of Fe2+, MDA and GSSG, and reversed the decrease in the levels of GSH and GSH/GSSH in lung tissue. Metabolic pathways such as glycerophospholipid metabolism, phospholipid metabolism, and lipid metabolism associated with ferroptosis were obtained by lipidomic analysis of differential lipid metabolite enrichment, suggesting that ferroptosis occurs in septic rats, and that XBJ inhibits ferroptosis and thereby improves sepsis-induced ALI. Furthermore, XBJ optimises iron metabolism and lipid oxide metabolism by regulating the expression of a series of proteins that are closely related to ferroptosis, such as GPX4, ACSL4, x-CT, and FTH1. CONCLUSIONS Our findings, initially, indicated that XBJ ameliorates sepsis-induced ALI by reducing oxidative stress and ferroptosis, revealing a previously unrecognised mechanism by which XBJ ameliorates sepsis-induced ALI.
Collapse
Affiliation(s)
- Fanmei Zou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Henan Province, 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Henan Province, 450052, China
| | - Jing Zou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Henan Province, 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Henan Province, 450052, China
| | - Qiuzheng Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Henan Province, 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Henan Province, 450052, China
| | - Liwei Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Henan Province, 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Henan Province, 450052, China
| | - Ding Li
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Lingguo Zhao
- Baoan District Center for Disease Control and Prevention of Shenzhen City, Guangdong Province, 518109, China
| | - Meng Tang
- Zhengzhou Central Hospital Affiliated to Zhengzhou University, Henan Province, 450052, China
| | - Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Henan Province, 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Henan Province, 450052, China.
| | - Zhi Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Henan Province, 450052, China; Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Henan Province, 450052, China.
| |
Collapse
|
42
|
Man C, An Y, Wang GX, Mao EQ, Ma L. Recent Advances in Pathogenesis and Anticoagulation Treatment of Sepsis-Induced Coagulopathy. J Inflamm Res 2025; 18:737-750. [PMID: 39845020 PMCID: PMC11752821 DOI: 10.2147/jir.s495223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025] Open
Abstract
Coagulopathy in sepsis is common and is associated with high mortality. Although immunothrombosis is necessary for infection control, excessive thrombus formation can trigger a systemic thrombo-inflammatory response. Immunothrombosis plays a core role in sepsis-induced coagulopathy, and research has revealed a complex interplay between inflammation and coagulation. Different mechanisms underlying sepsis-related coagulopathy are discussed, including factors contributing to the imbalance of pro- and anticoagulation relevant to endothelial cells. The potential therapeutic implications of anticoagulants on these mechanisms are discussed. This review contributes to our understanding of the pathogenesis of coagulopathy in patients with sepsis. Recent studies suggest that endothelial cells play an important role in immunoregulation and hemostasis. Meanwhile, the non-anticoagulation effects of anticoagulants, especially heparin, which act in the pathogenesis of coagulopathy in septic patients, have been partially revealed. We believe that further insights into the pathogenesis of sepsis-induced coagulopathy will help physicians evaluate patient conditions effectively, leading to advanced early recognition and better decision-making in the treatment of sepsis.
Collapse
Affiliation(s)
- Chit Man
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Yuan An
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Guo-Xin Wang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - En-Qiang Mao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Li Ma
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| |
Collapse
|
43
|
Xu T, Song S, Zhu K, Yang Y, Wu C, Wang N, Lu S. Systemic inflammatory response index improves prognostic predictive value in intensive care unit patients with sepsis. Sci Rep 2025; 15:1908. [PMID: 39809872 PMCID: PMC11732978 DOI: 10.1038/s41598-024-81860-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Sepsis is a severe infectious disease with high mortality. However, the indicators used to evaluate its severity and prognosis are relatively complicated. The systemic inflammatory response index (SIRI), a new inflammatory indicator, has shown good predictive value in chronic infection, stroke, and cancer. The purpose of this study was to investigate the connection between sepsis and SIRI and evaluate its predictive usefulness. A total of 401 patients with sepsis were included in this study. Multiple linear regression and logistic regression analyses were performed to evaluate the relationship between SIRI and sepsis. The restricted cubic spline (RCS) method was employed to illustrate the dose-response relationship. The area under the curve (AUC) and decision curve analysis (DCA) were used to evaluate the prognostic value of SIRI. Multiple linear regression analysis revealed a significant positive correlation between SIRI and both blood cell count and Sequential Organ Failure Assessment (SOFA) score. Additionally, higher SIRI levels were significantly linked to a higher risk of sepsis worsening, according to logistic regression analysis. The RCS curve demonstrated that the risk of poor prognosis rose with increasing SIRI, particularly when SIRI exceeded 6.1. Furthermore, AUC and DCA results showed that SIRI had superior predictive value compared to traditional indicators. A higher SIRI is linked to a worse prognosis and more severe sepsis. SIRI may serve as a novel prognostic indicator in sepsis, though further clinical studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- Tuo Xu
- Xinxiang Central Hospital, Xinxiang, Henan, People's Republic of China
- The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
- Nantong University Hospital, Nantong, Jiangsu, People's Republic of China
| | - Shuaiwei Song
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, People's Republic of China
| | - Ke Zhu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yin Yang
- The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Chengyu Wu
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, People's Republic of China
| | - Naixue Wang
- Nantong Sixth People's Hospital, Nantong, Jiangsu, People's Republic of China
| | - Shu Lu
- Nantong University Hospital, Nantong, Jiangsu, People's Republic of China.
- Member of the Critical Care Medicine Branch of Jiangsu Physicians Association, Member of the Critical Care Ultrasound Group of Jiangsu Medical Association, Member of the Critical Care Medicine Branch of Nantong Medical Association, Member of Nantong Critical Care Medicine Quality Control Center, Affiliated Hospital of Nantong University, No. 20 Xisi Road, Nantong, 226000, Jiangsu, People's Republic of China.
| |
Collapse
|
44
|
Huang C, Tong Q, Zhang W, Pan Z. Association of early aspirin use with 90-day mortality in patients with sepsis: an PSM analysis of the MIMIC-IV database. Front Pharmacol 2025; 15:1475414. [PMID: 39850571 PMCID: PMC11754289 DOI: 10.3389/fphar.2024.1475414] [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: 08/03/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025] Open
Abstract
Objective In addition to its antiplatelet and anti-inflammatory properties, aspirin inhibits bacterial proliferation directly. The potential benefits of aspirin may enhance the prognosis for sepsis patients. However, little is known about the effects of early aspirin administration. This study aimed to examine the correlation between the administration of aspirin at an early stage and the 90-day mortality rate among sepsis patients. Methods In order to distinguish between septic patients who received early aspirin treatment and those who did not, queries were conducted on the Medical Information Mart for Intensive Care IV (MIMIC-IV) database. The principal metric utilized was 90-day mortality. We determined the association between early aspirin use and 90-day mortality using multivariate Cox regression, and propensity score matching (PSM) was utilized to validate our findings. The analyses of the subgroups have been completed. Results Our analysis comprised 28,425 septic patients, of whom 7,568 (26.6%) received aspirin within 24 h of intensive care unit (ICU) admission. The aspirin users group had a lower 90-day mortality than the aspirin nonusers group [1,624 (21.8%) vs. 2,035 (27.3%), P < 0.001]. The logistic regression showed that early aspirin use was associated with a lower 90-day mortality (OR, 0.74, 95% CI, 0.69-0.80, P < 0.001). K-M curve analysis showed that the 90-day mortality of the aspirin users group was significantly lower than that of the aspirin nonusers group (P < 0.001). Subgroup analysis revealed comparable relationships between early aspirin use and 90-day mortality among individuals. Conclusion In conclusion, early aspirin use was associated with decreased in-hospital and 90-day mortality in septic patients, emphasizing the significance of early aspirin use administration in the ICU.
Collapse
Affiliation(s)
- Chunsheng Huang
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, China
| | - Qiaoling Tong
- Department of Otolaryngology, Ningbo No.2 Hospital, Ningbo, China
| | - Wenyuan Zhang
- The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhihao Pan
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, China
| |
Collapse
|
45
|
Zeng Y, Fu BM. Angiogenesis and Microvascular Permeability. Cold Spring Harb Perspect Med 2025; 15:a041163. [PMID: 38692737 PMCID: PMC11694756 DOI: 10.1101/cshperspect.a041163] [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: 05/03/2024]
Abstract
Angiogenesis, the formation of new blood microvessels, is a necessary physiological process for tissue generation and repair. Sufficient blood supply to the tissue is dependent on microvascular density, while the material exchange between the circulating blood and the surrounding tissue is controlled by microvascular permeability. We thus begin this article by reviewing the key signaling factors, particularly vascular endothelial growth factor (VEGF), which regulates both angiogenesis and microvascular permeability. We then review the role of angiogenesis in tissue growth (bone regeneration) and wound healing. Finally, we review angiogenesis as a pathological process in tumorigenesis, intraplaque hemorrhage, cerebral microhemorrhage, pulmonary fibrosis, and hepatic fibrosis. Since the glycocalyx is important for both angiogenesis and microvascular permeability, we highlight the role of the glycocalyx in regulating the interaction between tumor cells and endothelial cells (ECs) and VEGF-containing exosome release and uptake by tumor-associated ECs, all of which contribute to tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Bingmei M Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York 10031, USA
| |
Collapse
|
46
|
Li Y, Sun Z, Li Y, Sun J, Chen B. IL-1β-Stimulated Bone Mesenchymal Stem Cell-Derived Exosomes Mitigate Sepsis through Modulation of HMGB1/AKT Pathway and M2 Macrophage Polarization. Curr Mol Med 2025; 25:79-89. [PMID: 38173202 DOI: 10.2174/0115665240277763231206051401] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Sepsis is a life-threatening disease caused by infection, and developing novel strategies against sepsis is still required. Exosomes derived from mesenchymal stem cells (MSCs) have shown promising therapeutic potential for various diseases. In this study, we aimed to investigate the action and mechanism of exosomes derived from IL-1β-pre-conditioned bone marrow-derived mesenchymal stromal cells (BMSCs) in sepsis. METHODS Exosomes were isolated from BMSCs that were pretreated with (IL-1β- BMSC/exos) or without IL-1β (BMSC/exos). In vitro, a cell model of sepsis was induced by treating human umbilical vein endothelial cells (HUVECs) with lipopolysaccharide (LPS), while in vivo, a sepsis model was established through cecal ligation and puncture (CLP) operation. Immunofluorescence staining was used to detect the uptake of exosomes by HUVECs. The effects of exosomes on the cellular function of HUVECs were determined through EDU proliferation assay, migration assay, and tube formation assay. Gene and protein expression were analyzed using qRT-PCR, Western blot, ELISA, immunofluorescence staining, and immunohistochemistry staining. RESULTS IL-1β-BMSC/exos significantly enhanced the proliferation, migration, and tube formation of HUVECs. Treatment with LPS induced the expression of high mobility group box 1 (HMGB1) and the phosphorylation of AKT in HUVECs, but these effects were counteracted by the treatment of IL-1β-BMSC/exos. The protective effect of IL-1β-BMSC/exos on the viability and tube formation ability of HUVECs was reversed by overexpression of HMGB1. Moreover, IL-1β-BMSC/exos promoted the polarization of M2 macrophages and reduced the secretion of inflammatory chemokines. Additionally, IL-1β-BMSC/exos alleviated cecal ligation and puncture (CLP)-induced sepsis in vivo. CONCLUSION IL-1β-BMSC/exos alleviates sepsis by modulating the HMGB1/AKT pathway and triggering M2 macrophage polarization.
Collapse
Affiliation(s)
- Yang Li
- Department of Infectious Diseases, Anhui Provincial Children's Hospital, Hefei, 230022, China
| | - Zifa Sun
- Department of Infectious Diseases, Anhui Provincial Children's Hospital, Hefei, 230022, China
| | - Yuanyuan Li
- Department of Infectious Diseases, Anhui Provincial Children's Hospital, Hefei, 230022, China
| | - Jing Sun
- Department of Infectious Diseases, Anhui Provincial Children's Hospital, Hefei, 230022, China
| | - Biquan Chen
- Department of Infectious Diseases, Anhui Provincial Children's Hospital, Hefei, 230022, China
| |
Collapse
|
47
|
Wang H, Chen D, He M. Efficacy and Safety of Recombinant Human Thrombopoietin (rhTPO) on Coagulation Function and Inflammatory Factors in the Treatment of Patients with Sepsis-Related Thrombocytopenia. Clin Appl Thromb Hemost 2025; 31:10760296251315173. [PMID: 39901742 PMCID: PMC11792032 DOI: 10.1177/10760296251315173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND this study aimed to investigate the efficacy of recombinant human thrombopoietin (rhTPO) in the treatment of sepsis-associated thrombocytopenia, and to evaluate its impact on coagulation function, inflammatory markers, platelet (Plt) count, and patient prognosis. METHODS a total of 144 patients with sepsis-associated thrombocytopenia, admitted to our hospital between 2022 and 2023, were selected for the study. The patients were randomly divided into two groups using a random number table: the control group (Group C, n = 72) and the research group (Group R, n = 72). The Group C received standard treatment, while the Group R received rhTPO in addition to standard care. We compared the general demographic data, Acute Physiology and Chronic Health Evaluation II (APACHE II) scores, coagulation parameters, serum levels of Toll-like receptor 4 (TLR4), interleukin-6 (IL-6), serum creatinine (SCr), tumor necrosis factor-alpha (TNF-α), Plt count, transfusion volume, treatment duration, incidence of complications, and mortality rates between the two groups. RESULTS there were no significant differences in the general demographic characteristics between the two groups (P > 0.05). After treatment, the APACHE II scores in both groups significantly decreased, with a more pronounced reduction observed in the Group R. Coagulation function indicators, including activated partial thromboplastin time (APTT), fibrinogen (FIB), plasminogen activator inhibitor-1 (PAI-1), antithrombin III (AT-III), protein C, thrombomodulin (TM), and Plt factor 4 (PF4), showed greater improvement in the Group R compared to the Group C (P < 0.05). The serum levels of TLR4, IL-6, and TNF-α in the Group R were significantly lower than those in the Group C (P < 0.05), whereas no significant difference in SCr levels was observed between the groups (P > 0.05). The Plt count in the Group R began to significantly increase on day 3 of treatment, and was consistently higher than that in the Group C on days 3, 5, and 7 (P < 0.05). The Group R required significantly fewer red blood cell transfusions compared to the Group C and did not require Plt suspension (P < 0.05). No significant differences were found between the groups in terms of mechanical ventilation time, intensive care unit (ICU) length of stay, and total hospital stay (P > 0.05). However, the ICU and overall hospital mortality rates were significantly lower in the Group R than in the Group C (P < 0.05). Multivariate logistic regression analysis indicated that rhTPO treatment was an independent protective factor for reducing mortality (OR = 0.475, P = 0.042). CONCLUSION rhTPO treatment effectively improves coagulation function and inflammatory status in patients with sepsis-associated thrombocytopenia, increases Plt count, reduces transfusion requirements, and lowers mortality. These findings suggest that rhTPO has significant clinical application value in the management of this condition.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Intensive Care Unit (ICU), First People's Hospital of Linping District, Hangzhou, China
| | - Dong Chen
- Department of Colorectal Surgery, First People's Hospital of Linping District, Hangzhou, China
| | - Ming He
- Department of Intensive Care Unit (ICU), First People's Hospital of Linping District, Hangzhou, China
| |
Collapse
|
48
|
Milivojac T, Grabež M, Krivokuća A, Maličević U, Gajić Bojić M, Đukanović Đ, Uletilović S, Mandić-Kovačević N, Cvjetković T, Barudžija M, Vojinović N, Šmitran A, Amidžić L, Stojiljković MP, Čolić M, Mikov M, Škrbić R. Ursodeoxycholic and chenodeoxycholic bile acids attenuate systemic and liver inflammation induced by lipopolysaccharide in rats. Mol Cell Biochem 2025; 480:563-576. [PMID: 38578526 PMCID: PMC11695453 DOI: 10.1007/s11010-024-04994-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/17/2024] [Indexed: 04/06/2024]
Abstract
Bacterial lipopolysaccharide (LPS) induces general inflammation, by activating pathways involving cytokine production, blood coagulation, complement system activation, and acute phase protein release. The key cellular players are leukocytes and endothelial cells, that lead to tissue injury and organ failure. The aim of this study was to explore the anti-inflammatory, antioxidant, and cytoprotective properties of two bile acids, ursodeoxycholic acid (UDCA) and chenodeoxycholic acid (CDCA) in LPS-induced endotoxemia in rats. The experiment involved six distinct groups of Wistar rats, each subjected to different pretreatment conditions: control and LPS groups were pretreated with propylene glycol, as a bile acid solvent, while the other groups were pretreated with UDCA or CDCA for 10 days followed by an LPS injection on day 10. The results showed that both UDCA and CDCA reduced the production of pro-inflammatory cytokines: TNF-α, GM-CSF, IL-2, IFNγ, IL-6, and IL-1β and expression of nuclear factor-κB (NF-κB) induced by LPS. In addition, pretreatment with these bile acids showed a positive impact on lipid profiles, a decrease in ICAM levels, an increase in antioxidant activity (SOD, |CAT, GSH), and a decrease in prooxidant markers (H2O2 and O2-). Furthermore, both bile acids alleviated LPS-induced liver injury. While UDCA and CDCA pretreatment attenuated homocysteine levels in LPS-treated rats, only UDCA pretreatment showed reductions in other serum biochemical markers, including creatine kinase, lactate dehydrogenase, and high-sensitivity troponin I. It can be concluded that both, UDCA and CDCA, although exerted slightly different effects, can prevent the inflammatory responses induced by LPS, improve oxidative stress status, and attenuate LPS-induced liver injury.
Collapse
Affiliation(s)
- T Milivojac
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - M Grabež
- Department of Hygiene, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - A Krivokuća
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Pathophysiology, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - U Maličević
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Pathophysiology, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - M Gajić Bojić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - Đ Đukanović
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacy, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - S Uletilović
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Medical Biochemistry and Chemistry, Faculty of Medicine, The Republic of Srpska, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - N Mandić-Kovačević
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacy, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - T Cvjetković
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Medical Biochemistry and Chemistry, Faculty of Medicine, The Republic of Srpska, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - M Barudžija
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Histology and Embryology, Faculty of Medicine, The Republic of Srpska, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - N Vojinović
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - A Šmitran
- Department of Microbiology and Immunology, Faculty of Medicine, The Republic of Srpska, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Lj Amidžić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - M P Stojiljković
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - M Čolić
- Medical Faculty Foča, University of East Sarajevo, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - M Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - R Škrbić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina.
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, The Republic of Srpska, Banja Luka, Bosnia and Herzegovina.
| |
Collapse
|
49
|
Saxena J, Agarwal G, Das S, Kumar A, Thakkar K, Kaushik S, Srivatsava VK, Siddiqui AJ, Jyoti A. Immunopharmacological Insights into Cordyceps spp.: Harnessing Therapeutic Potential for Sepsis. Curr Pharm Des 2025; 31:823-842. [PMID: 39694962 DOI: 10.2174/0113816128326301240920040036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 12/20/2024]
Abstract
Cordyceps spp. (CS), a well-known medicinal mushroom that belongs to Tibetan medicine and is predominantly found in the high altitudes in the Himalayas. CS is a rich reservoir of various bioactive substances including nucleosides, sterols flavonoids, peptides, and phenolic compounds. The bioactive compounds and CS extract have antibacterial, antioxidant, immunomodulatory, and inflammatory properties in addition to organ protection properties across a range of disease states. The study aimed to review the potential of CS, a medicinal mushroom, as a treatment for sepsis. While current sepsis drugs have side effects, CS shows promise due to its anti-inflammatory, antioxidant, and antibacterial properties. We have performed an extensive literature search based on published original and review articles in Scopus and PubMed. The keywords used were Cordyceps, sepsis, and inflammation. Studies indicate that CS extract and bioactive compounds target free radicals including oxidative as well as nitrosative stress, lower inflammation, and modulate the immune system, all of which are critical components in sepsis. The brain, liver, kidneys, lungs, and heart are among the organs that CS extracts may be able to shield against harm during sepsis. Traditional remedies with anti-inflammatory and protective qualities, such as Cordyceps mushrooms, are promising in sepsis. However, more research including clinical trials is required to validate the usefulness of CS metabolites in terms of organ protection and fight infections in sepsis.
Collapse
Affiliation(s)
- Juhi Saxena
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara, Gujarat, India
| | - Gaurang Agarwal
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Sarvjeet Das
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Anshu Kumar
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Krish Thakkar
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara, Gujarat, India
| | - Sanket Kaushik
- Amity Institute of Biotechnology, Amity University, Jaipur, Rajasthan, India
| | | | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Anupam Jyoti
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
50
|
Yu Z, Li Y, Bai L, Zheng Y, Liu X, Zhen Y. The triple combination DBDx alleviates cytokine storm and related lung injury. Int Immunopharmacol 2024; 143:113431. [PMID: 39454409 DOI: 10.1016/j.intimp.2024.113431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Cytokine storm is a life-threatening disorder, and therapeutic treatments are urgently needed. Here, we investigated the anti-cytokine storm efficacy of DBDx, a triple drug combination composed of dipyridamole, ubenimex and dexamethasone. Evaluated by lipopolysaccharide (LPS)-induced cytokine storm murine model, DBDx significantly improved survival rate and prolonged survival time of the model mice. Notably, the efficacy of DBDx was higher than that of dipyridamole, ubenimex and dexamethasone. Determined by ELISA, DBDx significantly reduced the LPS-stimulated serum levels of TNF-α, IL-6 and IL-1β in mice. Luminex assay showed that DBDx suppressed the serum levels of a wide variety of inflammatory cytokines and chemokines, which was more potent than dexamethasone alone. Otherwise, DBDx exerted similar inhibitory effects on cytokine profiles in bronchoalveolar lavage fluid. Histopathological observation showed that DBDx significantly reduced the LPS-induced thickening of alveolar septum, indicating its suppression of capillary congestion, edema and neutrophil infiltration in the lung. Ultra-structure analysis showed that DBDx suppressed the LPS-induced morphological changes of microvilli in type II pneumocytes. In vitro experiment showed that DBDx inhibited IL-6 and TNF-α secretion in THP-1 cells, and downregulated TLR4/NF-κB/HIF-1α signaling pathway. All of these results demonstrate that DBDx, a triple combination of clinical orally-administered drugs, can alleviate cytokine storm and related lung injury. DBDx is beneficial for treating cytokine storm disorders.
Collapse
Affiliation(s)
- Zhuojun Yu
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Li
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liping Bai
- NHC Key Laboratory of Biotechnology of Antibiotics, CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanbo Zheng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiujun Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yongsu Zhen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|