1
|
Curtis SN, Mayer CA, Bonfield TL, Raffay TM, DiFiore JM, Martin RJ, Hoffman AC, Folz MA, Bavis RW, Dutschmann M, MacFarlane PM. Unique infrared thermographic profiles and altered hypothalamic neurochemistry associated with mortality in endotoxic shock. Exp Neurol 2025; 385:115130. [PMID: 39732274 DOI: 10.1016/j.expneurol.2024.115130] [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: 08/15/2024] [Revised: 12/13/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Neonatal sepsis results in significant morbidity and mortality, but early detection is clinically challenging. In a neonatal rat model of endotoxic shock, we characterised unique infrared thermographic (IRT) profiles in skin temperature that could identify risk of later mortality. Ten-day old rats were placed in a thermally stable isolette and IRT images of cranial (TCR), scapula (TSC) and rump (TRU) skin temperature were obtained continuously for 8 h following an intraperitoneal injection of lipopolysaccharide (LPS) or saline. LPS resulted in ∼74 % mortality (designated as non-survivors, LPSNS) between 4.5 and 7.5 h post-injection. LPSNS and survivors of LPS (LPSS) rats displayed hypothermic tendencies with TCR, TSC and TRU decreasing at ∼80-100 min (T80-100) post-injection. Compared to LPSS rats, however, the hypothermia of LPSNS rats occurred slightly earlier (T80), was more severe, and failed to recover. The TCR, TSC and TRU of LPSS rats fully recovered by 4 h (T240) post-injection. In separate rats, hypothalamic microglia and extracellular matrix (ECM) expression at T240 post-injection were increased in putatively identified LPSNS rats (but not LPSS rats) and negatively correlated with IR temperatures. IRT could be a useful early identifier of infants at risk of death from endotoxic shock, which may be related to early failure of central nervous system (CNS) thermogenic mechanisms mediated by unique hypothalamic changes in inflammatory (microglia) and ECM neurochemistry.
Collapse
Affiliation(s)
- Sean N Curtis
- Department of Pediatrics, UH Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Catherine A Mayer
- Department of Pediatrics, UH Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Tracey L Bonfield
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, United States of America
| | - Thomas M Raffay
- Department of Pediatrics, UH Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Juliann M DiFiore
- Department of Pediatrics, UH Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Richard J Martin
- Department of Pediatrics, UH Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Adriana C Hoffman
- Department of Pediatrics, UH Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Michael A Folz
- School of Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Ryan W Bavis
- Department of Biology, Bates College, Lewiston, ME, United States of America
| | - Mathias Dutschmann
- Pulmonary, Critical, and Sleep Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Peter M MacFarlane
- Department of Pediatrics, UH Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America.
| |
Collapse
|
2
|
Hou FF, Mi JH, Wang Q, Tao YL, Guo SB, Ran GH, Wang JC. Macrophage polarization in sepsis: Emerging role and clinical application prospect. Int Immunopharmacol 2025; 144:113715. [PMID: 39626538 DOI: 10.1016/j.intimp.2024.113715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/15/2024]
Abstract
Sepsis is a severe, potentially fatal condition defined by organ dysfunction due to excessive inflammation. Its complex pathogenesis and poor therapeutic outcomes pose significant challenges in treatment. Macrophages, with their high heterogeneity and plasticity, play crucial roles in both the innate and adaptive immune systems. They can polarize into M1-like macrophages, which promote pro-inflammatory responses, or M2-like macrophages, which mediate anti-inflammatory responses, positioning them as critical mediators in the immune response during sepsis.Macrophages are the main regulators of inflammatory responses, and their polarization is also regulated by inflammatory signaling pathways. This review highlights recent advances in the inflammatory signaling pathways involved in sepsis, mechanism of macrophage polarization mediated by inflammation-related signaling pathways in sepsis, and the role of signaling pathway mediated macrophage polarization in organ dysfunction involved in sepsis. We also explore the therapeutic potential of targeting macrophage polarization for immunotherapy, offering new perspectives on macrophage-targeted treatments for sepsis.
Collapse
Affiliation(s)
- Fei Fei Hou
- Intensive Care Unit Inner Mongolia Medical University Affiliated Hospital, Hohhot 010050, China
| | - Jun Hao Mi
- Liuzhou Maternity and Child Healthcare Hospital, Liuzhou 545001, China
| | - Qiong Wang
- Burn and Plastic Surgery Department of Hohhot First Hospital, Hohhot 010030, China
| | - Yan Lin Tao
- Intensive Care Unit Inner Mongolia Medical University Affiliated Hospital, Hohhot 010050, China
| | - Shuai Bin Guo
- Intensive Care Unit Inner Mongolia Medical University Affiliated Hospital, Hohhot 010050, China
| | - Guang He Ran
- Chongqing Changshou Traditional Cinese Medicine Hospital, 401200 Chongqing, China.
| | - Jing Chao Wang
- Intensive Care Unit Inner Mongolia Medical University Affiliated Hospital, Hohhot 010050, China.
| |
Collapse
|
3
|
Grünewald B, Wickel J, Hahn N, Rahmati V, Rupp H, Chung HY, Haselmann H, Strauss AS, Schmidl L, Hempel N, Grünewald L, Urbach A, Bauer M, Toyka KV, Blaess M, Claus RA, König R, Geis C. Targeted rescue of synaptic plasticity improves cognitive decline in sepsis-associated encephalopathy. Mol Ther 2024; 32:2113-2129. [PMID: 38788710 PMCID: PMC11286813 DOI: 10.1016/j.ymthe.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 04/02/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a frequent complication of severe systemic infection resulting in delirium, premature death, and long-term cognitive impairment. We closely mimicked SAE in a murine peritoneal contamination and infection (PCI) model. We found long-lasting synaptic pathology in the hippocampus including defective long-term synaptic plasticity, reduction of mature neuronal dendritic spines, and severely affected excitatory neurotransmission. Genes related to synaptic signaling, including the gene for activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) and members of the transcription-regulatory EGR gene family, were downregulated. At the protein level, ARC expression and mitogen-activated protein kinase signaling in the brain were affected. For targeted rescue we used adeno-associated virus-mediated overexpression of ARC in the hippocampus in vivo. This recovered defective synaptic plasticity and improved memory dysfunction. Using the enriched environment paradigm as a non-invasive rescue intervention, we found improvement of defective long-term potentiation, memory, and anxiety. The beneficial effects of an enriched environment were accompanied by an increase in brain-derived neurotrophic factor (BDNF) and ARC expression in the hippocampus, suggesting that activation of the BDNF-TrkB pathway leads to restoration of the PCI-induced reduction of ARC. Collectively, our findings identify synaptic pathomechanisms underlying SAE and provide a conceptual approach to target SAE-induced synaptic dysfunction with potential therapeutic applications to patients with SAE.
Collapse
Affiliation(s)
- Benedikt Grünewald
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Institute of Pathophysiology and Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Jonathan Wickel
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Nina Hahn
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Vahid Rahmati
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Hanna Rupp
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Ha-Yeun Chung
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Holger Haselmann
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Anja S Strauss
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Lars Schmidl
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Nina Hempel
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Lena Grünewald
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, 60528 Frankfurt, Germany
| | - Anja Urbach
- Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Jena Center for Healthy Aging, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Leibniz Institute on Aging, Aging Research Center Jena, Beutenbergstr. 11, 07745 Jena, Germany
| | - Michael Bauer
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Department of Anesthesiology and Intensive Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Klaus V Toyka
- Department of Neurology, University of Würzburg, 97080 Würzburg, Germany
| | - Markus Blaess
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, 78054 Villingen-Schwenningen, Germany
| | - Ralf A Claus
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Department of Anesthesiology and Intensive Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Rainer König
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Department of Anesthesiology and Intensive Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Christian Geis
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Jena, Germany.
| |
Collapse
|
4
|
Dal-Pizzol F, Coelho A, Simon CS, Michels M, Corneo E, Jeremias A, Damásio D, Ritter C. Prophylactic Minocycline for Delirium in Critically Ill Patients: A Randomized Controlled Trial. Chest 2024; 165:1129-1138. [PMID: 38043911 DOI: 10.1016/j.chest.2023.11.041] [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: 07/13/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Delirium is a potentially severe form of acute encephalopathy. Minocycline has neuroprotective effects in animal models of neurologic diseases; however, data from human studies remain scarce. RESEARCH QUESTION Does the neuroprotective effect of minocycline prevent delirium occurrence in critically ill patients? STUDY DESIGN AND METHODS This study was a randomized, placebo-controlled, double-anonymized trial conducted in four ICUs. Patients aged 18 years or older were eligible and randomized to receive minocycline (100 mg, twice daily) or placebo. The primary outcome was delirium incidence within 28 days or before ICU discharge. Secondary outcomes included days in delirium during ICU stay, delirium/coma-free days, length of mechanical ventilation, ICU length of stay, ICU mortality, and hospital mortality. The kinetics of various inflammatory (IL-1β, IL-6, IL-10, and C-reactive protein) and brain-related biomarkers (brain-derived neurotrophic factor and S100B) were used as exploratory outcomes. RESULTS A total of 160 patients were randomized, but one patient in the placebo group died before treatment; thus the data from 159 patients were analyzed (minocycline, n = 84; placebo, n = 75). After the COVID-19 pandemic it was decided to stop patient inclusion early. There was a small but significant decrease in delirium incidence: 17 patients (20%) in the minocycline arm compared with 26 patients (35%) in the placebo arm (P = .043). No other delirium-related outcomes were modified by minocycline treatment. Unexpectedly, there was a significant decrease in hospital mortality (39% vs. 23%; P = .029). Among all analyzed biomarkers, only plasma levels of C-reactive protein decreased significantly after minocycline treatment (F = 0.75, P = .78, within time; F = 4.09, P = .045, group × time). INTERPRETATION Our findings in this rather small study signal a possible positive effect of minocycline on delirium incidence. Further studies are needed to confirm the benefits of this drug as a preventive measure in critically ill patients. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov; No.: NCT04219735; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil; Intensive Care Unit, São José Hospital, Criciúma, Brazil; São José Hospital Research Center, Criciúma, Brazil.
| | - André Coelho
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil; Intensive Care Unit, São José Hospital, Criciúma, Brazil
| | - Carla S Simon
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Emily Corneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | | | | | - Cristiane Ritter
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil; Intensive Care Unit, São José Hospital, Criciúma, Brazil
| |
Collapse
|
5
|
Yu Z, Shi H, Zhang J, Ma C, He C, Yang F, Zhao L. ROLE OF MICROGLIA IN SEPSIS-ASSOCIATED ENCEPHALOPATHY PATHOGENESIS: AN UPDATE. Shock 2024; 61:498-508. [PMID: 38150368 DOI: 10.1097/shk.0000000000002296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
ABSTRACT Sepsis-associated encephalopathy (SAE) is a serious complication of sepsis, which is characterized by cognitive dysfunction, a poor prognosis, and high incidences of morbidity and mortality. Substantial levels of systemic inflammatory factors induce neuroinflammatory responses during sepsis, ultimately disrupting the central nervous system's (CNS) homeostasis. This disruption results in brain dysfunction through various underlying mechanisms, contributing further to SAE's development. Microglia, the most important macrophage in the CNS, can induce neuroinflammatory responses, brain tissue injury, and neuronal dysregulation, resulting in brain dysfunction. They serve an important regulatory role in CNS homeostasis and can be activated through multiple pathways. Consequently, activated microglia are involved in several pathogenic mechanisms related to SAE and play a crucial role in its development. This article discusses the role of microglia in neuroinflammation, dysfunction of neurotransmitters, disruption of the blood-brain barrier, abnormal control of cerebral blood flow, mitochondrial dysfunction, and reduction in the number of good bacteria in the gut as main pathogenic mechanisms of SAE and focuses on studies targeting microglia to ameliorate SAE to provide a theoretical basis for targeted microglial therapy for SAE.
Collapse
Affiliation(s)
| | - Hui Shi
- Department of Critical Care Medicine, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Jingjing Zhang
- Department of Central Laboratory, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Chunhan Ma
- Chifeng Clinical Medical College of Inner Mongolia Medical University, Hohhot, China
| | - Chen He
- Chifeng Clinical Medical College of Inner Mongolia Medical University, Hohhot, China
| | - Fei Yang
- Department of Critical Care Medicine, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Lina Zhao
- Department of Critical Care Medicine, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
6
|
Ji Y, Ma Y, Ma Y, Wang Y, Zhao X, Jin D, Xu L, Ge S. Rutin prevents pyroptosis and M1 microglia via Nrf2/Mac-1/caspase-1-mediated inflammasome axis to improve POCD. Int Immunopharmacol 2024; 127:111290. [PMID: 38064815 DOI: 10.1016/j.intimp.2023.111290] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/05/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Neuroinflammation following peripheral surgery plays a key role in postoperative cognitive dysfunction (POCD) development and there is no effective therapy to inflammation-mediated cognitive impairment. Recent studies showed that rutin, a natural flavonoid compound, conferred neuroprotection. However, the effects and mechanisms of rutin on cognition of surgical and aged mice and LPS-induced BV2 need deeper exploration. METHODS The effect of rutin in vivo and vitro were evaluated by Morris water maze test, HE stainin, Golgi-Cox staining, IF, IHC, RT-PCR, Flow Cytometer and Western blotting. In vivo, aged mice were treated with rutin and surgery. In vitro, rutin, Nrf2 knockdown, MAC-1 overexpression and VX765, a caspase-1 inhibitor, were administration on BV2 microglial cells. RESULTS Surgery led to compensatory increase in nuclear Nrf2 and rutin could further increase it. Neural damage was accompanied with high level in MAC-1, caspase-1-mediated pyroptosis and M1 microglia, while rutin recovered the process. Nrf2 inhibition abolished the effect of rutin with the increase of MAC-1, caspase-1-mediated pyroptosis and M1 microglia. Activation of MAC-1 abrogated protection of rutin by increase in pyroptosis and M1 microglia. Finally, we found that treatment with VX765 improved injury and increased M2 microglia against overexpression of MAC-1. CONCLUSIONS Our study indicated that rutin may be a potential therapy in POCD and exerted neural protection via Nrf2/ Mac-1/ caspase-1-mediated inflammasome axis to regulate pyroptosis and microglial polarization.
Collapse
Affiliation(s)
- Yelong Ji
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Yuanyuan Ma
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Yimei Ma
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Ying Wang
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Xining Zhao
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Danfeng Jin
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Li Xu
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China
| | - Shengjin Ge
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032 China.
| |
Collapse
|
7
|
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
|
8
|
Xin Y, Tian M, Deng S, Li J, Yang M, Gao J, Pei X, Wang Y, Tan J, Zhao F, Gao Y, Gong Y. The Key Drivers of Brain Injury by Systemic Inflammatory Responses after Sepsis: Microglia and Neuroinflammation. Mol Neurobiol 2023; 60:1369-1390. [PMID: 36445634 PMCID: PMC9899199 DOI: 10.1007/s12035-022-03148-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022]
Abstract
Sepsis is a leading cause of intensive care unit admission and death worldwide. Most surviving patients show acute or chronic mental disorders, which are known as sepsis-associated encephalopathy (SAE). Although accumulating studies in the past two decades focused on the pathogenesis of SAE, a systematic review of retrospective studies which exclusively focuses on the inflammatory mechanisms of SAE has been lacking yet. This review summarizes the recent advance in the field of neuroinflammation and sheds light on the activation of microglia in SAE. Activation of microglia predominates neuroinflammation. As the gene expression profile changes, microglia show heterogeneous characterizations throughout all stages of SAE. Here, we summarize the systemic inflammation following sepsis and also the relationship of microglial diversity and neuroinflammation. Moreover, a collection of neuroinflammation-related dysfunction has also been reviewed to illustrate the possible mechanisms for SAE. In addition, promising pharmacological or non-pharmacological therapeutic strategies, especially those which target neuroinflammation or microglia, are also concluded in the final part of this review. Collectively, clarification of the vital relationship between neuroinflammation and SAE-related mental disorders would significantly improve our understanding of the pathophysiological mechanisms in SAE and therefore provide potential targets for therapies of SAE aimed at inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Yuewen Xin
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Mi Tian
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Shuixiang Deng
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Li
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Miaoxian Yang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jianpeng Gao
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xu Pei
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yao Wang
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Tan
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Feng Zhao
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yanqin Gao
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Ye Gong
- Department of Critical Care Medicine of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Dominguini D, Michels M, Wessler LB, Streck EL, Barichello T, Dal-Pizzol F. Mitochondrial protective effects caused by the administration of mefenamic acid in sepsis. J Neuroinflammation 2022; 19:268. [PMID: 36333747 PMCID: PMC9636698 DOI: 10.1186/s12974-022-02616-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
The pathophysiology of sepsis may involve the activation of the NOD-type receptor containing the pyrin-3 domain (NLPR-3), mitochondrial and oxidative damages. One of the primary essential oxidation products is 8-oxoguanine (8-oxoG), and its accumulation in mitochondrial DNA (mtDNA) induces cell dysfunction and death, leading to the hypothesis that mtDNA integrity is crucial for maintaining neuronal function during sepsis. In sepsis, the modulation of NLRP-3 activation is critical, and mefenamic acid (MFA) is a potent drug that can reduce inflammasome activity, attenuating the acute cerebral inflammatory process. Thus, this study aimed to evaluate the administration of MFA and its implications for the reduction of inflammatory parameters and mitochondrial damage in animals submitted to polymicrobial sepsis. To test our hypothesis, adult male Wistar rats were submitted to the cecal ligation and perforation (CLP) model for sepsis induction and after receiving an injection of MFA (doses of 10, 30, and 50 mg/kg) or sterile saline (1 mL/kg). At 24 h after sepsis induction, the frontal cortex and hippocampus were dissected to analyze the levels of TNF-α, IL-1β, and IL-18; oxidative damage (thiobarbituric acid reactive substances (TBARS), carbonyl, and DCF-DA (oxidative parameters); protein expression (mitochondrial transcription factor A (TFAM), NLRP-3, 8-oxoG; Bax, Bcl-2 and (ionized calcium-binding adaptor molecule 1 (IBA-1)); and the activity of mitochondrial respiratory chain complexes. It was observed that the septic group in both structures studied showed an increase in proinflammatory cytokines mediated by increased activity in NLRP-3, with more significant oxidative damage and higher production of reactive oxygen species (ROS) by mitochondria. Damage to mtDNA it was also observed with an increase in 8-oxoG levels and lower levels of TFAM and NGF-1. In addition, this group had an increase in pro-apoptotic proteins and IBA-1 positive cells. However, MFA at doses of 30 and 50 mg/kg decreased inflammasome activity, reduced levels of cytokines and oxidative damage, increased bioenergetic efficacy and reduced production of ROS and 8-oxoG, and increased levels of TFAM, NGF-1, Bcl-2, reducing microglial activation. As a result, it is suggested that MFA induces protection in the central nervous system early after the onset of 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.
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Leticia B Wessler
- Laboratory of Bioenergetics, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Emilio L Streck
- Laboratory of Bioenergetics, 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, 77054, 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
|
10
|
Jing G, Zuo J, Fang Q, Yuan M, Xia Y, Jin Q, Liu Y, Wang Y, Zhang Z, Liu W, Wu X, Song X. Erbin protects against sepsis-associated encephalopathy by attenuating microglia pyroptosis via IRE1α/Xbp1s-Ca 2+ axis. J Neuroinflammation 2022; 19:237. [PMID: 36171629 PMCID: PMC9520943 DOI: 10.1186/s12974-022-02598-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/16/2022] [Indexed: 06/27/2024] Open
Abstract
Background Microglia pyroptosis-mediated neuroinflammation is thought to be the crucial pathogenesis of sepsis-associated encephalopathy (SAE). Erbin has been reported to be associated with various inflammatory diseases. However, the role of Erbin in SAE and the relationship between Erbin and microglia pyroptosis are unknown. In this study, we investigated the promising role and underlying molecular mechanism of Erbin in the regulation of microglia pyroptosis. Methods WT and Erbin knockout mice underwent cecum ligation perforation (CLP) to induce SAE. Primary mouse microglia and BV2 cells were treated with LPS/nigericin in vitro. Behavioral tests were performed to evaluate cognitive function. Nissl staining and transmission electron microscopy were used to assess histological and structural lesions. ELISA and qPCR were carried out to detect neuroinflammation. Western blot and immunofluorescence were used to analyze protein expression. Flow cytometry and confocal microscopy were utilized to observe the Ca2+ changes in the cytoplasm and endoplasmic reticulum (ER). To further explore the underlying mechanism, STF083010 was administered to block the IRE1α/Xbp1s pathway. Results Erbin deletion resulted in more pronounced neuronal damage and cognitive impairment in mice that underwent CLP. Erbin knockout promoted microglial pyroptosis and inflammatory cytokines secretion in vivo and in vitro, which was mediated by activation of the IRE1α/Xbp1s. Treatment with the selective inhibitor STF083010 significantly inhibited IRE1α/Xbp1s pathway activity, decreased intracytoplasmic Ca2+, attenuated microglial pyroptosis, reduced pro-inflammatory cytokine secretion, lessened neuronal damage, and improved cognitive function. Conclusions In SAE, Erbin inhibits IRE1/Xbp1s pathway activity and reduces the ER Ca2+ influx to the cytoplasm, reducing microglial pyroptosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02598-5.
Collapse
Affiliation(s)
- Guoqing Jing
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Zuo
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qing Fang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yun Xia
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiyan Jin
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuping Liu
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yanlin Wang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zongze Zhang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wanhong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xuemin Song
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
11
|
Li JM, Hu T, Jiang CL, Wang W. Pinocembrin ameliorates depressive-like behaviors by regulating P2X7/TRL4 receptors expression in mouse hippocampus. Behav Pharmacol 2022; 33:301-308. [PMID: 35621136 DOI: 10.1097/fbp.0000000000000677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mounting evidence indicates that immune dysfunction may contribute to the neurobiology of major depressive disorder (MDD). Toll-like receptor 4 (TLR4) and P2X7 receptor (P2X7R) were recently reckoned pivotally to regulate NOD-like receptor protein 3 (NLRP3) in microglia. Pinocembrin, one of the primary flavonoids from Pinus heartwood and Eucalyptus, has been studied in various animal models of human disease with anti-inflammatory and antioxidant activities. Herein, we investigated the potential antineuroinflammatory effects of pinocembrin on chronic unpredictable mild stress (CUMS)-induced depressive-like behavior. Male C57BL/6J mice were subjected to CUMS for 4 weeks, treatment group was injected with pinocembrin at a dose of 20 mg/kg. After the stress procedure, behavioral tests, including sucrose preference tests (SPTs) and tail suspension tests (TSTs) were performed to evaluate depressive-like phenotype. Subsequently, the expression of cytokines and microglia-related inflammatory biomarkers were assessed. In the study, we found that pinocembrin significantly blocked the declination of SPT percentage and the extension of TST immobility durations in the depression mouse model. Also, we observed that pinocembrin significantly suppressed microglial activation in the hippocampus. Additionally, pinocembrin downregulated hippocampal NLRP3 through P2X7/TLR4 pathway, and also regulated the CUMS-induced imbalance of pro-inflammatory cytokines, including interleukin-1beta, tumor necrosis factor-alpha and interleukin-6. In conclusion, pinocembrin ameliorates CUMS-induced depressive-like behaviors possibly through downregulating P2X7/TLR4 pathway, providing the mechanism of antidepressant treatment.
Collapse
Affiliation(s)
- Jia-Mei Li
- Department of Stress Medicine, Faculty of Psychology, Navy Medical University, Shanghai, China
| | | | | | | |
Collapse
|
12
|
Pre-treatment and continuous administration of simvastatin during sepsis improve metabolic parameters and prevent CNS injuries in survivor rats. Mol Cell Biochem 2022; 477:2657-2667. [DOI: 10.1007/s11010-022-04463-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
|
13
|
Xu C, Zhou G, Wang X, Zhang B, Zhao T, Wu L. Correlation analysis of serum miR-21 and miR-210 with hs-CRP, TNF-α, IL-6, and ICAM-1 in patients with sepsis after burns. Burns 2022; 48:633-638. [PMID: 34272079 DOI: 10.1016/j.burns.2021.05.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/19/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The aim of this study was to explore expression levels and clinical values of miR-21 and miR-210 in patients with sepsis after burns. METHODS One hundred and twenty-eight burn patients who were treated in Binzhou Medical University Hospital were selected as research objects, among which 69 complicated with sepsis were in an observation group and 59 complicated with infection were in a control group. MiR-21 and miR-210 expression in the patients' serum was detected by RT-PCR. Serum inflammatory cytokines were detected by an enzyme-linked immunosorbent assay (ELISA). Correlation analysis was used for the correlations of the miR-21 and miR-210 with the cytokines. Receiver operating characteristic (ROC) curves were plotted to analyze the diagnostic values of the miR-21 and miR-210 for sepsis after burns and their predictive values for the poor prognosis of sepsis patients. RESULTS MiR-21 expression reduced remarkably and miR-210 expression rose remarkably in the serum of patients with sepsis after burns. According to the analysis of the ROC curves, both of the miR-21 and miR-210 had relatively high diagnostic sensitivity for the disease, but the diagnostic value of their combined detection was higher. Contents of hs-CRP, TNF-α, IL-6, and ICAM-1 in serum were remarkably higher in the observation group than those in the control group. According to the correlation analysis, miR-21 was negatively correlated with the expression of the four cytokines, while miR-210 was positively correlated with that. The predictive value of miR-21 for the prognosis of sepsis patients was not high, but miR-210 had a certain predictive value, and their combined detection had a higher value. CONCLUSION In serum of patients with sepsis after burns, miR-21 expression reduces remarkably and miR-210 expression rises. The miR-21 and miR-210 are related to the degree of inflammatory responses in septic patients, and their combined detection has a certain value for diagnosing the disease and predicting its prognosis.
Collapse
Affiliation(s)
- Chuanzhen Xu
- Department of Burn and Plastic Surgery, Binzhou Medical University Hospital, Binzhou 256603, Shandong Province, China
| | - Guojun Zhou
- Department of Emergency Medicine, Binzhou Medical University Hospital, Binzhou 256603, Shandong Province, China
| | - Xiaoyu Wang
- Department of Emergency Medicine, Binzhou Medical University Hospital, Binzhou 256603, Shandong Province, China
| | - Bing Zhang
- Department of Emergency Medicine, Binzhou Medical University Hospital, Binzhou 256603, Shandong Province, China
| | - Tongzhen Zhao
- Department of Oncology, Binzhou Municipal Hospital of Traditional Chinese Medicine, Binzhou 256603, Shandong Province, China
| | - Liqiang Wu
- Department of Emergency Medicine, Binzhou Medical University Hospital, Binzhou 256603, Shandong Province, China.
| |
Collapse
|
14
|
Jiang S, Bai L, Zhang X, Zhou X, Liu Y. Preexposure to heat stress attenuates sepsis-associated inflammation and cognitive decline in rats. Neurosci Lett 2022; 780:136647. [PMID: 35460838 DOI: 10.1016/j.neulet.2022.136647] [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: 11/12/2021] [Revised: 03/16/2022] [Accepted: 04/18/2022] [Indexed: 12/25/2022]
Abstract
Sepsis is a life-threatening organ dysfunction due to an infection, leading to cognitive impairments. Studies have shown that heat shock protein 70 (HSP70) exhibited a neuroprotective effect. In this study we used mild heat stress to induce expression of HSP70, aimimg to detect the effect of HSP70 on neurocognitive deficits associated with sepsis and explored the underlying mechanisms. Male rats were exposed to 42℃ for 15 min. After 12 hours, they were subjected to cecal ligation and puncture (CLP). HSP70 and brain-derived neurotrophic factor (BDNF) expression, nuclear level of NF-level of NF-trophic factor (BDNF)ed by western blot. Levels of inflammatory cytokines in circulation and hippocampus were measured by ELISA and RT-PCR. Neuronal morphology and damage of hippocampal neurons were assessed by Hematoxylin-Eosin (HE) and Nissl stainings. Microglial activation was determined by immunohistochemistry. Finally, neurologic and cognitive functions were evaluated using neurobehavioral scoring and morris water maze (MWM) test. Mild heat stress increased survival rate of sepsis rats. Mild heat stress upregulated HSP70, inhibited nuclear level of NF-κB p65 in hippocampus. Mild heat stress could diminish IL-1β and TNF-α levels in circulation and hippocampus. Furthermore, mild heat stress was able to enhance expression of BDNF and alleviate cognitive impairment after sepsis. Overall, these results indicated that mild heat stress showed protective effects on sepsis-associated encephalopathy rat model, which may be associated with upregulation of HSP70 and inhibition of NF-κB pathway.
Collapse
Affiliation(s)
- Sufang Jiang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China
| | - Long Bai
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China
| | - Xiaoxiao Zhang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China
| | - Xiaoyu Zhou
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China
| | - Ya Liu
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China.
| |
Collapse
|
15
|
Wang H, Wang H, Song Y, Liu C, Qian X, Zhang D, Jiang X, Zhang S. Overexpression of Foxc1 ameliorates sepsis‑associated encephalopathy by inhibiting microglial migration and neuroinflammation through the IκBα/NF‑κB pathway. Mol Med Rep 2022; 25:107. [PMID: 35103290 PMCID: PMC8822881 DOI: 10.3892/mmr.2022.12623] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/30/2021] [Indexed: 12/16/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a common and severe complication of sepsis. The cognitive dysfunction that ensues during SAE has been reported to be caused by impairments of the hippocampus. Microglia serves a key role in neuroinflammation during SAE through migration. Forkhead box C1 (Foxc1) is a member of the forkhead transcription factor family that has been found to regulate in cell migration. However, the role of Foxc1 in neuroinflammation during SAE remains unknown. In the present study, the mechanistic role of Foxc1 on microglial migration, neuroinflammation and neuronal apoptosis during the occurrence of cognitive dysfunction in SAE was investigated. A microglia-mediated inflammation model was induced by LPS in BV-2 microglial cells in vitro, whilst a SAE-related cognitive impairment model was established in mice using cecal ligation and perforation (CLP) surgery. Cognitive function in mice was evaluated using the Morris Water Maze (MWM) trial. Lipopolysaccharide (LPS) treatment was found to trigger BV-2 cell migration, inflammation and neuronal apoptosis. In addition, CLP surgery induced cognitive injury, which was indicated by longer latencies and shorter dwell times in the goal quadrant compared with those in the Sham group in the MWM trial. LPS treatment or CLP induction decreased the expression of Foxc1 and inhibitor of NF-κB (IκΒα) whilst increasing that of p65, IL-1β and TNF-α. After Foxc1 was overexpressed, the cognitive dysfunction of mice that underwent CLP surgery was improved, with the expression of IκBα also increased, microglial cell migration, the expression of p65, IL-1β and TNF-α and neuronal apoptosis were all decreased in vivo and in vitro, which were in turn reversed by the inhibition of IκBα in vitro. Overall, these results suggest that the overexpression of Foxc1 inhibited microglial migration whilst suppressing the inflammatory response and neuronal apoptosis by regulating the IκBα/NF-κB pathway, thereby improving cognitive dysfunction during SAE.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of Critical Care Medicine, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Hongwei Wang
- Department of Respiratory Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Yinsen Song
- Department of Emergency Medicine, People's Hospital of Henan University of Chinese Medicine/Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Congyan Liu
- Department of Critical Care Medicine, The Fifth Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan 450003, P.R. China
| | - Xinling Qian
- Department of Emergency Medicine, People's Hospital of Henan University of Chinese Medicine/Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Dalong Zhang
- Department of Emergency Medicine, People's Hospital of Henan University of Chinese Medicine/Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Xin Jiang
- Department of Emergency Medicine, People's Hospital of Henan University of Chinese Medicine/Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Sisen Zhang
- Department of Critical Care Medicine, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
16
|
Mo Y, Wang L, Ren M, Xie W, Ye X, Zhou B, Zhang A, Dai Q, Wang J. Electroacupuncture prevents LPS- induced neuroinflammation via upregulation of PICK-TLR4 complexes in the microglia of hippocampus. Brain Res Bull 2021; 177:295-304. [PMID: 34673136 DOI: 10.1016/j.brainresbull.2021.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/17/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is a common complication of sepsis caused by neuroinflammation. Electroacupuncture (EA) can be used to treat SAE, but the underlying mechanism is not clear. Lack of PICK1 further aggravates the inflammatory response in mice with sepsis. Therefore, we sought to investigate whether PICK1 is involved in the protective effects of electroacupuncture to SAE. In this study, mice were treated with EA after lipopolysaccharide (LPS) treatment. Behavioral tests; microglial activity of hippocampus; neuron survival and the inflammatory factors PICK1 and TLR4, as well as TLR4-related proteins, such as ERK, JNK, and P38, were assessed after EA treatment. PICK1, TLR4, and TLR4-related proteins, as well as PICK1-TLR4 complex levels were assessed in BV2 cells treated with LPS, PICK1 siRNA, or PICK1 polypeptide. The results indicated that EA could improve neurological assessment and reduce activation of microglial and TLR4 and expression of proinflammatory cytokines. EA also reduced the expression of TLR4 and phosphorylation of ERK/JNK/P38 while, increased the expression of PICK1 and TLR4 complexes. PICK1 knockdown further promoted the expression of TLR4 and phosphorylation of ERK/JNK/P38 in BV2 cells, but this effect was reversed by PICK1 polypeptides. These results suggest that EA may reduce neuroinflammation responses, decrease inflammatory factors, and finally, protect SAE by increasing the formation of PICK1-TLR4 complexes in microglia.
Collapse
Affiliation(s)
- Yunchang Mo
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lu Wang
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Miao Ren
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenjing Xie
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoxiao Ye
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bingbing Zhou
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Anqi Zhang
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qinxue Dai
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junlu Wang
- The department of Anesthesiology and Operation Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
17
|
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
|
18
|
Effects of S100B neutralization on the long-term cognitive impairment and neuroinflammatory response in an animal model of sepsis. Neurochem Int 2020; 142:104906. [PMID: 33232757 DOI: 10.1016/j.neuint.2020.104906] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/05/2020] [Accepted: 11/15/2020] [Indexed: 01/11/2023]
Abstract
The nervous system is one of the first systems to be affected during sepsis. Sepsis not only has a high risk of mortality, but could also lead to cerebral dysfunction and cognitive impairment in long-term survival patients. The receptor for advanced glycation end products (RAGE) can interact with several ligands, and its activation triggers a series of cell signaling events, resulting in the hyperinflammatory condition related to sepsis. Recent studies show that elevated levels of S100B (RAGE ligand) are associated with the pathophysiology of neurodegenerative disorders. They also participate in inflammatory brain diseases and may lead to an increased activation of microglia and astrocytes, leading to neuronal death. This study aimed to determine the effect of S100B inhibition on the neuroinflammatory response in sepsis. Sepsis was induced in Wistar rats by cecal ligation and perforation (CLP). There were three groups: Sham, CLP, and CLP +10 μg/kg of monoclonal antibody (Anti-S100B) administered intracerebroventricularly. The animals were killed 30 days after sepsis following behavioral evaluation by open field, novel object recognition, and splash test. The hippocampus, prefrontal cortex, and amydgala were used for the determination of S100B and RAGE proteins by western blotting and for the evaluation of cytokine levels and verification of the number of microglial cells by immunohistochemistry. On day 30, both the Sham and CLP + anti-S100B groups were capable of recovering the habitual memory in the open field task. Regarding novel object recognition, Sham and CLP + anti-S100B groups increased the recognition index during the test session in comparison to the training session. There was a significant increase in the time of grooming in CLP + anti-S100B in comparison to the CLP group. There was a modulation of cytokine levels and immunohistochemistry showed that the CLP + anti-S100B group had a decrease in the number of microglial cells only in the hippocampus. These results helped to understand the role of S100B protein in the pathophysiology of sepsis-associated encephalopathy and could be helpful to further experimental studies regarding this subject.
Collapse
|