Pub Date : 2024-12-31DOI: 10.1097/SHK.0000000000002547
Hang Yang, Lin Feng, Zhenjie Jiang, Yirong Xiao, Kai Zeng
Abstract: Background Sepsis is a life-threatening condition characterized by multiple organ dysfunction. Blood cells abnormalities play a significant role in the onset and progression of sepsis; however, the potential causal relationship between platelets and sepsis remains unclear, as does whether immune cells mediate the interaction between platelets and sepsis. This study aims to explore the potential causal relationship between platelets and sepsis and analyze the mediating effect of immune cells. In addition, cell-to-cell communication was analyzed to explore the interaction between blood cells and immune cells.Material and methods In this study, genome-wide association study (GWAS) data were utilized to examine the association between blood cells and sepsis. Two-sample mendelian randomisation (MR) and reverse MR were performed to investigate the potential causal relationship between blood cells and sepsis, with a specific focus on the relationship between platelets and sepsis. Subsequently, two-step MR was employed to identify the immune cells that mediate the interaction between platelets and sepsis and to assess their potential mediating effects. Cellchat software was used to analyze cell-to-cell communication.Results The results of two-sample MR indicated that platelets were negatively correlated with sepsis (OR = 0.976, 95% CI 0.959-0.993, p = 0.005), suggesting that platelets have a protective effect against sepsis. Additionally, reverse MR demonstrated that sepsis had no significant effect on platelets (OR = 0.909, 95% CI 0.156-5.296, p = 0.916). The mediating effect analysis revealed that monocytes and B cells were important mediators in the relationship between platelets and sepsis. Notably, the correlation between platelets and sepsis shifted from negative to positive with the involvement of monocytes and B cells. The number and strength of cell-cell interactions were decreased in sepsis. Monocytes and B cells primarily regulate platelets through the CLEC signaling pathway, contributing to the pathogenesis of sepsis.Conclusion This study confirmed the protective role of platelets in sepsis. Monocytes and B cells mediate changes in the genetic association between platelets and sepsis. Monocytes and B cells primarily interact with platelets via the CLEC pathway, thereby modulating the genetic association between platelets and sepsis. These findings indicate that thrombocytopenia, especially when accompanied by elevated monocytes and B cells, may serve as a potential marker for sepsis.
摘要:脓毒症是一种以多器官功能障碍为特征的危及生命的疾病。血细胞异常在脓毒症的发生和发展中起重要作用;然而,血小板与败血症之间的潜在因果关系尚不清楚,免疫细胞是否介导血小板与败血症之间的相互作用也不清楚。本研究旨在探讨血小板与脓毒症之间的潜在因果关系,并分析免疫细胞的介导作用。此外,还分析了细胞间的通讯,以探索血细胞和免疫细胞之间的相互作用。材料和方法在本研究中,利用全基因组关联研究(GWAS)数据来研究血细胞与败血症之间的关系。进行双样本孟德尔随机化(MR)和反向MR来研究血细胞与败血症之间的潜在因果关系,特别关注血小板与败血症之间的关系。随后,采用两步磁共振鉴定介导血小板与败血症相互作用的免疫细胞,并评估其潜在的介导作用。使用Cellchat软件分析细胞间通信。结果两样本MR结果显示血小板与脓毒症呈负相关(OR = 0.976, 95% CI 0.959-0.993, p = 0.005),提示血小板对脓毒症具有保护作用。此外,反向MR显示脓毒症对血小板无显著影响(OR = 0.909, 95% CI 0.156-5.296, p = 0.916)。介导效应分析显示单核细胞和B细胞是血小板与败血症关系的重要介质。值得注意的是,随着单核细胞和B细胞的参与,血小板与败血症之间的相关性从负向正转变。脓毒症患者细胞间相互作用的数量和强度下降。单核细胞和B细胞主要通过CLEC信号通路调节血小板,参与脓毒症的发病机制。结论血小板在脓毒症中的保护作用。单核细胞和B细胞介导血小板和败血症之间遗传关联的变化。单核细胞和B细胞主要通过CLEC途径与血小板相互作用,从而调节血小板与败血症之间的遗传关联。这些发现表明,血小板减少症,特别是当伴有单核细胞和B细胞升高时,可能作为败血症的潜在标志。
{"title":"Monocytes and B cells Mediate Alterations in the Genetic Association Between Platelets and Sepsis via CLEC signaling pathway.","authors":"Hang Yang, Lin Feng, Zhenjie Jiang, Yirong Xiao, Kai Zeng","doi":"10.1097/SHK.0000000000002547","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002547","url":null,"abstract":"<p><strong>Abstract: </strong>Background Sepsis is a life-threatening condition characterized by multiple organ dysfunction. Blood cells abnormalities play a significant role in the onset and progression of sepsis; however, the potential causal relationship between platelets and sepsis remains unclear, as does whether immune cells mediate the interaction between platelets and sepsis. This study aims to explore the potential causal relationship between platelets and sepsis and analyze the mediating effect of immune cells. In addition, cell-to-cell communication was analyzed to explore the interaction between blood cells and immune cells.Material and methods In this study, genome-wide association study (GWAS) data were utilized to examine the association between blood cells and sepsis. Two-sample mendelian randomisation (MR) and reverse MR were performed to investigate the potential causal relationship between blood cells and sepsis, with a specific focus on the relationship between platelets and sepsis. Subsequently, two-step MR was employed to identify the immune cells that mediate the interaction between platelets and sepsis and to assess their potential mediating effects. Cellchat software was used to analyze cell-to-cell communication.Results The results of two-sample MR indicated that platelets were negatively correlated with sepsis (OR = 0.976, 95% CI 0.959-0.993, p = 0.005), suggesting that platelets have a protective effect against sepsis. Additionally, reverse MR demonstrated that sepsis had no significant effect on platelets (OR = 0.909, 95% CI 0.156-5.296, p = 0.916). The mediating effect analysis revealed that monocytes and B cells were important mediators in the relationship between platelets and sepsis. Notably, the correlation between platelets and sepsis shifted from negative to positive with the involvement of monocytes and B cells. The number and strength of cell-cell interactions were decreased in sepsis. Monocytes and B cells primarily regulate platelets through the CLEC signaling pathway, contributing to the pathogenesis of sepsis.Conclusion This study confirmed the protective role of platelets in sepsis. Monocytes and B cells mediate changes in the genetic association between platelets and sepsis. Monocytes and B cells primarily interact with platelets via the CLEC pathway, thereby modulating the genetic association between platelets and sepsis. These findings indicate that thrombocytopenia, especially when accompanied by elevated monocytes and B cells, may serve as a potential marker for sepsis.</p>","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142922556","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-24DOI: 10.1097/SHK.0000000000002545
Jianxiong Liu, Li Zhen, Dihua Yu, Weiqing Wang
Aims: Brain injury occupies the predominant cause of neurological dysfunction and mortality after successful cardiopulmonary resuscitation (CPR) from cardiac arrest (CA). This study investigates the role and mechanism of Sirtuin 6 (SIRT6) in post-cardiac arrest brain injury in rats.
Methods: All rats were subjected to asphyxial CA followed by CPR. Two weeks before modeling, rats were infected with lentivirus containing oe-SIRT6 and oe-FOXO1 through lateral ventricular injection. qRT-PCR and Western blot quantified SIRT6 and FOXO1 expressions in brain tissues. Neurological deficit scores evaluated the neural function of rats at different time points, and water maze test assessed the changes in short-term learning and memory abilities. The survival status of rats 7 days after modeling was recorded. The pathological changes in brain tissues, inflammatory factors, and apoptosis were evaluated by H&E staining, ELISA, and TUNEL, respectively. Ch-IP measured the enrichment of SIRT6 and H3K9ac in the FOXO1 promoter.
Results: SIRT6 was poorly expressed while FOXO1 was highly expressed in CA/CPR rats. Elevation of SIRT6 expression alleviated neural function, behavioral ability, and survival rate, as well as abated pathological damage, inflammatory responses, and cell apoptosis in CA/CPR rats. Mechanistically, SIRT6 curbed FOXO1 transcription and expression by lowering the H3K9ac level in the FOXO1 promoter; FOXO1 overexpression abolished the improvement effect of SIRT6 overexpression on brain injury in CA/CPR rats.
Conclusions: Elevation of SIRT6 expression restrained the FOXO1 expression by diminishing the H3K9ac level in the FOXO1 promoter, thereby mitigating post-cardiac arrest brain injury in rats.
{"title":"Effect and regulatory mechanism of SIRT6 on post-cardiac arrest brain injury in rats.","authors":"Jianxiong Liu, Li Zhen, Dihua Yu, Weiqing Wang","doi":"10.1097/SHK.0000000000002545","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002545","url":null,"abstract":"<p><strong>Aims: </strong>Brain injury occupies the predominant cause of neurological dysfunction and mortality after successful cardiopulmonary resuscitation (CPR) from cardiac arrest (CA). This study investigates the role and mechanism of Sirtuin 6 (SIRT6) in post-cardiac arrest brain injury in rats.</p><p><strong>Methods: </strong>All rats were subjected to asphyxial CA followed by CPR. Two weeks before modeling, rats were infected with lentivirus containing oe-SIRT6 and oe-FOXO1 through lateral ventricular injection. qRT-PCR and Western blot quantified SIRT6 and FOXO1 expressions in brain tissues. Neurological deficit scores evaluated the neural function of rats at different time points, and water maze test assessed the changes in short-term learning and memory abilities. The survival status of rats 7 days after modeling was recorded. The pathological changes in brain tissues, inflammatory factors, and apoptosis were evaluated by H&E staining, ELISA, and TUNEL, respectively. Ch-IP measured the enrichment of SIRT6 and H3K9ac in the FOXO1 promoter.</p><p><strong>Results: </strong>SIRT6 was poorly expressed while FOXO1 was highly expressed in CA/CPR rats. Elevation of SIRT6 expression alleviated neural function, behavioral ability, and survival rate, as well as abated pathological damage, inflammatory responses, and cell apoptosis in CA/CPR rats. Mechanistically, SIRT6 curbed FOXO1 transcription and expression by lowering the H3K9ac level in the FOXO1 promoter; FOXO1 overexpression abolished the improvement effect of SIRT6 overexpression on brain injury in CA/CPR rats.</p><p><strong>Conclusions: </strong>Elevation of SIRT6 expression restrained the FOXO1 expression by diminishing the H3K9ac level in the FOXO1 promoter, thereby mitigating post-cardiac arrest brain injury in rats.</p>","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142922223","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Death in the early phase of trauma is primarily attributable to uncontrolled bleeding exacerbated by trauma-induced coagulopathy (TIC). A comprehensive synthesis of the available evidence on interventions for TIC is needed.
Methods: We conducted a systematic review and meta-analysis of blood component products and tranexamic acid administrations for severe trauma patients with TIC. We included randomized and non-randomized controlled trials. We included studies with patients who required transfusion with any coagulopathy associated with trauma and a detailed definition. The intervention was administration of blood component products and tranexamic acid. The primary outcome of the study was all-cause mortality and transfusion quantity.
Results: Four randomized controlled trials and seven observational studies were included in the qualitative synthesis. In this study, Fibrinogen concentrate (FC), Prothrombin coagulation cofactor (PCC), and Combination administrations of FC and PCC (FC + PCC) administration did not significantly reduce mortality rates. FC, PCC, and FC + PCC administrations significantly reduced RBC transfusions after admission. In addition, PCC administration reduced FFP transfusions during hospital admission. The incidence of thrombotic events was not significantly higher in the FC + PCC, PCC, and rFVIIa groups. Although statistically nonsignificant, multiple organ failure was lower in the FC and FC + PCC groups.
Conclusions: FC and PCC administrations did not significantly reduce mortality. However, FC, PCC, and FC + PCC reduced transfusion rates and complications in patients with coagulopathy-associated trauma. However, the definition of TIC is quite heterogeneous. Thus, the definition of TIC should be defined universally. Furthermore, due to the lack of high certainty of evidence, further well-constructed trials are warranted to investigate the efficacy of blood component products, specifically FC and PCC supplementation for TIC.
{"title":"The efficacy of coagulation factor concentrates in the management of patients with trauma-induced coagulopathy: a systematic review and meta-analysis.","authors":"Yuki Itagaki, Mineji Hayakawa, Yuki Takahashi, Shigeki Kushimoto, Yuichiro Sakamoto, Yoshinobu Seki, Kohji Okamoto","doi":"10.1097/SHK.0000000000002534","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002534","url":null,"abstract":"<p><strong>Background: </strong>Death in the early phase of trauma is primarily attributable to uncontrolled bleeding exacerbated by trauma-induced coagulopathy (TIC). A comprehensive synthesis of the available evidence on interventions for TIC is needed.</p><p><strong>Methods: </strong>We conducted a systematic review and meta-analysis of blood component products and tranexamic acid administrations for severe trauma patients with TIC. We included randomized and non-randomized controlled trials. We included studies with patients who required transfusion with any coagulopathy associated with trauma and a detailed definition. The intervention was administration of blood component products and tranexamic acid. The primary outcome of the study was all-cause mortality and transfusion quantity.</p><p><strong>Results: </strong>Four randomized controlled trials and seven observational studies were included in the qualitative synthesis. In this study, Fibrinogen concentrate (FC), Prothrombin coagulation cofactor (PCC), and Combination administrations of FC and PCC (FC + PCC) administration did not significantly reduce mortality rates. FC, PCC, and FC + PCC administrations significantly reduced RBC transfusions after admission. In addition, PCC administration reduced FFP transfusions during hospital admission. The incidence of thrombotic events was not significantly higher in the FC + PCC, PCC, and rFVIIa groups. Although statistically nonsignificant, multiple organ failure was lower in the FC and FC + PCC groups.</p><p><strong>Conclusions: </strong>FC and PCC administrations did not significantly reduce mortality. However, FC, PCC, and FC + PCC reduced transfusion rates and complications in patients with coagulopathy-associated trauma. However, the definition of TIC is quite heterogeneous. Thus, the definition of TIC should be defined universally. Furthermore, due to the lack of high certainty of evidence, further well-constructed trials are warranted to investigate the efficacy of blood component products, specifically FC and PCC supplementation for TIC.</p>","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142882816","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p><strong>Introduction: </strong>Dl-3-n-butylphthalide (NBP), a small molecular compound extracted from celery seeds, has been shown to exhibit diverse pharmacological activities, including anti-inflammatory, antioxidative, and anti-apoptotic effects. Recent studies have highlighted its efficacy in treating various cardiovascular conditions, such as myocardial infarction, hypertrophy, heart failure, and cardiotoxicity. This study aimed to investigate whether NBP could alleviate cardiac dysfunction and injury following hemorrhage-induced cardiac arrest (HCA) in a porcine model and elucidate its potential mechanisms.</p><p><strong>Methods: </strong>Seventeen pigs were randomized into three groups: Sham (n = 5), HCA + vehicle (n = 5), and HCA + NBP (n = 7). In the HCA + vehicle and HCA + NBP groups, the HCA model was established by continuous bleeding at a rate of 2 mL/kg/min to induce cardiac arrest. Cardiac arrest was maintained for 7 min, followed by the reinfusion of 50% of the shed blood at a rate of 5 mL/kg/min. After successful resuscitation, the HCA + NBP group received an intravenous dose of 2.5 mg/kg of NBP within 120 min. Post-resuscitation cardiac function (stroke volume, global ejection fraction) and injury biomarkers (cardiac troponin I, creatine kinase-MB) were assessed at regular intervals. At the end of the post-resuscitation observation, cardiac tissue samples were collected to assess: 1) histopathological injury, 2) cellular apoptosis, 3) levels of pro-inflammatory cytokines, including tumor necrosis factor-a (TNF-a), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-18 (IL-18), 4) the expression levels of NOD-like receptor pyrin domain 3 (NLRP3), caspase 1, gasdermin D (GSDMD), cyclic-GMP-AMP synthase (cGAS), stimulator of interferon genes (STING), and tank-binding kinase 1 (TBK1), and 5) the integrated optical density (IOD) of GSDMD N-terminal (GSDMD-N), phosphorylated STING (p-STING), and phosphorylated TBK1 (p-TBK1).</p><p><strong>Results: </strong>Following resuscitation, both stroke volume and global ejection fraction were significantly reduced, while serum levels of cardiac troponin I and creatine kinase-MB were markedly elevated in the HCA + vehicle and HCA + NBP groups compared with the Sham group. However, the extent of cardiac dysfunction and injury was significantly attenuated in the HCA + NBP group relative to the HCA + vehicle group. At 24 h post-resuscitation, substantial cardiac pathological injury and apoptosis were observed. Additionally, pyroptosis-related proteins (NLRP3, caspase-1, GSDMD, GSDMD-N) were upregulated, inflammatory markers (TNF-α, IL-1β, IL-6, IL-18) were elevated, and the activation of the cGAS-STING-TBK1 pathway (cGAS, STING, TBK1, p-STING, p-TBK1) were noted in both the HCA + vehicle and HCA + NBP groups compared to the Sham group. Notably, these pathological changes were significantly attenuated in the HCA + NBP group compared to the HCA + vehicle group.</p><p><strong>Conclusions:
{"title":"Dl-3-n-butylphthalide Alleviates Cardiac Dysfunction and Injury Possibly by Inhibiting Cell Pyroptosis and Inflammation via the cGAS-STING-TBK1 Pathway in a Porcine Model of Hemorrhage-Induced Cardiac Arrest.","authors":"Ting Zhou, Yong Liu, Lijun Zhu, Jian Jiang, Qijiang Chen, Lulu Li, Xianlong Wu, Jiefeng Xu, Jianjiang Fang","doi":"10.1097/SHK.0000000000002539","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002539","url":null,"abstract":"<p><strong>Introduction: </strong>Dl-3-n-butylphthalide (NBP), a small molecular compound extracted from celery seeds, has been shown to exhibit diverse pharmacological activities, including anti-inflammatory, antioxidative, and anti-apoptotic effects. Recent studies have highlighted its efficacy in treating various cardiovascular conditions, such as myocardial infarction, hypertrophy, heart failure, and cardiotoxicity. This study aimed to investigate whether NBP could alleviate cardiac dysfunction and injury following hemorrhage-induced cardiac arrest (HCA) in a porcine model and elucidate its potential mechanisms.</p><p><strong>Methods: </strong>Seventeen pigs were randomized into three groups: Sham (n = 5), HCA + vehicle (n = 5), and HCA + NBP (n = 7). In the HCA + vehicle and HCA + NBP groups, the HCA model was established by continuous bleeding at a rate of 2 mL/kg/min to induce cardiac arrest. Cardiac arrest was maintained for 7 min, followed by the reinfusion of 50% of the shed blood at a rate of 5 mL/kg/min. After successful resuscitation, the HCA + NBP group received an intravenous dose of 2.5 mg/kg of NBP within 120 min. Post-resuscitation cardiac function (stroke volume, global ejection fraction) and injury biomarkers (cardiac troponin I, creatine kinase-MB) were assessed at regular intervals. At the end of the post-resuscitation observation, cardiac tissue samples were collected to assess: 1) histopathological injury, 2) cellular apoptosis, 3) levels of pro-inflammatory cytokines, including tumor necrosis factor-a (TNF-a), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-18 (IL-18), 4) the expression levels of NOD-like receptor pyrin domain 3 (NLRP3), caspase 1, gasdermin D (GSDMD), cyclic-GMP-AMP synthase (cGAS), stimulator of interferon genes (STING), and tank-binding kinase 1 (TBK1), and 5) the integrated optical density (IOD) of GSDMD N-terminal (GSDMD-N), phosphorylated STING (p-STING), and phosphorylated TBK1 (p-TBK1).</p><p><strong>Results: </strong>Following resuscitation, both stroke volume and global ejection fraction were significantly reduced, while serum levels of cardiac troponin I and creatine kinase-MB were markedly elevated in the HCA + vehicle and HCA + NBP groups compared with the Sham group. However, the extent of cardiac dysfunction and injury was significantly attenuated in the HCA + NBP group relative to the HCA + vehicle group. At 24 h post-resuscitation, substantial cardiac pathological injury and apoptosis were observed. Additionally, pyroptosis-related proteins (NLRP3, caspase-1, GSDMD, GSDMD-N) were upregulated, inflammatory markers (TNF-α, IL-1β, IL-6, IL-18) were elevated, and the activation of the cGAS-STING-TBK1 pathway (cGAS, STING, TBK1, p-STING, p-TBK1) were noted in both the HCA + vehicle and HCA + NBP groups compared to the Sham group. Notably, these pathological changes were significantly attenuated in the HCA + NBP group compared to the HCA + vehicle group.</p><p><strong>Conclusions:","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142922221","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-17DOI: 10.1097/SHK.0000000000002542
Yunfei Xu, Jing Chen, Lin Zhou, Yao Zhao, Nina He, Qing Xu, Jie Zhao, Ying Liu
Background: Sepsis-associated encephalopathy (SAE) represents a severe complication of sepsis, substantially elevating both mortality and healthcare costs for patients. Gastrodin (GAS), a principal bioactive constituent of Gastrodia elata Blume, is neuroprotective in various neurological disorders, including ischemic stroke, epilepsy, Alzheimer's disease, and neuropathic pain. In this study, we sought to investigate whether GAS could serve as a protective agent against SAE.
Methods: Mice were subjected to cecal ligation and puncture (CLP) or the murine brain microvascular endothelial cell bEnd.3 was exposed to lipopolysaccharide (LPS) and subsequently treated with GAS. We assessed neurological deficits, blood-brain barrier (BBB) integrity, neuroinflammation, and the state of ferroptosis to evaluate the regulation of GAS on SAE. Mechanistically, we utilized glutathione peroxidase 4 (GPX4) knockout mice to delineate the crucial role of GPX4 and examined the cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) pathway to uncover the upstream signaling of GPX4.
Results: GAS mitigated neurological deficits in SAE mice and reduced BBB disruption and neuroinflammation both in vivo and in vitro. Functionally, the neuroprotective effects of GAS were realized through the inhibition of ferroptosis. Furthermore, we demonstrated that GPX4 played a pivotal role in this process. Lastly, we found that the COX-2/PGE2 pathway was activated following GAS treatment in SAE mice, thereby increasing the expression level of GPX4.
Conclusions: Our study elucidated that GAS offers protection against SAE by suppressing ferroptosis through the activation of the COX-2/PGE2/GPX4 axis. This research validates the therapeutic potential of GAS and provides novel insights into potential therapeutic strategies for the management of SAE.
{"title":"Gastrodin protects against sepsis-associated encephalopathy by suppressing ferroptosis.","authors":"Yunfei Xu, Jing Chen, Lin Zhou, Yao Zhao, Nina He, Qing Xu, Jie Zhao, Ying Liu","doi":"10.1097/SHK.0000000000002542","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002542","url":null,"abstract":"<p><strong>Background: </strong>Sepsis-associated encephalopathy (SAE) represents a severe complication of sepsis, substantially elevating both mortality and healthcare costs for patients. Gastrodin (GAS), a principal bioactive constituent of Gastrodia elata Blume, is neuroprotective in various neurological disorders, including ischemic stroke, epilepsy, Alzheimer's disease, and neuropathic pain. In this study, we sought to investigate whether GAS could serve as a protective agent against SAE.</p><p><strong>Methods: </strong>Mice were subjected to cecal ligation and puncture (CLP) or the murine brain microvascular endothelial cell bEnd.3 was exposed to lipopolysaccharide (LPS) and subsequently treated with GAS. We assessed neurological deficits, blood-brain barrier (BBB) integrity, neuroinflammation, and the state of ferroptosis to evaluate the regulation of GAS on SAE. Mechanistically, we utilized glutathione peroxidase 4 (GPX4) knockout mice to delineate the crucial role of GPX4 and examined the cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) pathway to uncover the upstream signaling of GPX4.</p><p><strong>Results: </strong>GAS mitigated neurological deficits in SAE mice and reduced BBB disruption and neuroinflammation both in vivo and in vitro. Functionally, the neuroprotective effects of GAS were realized through the inhibition of ferroptosis. Furthermore, we demonstrated that GPX4 played a pivotal role in this process. Lastly, we found that the COX-2/PGE2 pathway was activated following GAS treatment in SAE mice, thereby increasing the expression level of GPX4.</p><p><strong>Conclusions: </strong>Our study elucidated that GAS offers protection against SAE by suppressing ferroptosis through the activation of the COX-2/PGE2/GPX4 axis. This research validates the therapeutic potential of GAS and provides novel insights into potential therapeutic strategies for the management of SAE.</p>","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142922343","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-17DOI: 10.1097/SHK.0000000000002528
Haoran Chen, Xinyi Tang, Xiaomin Li, Yongpeng Xie
Background: The relationship between the partial pressure of oxygen in arterial blood (PaO2) and the prognosis of sepsis patients, and its potential variation over time, remains unclear. The optimal PaO2 range for sepsis patients has always been a contentious issue, with no consensus. We aimed to explore the association between different levels of PaO2 exposure over time and the 28-day mortality of sepsis patients, and to identify the optimal PaO2 range for sepsis patients within a specific time frame.
Methods: We retrieved data on adult patients diagnosed with sepsis within 24 hours before or after ICU admission from the Medical Information Mart for Intensive Care IV (MIMIC-IV; version 2.2) database. We excluded patients who were not admitted to the ICU for the first time, those with ICU stay <24 hours, and those without PaO2 results during their ICU stay. We calculated the time-weighted average (TWA) of PaO2 and used piece-wise exponential additive mixed models (PAMMs) to estimate the time-dependent changes in the association between TWA-PaO2 and patient prognosis.
Results: A total of 16,880 sepsis patients were included in the MIMIC cohort. Results indicated that patients' TWA-PaO2 correlates with increased 28-day mortality after intensive care unit (ICU) admission in sepsis patients, and this association was mainly manifested in the early course of the disease. With a time window of the first 1-7 days after ICU admission, the optimal TWA-PaO2 range for sepsis patients was ≥130 mmHg and ≤ 160 mmHg. Increased exposure time, proportion of exposure time, and exposure dose of high-risk PaO2 outside the range were all associated with an increased risk of 28-day mortality.
Conclusion: PaO2 in sepsis patients should be closely monitored. During the first 1-7 days of ICU admission, PaO2 should be maintained within the range of ≥130 mmHg and ≤ 160 mmHg. A dose-dependent relationship exists between high-risk PaO2 outside the range and patient outcome.
{"title":"Optimal PaO2 is 130-160 mmHg in the first week for sepsis patients in ICU: a retrospective cohort study based on MIMIC-IV database.","authors":"Haoran Chen, Xinyi Tang, Xiaomin Li, Yongpeng Xie","doi":"10.1097/SHK.0000000000002528","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002528","url":null,"abstract":"<p><strong>Background: </strong>The relationship between the partial pressure of oxygen in arterial blood (PaO2) and the prognosis of sepsis patients, and its potential variation over time, remains unclear. The optimal PaO2 range for sepsis patients has always been a contentious issue, with no consensus. We aimed to explore the association between different levels of PaO2 exposure over time and the 28-day mortality of sepsis patients, and to identify the optimal PaO2 range for sepsis patients within a specific time frame.</p><p><strong>Methods: </strong>We retrieved data on adult patients diagnosed with sepsis within 24 hours before or after ICU admission from the Medical Information Mart for Intensive Care IV (MIMIC-IV; version 2.2) database. We excluded patients who were not admitted to the ICU for the first time, those with ICU stay <24 hours, and those without PaO2 results during their ICU stay. We calculated the time-weighted average (TWA) of PaO2 and used piece-wise exponential additive mixed models (PAMMs) to estimate the time-dependent changes in the association between TWA-PaO2 and patient prognosis.</p><p><strong>Results: </strong>A total of 16,880 sepsis patients were included in the MIMIC cohort. Results indicated that patients' TWA-PaO2 correlates with increased 28-day mortality after intensive care unit (ICU) admission in sepsis patients, and this association was mainly manifested in the early course of the disease. With a time window of the first 1-7 days after ICU admission, the optimal TWA-PaO2 range for sepsis patients was ≥130 mmHg and ≤ 160 mmHg. Increased exposure time, proportion of exposure time, and exposure dose of high-risk PaO2 outside the range were all associated with an increased risk of 28-day mortality.</p><p><strong>Conclusion: </strong>PaO2 in sepsis patients should be closely monitored. During the first 1-7 days of ICU admission, PaO2 should be maintained within the range of ≥130 mmHg and ≤ 160 mmHg. A dose-dependent relationship exists between high-risk PaO2 outside the range and patient outcome.</p>","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142882883","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-16DOI: 10.1097/SHK.0000000000002541
Alen Palackic, Amina El Ayadi, Charles E Wade, Lisa A Baer, Ludwik K Branski, Gabriel Hundeshagen, Julia Kleinhapl, Steven E Wolf, Juquan Song
Background: Loss of muscle mass and strength in patients who have experienced severe burns is dramatic and associated with subsequent functional impairment. Past work has shown that exercise and oxandrolone, an anabolic steroid, individually improve muscle function and muscle mass in severely burned patients. This study aims to evaluate the effect of oxandrolone treatment combined with resistance exercise on muscle atrophy and investigate the protein synthesis and mitochondrial biogenesis pathways in a hindlimb suspension model.
Methods: Twenty-four Sprague Dawley rats received 40% total body surface area (%TBSA) scald burns and were then placed for hindlimb unloading. All animals were randomly grouped into vehicle (corn oil) without exercise (V/NEX), oxandrolone administration (0.1 mg/kg/day) without exercise (OX/NEX), vehicle with exercise (V/EX), or oxandrolone with exercise (OX/EX). (n = 6/group). On day 14 isometric forces of the left plantaris and soleus muscle were measured by using a muscle lever system with dynamic muscle control and analysis software. Fatigue measurement was only performed in the soleus muscle. The tissue of the muscle was then collected for protein extraction. Western blots were performed to study signal alternations and mitochondrial biogenesis pathways.
Results: Tetanic force (Po) was significantly increased in the plantaris with exercise, rather than with oxandrolone treatment. Fatigue index (FI) was lower and integration was significantly elevated in the soleus with exercise but not with oxandrolone treatment. Fatigue curve in the soleus further revealed the average maximum force were achieved in soleus with either oxandrolone treatment or exercise alone independently. Raptor and p-Akt levels are elevated in the OX/EX group while PGC1a expression was not altered.
Conclusion: Oxandrolone and resistance exercise have independent positive effects on muscle function recovery in this clinically relevant rodent model of severe burn. Both treatments combined increased signaling pathways by increasing protein synthesis.
{"title":"Combined Effects of Oxandrolone and Exercise on Muscle Recovery in Rats with Severe Burn and Hindlimb Unloading.","authors":"Alen Palackic, Amina El Ayadi, Charles E Wade, Lisa A Baer, Ludwik K Branski, Gabriel Hundeshagen, Julia Kleinhapl, Steven E Wolf, Juquan Song","doi":"10.1097/SHK.0000000000002541","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002541","url":null,"abstract":"<p><strong>Background: </strong>Loss of muscle mass and strength in patients who have experienced severe burns is dramatic and associated with subsequent functional impairment. Past work has shown that exercise and oxandrolone, an anabolic steroid, individually improve muscle function and muscle mass in severely burned patients. This study aims to evaluate the effect of oxandrolone treatment combined with resistance exercise on muscle atrophy and investigate the protein synthesis and mitochondrial biogenesis pathways in a hindlimb suspension model.</p><p><strong>Methods: </strong>Twenty-four Sprague Dawley rats received 40% total body surface area (%TBSA) scald burns and were then placed for hindlimb unloading. All animals were randomly grouped into vehicle (corn oil) without exercise (V/NEX), oxandrolone administration (0.1 mg/kg/day) without exercise (OX/NEX), vehicle with exercise (V/EX), or oxandrolone with exercise (OX/EX). (n = 6/group). On day 14 isometric forces of the left plantaris and soleus muscle were measured by using a muscle lever system with dynamic muscle control and analysis software. Fatigue measurement was only performed in the soleus muscle. The tissue of the muscle was then collected for protein extraction. Western blots were performed to study signal alternations and mitochondrial biogenesis pathways.</p><p><strong>Results: </strong>Tetanic force (Po) was significantly increased in the plantaris with exercise, rather than with oxandrolone treatment. Fatigue index (FI) was lower and integration was significantly elevated in the soleus with exercise but not with oxandrolone treatment. Fatigue curve in the soleus further revealed the average maximum force were achieved in soleus with either oxandrolone treatment or exercise alone independently. Raptor and p-Akt levels are elevated in the OX/EX group while PGC1a expression was not altered.</p><p><strong>Conclusion: </strong>Oxandrolone and resistance exercise have independent positive effects on muscle function recovery in this clinically relevant rodent model of severe burn. Both treatments combined increased signaling pathways by increasing protein synthesis.</p>","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142922129","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-10DOI: 10.1097/SHK.0000000000002535
Xiaoke Wang, Xiaojuan Fan, Taibo Wu, Shaopeng Che, Xue Shi, Peining Liu, Junhui Liu, Yongbai Luo, Yue Wu, Beidi Lan
Background: While acute myocardial infarction (AMI) is widely recognized as the primary cause of Cardiogenic Shock (CS), Non-AMI related CS has been excluded from the majority of CS studies. Information on its prognostic factors remains largely understudied, and it is necessary to focus on these patients to identify the specific risk factors. In this study, we aimed to build and validate a predictive nomogram and risk classification system.
Methods: 1298 patients and 548 patients with CS from the Medical Information Mart for Intensive Care IV (MIMIC-IV) and MIMIC-III databases were included in the study after excluding patients with acute myocardial infarction. Lasson and logistic regression analysis were used to identify statistically significant predictors which were finally involved in the nomogram. The predictive performance of the nomogram was validated by calibration plots and was compared with other scoring systems by AUC and DCA curves.
Results: Age, heart rate, WBC count, albumin level, lactic acid level, GCS Score, 24 h urine output, and vasopressor use were identified as the most critical factors for in-hospital death. Based on these results, a nomogram was established for predicting in-hospital mortality. The AUC value of the nomogram was 0.806 in the training group and 0.814 and 0.730 in the internal and external validation sets, respectively, which were significantly higher than those of other commonly used Intensive Care Unit scoring systems (SAPSII, APSIII, and SOFA).In addition, the survival curve showed significant differences in the 30-day survival of the three risk subgroups divided by the nomogram.
Conclusion: For non-AMI associated CS, a predictive nomogram and risk classification system were developed and validated, and the nomogram demonstrated good performance in prognostic prediction and risk stratification.
{"title":"A Clinical Prediction Model For Short-Term Prognosis In Patients With Non Acute Myocardial Infarction Related Cardiogenic Shock.","authors":"Xiaoke Wang, Xiaojuan Fan, Taibo Wu, Shaopeng Che, Xue Shi, Peining Liu, Junhui Liu, Yongbai Luo, Yue Wu, Beidi Lan","doi":"10.1097/SHK.0000000000002535","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002535","url":null,"abstract":"<p><strong>Background: </strong>While acute myocardial infarction (AMI) is widely recognized as the primary cause of Cardiogenic Shock (CS), Non-AMI related CS has been excluded from the majority of CS studies. Information on its prognostic factors remains largely understudied, and it is necessary to focus on these patients to identify the specific risk factors. In this study, we aimed to build and validate a predictive nomogram and risk classification system.</p><p><strong>Methods: </strong>1298 patients and 548 patients with CS from the Medical Information Mart for Intensive Care IV (MIMIC-IV) and MIMIC-III databases were included in the study after excluding patients with acute myocardial infarction. Lasson and logistic regression analysis were used to identify statistically significant predictors which were finally involved in the nomogram. The predictive performance of the nomogram was validated by calibration plots and was compared with other scoring systems by AUC and DCA curves.</p><p><strong>Results: </strong>Age, heart rate, WBC count, albumin level, lactic acid level, GCS Score, 24 h urine output, and vasopressor use were identified as the most critical factors for in-hospital death. Based on these results, a nomogram was established for predicting in-hospital mortality. The AUC value of the nomogram was 0.806 in the training group and 0.814 and 0.730 in the internal and external validation sets, respectively, which were significantly higher than those of other commonly used Intensive Care Unit scoring systems (SAPSII, APSIII, and SOFA).In addition, the survival curve showed significant differences in the 30-day survival of the three risk subgroups divided by the nomogram.</p><p><strong>Conclusion: </strong>For non-AMI associated CS, a predictive nomogram and risk classification system were developed and validated, and the nomogram demonstrated good performance in prognostic prediction and risk stratification.</p>","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142814215","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-05DOI: 10.1097/SHK.0000000000002533
Stéphane Bar, John Diaper, Fabienne Fontao, Xavier Belin, Stanislas Abrard, Gergely Albu, Hervé Dupont, Walid Habre, Eduardo Schiffer
Background: Intestinal ischemia-reperfusion injury is associated with both macrocirculatory and microcirculatory failure. Association of a vasoconstrictor in combination with a vasodilator such as ilomedin may improve macrocirculation parameters, microcirculation perfusion and reduce endothelial dysfunction. The primary objective was to demonstrate a difference in mean arterial pressure (MAP) after intestinal reperfusion with the concomitant administration of norepinephrine and ilomedin during ischemia compared with traditional hemodynamic treatment strategies (fluid resuscitation and vasopressors only). Secondary objectives were to demonstrate an improvement in peripheral and intestinal microcirculatory perfusion and endothelial dysfunction after intestinal reperfusion using this association.
Methods: We conducted a randomized preclinical trial in twenty-one large white pigs, in which a 2-hour small bowel ischemia was performed using a segmental mesenteric occlusion model, followed by a 2-hour reperfusion. Pigs were randomized into 3 groups: goal directed fluid therapy, early administration of norepinephrine before reperfusion and early administration of ilomedin and norepinephrine before reperfusion. Macrocirculatory (MAP and Cardiac Index (CI)), microcirculatory (Sublingual with SideStream Dark Field system and intestinal hemoglobin oxygen saturation with hyperspectral imaging (HSI)) measurements and biological analysis (biomarkers of endothelial dysfunction) were performed.
Results: There were no significant differences in the MAP (p = 0.499) and the CI (p = 0.659) between the 3 groups. Perfused Vessel Density (PVD) in sublingual microcirculation was significantly higher immediately after reperfusion and 2 hours after reperfusion in the early administration of ilomedin and norepinephrine group compared with the other 2 groups (p < 0.05). Hemoglobin oxygen saturation measured at the intestinal level was significantly higher immediately after reperfusion in the early administration of ilomedin and norepinephrine group compared with the other 2 groups (p < 0.01). There were no significant differences in biomarkers of endothelial dysfunction between the 3 groups. Creatinine, AST and alkaline phosphatases increased significantly 2 hours after reperfusion in the early administration of ilomedin and norepinephrine group compared with baseline (p < 0.05).
Conclusions: Early administration of norepinephrine and ilomedin during ischemia improved short-term post-reperfusion sublingual and intestinal microcirculation without worsening macrocirculatory parameters in an intestinal ischemia-reperfusion injury model. However, use of this strategy seemed to worsen both liver and kidney function.
{"title":"Early and concomitant administration of norepinephrine and ilomedin improves microcirculatory perfusion without impairing macrocirculation in an intestinal ischemia-reperfusion injury swine model : a randomized experimental trial.","authors":"Stéphane Bar, John Diaper, Fabienne Fontao, Xavier Belin, Stanislas Abrard, Gergely Albu, Hervé Dupont, Walid Habre, Eduardo Schiffer","doi":"10.1097/SHK.0000000000002533","DOIUrl":"https://doi.org/10.1097/SHK.0000000000002533","url":null,"abstract":"<p><strong>Background: </strong>Intestinal ischemia-reperfusion injury is associated with both macrocirculatory and microcirculatory failure. Association of a vasoconstrictor in combination with a vasodilator such as ilomedin may improve macrocirculation parameters, microcirculation perfusion and reduce endothelial dysfunction. The primary objective was to demonstrate a difference in mean arterial pressure (MAP) after intestinal reperfusion with the concomitant administration of norepinephrine and ilomedin during ischemia compared with traditional hemodynamic treatment strategies (fluid resuscitation and vasopressors only). Secondary objectives were to demonstrate an improvement in peripheral and intestinal microcirculatory perfusion and endothelial dysfunction after intestinal reperfusion using this association.</p><p><strong>Methods: </strong>We conducted a randomized preclinical trial in twenty-one large white pigs, in which a 2-hour small bowel ischemia was performed using a segmental mesenteric occlusion model, followed by a 2-hour reperfusion. Pigs were randomized into 3 groups: goal directed fluid therapy, early administration of norepinephrine before reperfusion and early administration of ilomedin and norepinephrine before reperfusion. Macrocirculatory (MAP and Cardiac Index (CI)), microcirculatory (Sublingual with SideStream Dark Field system and intestinal hemoglobin oxygen saturation with hyperspectral imaging (HSI)) measurements and biological analysis (biomarkers of endothelial dysfunction) were performed.</p><p><strong>Results: </strong>There were no significant differences in the MAP (p = 0.499) and the CI (p = 0.659) between the 3 groups. Perfused Vessel Density (PVD) in sublingual microcirculation was significantly higher immediately after reperfusion and 2 hours after reperfusion in the early administration of ilomedin and norepinephrine group compared with the other 2 groups (p < 0.05). Hemoglobin oxygen saturation measured at the intestinal level was significantly higher immediately after reperfusion in the early administration of ilomedin and norepinephrine group compared with the other 2 groups (p < 0.01). There were no significant differences in biomarkers of endothelial dysfunction between the 3 groups. Creatinine, AST and alkaline phosphatases increased significantly 2 hours after reperfusion in the early administration of ilomedin and norepinephrine group compared with baseline (p < 0.05).</p><p><strong>Conclusions: </strong>Early administration of norepinephrine and ilomedin during ischemia improved short-term post-reperfusion sublingual and intestinal microcirculation without worsening macrocirculatory parameters in an intestinal ischemia-reperfusion injury model. However, use of this strategy seemed to worsen both liver and kidney function.</p>","PeriodicalId":21667,"journal":{"name":"SHOCK","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142786939","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}