1
|
Deng X, Zhou J, Fang W, Sun R, Yan G, Sun Y. pH-triggered small molecule nanodrugs self-assembled from tryptamine-cinnamaldehyde and fisetin for targeted sepsis-associated encephalopathy therapy. J Biomater Appl 2025; 39:1165-1176. [PMID: 39894462 DOI: 10.1177/08853282251318052] [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: 02/04/2025]
Abstract
Sepsis-associated encephalopathy (SAE) is an acute diffuse brain dysfunction, but its clinical treatment just focuses on antibiotics and supportive therapy, which fail to directly limit the development of SAE. Herein, this work highlights the development of pH-triggered small molecule nanodrugs self-assembled from tryptamine (Try)-cinnamaldehyde (CA) and fisetin for targeted SAE therapy. The imine linkage in Try-CA and acid-dependent protonation of Try and fisetin endow the nanodrugs with pH-triggered dynamic changes of particle sizes, surficial charges, and drug release. Moreover, the combined use of Try-CA and fisetin also endows the nanodrugs with superior antioxidative, anti-inflammatory and antibacterial capabilities compared to their individual use. These characteristics of the nanodrugs facilitate long-term circulation stability, effective penetration through BBB, selective accumulation in the brain, and target to central and peripheral focal areas, thereby achieving comprehensive treatment or relief of SAE. Thus, these attractive experimental results illuminate the enormous potential of such pH-triggered small molecule nanodrugs for targeted SAE therapy, advancing their use in clinics.
Collapse
Affiliation(s)
- Ximing Deng
- Department of Critical Care Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jinyao Zhou
- Department of Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Wei Fang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Rao Sun
- Department of Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Guoqing Yan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei, China
| | - Yun Sun
- Department of Critical Care Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Wang T, Huang X, Sun S, Wang Y, Han L, Zhang T, Zhang T, Chen X. Recent Advances in the Mechanisms of Postoperative Neurocognitive Dysfunction: A Narrative Review. Biomedicines 2025; 13:115. [PMID: 39857699 PMCID: PMC11762480 DOI: 10.3390/biomedicines13010115] [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: 12/03/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Postoperative neurocognitive dysfunction (PND) is a prevalent and debilitating complication in elderly surgical patients, characterized by persistent cognitive decline that negatively affects recovery and quality of life. As the aging population grows, the rising number of elderly surgical patients has made PND an urgent clinical challenge. Despite increasing research efforts, the pathophysiological mechanisms underlying PND remain inadequately characterized, underscoring the need for a more integrated framework to guide targeted interventions. To better understand the molecular mechanisms and therapeutic targets of PND, this narrative review synthesized evidence from peer-reviewed studies, identified through comprehensive searches of PubMed, Embase, Cochrane Library, and Web of Science. Key findings highlight neuroinflammation, oxidative stress, mitochondrial dysfunction, neurotransmitter imbalances, microvascular changes, and white matter lesions as central to PND pathophysiology, with particular parallels to encephalocele- and sepsis-associated cognitive impairments. Among these, neuroinflammation, mediated by pathways such as the NLRP3 inflammasome and blood-brain barrier disruption, emerges as a pivotal driver, triggering cascades that exacerbate neuronal injury. Oxidative stress and mitochondrial dysfunction synergistically amplify these effects, while neurotransmitter imbalances and microvascular alterations, including white matter lesions, contribute to synaptic dysfunction and cognitive decline. Anesthetic agents modulate these interconnected pathways, exhibiting both protective and detrimental effects. Propofol and dexmedetomidine demonstrate neuroprotective properties by suppressing neuroinflammation and microglial activation, whereas inhalational anesthetics like sevoflurane intensify oxidative stress and inflammatory responses. Ketamine, with its anti-inflammatory potential, offers promise but requires further evaluation to determine its long-term safety and efficacy. By bridging molecular insights with clinical practice, this review highlights the critical role of personalized anesthetic strategies in mitigating PND and improving cognitive recovery in elderly surgical patients. It aims to inform future research and clinical decision-making to address this multifaceted challenge.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xin Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| |
Collapse
|
3
|
Li Y, Zhou F, You J, Gong X. Annexin A1 Mitigates Blood-Brain Barrier Disruption in a Sepsis-Associated Encephalopathy Model by Enhancing the Expression of Occludin and Zonula Occludens-1 (ZO-1). CNS Neurosci Ther 2024; 30:e70173. [PMID: 39727329 DOI: 10.1111/cns.70173] [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: 09/02/2024] [Revised: 11/15/2024] [Accepted: 12/01/2024] [Indexed: 12/28/2024] Open
Abstract
AIMS This study investigated the protective role of Annexin A1 (ANXA1) in sepsis-associated encephalopathy (SAE) by examining its effects on brain vascular endothelium and blood-brain barrier (BBB) integrity. METHODS Mice were divided into four groups: wild type (WT), cecal ligation and puncture (CLP), ANXA1 knockout (ANXA1[-/-]), and ANXA1(-/-) with CLP. Neurobehavioral changes were assessed using the Y-maze test, while BBB integrity was evaluated through Evans blue dye (EBD) staining and permeability tests with fluorescein isothiocyanate (FITC)-dextran. RESULTS Results showed that ANXA1 levels were reduced in septic mice, and its deficiency exacerbated cognitive impairment and survival rate reduction. ANXA1 deficiency also upregulated proinflammatory cytokines and adhesion molecules, worsened BBB impairment, and altered expression of tight junction proteins and VEGF-A/VEGF-R2. In vitro, ANXA1 Ac2-26 prevented LPS-induced increased permeability in bEnd.3 cells by restoring tight junction proteins and reducing VEGF-A/VEGF-R2 expression. Notably, VEGF-A negated the protective effects of ANXA1 Ac2-26. CONCLUSION The study concludes that ANXA1 reduces BBB permeability to protect against sepsis-induced brain dysfunction via VEGF-A/VEGF-R2 regulation of tight junction proteins, suggesting ANXA1 as a potential therapeutic for SAE.
Collapse
Affiliation(s)
- Yao Li
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Fang Zhou
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiyue You
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinran Gong
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Liu Y, Wu W, Fu D. Genetic differences in hippocampus of mice susceptible to sepsis-associated encephalopathy. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:1777-1789. [PMID: 40177761 PMCID: PMC11964805 DOI: 10.11817/j.issn.1672-7347.2024.240045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Indexed: 04/05/2025]
Abstract
OBJECTIVES Sepsis-associated encephalopathy (SAE) is a common complication of sepsis, which can lead to long-term cognitive impairment and anxiety in patients, and may even contribute to mortality in septic individuals. There is substantial individual variability in the incidence and severity and susceptibility of SAE, but the mechanisms regulating susceptibility remain unclear. Previous studies have shown that hippocampal damage is directly associated with cognitive and emotional disturbances in SAE. This study aims to explore the impact of hippocampal differentially expressed genes on SAE susceptibility in a mouse model. METHODS Male specific pathogen-free (SPF)-grade C57BL/6 mice (6-8 weeks old) were randomly divided into a saline control group (Con group) and an SAE model group. SAE was induced by intraperitoneal injection of 10 mg/kg lipopolysaccharide (LPS), while control mice received an equivalent volume of saline. Cognitive and anxiety-like behaviors were assessed using the open field test (OFT), novel object recognition (NOR), and Y-maze test. Based on mean±standard deviation of behavioral results from the Con group, SAE mice were further classified into high-sensitivity (HS) and low-sensitivity (LS) subgroups. Immunohistochemistry was performed to detect the expression of immediate early gene c-Fos and neuronal marker neuronal nuclei (NeuN). Nissl staining was used to assess neuronal injury in the dentate gyrus (DG), cornu ammonis 1 (CA1), and cornu ammonis 3 (CA3) regions of the hippocampus. RNA sequencing (RNA-seq) was conducted on hippocampal tissues from HS and LS mice to identify differentially expressed genes, followed by pathway enrichment analysis. RESULTS No significant behavioral susceptibility differences were observed between the overall SAE group and controls. However, HS mice showed severer cognitive deficits and anxiety-like behavior compared to LS mice. Immunohistochemistry revealed significantly higher expression of c-Fos in the hippocampus of LS mice (P<0.05), while Nissl and NeuN staining revealed milder neuronal damage in the hippocampus of LS mice than that of HS mice (both P<0.05). RNA-seq analysis identified 130 upregulated and 142 downregulated DEGs in LS and HS mice, respectively. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that upregulated genes in LS mice were primarily involved in pluripotency regulation, cyclic adenosine monophosphate (cAMP) signaling, and Wnt signaling pathways, in contrast, the downregulated genes were mainly related to cell adhesion, neuroactive ligand-receptor interaction, and calcium signaling pathways. CONCLUSIONS Differential gene expression in the hippocampus may contribute to individual susceptibility to cognitive and emotional dysfunction in SAE, suggesting potential genetic targets for individualized intervention.
Collapse
Affiliation(s)
- Yu Liu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008.
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha 410011.
- Department of Human Anatomy, School of Basic Medical Sciences, Central South University, Changsha 410013, China.
| | - Weiwei Wu
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha 410011.
| | - Di Fu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008.
| |
Collapse
|
5
|
Li X, Xu X, Zhang J, Wang X, Zhao C, Liu Q, Fan K. Review of the therapeutic effects of traditional Chinese medicine in sepsis-associated encephalopathy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118588. [PMID: 39029543 DOI: 10.1016/j.jep.2024.118588] [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: 02/22/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sepsis-associated encephalopathy (SAE) is a common and serious complication during the acute phase of and after recovery from sepsis that seriously affects the quality of life of patients. Traditional Chinese medicine (TCM) has been widely used in modern medicine for neurological anomalies and has become a therapeutic tool for the treatment of SAE due to its multitargeting effects and low toxicity and side effects. AIMS OF THE STUDY This review provides insights into the pathogenesis and treatments of SAE, focusing on the clinical and experimental impacts of TCM formulations and their single components. METHODS Several known databases such as PubMed, Web of Science, Google Scholar, China National Knowledge Infrastructure (CNKI), and others were extensively explored with keywords and phrases such as "sepsis-associated encephalopathy", "traditional Chinese medicine", "herbs", "SAE", "sepsis", "cerebral" or other relevant terms to obtain literature between 2018 and 2024. RESULTS Extensive evidence indicated that TCM could decrease mortality and normalize neurological function in patients with sepsis; these effects might be associated with factors such as reduced oxidative stress and downregulated expression of inflammatory factors. CONCLUSIONS TCM shows notable efficacy in treating SAE, warranting deeper mechanistic studies to optimize its clinical application.
Collapse
Affiliation(s)
- Xingyao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Jun Zhang
- Intensive Care Unit, Wuhan Hospital of Traditional Chinese Medicine, Wu Han, 430014, China.
| | - Xuerui Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Chunming Zhao
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Kai Fan
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
6
|
Peng S, Sun T, Yang D, Zhao H, Lin L, Xia B, Li M, Piao M, Shi Z, Tuo Q. Dipsacoside B ameliorates cognitive impairment in sepsis-associated encephalopathy by reducing Th17 cell infiltration and neuroinflammation. Biochem Pharmacol 2024; 227:116428. [PMID: 39009096 DOI: 10.1016/j.bcp.2024.116428] [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: 04/16/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is the main cause of cognitive impairment in patients with sepsis. The infiltration of inflammatory signals into the central nervous system (CNS) via the compromised blood-brain barrier (BBB) represents a crucial step in the pathological progression of SAE. In particular, T-helper 17 cell (Th17 cells) has been suggested to be highly correlated with the activation of central immune responses. Thus, preventing Th17 cell infiltration into the CNS may be a possible strategy to alleviate cognitive decline in SAE. Dipsacoside B (DB) is one of the primary active components in Chuan Xu Duan (Dipsacus asper Wall). We speculate that DB may be a potential candidate for the treatment of SAE-related cognitive deficits. In the present study, we demonstrated that DB could effectively alleviate cognitive impairment in SAE mice. DB significantly suppressed the central inflammatory response induced by repeated lipopolysaccharide (LPS) injection. The mechanism underlying its therapeutic effect should be attributed to the reduction of BBB impairment and pathogenic Th17 cell infiltration into the CNS by inhibition of vascular endothelial growth factor A (VEGFA)/ Vascular endothelial growth factor receptor 2(VEGFR2)/ Endothelial nitric oxide synthase (eNOS) signaling. Our findings suggest that DB is a potential candidate for the treatment of SAE-related cognitive dysfunction.
Collapse
Affiliation(s)
- Sha Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China; Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
| | - Taoli Sun
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China; Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
| | - Dongmei Yang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China; Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China; Basic Research Center of Integrated Chinese and Western medicine on prevention and treatment of vascular diseases, Medical School, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
| | - Hongqing Zhao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
| | - Limei Lin
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
| | - Bohou Xia
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
| | - Minjie Li
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
| | - Meihong Piao
- Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China; Basic Research Center of Integrated Chinese and Western medicine on prevention and treatment of vascular diseases, Medical School, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
| | - Zhe Shi
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China.
| | - Qinhui Tuo
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China; Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China; Basic Research Center of Integrated Chinese and Western medicine on prevention and treatment of vascular diseases, Medical School, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China.
| |
Collapse
|
7
|
Fang Y, Dou A, Shen Y, Li T, Liu H, Cui Y, Xie K. Association of triglyceride-glucose index and delirium in patients with sepsis: a retrospective study. Lipids Health Dis 2024; 23:227. [PMID: 39054513 PMCID: PMC11271053 DOI: 10.1186/s12944-024-02213-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: 06/03/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE It is well known that glucose and lipid metabolism disorders and insulin resistance are common in sepsis, which affect the occurrence and prognosis of multiple organ dysfunction in septic patients. Previous study reported the predictive value of triglyceride-glucose index (TyG), a clinical indicator for insulin resistance, in postoperative delirium patients. However, it remains unclear whether the TyG index is a novel predictive biomarker for sepsis-associated delirium. The aim of this study is to explore the relationship between TyG index and the risk of delirium in patients with sepsis. METHODS Adult septic patients were identified from the MIMIC-IV database and divided into four groups based on the mean value of TyG. The primary outcome was the incidence of delirium. The association between TyG and the risk of developing delirium was evaluated by restricted cubic spline (RCS), multivariate logistic regression and subgroup analysis. Propensity Score Matching (PSM) method was used to balance the baseline data. RESULTS A total of 3,331 septic patients were included in the analysis, and further divided into four groups: Q1 (TyG ≤ 8.67), Q2 (8.67 < TyG ≤ 9.08), Q3 (9.08 < TyG ≤ 9.61), and Q4 (TyG > 9.61). The RCS curves demonstrated a non-linear positive relationship between TyG index and the risk of developing delirium, and an optimal cut-of value 9.09 was recommended. After balancing the baseline information by PSM, patients in the TyG > 9.09 group had a significant higher incidence of delirium compared with those in the TyG ≤ 9.09 group. In logistic regression analysis, TyG > 9.09 was significantly associated with lower risk of developing delirium in both original cohort (OR 1.54-1.78, all P < 0.001) and the PSM cohort (OR 1.41-1.48, all P < 0.001). No association was found between the TyG index and mortality (all P > 0.05). In subgroup analysis, our findings were consistent (all OR > 1 in all subgroups). CONCLUSION Our study demonstrated an independent association between TyG index and increased risk of delirium in septic patients, indicating that TyG index can serve as a biomarker for delirium in sepsis.
Collapse
Affiliation(s)
- Yipeng Fang
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Aizhen Dou
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Yuehao Shen
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Tianyu Li
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Haiying Liu
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Yan Cui
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China.
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22, Qixiangtai Road, Heping District, Tianjin, 300070, China.
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China.
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
8
|
Abdalla MMI. Insulin resistance as the molecular link between diabetes and Alzheimer's disease. World J Diabetes 2024; 15:1430-1447. [PMID: 39099819 PMCID: PMC11292327 DOI: 10.4239/wjd.v15.i7.1430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 07/08/2024] Open
Abstract
Diabetes mellitus (DM) and Alzheimer's disease (AD) are two major health concerns that have seen a rising prevalence worldwide. Recent studies have indicated a possible link between DM and an increased risk of developing AD. Insulin, while primarily known for its role in regulating blood sugar, also plays a vital role in protecting brain functions. Insulin resistance (IR), especially prevalent in type 2 diabetes, is believed to play a significant role in AD's development. When insulin signalling becomes dysfunctional, it can negatively affect various brain functions, making individuals more susceptible to AD's defining features, such as the buildup of beta-amyloid plaques and tau protein tangles. Emerging research suggests that addressing insulin-related issues might help reduce or even reverse the brain changes linked to AD. This review aims to explore the rela-tionship between DM and AD, with a focus on the role of IR. It also explores the molecular mechanisms by which IR might lead to brain changes and assesses current treatments that target IR. Understanding IR's role in the connection between DM and AD offers new possibilities for treatments and highlights the importance of continued research in this interdisciplinary field.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Department of Human Biology, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
9
|
Zhou Y, Bai L, Tang W, Yang W, Sun L. Research progress in the pathogenesis of sepsis-associated encephalopathy. Heliyon 2024; 10:e33458. [PMID: 39027435 PMCID: PMC11254713 DOI: 10.1016/j.heliyon.2024.e33458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
Sepsis is a syndrome that causes dysfunction of multiple organs due to the host's uncontrolled response to infection and is a significant contributor to morbidity and mortality in intensive care units worldwide. Surviving patients are often left with acute brain injury and long-term cognitive impairment, known as sepsis-associated encephalopathy (SAE). In recent years, researchers have directed their focus towards the pathogenesis of SAE. However, due to the complexity of its development, there remains a lack of effective treatment measures that arise as a serious issue affecting the prognosis of sepsis patients. Further research on the possible causes of SAE aims to provide clinicians with potential therapeutic targets and help develop targeted prevention strategies. This paper aims to review recent research on the pathogenesis of SAE, in order to enhance our understanding of this syndrome.
Collapse
Affiliation(s)
- Yue Zhou
- Teaching Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Lu Bai
- Department of Medical Oncology, Dalian NO.3 People's Hospital, Dalian, 116091, China
| | - Wenjing Tang
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun, 130021, China
| | - Weiying Yang
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun, 130021, China
| | - Lichao Sun
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
10
|
Nwafor DC, Obiri-Yeboah D, Fazad F, Blanks W, Mut M. Focused ultrasound as a treatment modality for gliomas. Front Neurol 2024; 15:1387986. [PMID: 38813245 PMCID: PMC11135048 DOI: 10.3389/fneur.2024.1387986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Ultrasound waves were initially used as a diagnostic tool that provided critical insights into several pathological conditions (e.g., gallstones, ascites, pneumothorax, etc.) at the bedside. Over the past decade, advancements in technology have led to the use of ultrasound waves in treating many neurological conditions, such as essential tremor and Parkinson's disease, with high specificity. The convergence of ultrasound waves at a specific region of interest/target while avoiding surrounding tissue has led to the coined term "focused ultrasound (FUS)." In tumor research, ultrasound technology was initially used as an intraoperative guidance tool for tumor resection. However, in recent years, there has been growing interest in utilizing FUS as a therapeutic tool in the management of brain tumors such as gliomas. This mini-review highlights the current knowledge surrounding using FUS as a treatment modality for gliomas. Furthermore, we discuss the utility of FUS in enhanced drug delivery to the central nervous system (CNS) and highlight promising clinical trials that utilize FUS as a treatment modality for gliomas.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, United States
| | - Derrick Obiri-Yeboah
- Department of Neurological Surgery, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
| | - Faraz Fazad
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, United States
| | - William Blanks
- Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Melike Mut
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
11
|
LI Z, WANG X, Luis U, Ayman Y, BAI Y, XU X, LIU Q. Complementary and alternative medicine on cognitive defects and neuroinflammation after sepsis. J TRADIT CHIN MED 2024; 44:408-416. [PMID: 38504548 PMCID: PMC10927414 DOI: 10.19852/j.cnki.jtcm.20240203.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/27/2023] [Indexed: 03/21/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is a common manifestation of sepsis, ranging from mild confusion and delirium to severe cognitive impairment and deep coma. SAE is associated with higher mortality and long-term outcomes, particularly substantial declines in cognitive function. The mechanisms of SAE probably include neuroinflammation that is mediated by systemic inflammation and ischemic lesions in the brain, a disrupted blood-brain barrier, oxidative stress, neurotransmitter dysfunction, and severe microglial activation. Increasing evidence suggests that complementary and alternative medicine, especially Traditional Chinese Medicine (TCM), is favorable in alleviating cognitive decline after sepsis. Here, we summarized the studies of traditional herbal remedies, TCM formulas and acupuncture therapy in animal models of neurological dysfunctions after sepsis in recent decades and reviewed their potential mechanisms.
Collapse
Affiliation(s)
- Zhenxuan LI
- 1 Department of Infectious Diseases, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- 2 Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China
- 3 Infection immunity laboratory, Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
- 4 Clinical College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Xuerui WANG
- 1 Department of Infectious Diseases, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- 2 Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China
- 3 Infection immunity laboratory, Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
- 4 Clinical College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Ulloa Luis
- 5 Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Youssef Ayman
- 5 Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Yunjing BAI
- 1 Department of Infectious Diseases, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- 2 Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China
- 3 Infection immunity laboratory, Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
- 4 Clinical College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Xiaolong XU
- 1 Department of Infectious Diseases, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- 2 Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China
- 3 Infection immunity laboratory, Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
- 4 Clinical College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Qingquan LIU
- 1 Department of Infectious Diseases, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- 2 Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, China
- 3 Infection immunity laboratory, Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
- 4 Clinical College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
12
|
Li Z, Wang H, Yin Y. Peripheral inflammation is a potential etiological factor in Alzheimer's disease. Rev Neurosci 2024; 35:99-120. [PMID: 37602685 DOI: 10.1515/revneuro-2023-0049] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023]
Abstract
Peripheral inflammation could constitute a risk factor for AD. This review summarizes the research related to peripheral inflammation that appears to have a relationship with Alzheimer's disease. We find there are significant associations between AD and peripheral infection induced by various pathogens, including herpes simplex virus type 1, cytomegalovirus, Epstein-Barr virus, human immunodeficiency virus, severe acute respiratory syndrome coronavirus 2, Porphyromonas gingivalis, Helicobacter pylori, and Toxoplasma gondii. Chronic inflammatory diseases are also reported to contribute to the pathophysiology of AD. The mechanisms by which peripheral inflammation affects the pathophysiology of AD are complex. Pathogen-derived neurotoxic molecule composition, disrupted BBB, and dysfunctional neurogenesis may all play a role in peripheral inflammation, promoting the development of AD. Anti-pathogenic medications and anti-inflammatory treatments are reported to decrease the risk of AD. Studies that could improve understanding the associations between AD and peripheral inflammation are needed. If our assumption is correct, early intervention against inflammation may be a potential method of preventing and treating AD.
Collapse
Affiliation(s)
- Ziyuan Li
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| | - Yafu Yin
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| |
Collapse
|
13
|
Ayieng'a EO, Afify EA, Abuiessa SA, Elblehi SS, El-Gowilly SM, El-Mas MM. Morphine aggravates inflammatory, behavioral, and hippocampal structural deficits in septic rats. Sci Rep 2023; 13:21460. [PMID: 38052832 PMCID: PMC10697987 DOI: 10.1038/s41598-023-46427-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: 07/20/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023] Open
Abstract
Although pain and sepsis are comorbidities of intensive care units, reported data on whether pain control by opioid analgesics could alter inflammatory and end-organ damage caused by sepsis remain inconclusive. Here, we tested the hypothesis that morphine, the gold standard narcotic analgesic, modifies behavioral and hippocampal structural defects induced by sepsis in male rats. Sepsis was induced with cecal ligation and puncture (CLP) and behavioral studies were undertaken 24 h later in septic and/or morphine-treated animals. The induction of sepsis or exposure to morphine (7 mg/kg) elicited similar: (i) falls in systolic blood pressure, (ii) alterations in spatial memory and learning tested by the Morris water maze, and (iii) depression of exploratory behavior measured by the new object recognition test. These hemodynamic and cognitive defects were significantly exaggerated in septic rats treated with morphine compared with individual interventions. Similar patterns of amplified inflammatory (IL-1β) and histopathological signs of hippocampal damage were noted in morphine-treated septic rats. Additionally, the presence of intact opioid receptors is mandatory for the induction of behavioral and hemodynamic effects of morphine because no such effects were observed when the receptors were blocked by naloxone. That said, our findings suggest that morphine provokes sepsis manifestations of inflammation and interrelated hemodynamic, behavioral, and hippocampal deficits.
Collapse
Affiliation(s)
- Evans O Ayieng'a
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, 1-El-Khartoum Square-Azarita, Alexandria, 21521, Egypt
| | - Elham A Afify
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, 1-El-Khartoum Square-Azarita, Alexandria, 21521, Egypt.
| | - Salwa A Abuiessa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, 1-El-Khartoum Square-Azarita, Alexandria, 21521, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Sahar M El-Gowilly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, 1-El-Khartoum Square-Azarita, Alexandria, 21521, Egypt
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, 1-El-Khartoum Square-Azarita, Alexandria, 21521, Egypt
- Department of Pharmacology and Toxicology, College of Medicine, Kuwait University, Kuwait, Kuwait
| |
Collapse
|
14
|
Hong Y, Chen P, Gao J, Lin Y, Chen L, Shang X. Sepsis-associated encephalopathy: From pathophysiology to clinical management. Int Immunopharmacol 2023; 124:110800. [PMID: 37619410 DOI: 10.1016/j.intimp.2023.110800] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/20/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023]
Abstract
Sepsis-associated encephalopathy, which presents as delirium and coma, is a significant complication of sepsis characterized by acute brain dysfunction. The presence of inflammatory pathological changes in the brain of sepsis patients and animal models has been recognized since the 1920 s, initially attributed to the entry of microbial toxins into the brain. In the early 2000 s, attention shifted towards the impact of oxidative stress, the cholinergic system, and cytokines on brain function following sepsis onset. More recently, sepsis-associated encephalopathy has been defined as a diffuse brain dysfunction not directly caused by pathogenic infection of the brain. Currently, there is no evidence-based standard for diagnosing sepsis-associated encephalopathy, and clinical management is primarily focused on symptomatic and supportive measures. This review aims to explore the pathophysiology of sepsis-associated encephalopathy and establish the connection between pathophysiological mechanisms and clinical characteristics. We hope that this work will spark the interest of researchers from various fields and contribute to the advancement of sepsis-associated encephalopathy research.
Collapse
Affiliation(s)
- Yixiao Hong
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Peiling Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Jingqi Gao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Yingying Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Linfang Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Xiuling Shang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China.
| |
Collapse
|
15
|
Dumbuya JS, Chen X, Du J, Li S, Liang L, Xie H, Zeng Q. Hydrogen-rich saline regulates NLRP3 inflammasome activation in sepsis-associated encephalopathy rat model. Int Immunopharmacol 2023; 123:110758. [PMID: 37556997 DOI: 10.1016/j.intimp.2023.110758] [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: 06/10/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is characterised by long-term cognitive impairment and psychiatric illness in sepsis survivors, associated with increased morbidity and mortality. There is a lack of effective therapeutics for SAE. Molecular hydrogen (H2) plays multiple roles in septic diseases by regulating neuroinflammation, reducing oxidative stress parameters, regulating signalling pathways, improving mitochondrial dysfunction, and regulating astrocyte and microglia activation. Here we report the protective effect of hydrogen-rich saline in the juvenile SAE rat model and its possible underlying mechanisms. Rats were injected intraperitoneally with lipopolysaccharide at a dose of 5 mg/kg to induce sepsis; Hydrogen-rich saline (HRS) was administered 1 h after LPS induction at a dose of 5 ml/kg and nigericin at 1 mg/kg 1 h before LPS injection. H&E staining for neuronal damage, TUNEL assay for detection of apoptotic cells, immunofluorescence, ELISA protocol for inflammatory cytokines and 8-OHdG determination and western blot analysis to determine the effect of HRS in LPS-induced septic rats. Rats treated with HRS showed decreased TNF-α and IL-1β expression levels. HRS treatment enhanced the activities of antioxidant enzymes (SOD, CAT and GPX) and decreased MDA and MPO activities. The number of MMP-9 and NLRP3 positive immunoreactivity cells decreased in the HRS-treated group. Subsequently, GFAP, IBA-1 and CD86 immunoreactivity were reduced, and CD206 increased after HRS treatment. 8-OHdG expression was decreased in the HRS-treated rats. Western blot analysis showed decreased NLRP3, ASC, caspase-1, MMP-2/9, TLR4 and Bax protein levels after HRS treatment, while Bcl-2 expression increased after HRS treatment. These data demonstrated that HRS attenuated neuroinflammation, NLRP3 inflammasome activation, neuronal injury, and mitochondrial damage via NLRP3/Caspase-1/TLR4 signalling in the juvenile rat model, making it a potential therapeutic agent in the treatment of paediatric SAE.
Collapse
Affiliation(s)
- John Sieh Dumbuya
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Xinxin Chen
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Jiang Du
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Siqi Li
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Lili Liang
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China
| | - Hairui Xie
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China.
| | - Qiyi Zeng
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282 PR China.
| |
Collapse
|
16
|
Liu Y, Yang H, Luo N, Fu Y, Qiu F, Pan Z, Li X, Jian W, Yang X, Xue Q, Luo Y, Yu B, Liu Z. An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway. J Transl Med 2023; 21:486. [PMID: 37475042 PMCID: PMC10360347 DOI: 10.1186/s12967-023-04345-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is characterized by diffuse brain dysfunction, long-term cognitive impairment, and increased morbidity and mortality. The current treatment for SAE is mainly symptomatic; the lack of specific treatment options and a poor understanding of the underlying mechanism of disease are responsible for poor patient outcomes. Fgr is a member of the Src family of tyrosine kinases and is involved in the innate immune response, hematologic cancer, diet-induced obesity, and hemorrhage-induced thalamic pain. This study investigated the protection provided by an Fgr kinase inhibitor in SAE and the underlying mechanism(s) of action. METHODS A cecal ligation and puncture (CLP)-induced mouse sepsis model was established. Mice were treated with or without an Fgr inhibitor and a PGC-1α inhibitor/activator. An open field test, a novel object recognition test, and an elevated plus maze were used to assess neurobehavioral changes in the mice. Western blotting and immunofluorescence were used to measure protein expression, and mRNA levels were measured using quantitative PCR (qPCR). An enzyme-linked immunosorbent assay was performed to quantify inflammatory cytokines. Mitochondrial membrane potential and morphology were measured by JC-1, electron microscopy, and the MitoTracker Deep Red probe. Oxidative stress and mitochondrial dysfunction were analyzed. In addition, the regulatory effect of Fgr on sirtuin 1 (SIRT1) was assessed. RESULTS CLP-induced sepsis increased the expression of Fgr in the hippocampal neurons. Pharmacological inhibition of Fgr attenuated CLP-induced neuroinflammation, the survival rate, cognitive and emotional dysfunction, oxidative stress, and mitochondrial dysfunction. Moreover, Fgr interacted with SIRT1 and reduced its activity and expression. In addition, activation of SIRT1/PGC-1α promoted the protective effects of the Fgr inhibitor on CLP-induced brain dysfunction, while inactivation of SIRT1/PGC-1α counteracted the benefits of the Fgr inhibitor. CONCLUSIONS To our knowledge, this is the first report of Fgr kinase inhibition markedly ameliorating SAE through activation of the SIRT1/PGC-1α pathway, and this may be a promising therapeutic target for SAE.
Collapse
Affiliation(s)
- Yuqiang Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Han Yang
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Nanbo Luo
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yifei Fu
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Fang Qiu
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Zhenglong Pan
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiongjuan Li
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wenling Jian
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xinping Yang
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qingsheng Xue
- Department of Anesthesiology, Ruijin Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Buwei Yu
- Department of Anesthesiology, Ruijin Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Zhiheng Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| |
Collapse
|
17
|
Ji MH, Gao YZ, Shi CN, Wu XM, Yang JJ. Acute and long-term cognitive impairment following sepsis: mechanism and prevention. Expert Rev Neurother 2023; 23:931-943. [PMID: 37615511 DOI: 10.1080/14737175.2023.2250917] [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: 05/28/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
INTRODUCTION Sepsis is a severe host response to infection, which induces both acute and long-term cognitive impairment. Despite its high incidence following sepsis, the underlying mechanisms remain elusive and effective treatments are not available clinically. AREA COVERED This review focuses on elucidating the pathological mechanisms underlying cognitive impairment following sepsis. Specifically, the authors discuss the role of systemic inflammation response, blood-brain barrier disruption, neuroinflammation, mitochondrial dysfunction, neuronal dysfunction, and Aβ accumulation and tau phosphorylation in cognitive impairment after sepsis. Additionally, they review current strategies to ameliorate cognitive impairment. EXPERT OPINION Potential interventions to reduce cognitive impairment after sepsis include earlier diagnosis and effective infection control, hemodynamic homeostasis, and adequate brain perfusion. Furthermore, interventions to reduce inflammatory response, reactive oxygen species, blood-brain barrier disruption, mitochondrial dysfunction, neuronal injury or death could be beneficial. Implementing strategies to minimize delirium, sleep disturbance, stress factors, and immobility are also recommended. Furthermore, avoiding neurotoxins and implementing early rehabilitation may also be important for preventing cognitive impairment after sepsis.
Collapse
Affiliation(s)
- Mu-Huo Ji
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Zhu Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cui-Na Shi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Miao Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Dumbuya JS, Li S, Liang L, Zeng Q. Paediatric sepsis-associated encephalopathy (SAE): a comprehensive review. Mol Med 2023; 29:27. [PMID: 36823611 PMCID: PMC9951490 DOI: 10.1186/s10020-023-00621-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) is one of the most common types of organ dysfunction without overt central nervous system (CNS) infection. It is associated with higher mortality, low quality of life, and long-term neurological sequelae, its mortality in patients diagnosed with sepsis, progressing to SAE, is 9% to 76%. The pathophysiology of SAE is still unknown, but its mechanisms are well elaborated, including oxidative stress, increased cytokines and proinflammatory factors levels, disturbances in the cerebral circulation, changes in blood-brain barrier permeability, injury to the brain's vascular endothelium, altered levels of neurotransmitters, changes in amino acid levels, dysfunction of cerebral microvascular cells, mitochondria dysfunction, activation of microglia and astrocytes, and neuronal death. The diagnosis of SAE involves excluding direct CNS infection or other types of encephalopathies, which might hinder its early detection and appropriate implementation of management protocols, especially in paediatric patients where only a few cases have been reported in the literature. The most commonly applied diagnostic tools include electroencephalography, neurological imaging, and biomarker detection. SAE treatment mainly focuses on managing underlying conditions and using antibiotics and supportive therapy. In contrast, sedative medication is used judiciously to treat those showing features such as agitation. The most widely used medication is dexmedetomidine which is neuroprotective by inhibiting neuronal apoptosis and reducing a sepsis-associated inflammatory response, resulting in improved short-term mortality and shorter time on a ventilator. Other agents, such as dexamethasone, melatonin, and magnesium, are also being explored in vivo and ex vivo with encouraging results. Managing modifiable factors associated with SAE is crucial in improving generalised neurological outcomes. From those mentioned above, there are still only a few experimentation models of paediatric SAE and its treatment strategies. Extrapolation of adult SAE models is challenging because of the evolving brain and technical complexity of the model being investigated. Here, we reviewed the current understanding of paediatric SAE, its pathophysiological mechanisms, diagnostic methods, therapeutic interventions, and potential emerging neuroprotective agents.
Collapse
Affiliation(s)
- John Sieh Dumbuya
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Siqi Li
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Lili Liang
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Qiyi Zeng
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China.
| |
Collapse
|
19
|
Baby S, Reljic T, Villalba N, Kumar A, Yuan SY. Endothelial glycocalyx-associated molecules as potential serological markers for sepsis-associated encephalopathy: A systematic review and meta-analysis. PLoS One 2023; 18:e0281941. [PMID: 36802387 PMCID: PMC9942976 DOI: 10.1371/journal.pone.0281941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/03/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is characterized by a diffuse cerebral dysfunction that accompanies sepsis in the absence of direct central nervous system infection. The endothelial glycocalyx is a dynamic mesh containing heparan sulfate linked to proteoglycans and glycoproteins, including selectins and vascular/intercellular adhesion molecules (V/I-CAMs), which protects the endothelium while mediating mechano-signal transduction between the blood and vascular wall. During severe inflammatory states, components of the glycocalyx are shed into the circulation and can be detected in soluble forms. Currently, SAE remains a diagnosis of exclusion and limited information is available on the utility of glycocalyx-associated molecules as biomarkers for SAE. We set out to synthesize all available evidence on the association between circulating molecules released from the endothelial glycocalyx surface during sepsis and sepsis-associated encephalopathy. METHODS MEDLINE (PubMed) and EMBASE were searched since inception until May 2, 2022 to identify eligible studies. Any comparative observational study: i) evaluating the association between sepsis and cognitive decline and ii) providing information on level of circulating glycocalyx-associated molecules was eligible for inclusion. RESULTS Four case-control studies with 160 patients met the inclusion criteria. Meta-analysis of biomarkers ICAM-1 (SMD 0.41; 95% CI 0.05-0.76; p = 0.03; I2 = 50%) and VCAM-1 (SMD 0.55; 95% CI 0.12-0.98; p = 0.01; I2 = 82%) revealed higher pooled mean concentration in patients with SAE compared to the patients with sepsis alone. Single studies reported elevated levels of P-selectin (MD 0.80; 95% CI -17.77-19.37), E-selectin (MD 96.40; 95% Cl 37.90-154.90), heparan sulfate NS2S (MD 19.41; 95% CI 13.37-25.46), and heparan sulfate NS+NS2S+NS6S (MD 67.00; 95% CI 31.00-103.00) in patients with SAE compared to the patients with sepsis alone. CONCLUSION Plasma glycocalyx-associated molecules are elevated in SAE and may be useful for early identification of cognitive decline in sepsis patients.
Collapse
Affiliation(s)
- Sheon Baby
- Morsani College of Medicine, University of South Florida, Tampa, FL, United States of America
| | - Tea Reljic
- Department of Evidence Based Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Nuria Villalba
- Department of Molecular Pharmacology & Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Ambuj Kumar
- Department of Evidence Based Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Sarah Y. Yuan
- Department of Molecular Pharmacology & Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| |
Collapse
|
20
|
Li Y, Ma J, Diao J, Chen W, Wang Z. Esmolol inhibits cognitive impairment and neuronal inflammation in mice with sepsis-induced brain injury. Transl Neurosci 2023; 14:20220297. [PMID: 37529169 PMCID: PMC10388135 DOI: 10.1515/tnsci-2022-0297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Sepsis is a potentially fatal organ failure resulting from a dysregulated host response to infection. It can be a substantial financial burden on families and society due to the high cost of medical care. The study aims to investigate the protective roles of Esmolol in mice with sepsis-induced brain injuries against cognitive dysfunction and neuronal inflammation. Male C57BL/6J mice were intraperitoneally injected with LPS (10 mg/kg, L2630, Sigma) to establish a septic encephalopathy model. Esmolol (15 mg/kg/h, HY-B1392, MedChemExpress) was subcutaneously infused using osmotic mini-pumps for 6 h before LPS injection. Morris water maze and novel object recognition tests evaluated LPS-induced cognitive impairment and behavioral phenotypes. Cytokines and protein expression were assessed using ELISA assay and RT-qPCR. Esmolol treatment potentially improved cognitive impairment in septic mice. Esmolol administration markedly diminished the abnormal hippocampal neuronal structure, and the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α was significantly downregulated in the hippocampal tissue. Esmolol treatment significantly reduced apoptotic TUNEL-positive cells and reversed the related gene expression (BAX and BCL-2). The effects of esmolol on the reactive oxidative species and oxidative stress markedly reduce malondialdehyde MDA content and increase superoxide dismutase and catalase in hippocampal tissues. In addition, esmolol significantly reduced the percentage and density of Iba-1 + microglia in septic mice. Our results demonstrated that esmolol potentially improved cognitive impairment and neuronal inflammation in mice with sepsis-induced brain injury.
Collapse
Affiliation(s)
- Yanpeng Li
- Department of Emergency, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai201399, China
| | - Junli Ma
- Department of Cardiovascular Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai201399, China
| | - Jianjun Diao
- Department of Emergency, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai201399, China
| | - Wei Chen
- Department of Emergency, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai201399, China
| | - Zhihua Wang
- Department of Emergency, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai201399, China
| |
Collapse
|
21
|
Yue J, Tan Y, Huan R, Guo J, Yang S, Deng M, Xiong Y, Han G, Liu L, Liu J, Cheng Y, Zha Y, Zhang J. Mast cell activation mediates blood-brain barrier impairment and cognitive dysfunction in septic mice in a histamine-dependent pathway. Front Immunol 2023; 14:1090288. [PMID: 36817492 PMCID: PMC9929573 DOI: 10.3389/fimmu.2023.1090288] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Introduction Sepsis-associated encephalopathy (SAE) is a diffuse cerebral dysfunction resulting from a systemic inflammatory response to infection; however, its pathophysiology remains unclear. Sepsis-induced neuroinflammation and blood-brain barrier (BBB) disruption are crucial factors in brain function disturbance in SAE. Mast cells (MCs) activation plays an important role in several neuroinflammation models; however, its role in SAE has not been comprehensively investigated. Methods We first established a SAE model by cecal ligation puncture (CLP) surgery and checked the activation of MCs. MCs activation was checked using immumohistochemical staining and Toluidine Blue staining. We administrated cromolyn (10mg/ml), a MC stabilizer, to rescue the septic mice. Brain cytokines levels were measured using biochemical assays. BBB disruption was assessed by measuring levels of key tight-junction (TJ) proteins. Cognitive function of mice was analyzed by Y maze and open field test. Transwell cultures of brain microvascular endothelial cells (BMVECs) co-cultured with MCs were used to assess the interaction of BMVECs and MCs. Results Results showed that MCs were overactivated in the hippocampus of CLP-induced SAE mice. Cromolyn intracerebroventricular (i.c.v) injection substantially inhibited the MCs activation and neuroinflammation responses, ameliorated BBB impairment, improved the survival rate and alleviated cognitive dysfunction in septic mice. In vitro experiments, we revealed that MCs activation increased the sensitivity of BMVECs against to lipopolysaccharide (LPS) challenge. Furthermore, we found that the histamine/histamine 1 receptor (H1R) mediated the interaction between MCs and BMVECs, and amplifies the LPS-induced inflammatory responses in BMVECs by modulating the TLR2/4-MAPK signaling pathway. Conclusions MCs activation could mediate BBB impairment and cognitive dysfunction in septic mice in a histamine-dependent pathway.
Collapse
Affiliation(s)
- Jianhe Yue
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Renzheng Huan
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Guo
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Sha Yang
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Mei Deng
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yunbiao Xiong
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guoqiang Han
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Lin Liu
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jian Liu
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Zha
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jiqin Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
22
|
Tang C, Jin Y, Wang H. The biological alterations of synapse/synapse formation in sepsis-associated encephalopathy. Front Synaptic Neurosci 2022; 14:1054605. [PMID: 36530954 PMCID: PMC9755596 DOI: 10.3389/fnsyn.2022.1054605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/18/2022] [Indexed: 06/12/2024] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a common complication caused by sepsis, and is responsible for increased mortality and poor outcomes in septic patients. Neurological dysfunction is one of the main manifestations of SAE patients. Patients may still have long-term cognitive impairment after hospital discharge, and the underlying mechanism is still unclear. Here, we first outline the pathophysiological changes of SAE, including neuroinflammation, glial activation, and blood-brain barrier (BBB) breakdown. Synapse dysfunction is one of the main contributors leading to neurological impairment. Therefore, we summarized SAE-induced synaptic dysfunction, such as synaptic plasticity inhibition, neurotransmitter imbalance, and synapses loss. Finally, we discuss the alterations in the synapse, synapse formation, and mediators associated with synapse formation during SAE. In this review, we focus on the changes in synapse/synapse formation caused by SAE, which can further understand the synaptic dysfunction associated with neurological impairment in SAE and provide important insights for exploring appropriate therapeutic targets of SAE.
Collapse
Affiliation(s)
| | | | - Huan Wang
- College of Life and Health, Dalian University, Dalian, China
| |
Collapse
|
23
|
Dumbuya JS, Li S, Liang L, Chen Y, Du J, Zeng Q. Effects of hydrogen-rich saline in neuroinflammation and mitochondrial dysfunction in rat model of sepsis-associated encephalopathy. J Transl Med 2022; 20:546. [DOI: 10.1186/s12967-022-03746-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/31/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract
Background
Sepsis-associated encephalopathy (SAE) is one of the most common types of sepsis-related organ dysfunction without overt central nervous system (CNS) infection. It is associated with higher mortality, low quality of life, and long-term neurological sequelae in suspected patients. At present there is no specific treatment for SAE rather than supportive therapy and judicious use of antibiotics, which are sometimes associated with adverse effects. Molecular hydrogen (H2) has been reported to play crucial role in regulating inflammatory responses, neuronal injury, apoptosis and mitochondrial dysfunction in adult models of SAE. Here we report the protective effect of hydrogen-rich saline in juvenile SAE rat model and its possible underling mechanism(s).
Materials and methods
Rats were challenged with lipopolysaccharide (LPS) at a dose of 8 mg/kg injected intraperitoneally to induce sepsis and hydrogen-rich saline (HRS) administered 1 h following LPS induction at a dose of 5 ml/kg. Rats were divided into: sham, sham + HRS, LPS and LPS + HRS. At 48 h, rats were sacrificed and Nissl staining for neuronal injury, TUNEL assay for apoptotic cells detection, immunohistochemistry, and ELISA protocol for inflammatory cytokines determination, mitochondrial dysfunction parameters, electron microscopy and western blot analysis were studied to examine the effect of HRS in LPS-induced septic rats.
Results
Rats treated with HRS improved neuronal injury, improvement in rats’ survival rate. ELISA analysis showed decreased TNF-α and IL-1β and increased IL-10 expression levels in the HRS-treated group. Apoptotic cells were decreased after HRS administration in septic rats. The numbers of GFAP and IBA-1positive cells were attenuated in the HRS-treated group when compared to the LPS group. Subsequently, GFAP and IBA-1 immunoreactivity were decreased after HRS treatment. Mitochondrial membrane potential detected by JC-1 dye and ATP content were decreased in septic rats, which were improved after HRS treatment, while release of ROS was increased in the LPS group reverted by HRS treatment, ameliorating mitochondrial dysfunction. Further analysis by transmission electron microscopy showed decreased number of mitochondria and synapses, and disrupted mitochondrial membrane ultrastructure in the LPS group, while HRS administration increased mitochondria and synapses number.
Conclusion
These data demonstrated that HRS can improve survival rate, attenuate neuroinflammation, astrocyte and microglial activation, neuronal injury and mitochondrial dysfunction in juvenile SAE rat model, making it a potential therapeutic candidate in treating paediatric SAE.
Collapse
|
24
|
Bai Y, Li L, Dong B, Ma W, Chen H, Yu Y. Phosphorylation‐mediated PI3K‐Art
signalling pathway as a therapeutic mechanism in the
hydrogen‐induced
alleviation of brain injury in septic mice. J Cell Mol Med 2022; 26:5713-5727. [DOI: 10.1111/jcmm.17568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/13/2022] [Accepted: 09/17/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Yuanyuan Bai
- Department of Anesthesiology Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University Tianjin China
- Tianjin Research Institute of Anesthesiology Tianjin China
| | - Li Li
- Department of Anesthesiology, Huashan Hospital Fudan University Shanghai China
| | - Beibei Dong
- Department of Anesthesiology Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University Tianjin China
- Tianjin Research Institute of Anesthesiology Tianjin China
| | - Wanjie Ma
- Department of Anesthesiology Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University Tianjin China
- Tianjin Research Institute of Anesthesiology Tianjin China
| | - Hongguang Chen
- Department of Anesthesiology Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University Tianjin China
- Tianjin Research Institute of Anesthesiology Tianjin China
| | - Yonghao Yu
- Department of Anesthesiology Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University Tianjin China
- Tianjin Research Institute of Anesthesiology Tianjin China
| |
Collapse
|
25
|
Lei W, Ren Z, Su J, Zheng X, Gao L, Xu Y, Deng J, Xiao C, Sheng S, Cheng Y, Ma T, Liu Y, Wang P, Luo OJ, Chen G, Wang Z. Immunological risk factors for sepsis-associated delirium and mortality in ICU patients. Front Immunol 2022; 13:940779. [PMID: 36203605 PMCID: PMC9531264 DOI: 10.3389/fimmu.2022.940779] [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: 05/10/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
Background A major challenge in intervention of critical patients, especially sepsis-associated delirium (SAD) intervention, is the lack of predictive risk factors. As sepsis and SAD are heavily entangled with inflammatory and immunological processes, to identify the risk factors of SAD and mortality in the intensive care unit (ICU) and determine the underlying molecular mechanisms, the peripheral immune profiles of patients in the ICU were characterized. Methods This study contains a cohort of 52 critical patients who were admitted to the ICU of the First Affiliated Hospital of Jinan University. Comorbidity, including sepsis and SAD, of this cohort was diagnosed and recorded. Furthermore, peripheral blood samples were collected on days 1, 3, and 5 of admission for peripheral immune profiling with blood routine examination, flow cytometry, ELISA, RNA-seq, and qPCR. Results The patients with SAD had higher mortality during ICU admission and within 28 days of discharge. Compared with survivors, nonsurvivors had higher neutrophilic granulocyte percentage, higher CRP concentration, lower monocyte count, lower monocyte percentage, lower C3 complement level, higher CD14loCD16+ monocytes percentage, and higher levels of IL-6 and TNFα. The CD14hiCD16- monocyte percentage manifested favorable prediction values for the occurrence of SAD. Differentially expressed genes between the nonsurvival and survival groups were mainly associated with immune response and metabolism process. The longitudinal expression pattern of SLC2A1 and STIMATE were different between nonsurvivors and survivors, which were validated by qPCR. Conclusions Nonsurvival critical patients have a distinct immune profile when compared with survival patients. CD14hiCD16- monocyte prevalence and expression levels of SLC2A1 and STIMATE may be predictors of SAD and 28-day mortality in ICU patients.
Collapse
Affiliation(s)
- Wen Lei
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Zhiyao Ren
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- National Health Commission (NHC) Key Laboratory of Male Reproduction and Genetics, Guangzhou, China
- Department of Central Laboratory, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
| | - Jun Su
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Sonograph, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xinglong Zheng
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Critical Care Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Yudai Xu
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Jieping Deng
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Chanchan Xiao
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Shuai Sheng
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Yu Cheng
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Tianshun Ma
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Yu Liu
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Pengcheng Wang
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Oscar Junhong Luo
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Zhigang Wang, ; Guobing Chen, ; Oscar Junhong Luo,
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Sonograph, The First Affiliated Hospital, Jinan University, Guangzhou, China
- *Correspondence: Zhigang Wang, ; Guobing Chen, ; Oscar Junhong Luo,
| | - Zhigang Wang
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Critical Care Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China
- *Correspondence: Zhigang Wang, ; Guobing Chen, ; Oscar Junhong Luo,
| |
Collapse
|
26
|
Sepsis-Induced Brain Dysfunction: Pathogenesis, Diagnosis, and Treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1328729. [PMID: 36062193 PMCID: PMC9433216 DOI: 10.1155/2022/1328729] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/30/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022]
Abstract
Dysregulated host response to infection, which cause life-threatening organ dysfunction, was defined as sepsis. Sepsis can cause acute and long-term brain dysfunction, namely, sepsis-associated encephalopathy (SAE) and cognitive impairment. SAE refers to changes in consciousness without direct evidence of central nervous system infection. It is highly prevalent and may cause poor outcomes in sepsis patients. Cognitive impairment seriously affects the life quality of sepsis patients and increases the medical burden. The pathogenesis of sepsis-induced brain dysfunction is mainly characterized by the interaction of systemic inflammation, blood-brain barrier (BBB) dysfunction, neuroinflammation, microcirculation dysfunction, and brain dysfunction. Currently, the diagnosis of sepsis-induced brain dysfunction is based on clinical manifestation of altered consciousness along with neuropathological examination, and the treatment is mainly involves controlling sepsis. Although treatments for sepsis-induced brain dysfunction have been tested in animals, clinical treat sepsis-induced brain dysfunction is still difficult. Therefore, we review the underlying mechanisms of sepsis-induced brain injury, which mainly focus on the influence of systemic inflammation on BBB, neuroinflammation, brain microcirculation, and the brain function, which want to bring new mechanism-based directions for future basic and clinical research aimed at preventing or ameliorating brain dysfunction.
Collapse
|
27
|
Hobin K, Costas-Rodríguez M, Van Wonterghem E, Vandenbroucke RE, Vanhaecke F. High-Precision Isotopic Analysis of Cu and Fe via Multi-Collector Inductively Coupled Plasma-Mass Spectrometry Reveals Lipopolysaccharide-Induced Inflammatory Effects in Blood Plasma and Brain Tissues. Front Chem 2022; 10:896279. [PMID: 35783204 PMCID: PMC9241339 DOI: 10.3389/fchem.2022.896279] [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: 03/14/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
The concentration and the isotopic composition of the redox-active essential elements Cu and Fe were investigated in blood plasma and specific brain regions (hippocampus, cortex, brain stem and cerebellum) of mice to assess potential alterations associated with sepsis-associated encephalopathy induced by lipopolysaccharide (LPS) administration. Samples were collected from young (16-22 weeks) and aged (44-65 weeks) mice after intraperitoneal injection of the LPS, an endotoxin inducing neuroinflammation, and from age- and sex-matched controls, injected with phosphate-buffered saline solution. Sector-field single-collector inductively coupled plasma-mass spectrometry was relied upon for elemental analysis and multi-collector inductively coupled plasma-mass spectrometry for isotopic analysis. Significant variations were observed for the Cu concentration and for the Cu and Fe isotope ratios in the blood plasma. Concentrations and isotope ratios of Cu and Fe also varied across the brain tissues. An age- and an inflammatory-related effect was found affecting the isotopic compositions of blood plasma Cu and cerebellum Fe, whereas a regional Cu isotopic redistribution was found within the brain tissues. These findings demonstrate that isotopic analysis of essential mineral elements picks up metabolic changes not revealed by element quantification, making the two approaches complementary.
Collapse
Affiliation(s)
- Kasper Hobin
- Atomic and Mass Spectrometry - A&MS Research Unit, Department of Chemistry, Ghent University, Ghent, Belgium
| | - Marta Costas-Rodríguez
- Atomic and Mass Spectrometry - A&MS Research Unit, Department of Chemistry, Ghent University, Ghent, Belgium
| | - Elien Van Wonterghem
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Frank Vanhaecke
- Atomic and Mass Spectrometry - A&MS Research Unit, Department of Chemistry, Ghent University, Ghent, Belgium
| |
Collapse
|
28
|
de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol Rev 2022; 74:712-768. [PMID: 35738680 DOI: 10.1124/pharmrev.121.000349] [Citation(s) in RCA: 182] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The first matrix metalloproteinase (MMP) was discovered in 1962 from the tail of a tadpole by its ability to degrade collagen. As their name suggests, matrix metalloproteinases are proteases capable of remodeling the extracellular matrix. More recently, MMPs have been demonstrated to play numerous additional biologic roles in cell signaling, immune regulation, and transcriptional control, all of which are unrelated to the degradation of the extracellular matrix. In this review, we will present milestones and major discoveries of MMP research, including various clinical trials for the use of MMP inhibitors. We will discuss the reasons behind the failures of most MMP inhibitors for the treatment of cancer and inflammatory diseases. There are still misconceptions about the pathophysiological roles of MMPs and the best strategies to inhibit their detrimental functions. This review aims to discuss MMPs in preclinical models and human pathologies. We will discuss new biochemical tools to track their proteolytic activity in vivo and ex vivo, in addition to future pharmacological alternatives to inhibit their detrimental functions in diseases. SIGNIFICANCE STATEMENT: Matrix metalloproteinases (MMPs) have been implicated in most inflammatory, autoimmune, cancers, and pathogen-mediated diseases. Initially overlooked, MMP contributions can be both beneficial and detrimental in disease progression and resolution. Thousands of MMP substrates have been suggested, and a few hundred have been validated. After more than 60 years of MMP research, there remain intriguing enigmas to solve regarding their biological functions in diseases.
Collapse
Affiliation(s)
- Luiz G N de Almeida
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Hayley Thode
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Yekta Eslambolchi
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sameeksha Chopra
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Daniel Young
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sean Gill
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Laurent Devel
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Antoine Dufour
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| |
Collapse
|
29
|
Liu YX, Yu Y, Liu JP, Liu WJ, Cao Y, Yan RM, Yao YM. Neuroimmune Regulation in Sepsis-Associated Encephalopathy: The Interaction Between the Brain and Peripheral Immunity. Front Neurol 2022; 13:892480. [PMID: 35832175 PMCID: PMC9271799 DOI: 10.3389/fneur.2022.892480] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/27/2022] [Indexed: 11/15/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE), the most popular cause of coma in the intensive care unit (ICU), is the diffuse cerebral damage caused by the septic challenge. SAE is closely related to high mortality and extended cognitive impairment in patients in septic shock. At present, many studies have demonstrated that SAE might be mainly associated with blood–brain barrier damage, abnormal neurotransmitter secretion, oxidative stress, and neuroimmune dysfunction. Nevertheless, the precise mechanism which initiates SAE and contributes to the long-term cognitive impairment remains largely unknown. Recently, a growing body of evidence has indicated that there is close crosstalk between SAE and peripheral immunity. The excessive migration of peripheral immune cells to the brain, the activation of glia, and resulting dysfunction of the central immune system are the main causes of septic nerve damage. This study reviews the update on the pathogenesis of septic encephalopathy, focusing on the over-activation of immune cells in the central nervous system (CNS) and the “neurocentral–endocrine–immune” networks in the development of SAE, aiming to further understand the potential mechanism of SAE and provide new targets for diagnosis and management of septic complications.
Collapse
Affiliation(s)
- Yu-xiao Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
| | - Yang Yu
- Department of Traditional Chinese Medical Science, Sixth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Jing-peng Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Department of Traditional Chinese Medical Science, Sixth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Wen-jia Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Yang Cao
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
| | - Run-min Yan
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
- *Correspondence: Yong-ming Yao
| | - Yong-ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Run-min Yan
| |
Collapse
|
30
|
Ponce-Regalado MD, Salazar-Juárez A, Rojas-Espinosa O, Contis-Montes de Oca A, Hurtado-Alvarado G, Arce-Paredes P, Pérez-Sánchez G, Pavón L, Girón-Pérez MI, Hernández-Pando R, Alvarez-Sánchez ME, Becerril-Villanueva E. Development of Anxiolytic and Depression-like Behavior in Mice Infected with Mycobacterium lepraemurium. Neuroscience 2022; 493:15-30. [PMID: 35447197 DOI: 10.1016/j.neuroscience.2022.03.021] [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: 05/06/2021] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022]
Abstract
Murine leprosy is a systemic infectious disease of mice caused by Mycobacterium lepraemurium (MLM) in which the central nervous system (CNS) is not infected; nevertheless, diseased animals show measurable cognitive alterations. For this reason, in this study, we explored the neurobehavioral changes in mice chronically infected with MLM. BALB/c mice were infected with MLM, and 120 days later, the alterations in mice were evaluated based on immunologic, histologic, endocrine, neurochemical, and behavioral traits. We found increases in the levels of IL-4 and IL-10 associated with high bacillary loads. We also found increase in the serum levels of corticosterone, epinephrine, and norepinephrine in the adrenal gland, suggesting neuroendocrine deregulation. Mice exhibited depression-like behavior in the tail suspension and forced swimming tests and anxiolytic behavior in the open field and elevated plus maze tests. The neurobehavioral alterations of mice were correlated with the histologic damage in the prefrontal cortex, ventral hippocampus, and amygdala, as well as with a blood-brain barrier disruption in the hippocampus. These results reveal an interrelated response of the neuroimmune--endocrinological axis in unresolved chronic infections that result in neurocognitive deterioration.
Collapse
Affiliation(s)
- M D Ponce-Regalado
- Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara, Carretera a Yahualica, Km. 7.5 Tepatitlán de Morelos, Jalisco 47600, Mexico
| | - A Salazar-Juárez
- Branch Clinical Research. Laboratory of Molecular Neurobiology and Neurochemistry of Addiction, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - O Rojas-Espinosa
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala, Colonia Santo Tomás, 11340, Ciudad de México, Mexico.
| | - A Contis-Montes de Oca
- Sección de estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Diaz Miron y Plan de San Luis S/N, Miguel Hidalgo, 11340 Mexico City, Mexico
| | - G Hurtado-Alvarado
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - P Arce-Paredes
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
| | - G Pérez-Sánchez
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - L Pavón
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - M I Girón-Pérez
- Laboratorio de Inmunotoxicología, Secretaría de Investigación y Posgrado Universidad Autónoma de Nayarit, Boulevard Tepic-Xalisco s/n. Cd, de la Cultura Amado Nervo, C.P. 63000 Tepic, Nayarit, México
| | - R Hernández-Pando
- Experimental Pathology Section, Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Colonia Belisario Dominguez Seccion XVI, 14080, Deleg. Tlalpan, México City, Mexico
| | - M E Alvarez-Sánchez
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo # 290, Col. Del Valle, CP 03100 México City, México
| | - Enrique Becerril-Villanueva
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico.
| |
Collapse
|
31
|
Vijikumar A, Saralkar P, Saylor SD, Sullivan PG, Huber JD, Geldenhuys WJ. Novel mitoNEET ligand NL-1 improves therapeutic outcomes in an aged rat model of cerebral ischemia/reperfusion injury. Exp Neurol 2022; 355:114128. [PMID: 35662609 DOI: 10.1016/j.expneurol.2022.114128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 05/16/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Cerebral ischemic stroke is a leading cause of mortality and disability worldwide. Currently, there are a lack of drugs capable of reducing neuronal cell loss due to ischemia/reperfusion-injury after stroke. Previously, we identified mitoNEET, a [2Fe-2S] redox mitochondrial protein, as a putative drug target for ischemic stroke. In this study, we tested NL-1, a novel mitoNEET ligand, in a preclinical model of ischemic stroke with reperfusion using aged female rats. Using a transient middle cerebral artery occlusion (tMCAO), we induced a 2 h ischemic injury and then evaluated the effects of NL-1 treatment on ischemic/reperfusion brain injury at 24 and 72 h. Test compounds were administered at time of reperfusion via intravenous dosing. Results of the study demonstrated that NL-1 (10 mg/kg) treatment markedly improved survival and reduced infarct volume and hemispheric swelling in the brain as compared aged rats treated with vehicle or a lower dose of NL-1 (0.25 mg/kg). Interestingly, the protective effect of NL-1 was significantly improved when encapsulated in PLGA nanoparticles, where a 40-fold lesser dose (0.25 mg/kg) of NL-1 produced an equivalent effect as the 10 mg/kg dose. Evaluation of changes in blood-brain barrier permeability and lipid peroxidation corroborated the protective actions of NL-1 (10 mg/kg) or NL-1 NP treatment demonstrated a reduced accumulation of parenchymal IgG, decreased levels of 4-hydroxynonenal (4-HNE) and a decreased TUNEL positive cells in the brains of aged female rats at 72 h after tMCAO with reperfusion. Our studies indicate that targeting mitoNEET following ischemia/reperfusion-injury is a novel drug target pathway that warrants further investigation.
Collapse
Affiliation(s)
- Aruvi Vijikumar
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26505, United States of America
| | - Pushkar Saralkar
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26505, United States of America
| | - Scott D Saylor
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV 26505, United States of America
| | - Patrick G Sullivan
- Department of Neuroscience, Spinal and Brain Injury Research Center, School of Medicine, University of Kentucky, Lexington, KY 40536, United States of America
| | - Jason D Huber
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26505, United States of America; Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26505, United States of America.
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26505, United States of America; Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26505, United States of America; Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV 26505, United States of America
| |
Collapse
|
32
|
Pathogenesis of sepsis-associated encephalopathy: more than blood-brain barrier dysfunction. Mol Biol Rep 2022; 49:10091-10099. [PMID: 35639274 DOI: 10.1007/s11033-022-07592-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 05/11/2022] [Indexed: 12/19/2022]
Abstract
Sepsis-associated encephalopathy is a common neurological complication of sepsis and is responsible for higher mortality and poorer long-term outcomes in septic patients. Sepsis-associated encephalopathy symptoms can range from mild delirium to deep coma, which occurs in up to 70% of patients in intensive care units. The pathological changes in the brain associated with sepsis include cerebral ischaemia, cerebral haemorrhage, abscess and progressive multifocal necrotic leukoencephalopathy. Several mechanisms are involved in the pathogenesis of sepsis-associated encephalopathy, such as blood-brain barrier dysfunction, cerebral blood flow impairment, glial cell activation, leukocyte transmigration, and neurotransmitter disturbances. These events are interrelated and influence each other, therefore they do not act as independent factors. This review is focused on new evidence showing the pathological process of sepsis-associated encephalopathy.
Collapse
|
33
|
Nwafor DC, Brichacek AL, Foster CH, Lucke-Wold BP, Ali A, Colantonio MA, Brown CM, Qaiser R. Pediatric Traumatic Brain Injury: An Update on Preclinical Models, Clinical Biomarkers, and the Implications of Cerebrovascular Dysfunction. J Cent Nerv Syst Dis 2022; 14:11795735221098125. [PMID: 35620529 PMCID: PMC9127876 DOI: 10.1177/11795735221098125] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 04/14/2022] [Indexed: 11/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of pediatric morbidity and mortality. Recent studies suggest that children and adolescents have worse post-TBI outcomes and take longer to recover than adults. However, the pathophysiology and progression of TBI in the pediatric population are studied to a far lesser extent compared to the adult population. Common causes of TBI in children are falls, sports/recreation-related injuries, non-accidental trauma, and motor vehicle-related injuries. A fundamental understanding of TBI pathophysiology is crucial in preventing long-term brain injury sequelae. Animal models of TBI have played an essential role in addressing the knowledge gaps relating to pTBI pathophysiology. Moreover, a better understanding of clinical biomarkers is crucial to diagnose pTBI and accurately predict long-term outcomes. This review examines the current preclinical models of pTBI, the implications of pTBI on the brain’s vasculature, and clinical pTBI biomarkers. Finally, we conclude the review by speculating on the emerging role of the gut-brain axis in pTBI pathophysiology.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- West Virginia University School of Medicine, Morgantown, WV, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Allison L. Brichacek
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Chase H. Foster
- Department of Neurosurgery, George Washington University Hospital, Washington D.C., USA
| | | | - Ahsan Ali
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | | | - Candice M. Brown
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Rabia Qaiser
- Department of Neurosurgery, Baylor Scott and White, Temple, TX, USA
| |
Collapse
|
34
|
Li Y, Ji M, Yang J. Current Understanding of Long-Term Cognitive Impairment After Sepsis. Front Immunol 2022; 13:855006. [PMID: 35603184 PMCID: PMC9120941 DOI: 10.3389/fimmu.2022.855006] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is recognized as a life-threatening multi-organ dysfunction resulting from a dysregulated host response to infection. Although the incidence and mortality of sepsis decrease significantly due to timely implementation of anti-infective and support therapies, accumulating evidence suggests that a great proportion of survivors suffer from long-term cognitive impairment after hospital discharge, leading to decreased life quality and substantial caregiving burdens for family members. Several mechanisms have been proposed for long-term cognitive impairment after sepsis, which are not mutually exclusive, including blood-brain barrier disruption, neuroinflammation, neurotransmitter dysfunction, and neuronal loss. Targeting these critical processes might be effective in preventing and treating long-term cognitive impairment. However, future in-depth studies are required to facilitate preventive and/or treatment strategies for long-term cognitive impairment after sepsis.
Collapse
Affiliation(s)
- Ying Li
- Department of Anesthesiology, Jiangyin Hospital, Affiliated to Southeast University Medical School, Jiangyin, China
| | - Muhuo Ji
- Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
35
|
Sekino N, Selim M, Shehadah A. Sepsis-associated brain injury: underlying mechanisms and potential therapeutic strategies for acute and long-term cognitive impairments. J Neuroinflammation 2022; 19:101. [PMID: 35488237 PMCID: PMC9051822 DOI: 10.1186/s12974-022-02464-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/14/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis causes cerebral dysfunction in the short and long term and induces disruption of the blood–brain barrier (BBB), neuroinflammation, hypoperfusion, and accumulation of amyloid β (Aβ) and tau protein in the brain. White matter changes and brain atrophy can be detected using brain imaging, but unfortunately, there is no specific treatment that directly addresses the underlying mechanisms of cognitive impairments in sepsis. Here, we review the underlying mechanisms of sepsis-associated brain injury, with a focus on BBB dysfunction and Aβ and tau protein accumulation in the brain. We also describe the neurological manifestations and imaging findings of sepsis-associated brain injury, and finally, we propose potential therapeutic strategies for acute and long-term cognitive impairments associated with sepsis. In the acute phase of sepsis, we suggest using antibiotics (such as rifampicin), targeting proinflammatory cytokines, and preventing ischemic injuries and hypoperfusion. In the late phase of sepsis, we suggest targeting neuroinflammation, BBB dysfunction, Aβ and tau protein phosphorylation, glycogen synthase kinase-3 beta (GSK3β), and the receptor for advanced glycation end products (RAGE). These proposed strategies are meant to bring new mechanism-based directions for future basic and clinical research aimed at preventing or ameliorating acute and long-term cognitive impairments in patients with sepsis.
Collapse
Affiliation(s)
- Nobufumi Sekino
- Department of Medicine, Translational Therapeutics Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Magdy Selim
- Department of Neurology, Stroke and Cerebrovascular Diseases Division, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS-641, Boston, MA, 02215, USA
| | - Amjad Shehadah
- Department of Neurology, Stroke and Cerebrovascular Diseases Division, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS-641, Boston, MA, 02215, USA.
| |
Collapse
|
36
|
Neuropsychological Outcome of Critically Ill Patients with Severe Infection. Biomedicines 2022; 10:biomedicines10030526. [PMID: 35327328 PMCID: PMC8945835 DOI: 10.3390/biomedicines10030526] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis and septic shock represent important burdens of disease around the world. Sepsis-associated neurological consequences have a great impact on patients, both in the acute phase and in the long term. Sepsis-associated encephalopathy (SAE) is a severe brain dysfunction that may contribute to long-term cognitive impairment. Its pathophysiology recognizes the following two main mechanisms: neuroinflammation and hemodynamic impairment. Clinical manifestations include different forms of altered mental status, from agitation and restlessness to delirium and deep coma. A definite diagnosis is difficult because of the absence of specific radiological and biological criteria; clinical management is restricted to the treatment of sepsis, focusing on early detection of the infection source, maintenance of hemodynamic homeostasis, and avoidance of metabolic disturbances or neurotoxic drugs.
Collapse
|
37
|
The Improvement of Sepsis-Associated Encephalopathy by P2X7R Inhibitor through Inhibiting the Omi/HtrA2 Apoptotic Signaling Pathway. Behav Neurol 2022; 2022:3777351. [PMID: 35126784 PMCID: PMC8813303 DOI: 10.1155/2022/3777351] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/14/2021] [Accepted: 01/05/2022] [Indexed: 12/15/2022] Open
Abstract
The pathogenesis of sepsis-associated encephalopathy (SAE) involves many aspects, including intracellular peroxidative stress damage, mitochondrial dysfunction, and cell apoptosis. In this study, we mainly explored the influence of P2X7R on the cognitive function of SAE and its molecular mechanism. We established a sepsis model using lipopolysaccharide (LPS) stimulation, followed by an assessment of cognitive function using Morris water maze, and then Western Blot was used to analyze the expression of tight junction proteins ZO-1 and Occludin in the hippocampus of mice. TUNEL assay was used to analyze the apoptosis of brain cells in frozen brain slices of mice during sepsis. Human brain microvascular endothelial cells (HBMECs) were used to research the molecular mechanism of brain cell damage induced by P2X7R. The results showed that P2X7R inhibitors dramatically improved the survival rate of mice, relieved the cognitive dysfunction caused by LPS stimulation, and significantly reduced the brain cell apoptosis caused by LPS. In addition, the inhibition of P2X7R can also reduce the production and accumulation of reactive oxygen species (ROS) in HBMECs in vitro and inhibit the apoptosis signaling pathway associated with mitochondrial serine protease Omi/HtrA2 in HBMECs in vitro. These results suggest that P2X7R has strong value as a potential target for the treatment of SAE.
Collapse
|
38
|
Pu Y, Qian F, Guo J, Sha Y, Qian Y. Selegiline Protects Against Lipopolysaccharide (LPS)-Induced Impairment of the Blood-Brain Barrier Through Regulating the NF-κB/MLCK/p-MLC Signaling Pathway. Neurotox Res 2022; 40:267-275. [PMID: 34981455 DOI: 10.1007/s12640-021-00448-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 11/29/2022]
Abstract
Disruption of the blood-brain barrier (BBB) is an important hallmark of sepsis-associated encephalopathy (SAE). Selegiline, a selective and irreversible inhibitor of monoamine oxidase type B, has been applied for the treatment of nervous disorders. In this study, we aimed to investigate whether selegiline has a protective capacity in the impairment of the BBB in both in vivo and in vitro experiments. In a sepsis mouse model, administration of selegiline ameliorated lipopolysaccharide (LPS)-induced impairment of BBB integrity. Additionally, treatment with selegiline increased the expression of the tight junction protein junctional adhesion molecule A (JAM-A) against LPS. Also, we found that selegiline inhibited the production of the proinflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-1β. In an in vitro experimental model, bEnd.3 brain endothelial cells were exposed to LPS. Results indicate that stimulation with LPS significantly increased the permeability of bEnd.3 cells and reduced the expression of JAM-A, both of which were rescued by treatment with selegiline. Additionally, selegiline prevented the activation of the NF-κB/MLCK/p-MLC signaling pathway in LPS-challenged bEnd.3 cells. These results indicate that selegiline exerted a protective effect on BBB dysfunction, which might be attributed to the inhibition of the NF-κB/MLCK/p-MLC signaling pathway. These findings provide a basis for further research into the neuroprotective mechanism of selegiline.
Collapse
Affiliation(s)
- Yuehong Pu
- Department of Emergency Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Fenghua Qian
- Department of Emergency Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Jian Guo
- Department of Emergency Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Yuanyuan Sha
- Department of Emergency Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Yiming Qian
- Department of Emergency Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110 Ganhe Road, Hongkou District, Shanghai, 200437, China.
| |
Collapse
|
39
|
Xue W, Li Y, Zhang M. Pristimerin inhibits neuronal inflammation and protects cognitive function in mice with sepsis-induced brain injuries by regulating PI3K/Akt signalling. PHARMACEUTICAL BIOLOGY 2021; 59:1351-1358. [PMID: 34590530 PMCID: PMC8491740 DOI: 10.1080/13880209.2021.1981399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 09/10/2021] [Indexed: 06/05/2023]
Abstract
CONTEXT Sepsis is a systemic inflammatory disease; pristimerin exhibits strong antibacterial, anti-inflammatory and antioxidant properties. OBJECTIVES We explored whether pristimerin protected against cognitive dysfunction and neuroinflammation in C57BL/6 J mice with sepsis-induced brain injuries. MATERIALS AND METHODS Sepsis was induced by intraperitoneal administration of 2 mg/kg lipopolysaccharide (LPS). C57BL/6 J mice were separated into four groups (n = 10 per group): positive control, negative control, pristimerin 10 mg/kg and pristimerin 100 mg/kg. Pristimerin was administered orally for 28 days prior to LPS administration and for six days thereafter. Behavioural changes were assessed one day after LPS administration using the Morris water maze and via neurological dysfunction scoring. Molecular pathogenesis was explored by measurement of malondialdehyde, superoxide dismutase, reactive oxygen species and inflammatory cytokine levels in mouse brains. Neuronal apoptosis was evaluated using the TUNEL assay. The levels of p-Akt/Akt, p-PI3K/PI3K, mTOR, Bax, Bcl-2 and caspase-3 proteins were determined via Western blotting. RESULTS Pristimerin improved cognitive function and reduces the neurological score to 1.15 ± 0.03. Pristimerin significantly reduced all cytokine levels: TNF-α by 18 ± 0.6 pg/mg, IL-1β by 43 ± 1.3 pg/mg and IL-6 by 34 ± 1.12 pg/mg. There was significant (p < 0.01) improvement in PI3K/Akt signalling and histopathological changes in the brain tissue of sepsis induced brain injured rats. CONCLUSIONS Pristimerin ameliorated neuronal injury by regulating PI3K/Akt signalling in mice with sepsis-induced brain injuries. Pristimerin may merit further development for clinical applications.
Collapse
Affiliation(s)
- Weimin Xue
- Department of Neurology, First Affiliated Hospital of Anhui University of Science and Technology, First People's Hospital of Huainan, Huainan, China
| | - Yaqiang Li
- Department of Neurology, First Affiliated Hospital of Anhui University of Science and Technology, First People's Hospital of Huainan, Huainan, China
| | - Mei Zhang
- Department of Neurology, First Affiliated Hospital of Anhui University of Science and Technology, First People's Hospital of Huainan, Huainan, China
| |
Collapse
|
40
|
Huang B, Li X, Zhu X. The Role of GM130 in Nervous System Diseases. Front Neurol 2021; 12:743787. [PMID: 34777211 PMCID: PMC8581157 DOI: 10.3389/fneur.2021.743787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/29/2021] [Indexed: 11/24/2022] Open
Abstract
Golgi matrix protein 130 (GM130) is a Golgi-shaping protein located on the cis surface of the Golgi apparatus (GA). It is one of the most studied Golgin proteins so far. Its biological functions are involved in many aspects of life processes, including mitosis, autophagy, apoptosis, cell polarity, and directed migration at the cellular level, as well as intracellular lipid and protein transport, microtubule formation and assembly, lysosome function maintenance, and glycosylation modification. Mutation inactivation or loss of expression of GM130 has been detected in patients with different diseases. GM130 plays an important role in the development of the nervous system, but the studies on it are limited. This article reviewed the current research progress of GM130 in nervous system diseases. It summarized the physiological functions of GM130 in the occurrence and development of Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), microcephaly (MCPH), sepsis associated encephalopathy (SAE), and Ataxia, aiming to provide ideas for the further study of GM130 in nervous system disease detection and treatment.
Collapse
Affiliation(s)
- Bei Huang
- Operational Management Office, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.,Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaoshi Zhu
- Pediatric Intensive Care Unit, Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
41
|
Peng X, Luo Z, He S, Zhang L, Li Y. Blood-Brain Barrier Disruption by Lipopolysaccharide and Sepsis-Associated Encephalopathy. Front Cell Infect Microbiol 2021; 11:768108. [PMID: 34804998 PMCID: PMC8599158 DOI: 10.3389/fcimb.2021.768108] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
As a complex multicellular structure of the vascular system at the central nervous system (CNS), the blood-brain barrier (BBB) separates the CNS from the system circulation and regulates the influx and efflux of substances to maintain the steady-state environment of the CNS. Lipopolysaccharide (LPS), the cell wall component of Gram-negative bacteria, can damage the barrier function of BBB and further promote the occurrence and development of sepsis-associated encephalopathy (SAE). Here, we conduct a literature review of the direct and indirect damage mechanisms of LPS to BBB and the relationship between these processes and SAE. We believe that after LPS destroys BBB, a large number of inflammatory factors and neurotoxins will enter and damage the brain tissue, which will activate brain immune cells to mediate inflammatory response and in turn further destroys BBB. This vicious circle will ultimately lead to the progression of SAE. Finally, we present a succinct overview of the treatment of SAE by restoring the BBB barrier function and summarize novel opportunities in controlling the progression of SAE by targeting the BBB.
Collapse
Affiliation(s)
- Xiaoyao Peng
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Zhixuan Luo
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Shuang He
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Luhua Zhang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ying Li
- Department of Immunology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
42
|
Luo D, Han L, Gao S, Xiao Z, Zhou Q, Cheng X, Zhang Y, Zhou W. LINCS Dataset-Based Repositioning of Dutasteride as an Anti-Neuroinflammation Agent. Brain Sci 2021; 11:1411. [PMID: 34827410 PMCID: PMC8615696 DOI: 10.3390/brainsci11111411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation is often accompanied by central nervous system (CNS) injury seen in various CNS diseases, with no specific treatment. Drug repurposing is a strategy of finding new uses for approved or investigational drugs, and can be enabled by the Library of Integrated Network-based Cellular Signatures (LINCS), a large drug perturbation database. In this study, the signatures of Lipopolysaccharide (LPS) were compared with the signatures of compounds contained in the LINCS dataset. To the top 100 compounds obtained, the Quantitative Structure-Activity Relationship (QSAR)-based tool admetSAR was used to identify the top 10 candidate compounds with relatively high blood-brain barrier (BBB) penetration. Furthermore, the seventh-ranked compound, dutasteride, a 5-α-reductase inhibitor, was selected for in vitro and in vivo validation of its anti-neuroinflammation activity. The results showed that dutasteride significantly reduced the levels of IL-6 and TNF-α in the supernatants of LPS-stimulated BV2 cells, and decreased the levels of IL-6 in the hippocampus and plasma, and the number of activated microglia in the brain of LPS administration mice. Furthermore, dutasteride also attenuated the cognitive impairment caused by LPS stimulation in mice. Taken together, this study demonstrates that the LINCS dataset-based drug repurposing strategy is an effective approach, and the predicted candidate, dutasteride, has the potential to ameliorate LPS-induced neuroinflammation and cognitive impairment.
Collapse
Affiliation(s)
- Dan Luo
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Lu Han
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Shengqiao Gao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Qingru Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Xiaorui Cheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Yongxiang Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| |
Collapse
|
43
|
Archie SR, Al Shoyaib A, Cucullo L. Blood-Brain Barrier Dysfunction in CNS Disorders and Putative Therapeutic Targets: An Overview. Pharmaceutics 2021; 13:pharmaceutics13111779. [PMID: 34834200 PMCID: PMC8622070 DOI: 10.3390/pharmaceutics13111779] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 01/22/2023] Open
Abstract
The blood-brain barrier (BBB) is a fundamental component of the central nervous system (CNS). Its functional and structural integrity is vital to maintain the homeostasis of the brain microenvironment by controlling the passage of substances and regulating the trafficking of immune cells between the blood and the brain. The BBB is primarily composed of highly specialized microvascular endothelial cells. These cells’ special features and physiological properties are acquired and maintained through the concerted effort of hemodynamic and cellular cues from the surrounding environment. This complex multicellular system, comprising endothelial cells, astrocytes, pericytes, and neurons, is known as the neurovascular unit (NVU). The BBB strictly controls the transport of nutrients and metabolites into brain parenchyma through a tightly regulated transport system while limiting the access of potentially harmful substances via efflux transcytosis and metabolic mechanisms. Not surprisingly, a disruption of the BBB has been associated with the onset and/or progression of major neurological disorders. Although the association between disease and BBB disruption is clear, its nature is not always evident, specifically with regard to whether an impaired BBB function results from the pathological condition or whether the BBB damage is the primary pathogenic factor prodromal to the onset of the disease. In either case, repairing the barrier could be a viable option for treating and/or reducing the effects of CNS disorders. In this review, we describe the fundamental structure and function of the BBB in both healthy and altered/diseased conditions. Additionally, we provide an overview of the potential therapeutic targets that could be leveraged to restore the integrity of the BBB concomitant to the treatment of these brain disorders.
Collapse
Affiliation(s)
- Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (S.R.A.); (A.A.S.)
| | - Abdullah Al Shoyaib
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (S.R.A.); (A.A.S.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Correspondence: ; Tel.: +1-248-370-3884; Fax: +1-248-370-4060
| |
Collapse
|
44
|
Huang Y, Chen R, Jiang L, Li S, Xue Y. Basic research and clinical progress of sepsis-associated encephalopathy. JOURNAL OF INTENSIVE MEDICINE 2021; 1:90-95. [PMID: 36788800 PMCID: PMC9923961 DOI: 10.1016/j.jointm.2021.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/22/2021] [Accepted: 08/18/2021] [Indexed: 01/02/2023]
Abstract
Sepsis-associated encephalopathy (SAE), a major cerebral complication of sepsis, occurs in 70% of patients admitted to the intensive care unit (ICU). This condition can cause serious impairment of consciousness and is associated with a high mortality rate. Thus far, several experimental screenings and radiological techniques (e.g., electroencephalography) have been used for the non-invasive assessment of the structure and function of the brain in patients with SAE. Nevertheless, the pathogenesis of SAE is complicated and remains unclear. In the present article, we reviewed the currently available literature on the epidemiology, clinical manifestations, pathology, diagnosis, and management of SAE. However, currently, there is no ideal pharmacological treatment for SAE. Treatment targeting mitochondrial dysfunction may be useful in the management of SAE.
Collapse
Affiliation(s)
- Ying Huang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China,Corresponding author: Ying Huang, Department of Anesthesiology and Surgical Intensive Care Unit, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
| | - Ruman Chen
- Department of Blood Purification, Hainan General Hospital Affiliated to Hainan Medical University, Haikou, Hainan 570311, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Siyuan Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yuchen Xue
- Department of Anesthesiology and Surgical Intensive Care Unit, Xin-Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
45
|
Yao Mattisson I, Christoffersen C. Apolipoprotein M and its impact on endothelial dysfunction and inflammation in the cardiovascular system. Atherosclerosis 2021; 334:76-84. [PMID: 34482091 DOI: 10.1016/j.atherosclerosis.2021.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/09/2023]
Abstract
Apolipoprotein M (apoM) is a member of the lipocalin superfamily and is predominantly associated with high-density lipoprotein (HDL). It was found that apoM is the chaperon to the bioactive sphingolipid, sphingosine-1-phosphate (S1P). Several studies have since contributed to expand the knowledge on apoM, S1P, and the apoM/S1P-complex in cardiovascular diseases. For instance, the HDL-bound apoM/S1P complex serves as a bridge between HDL and endothelial cells, maintaining a healthy endothelial barrier. Evidence indicates, however, that the apoM/S1P complex may has both protective and harmful effects on the cardiovascular system, which suggests the need for more research to understand the interplay between these molecules. This review aims to shed light on the most recent findings on apoM/S1P-signaling and its impact on endothelial dysfunction, inflammation, and cardiovascular diseases. Finally, it will be discussed whether drugs that target apoM and/or S1P-signaling may be beneficial to patients with cardiovascular and inflammatory diseases.
Collapse
Affiliation(s)
- Ingrid Yao Mattisson
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark; Department of Clinical Sciences Malmö, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3A, 2200, Copenhagen, Denmark.
| |
Collapse
|
46
|
Kodali MC, Chen H, Liao FF. Temporal unsnarling of brain's acute neuroinflammatory transcriptional profiles reveals panendothelitis as the earliest event preceding microgliosis. Mol Psychiatry 2021; 26:3905-3919. [PMID: 33293688 PMCID: PMC7722246 DOI: 10.1038/s41380-020-00955-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is an acutely progressing brain dysfunction induced by systemic inflammation. The mechanism of initiation of neuroinflammation during SAE, which ultimately leads to delirium and cognitive dysfunction, remains elusive. We aimed to study the molecular events of SAE to capture its onset and progression into the central nervous system (CNS), and further identify the cellular players involved in mediating acute inflammatory signaling. Gene expression profiling on the cerebral vessels isolated from the brains of the mice treated with peripheral lipopolysaccharide (LPS) revealed that the cerebral vasculature responds within minutes to acute systemic inflammation by upregulating the expression of immediate early response genes, followed by activation of the nuclear factor-κB pathway. To identify the earliest responding cell type, we used fluorescence-activated cell sorting (FACS) to sort the glial and vascular cells from the brains of the mice treated with LPS at different time points, and RNA-seq was performed on microglia and cerebral endothelial cells (CECs). Bioinformatic analysis followed by further validation in all the cell types revealed that panendothelitis. i.e., the activation of CECs is the earliest event in the CNS during the inception of acute neuroinflammation. Microglial activation occurs later than that of CECs, suggesting that CECs are the most likely initial source of proinflammatory mediators, which could further initiate glial cell activation. This is then followed by the activation of apoptotic signaling in the CECs, which is known to lead to the blood-brain barrier disruption and allow peripheral cytokines to leak into the CNS, exacerbate the gliosis, and result in the vicious neuroinflammatory cascade. Together, our results model the earliest sequential events during the advancement of systemic inflammation into the CNS and facilitate to understand the interplay between the vascular and glial cells in initiating and driving acute neuroinflammation during SAE.
Collapse
Affiliation(s)
- Mahesh Chandra Kodali
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
- Integrated Biomedical Sciences Program, Molecular and Systems Pharmacology Track, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
| |
Collapse
|
47
|
Gao Q, Hernandes MS. Sepsis-Associated Encephalopathy and Blood-Brain Barrier Dysfunction. Inflammation 2021; 44:2143-2150. [PMID: 34291398 DOI: 10.1007/s10753-021-01501-3] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022]
Abstract
Sepsis is a life-threatening clinical condition caused by a dysregulated host response to infection. Sepsis-associated encephalopathy (SAE) is a common but poorly understood neurological complication of sepsis, which is associated with increased morbidity and mortality. SAE clinical presentation may range from mild confusion and delirium to severe cognitive impairment and deep coma. Important mechanisms associated with SAE include excessive microglial activation, impaired endothelial barrier function, and blood-brain barrier (BBB) dysfunction. Endotoxemia and pro-inflammatory cytokines produced systemically during sepsis lead to microglial and brain endothelial cell activation, tight junction downregulation, and increased leukocyte recruitment. The resulting neuroinflammation and BBB dysfunction exacerbate SAE pathology and aggravate sepsis-induced brain dysfunction. In this mini-review, recent literature surrounding some of the mediators of BBB dysfunction during sepsis is summarized. Modulation of microglial activation, endothelial cell dysfunction, and the consequent prevention of BBB permeability represent relevant therapeutic targets that may significantly impact SAE outcomes.
Collapse
Affiliation(s)
- Qingzeng Gao
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, WMB 308, Atlanta, GA, 30322, USA
| | - Marina Sorrentino Hernandes
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, WMB 308, Atlanta, GA, 30322, USA.
| |
Collapse
|
48
|
Zhang N, Zhao W, Hu ZJ, Ge SM, Huo Y, Liu LX, Gao BL. Protective effects and mechanisms of high-dose vitamin C on sepsis-associated cognitive impairment in rats. Sci Rep 2021; 11:14511. [PMID: 34267240 PMCID: PMC8282649 DOI: 10.1038/s41598-021-93861-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 06/30/2021] [Indexed: 01/29/2023] Open
Abstract
Sepsis survivors present long-term cognitive deficits. The present study was to investigate the effect of early administration of high-dose vitamin C on cognitive function in septic rats and explore its possible cerebral protective mechanism. Rat sepsis models were established by cecal ligation and puncture (CLP). Ten days after surgery, the Morris water maze test was performed to evaluate the behavior and cognitive function. Histopathologic changes in the hippocampus were evaluated by nissl staining. The inflammatory cytokines, activities of antioxidant enzymes (superoxide dismutase or SOD) and oxidative products (malondialdehyde or MDA) in the serum and hippocampus were tested 24 h after surgery. The activity of matrix metalloproteinase-9 (MMP-9) and expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1(HO-1) in the hippocampus were measured 24 h after surgery. Compared with the sham group in the Morris water maze test, the escape latency of sepsis rats was significantly (P = 0.001) prolonged in the navigation test, whereas the frequency to cross the platform and the time spent in the target quadrant were significantly (P = 0.003) reduced. High-dose vitamin C significantly decreased the escape latency (P = 0.01), but increased the time spent in the target quadrant (P = 0.04) and the frequency to cross the platform (P = 0.19). In the CLP+ saline group, the pyramidal neurons were reduced and distributed sparsely and disorderly, the levels of inflammatory cytokines of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10 in the serum and hippocampus were significantly increased (P = 0.000), the blood brain barrier (BBB) permeability in the hippocampus was significantly (P = 0.000) increased, the activities of SOD in the serum and hippocampus were significantly (P = 0.000 and P = 0.03, respectively) diminished while the levels of MDA in the serum and hippocampus were significantly (P = 0.007) increased. High-dose vitamin C mitigated hippocampus histopathologic changes, reduced systemic inflammation and neuroinflammation, attenuated BBB disruption, inhibited oxidative stress in brain tissue, and up-regulated the expression of nuclear and total Nrf2 and HO-1. High-dose vitamin C significantly (P < 0.05) decreased the levels of tumor necrosis factor- (TNF)-α, interleukin-6 (IL-6), MDA in the serum and hippocampus, and the activity of MMP-9 in the hippocampus, but significantly (P < 0.05) increased the levels of SOD, the anti-inflammatory cytokine (IL-10) in the serum and hippocampus, and nuclear and total Nrf2, and HO-1 in the hippocampus. In conclusion, high-dose vitamin C can improve cognition impairment in septic rats, and the possible protective mechanism may be related to inhibition of inflammatory factors, alleviation of oxidative stress, and activation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Wei Zhao
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Zhen-Jie Hu
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Sheng-Mei Ge
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Yan Huo
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Li-Xia Liu
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China.
| | - Bu-Lang Gao
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
49
|
Nong A, Li Q, Huang Z, Xu Y, He K, Jia Y, Cen Z, Liao L, Huang Y. MicroRNA miR-126 attenuates brain injury in septic rats via NF-κB signaling pathway. Bioengineered 2021; 12:2639-2648. [PMID: 34115555 PMCID: PMC8806573 DOI: 10.1080/21655979.2021.1937905] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The purpose of this study was to investigate the impact and mechanism of microRNA miR-126 on brain injury induced by blood-brain barrier (BBB) damage in septic rats. We used cecal ligation and perforation (CLP) to create a rat model of sepsis. The experimental rats were randomly divided into Control group, CLP group, CLP + miR-NC group, CLP + miR-126 group and CLP + miR-126 + NF-κB pathway agonist (PMA) group. MiR-126 expressed in the brain tissue of CLP rats was down-regulated by qRT-PCR. Upregulation of miR-126 in CLP rats could improve brain injury and BBB marker protein level, reduce brain water content, Evans blue extravasation, inflammation, and excessive oxidative stress. This could also result in an inhibition of NF-κB signaling pathway activity. In conclusion, miR-126 overexpression can prevent brain injury caused by BBB damage via the inhibition of NF-κB signaling pathway activity.
Collapse
Affiliation(s)
- Anna Nong
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Qingfeng Li
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Zhijing Huang
- Department of Pediatric Internal Medicine Ward 1, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Yunan Xu
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Kebin He
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Yuying Jia
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Zhenyi Cen
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Lianghua Liao
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Yueyan Huang
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi China
| |
Collapse
|
50
|
Dominguini D, Steckert AV, Abatti MR, Generoso JS, Barichello T, Dal-Pizzol F. The Protective Effect of PK-11195 on Cognitive Impairment in Rats Survived of Polymicrobial Sepsis. Mol Neurobiol 2021; 58:2724-2733. [PMID: 33495933 DOI: 10.1007/s12035-021-02294-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/13/2021] [Indexed: 10/22/2022]
Abstract
Sepsis is an organ dysfunction caused by a host's unregulated response to infection, causing long-term brain dysfunction with microglial activation, the release of inflammatory components, and mitochondrial changes. Neuroinflammation can increase the expression of the 18-kD translocator protein (TSPO) in the mitochondria, leading to the activation of the microglia and the release of inflammatory components. The antagonist PK-11195 can modulate TSPO and reduce microglial activation and cognitive damage presented in an animal model of sepsis. The aim of this was to evaluate the effects of PK-11195 on long-term brain inflammation and cognitive impairment in an animal model of sepsis. Wistar rats, 60 days old, were submitted to cecal ligation and puncture (CLP) surgery, divided into groups control/saline, control/PK-11195, sepsis/saline, and sepsis/PK-11195. Immediately after surgery, the antagonist PK-11195 was administered at a dose of 3 mg/kg. Ten days after CLP surgery, the animals were submitted to behavioral tests and determination of brain inflammatory parameters. The sepsis/saline group presented cognitive damage. However, there was damage prevention in animals that received PK-11195. Besides, the sepsis increased the levels of cytokines and M1 microglia markers and caused oxidative damage. However, PK-11195 had the potential to decrease inflammation. These events show that the modulation of neuroinflammation during sepsis by PK-11195, possibly related to changes in TSPO, improves mitochondrial function in the animals' brains. In conclusion, the antagonist PK-11195 attenuated brain inflammation and prevented cognitive impairment in animals subjected to sepsis.
Collapse
Affiliation(s)
- Diogo Dominguini
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil.
| | - Amanda V Steckert
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Mariane R Abatti
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Jaqueline S Generoso
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| |
Collapse
|