1
|
Denver P, Cunningham C. Microglial activation and neuroinflammation in acute and chronic cognitive deficits in sepsis. Neuropharmacology 2025; 267:110285. [PMID: 39746541 DOI: 10.1016/j.neuropharm.2024.110285] [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: 09/20/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Sepsis is characterised by dysregulated immune responses to infection, leading to multi-organ dysfunction and high rates of mortality. With increasing survival rates in recent years long-term neurological and psychiatric consequences have become more apparent in survivors. Many patients develop sepsis associated encephalopathy (SAE) which encompasses the profound but usually transient neuropsychiatric syndrome delirium but also new brain injury that emerges in the months and years post-sepsis. It is now clear that systemic inflammatory signals reach the brain during sepsis and that very significant neuroinflammation ensues. The major brain resident immune cell population, the microglia, has been implicated in acute and chronic cognitive dysfunction in animal models of sepsis based on a growing number of studies using bacterial endotoxin and in polymicrobial sepsis models such as cecal ligation and puncture. The current review explores the effects of sepsis on the brain, focussing on how systemic insults translate to microglial activation and neuroinflammation and how this disrupts neuronal function and integrity. We examine what has been demonstrated specifically with respect to microglial activation, revealing robust evidence for a role for neuroinflammation in sepsis-induced brain sequelae but less clear information on the extent of the specific microglial contribution to this, arising from findings using global knockout mice, non-selective drugs and treatments that equally target peripheral and central compartments. There is, nonetheless, clear evidence that microglia do become activated and do contribute to brain consequences of sepsis thus arguing for improved understanding of these neuroinflammatory processes toward the prevention and treatment of sepsis-induced brain dysfunction.
Collapse
Affiliation(s)
- Paul Denver
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland.
| |
Collapse
|
2
|
Zhu Y, Li J, Dai L, Feng W. Ginsenoside Rh2 Alleviate Sepsis-related Encephalopathy via Up-regulating Nrf2/HO-1 Pathway and Apoptosis Inhibition. Cell Biochem Biophys 2025; 83:587-597. [PMID: 39187743 DOI: 10.1007/s12013-024-01488-2] [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] [Accepted: 08/09/2024] [Indexed: 08/28/2024]
Abstract
Sepsis patients are highly prone to sepsis-associated encephalopathy (SAE) complications, resulting in a high mortality rate. Recently, there has been no specific treatment for long-term improvement of cerebral function. Ginsenoside Rh2 is a form of steroidal saponins isolated from plant ginseng and has been shown to possess anti-inflammatory as well as neuroprotective characteristics; yet, the effect of ginsenoside Rh2 on SAE treatment is obscure. Accordingly, we proposed to investigate the effect of ginsenoside Rh2 in alleviating SAE damage. We established and utilized the SAE mouse model to determine the effect of Rh2 treatment on alleviating SAE. We determined the expression levels of Heme oxygenase-1(HO-1) and Nuclear factor erythroid 2-related factor 2 (Nrf2) as well as measured neural apoptosis by flow cytometry. Also, we quantified the levels of caspase-3, malondialdehyde (MDA), GSH-Px superoxide dismutase (SOD) and evaluated the animals' neural reflex function. First, used Rh2 to treat microglia BV2 and mouse neuron MN-c whether LPS exist or not, and then measured expression level of Iba-1, apoptotic rate, and ROS content applying flow cytometry. Also, we quantified the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). In comparison with the Sham group, the SAE model exhibited an elevated MDA content, caspase-3 activity, and cell apoptosis. On the other hand, the GSH-Px activity and SOD level were decreased along with a decreased neural reflex score. Our investigation concluded that Rh2 treatment significantly alleviated SAE damage and inhibited LPS-induced response via up-regulation of the Nrf2/HO-1 pathway to promote anti-oxidative stress capacity and inhibit neural cell apoptosis.
Collapse
Affiliation(s)
- Yufeng Zhu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Jinhang Li
- Department of Hemodialysis Room, Shidong Hospital of Yangpu District, Shanghai, 200082, China
| | - Lijun Dai
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Wei Feng
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| |
Collapse
|
3
|
Chourpiliadis C, Lovik A, Seitz C, Hu Y, Wu J, Ljungman P, Press R, Samuelsson K, Ingre C, Fang F. Association between cardiometabolic diseases and the risk and progression of motor neuron diseases in Sweden: a population-based case-control study. THE LANCET REGIONAL HEALTH. EUROPE 2025; 49:101173. [PMID: 39759580 PMCID: PMC11697398 DOI: 10.1016/j.lanepe.2024.101173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025]
Abstract
Background The evidence on the link between cardiometabolic diseases (CMDs) and motor neuron diseases (MNDs) remains inconsistent. We aimed to determine whether there is an association of CMDs, namely, any cardiovascular disease, cardiac arrhythmia, heart failure, thromboembolic disease, hypertension, cerebrovascular disease, ischemic heart disease, diabetes mellitus type 2, and hypercholesterolemia with the risk and progression of MNDs. Methods We included 1463 MND patients (amyotrophic lateral sclerosis (ALS), primary lateral sclerosis (PLS), progressive spinal muscular atrophy (PSMA), and unspecified MND) diagnosed from January 1, 2015, to July 1, 2023, in Sweden according to the Swedish Motor Neuron Disease Quality Registry (i.e., cases), up to 5 MND-free population controls per case (N = 7311) who were individually matched to the cases on age and sex, and the full siblings (N = 2002) and spouses (N = 1220) of MND patients (i.e., relative controls). Conditional logistic regression models were used to estimate the risk of MND diagnosis in relation to previous CMDs, through comparing MND patients to population controls or relative controls. MND patients were followed from diagnosis to assess the role of pre-diagnostic CMDs on disease progression. A joint longitudinal-survival model was used to estimate risk of mortality (or use of invasive ventilation) in relation to CMDs after taking into account the longitudinal changes of ALS functional rating scale-revised (ALSFRS-R) in the time-to-event analysis. Hierarchical clustering with the Ward's linkage and a dissimilarity matrix created by Gower's method was used to identify clusters of MND patients with distinct phenotypes. Findings Among the CMDs studied, a history of diabetes mellitus type 2 (OR 0.75; 95% CI 0.62, 0.93) or hypercholesterolemia (OR 0.82; 95% CI 0.71, 0.94) more than one year before diagnosis was associated with a lower risk for MNDs. The associations persisted for more than five years before MND diagnosis. MND patients with a history of any cardiovascular disease (HR 1.43; 95% CI 1.13, 1.81), arrhythmia (HR 1.42; 95% CI 1.04, 1.93), heart failure (HR 1.79; 95% CI 1.02, 3.14), hypertension (HR 1.41; 95% CI 1.12, 1.77), or hypercholesterolemia (HR 1.28; 95% CI 1.01, 1.62) had an increased mortality risk, compared to others, after taking into consideration the longitudinal changes in ALSFRS-R. Cluster analysis identified two clusters of MND patients, where one cluster demonstrated higher age, worse functional status, and higher prevalence of CMDs at the time of diagnosis as well as a higher mortality and faster functional decline during follow-up, compared to the ones included in the other cluster. Interpretation Diabetes mellitus type 2 and hypercholesterolemia were associated with a lower future risk of MND. On the other hand, most of the CMDs were indicative of a poor disease progression after an MND diagnosis. Funding European Research Council, US Center for Disease Control and Prevention, Swedish Research Council.
Collapse
Affiliation(s)
| | - Anikó Lovik
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Institute of Psychology, Leiden University, Leiden, the Netherlands
| | - Christina Seitz
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yihan Hu
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jing Wu
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Petter Ljungman
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology and Clinical Physiology, Danderyd Hospital, Stockholm, Sweden
| | - Rayomand Press
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Kristin Samuelsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Ingre
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Dai M, Sun S, Dai Y, Dou X, Yang J, Chen X, Yang D, Lin Y. Maresin-1 Ameliorates Sepsis-Induced Microglial Activation Through Modulation of the P38 MAPK Pathway. Neurochem Res 2024; 50:26. [PMID: 39565476 DOI: 10.1007/s11064-024-04280-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/10/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024]
Abstract
Sepsis is a life-threatening disease characterized by a dysregulated immune response to infection, often leading to neuroinflammation. As a known immunomodulator, Maresin-1 (MaR1) may have potential applications in the treatment of sepsis-induced neuroinflammation, but its effects in this context are unknown. We used a mouse cecum ligation and puncture (CLP)-induced sepsis model and an in vitro lipopolysaccharide (LPS)-induced neuroinflammatory model of BV2 microglia. Expression of microglial cell markers (IBA1, CD11B, CD68, CD86 and CD206) and pro-inflammatory markers (iNOS and COX2) was assessed. The role of MaR1 in regulating the P38 MAPK pathway was explored using the P38 MAPK inhibitor SB203580. In the CLP model, an increased proportion of M1-type microglia was observed, and MaR1 was able to reverse it. However, the combination of SB203580 and MaR1 did not enhance the therapeutic effect compared to SB20580 alone. In vitro experiments, MaR1 inhibited LPS-induced P38 MAPK nuclear translocation and decreased the expression of pro-inflammatory markers such as iNOS and COX2. As with the animal results, no stacking effect could be obtained with the co-administration of SB203580 and MaR1. Our findings suggest that MaR1 attenuates sepsis-induced neuroinflammation mainly by inhibiting phosphorylation of P38 MAPK in microglial cells. This suggests that MaR1 may have a potential therapeutic role in the treatment of sepsis neuroinflammation.
Collapse
Affiliation(s)
- Maosha Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Yan Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiaoke Dou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Juexi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| | - Dong Yang
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
5
|
Wang P, Liang L, Ge Q, Liu S, Yang Z, Jiang L. Dichloroacetate attenuates brain injury through inhibiting neuroinflammation and mitochondrial fission in a rat model of sepsis-associated encephalopathy. Int Immunopharmacol 2024; 140:112840. [PMID: 39106713 DOI: 10.1016/j.intimp.2024.112840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/09/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is a severe complication of sepsis, characterized by neuroinflammation, mitochondrial dysfunction, and oxidative stress, leading to cognitive decline and high mortality. The effectiveness of dichloroacetate (DCA) in modulating mitochondrial function provides a novel therapeutic strategy for SAE. In this study, we evaluated the neuroprotective effects of DCA in a rat model of SAE induced by cecal ligation and puncture (CLP). Rats treated with DCA exhibited significant improvements in neurological function and survival, as evidenced by less neuron loss from histopathologic analysis, restored neurologic deficit scores, improved Y-maze alternation percentages, and enhanced recognition index performance. Biochemical analyses showed that DCA administration at 25 mg/kg and 100 mg/kg reduced astrocyte and microglial activation, indicating reduced neuroinflammation. Furthermore, DCA simultaneously reduced the production of circulating and cerebral inflammatory cytokines (including TNF-α, IL-1β, and IL-10), concomitant with mitigating oxidative stress through down-regulating expression of 8-Hydroxy-2'-deoxyguanosine (8-OHdG) and reactive oxygen species (ROS) in the brain. Mechanistically, DCA modulated mitochondrial dynamics by suppressing Drp1 and pDrp1 expression, which are indicators of mitochondrial fission. This was corroborated by transmission electron microscopy, quantification of mitochondrial area, and Western blot analyses. Furthermore, DCA treatment improved ATP levels, mitochondrial complex I activity, and NAD+/NADH ratio, indicating a significant attenuation of brain mitochondrial dysfunction. In conclusion, our findings suggest that DCA confers neuroprotection in SAE by curtailing neuroinflammation and mitochondrial fission, outlining a promising therapeutic strategy for treating SAE in critically ill patients.
Collapse
Affiliation(s)
- Peng Wang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou 510120, China
| | - Lian Liang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou 510120, China
| | - Qiulin Ge
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou 510120, China
| | - Siqi Liu
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhengfei Yang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou 510120, China.
| | - Longyuan Jiang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
6
|
Kuo PC, Weng WT, Scofield BA, Paraiso HC, Yu ICI, Yen JHJ. Ischemia-induced endogenous Nrf2/HO-1 axis activation modulates microglial polarization and restrains ischemic brain injury. Front Immunol 2024; 15:1440592. [PMID: 39469715 PMCID: PMC11513276 DOI: 10.3389/fimmu.2024.1440592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024] Open
Abstract
Cerebral ischemic stroke accounts for more than 80% of all stroke cases. During cerebral ischemia, reactive oxygen species produced in the ischemic brain induce oxidative stress and inflammatory responses. Nrf2 is a transcription factor responsible for regulating cellular redox balance through the induction of protective antioxidant and phase II detoxification responses. Although the induction of endogenous Nrf2/HO-1 axis activation has been observed in the ischemic brain, whether ischemia-induced endogenous Nrf2/HO-1 axis activation plays a role in modulating microglia (MG) phenotypes and restraining ischemic brain injury is not characterized and requires further exploration. To investigate that, we generated mice with Nrf2 knockdown specifically in MG to rigorously assess the role of endogenous Nrf2 activation in ischemic brain injury after stroke. Our results showed that MG-specific Nrf2 knockdown exacerbated ischemic brain injury after stroke. We found that Nrf2 knockdown altered MG phenotypes after stroke, in which increased frequency of inflammatory MG and decreased frequency of anti-inflammatory MG were detected in the ischemic brain. Moreover, we identified attenuated Nrf2/HO-1 axis activation led to increased CD68/IL-1β and suppressed CD206 expression in MG, resulting in aggravated inflammatory MG in MG-specific Nrf2 knockdown mice after stroke. Intriguingly, using type II diabetic preclinical models, we revealed that diabetic mice exhibited attenuated Nrf2/HO-1 axis activation in MG and exacerbated ischemic brain injury after stroke that phenocopy mice with MG-specific Nrf2 knockdown. Finally, the induction of exogenous Nrf2/HO-1 axis activation in MG through pharmacological approaches ameliorated ischemic brain injury in diabetic mice. In conclusion, our findings provide cellular and molecular insights demonstrating ischemia-induced endogenous Nrf2/HO-1 axis activation modulates MG phenotypes and restrains ischemic brain injury. These results further strengthen the therapeutic potential of targeting Nrf2/HO-1 axis in MG for the treatment of ischemic stroke and diabetic stroke.
Collapse
Affiliation(s)
- Ping-Chang Kuo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Wen-Tsan Weng
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Barbara A. Scofield
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Hallel C. Paraiso
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - I-Chen Ivorine Yu
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Jui-Hung Jimmy Yen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| |
Collapse
|
7
|
Zhou D, Wang W, Gu J, Lu Q. Causal effects of sepsis on structural changes in cerebral cortex: A Mendelian randomization investigation. Medicine (Baltimore) 2024; 103:e39404. [PMID: 39252275 PMCID: PMC11383497 DOI: 10.1097/md.0000000000039404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/01/2024] [Indexed: 09/11/2024] Open
Abstract
Previous research has shown a strong correlation between sepsis and brain structure. However, whether this relationship represents a causality remains elusive. In this study, we employed Mendelian randomization (MR) to probe the associations of genetically predicted sepsis and sepsis-related death with structural changes in specific brain regions. Genome-wide association study (GWAS) data for sepsis phenotypes (sepsis and sepsis-related death) were obtained from the IEU OpenGWAS. Correspondingly, GWAS data for brain structural traits (volume of the subcortical structure, cortical thickness, and surface area) were derived from the ENIGMA consortium. Inverse variance weighted was mainly utilized to assess the causal effects, while weighted median and MR-Egger regression served as complementary methods. Sensitivity analyses were implemented with Cochran Q test, MR-Egger regression, and MR-PRESSO. In addition, a reverse MR analysis was carried out to assess the possibility of reverse causation. We identified that genetic liability to sepsis was normally significantly associated with a reduced surface area of the postcentral gyrus (β = -35.5280, SE = 13.7465, P = .0096). The genetic liability to sepsis-related death showed a suggestive positive correlation with the surface area of fusiform gyrus (β = 11.0920, SE = 3.6412, P = .0023) and posterior cingulate gyrus (β = 3.6530, SE = 1.6684, P = .0286), While it presented a suggestive negative correlation with surface area of the caudal middle frontal gyrus (β = -11.4586, SE = 5.1501, P = .0261) and frontal pole (β = -1.0024, SE = 0.4329, P = .0206). We also indicated a possible bidirectional causal association between genetic liability to sepsis-related death and the thickness of the transverse temporal gyrus. Sensitivity analyses verified the robustness of the above associations. These findings suggested that genetically determined liability to sepsis might influence the specific brain structure in a causal way, offering new perspectives to investigate the mechanism of sepsis-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Dengfeng Zhou
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Weina Wang
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Jiaying Gu
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Qiaofa Lu
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| |
Collapse
|
8
|
Liang N, Li Y, Yuan C, Zhong X, Yang Y, Liang F, Zhao K, Yuan F, Shi J, Wang E, Zhong Y, Tian G, Lu B, Tang Y. Caspase-11 signaling promotes damage to hippocampal CA3 to enhance cognitive dysfunction in infection. Mol Med 2024; 30:127. [PMID: 39179968 PMCID: PMC11342519 DOI: 10.1186/s10020-024-00891-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Cognitive dysfunction caused by infection frequently emerges as a complication in sepsis survivor patients. However, a comprehensive understanding of its pathogenesis remains elusive. METHODS In our in vivo experiments, an animal model of endotoxemia was employed, utilizing the Novel Object Recognition Test and Morris Water Maze Test to assess cognitive function. Various techniques, including immunofluorescent staining, Western blotting, blood‒brain barrier permeability assessment, Limulus Amebocyte Lysate (LAL) assay, and Proximity-ligation assay, were employed to identify brain pathological injury and neuroinflammation. To discern the role of Caspase-11 (Casp11) in hematopoietic or non-hematopoietic cells in endotoxemia-induced cognitive decline, bone marrow chimeras were generated through bone marrow transplantation (BMT) using wild-type (WT) and Casp11-deficient mice. In vitro studies involved treating BV2 cells with E. coli-derived outer membrane vesicles to mimic in vivo conditions. RESULTS Our findings indicate that the deficiency of Casp11-GSDMD signaling pathways reverses infection-induced cognitive dysfunction. Moreover, cognitive dysfunction can be ameliorated by blocking the IL-1 effect. Mechanistically, the absence of Casp11 signaling significantly mitigated blood‒brain barrier leakage, microglial activation, and synaptic damage in the hippocampal CA3 region, ultimately leading to improved cognitive function. CONCLUSION This study unveils the crucial contribution of Casp11 and GSDMD to cognitive impairments and spatial memory loss in a murine sepsis model. Targeting Casp11 signaling emerges as a promising strategy for preventing or treating cognitive dysfunction in patients with severe infections.
Collapse
Affiliation(s)
- Ni Liang
- Department of Critical Care Medicine and Hematology, The 3Rd Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Yi Li
- Department of Critical Care Medicine and Hematology, The 3Rd Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Chuang Yuan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410000, People's Republic of China
| | - Xiaoli Zhong
- ICU Center, The Second Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Yanliang Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410000, People's Republic of China
| | - Fang Liang
- Department of Critical Care Medicine and Hematology, The 3Rd Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Kai Zhao
- Department of Critical Care Medicine and Hematology, The 3Rd Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Fangfang Yuan
- Department of Critical Care Medicine and Hematology, The 3Rd Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Jian Shi
- Department of Critical Care Medicine and Hematology, The 3Rd Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Erhua Wang
- Department of Critical Care Medicine and Hematology, The 3Rd Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Yanjun Zhong
- ICU Center, The Second Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
| | - Guixiang Tian
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410000, People's Republic of China
| | - Ben Lu
- Department of Critical Care Medicine and Hematology, The 3Rd Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
- Hunan Key Laboratory of Organ Fibrosis, Xiangya Hospital, Central South University, Changsha, 410000, People's Republic of China
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan Province, 410000, People's Republic of China
| | - Yiting Tang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410000, People's Republic of China.
| |
Collapse
|
9
|
Che J, Sun Y, Deng Y, Zhang J. Blood-brain barrier disruption: a culprit of cognitive decline? Fluids Barriers CNS 2024; 21:63. [PMID: 39113115 PMCID: PMC11305076 DOI: 10.1186/s12987-024-00563-3] [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: 01/14/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Cognitive decline covers a broad spectrum of disorders, not only resulting from brain diseases but also from systemic diseases, which seriously influence the quality of life and life expectancy of patients. As a highly selective anatomical and functional interface between the brain and systemic circulation, the blood-brain barrier (BBB) plays a pivotal role in maintaining brain homeostasis and normal function. The pathogenesis underlying cognitive decline may vary, nevertheless, accumulating evidences support the role of BBB disruption as the most prevalent contributing factor. This may mainly be attributed to inflammation, metabolic dysfunction, cell senescence, oxidative/nitrosative stress and excitotoxicity. However, direct evidence showing that BBB disruption causes cognitive decline is scarce, and interestingly, manipulation of the BBB opening alone may exert beneficial or detrimental neurological effects. A broad overview of the present literature shows a close relationship between BBB disruption and cognitive decline, the risk factors of BBB disruption, as well as the cellular and molecular mechanisms underlying BBB disruption. Additionally, we discussed the possible causes leading to cognitive decline by BBB disruption and potential therapeutic strategies to prevent BBB disruption or enhance BBB repair. This review aims to foster more investigations on early diagnosis, effective therapeutics, and rapid restoration against BBB disruption, which would yield better cognitive outcomes in patients with dysregulated BBB function, although their causative relationship has not yet been completely established.
Collapse
Affiliation(s)
- Ji Che
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yinying Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yixu Deng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
10
|
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
|
11
|
Zhu Q, Wan L, Huang H, Liao Z. IL-1β, the first piece to the puzzle of sepsis-related cognitive impairment? Front Neurosci 2024; 18:1370406. [PMID: 38665289 PMCID: PMC11043581 DOI: 10.3389/fnins.2024.1370406] [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: 01/14/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a leading cause of death resulting from an uncontrolled inflammatory response to an infectious agent. Multiple organ injuries, including brain injuries, are common in sepsis. The underlying mechanism of sepsis-associated encephalopathy (SAE), which is associated with neuroinflammation, is not yet fully understood. Recent studies suggest that the release of interleukin-1β (IL-1β) following activation of microglial cells plays a crucial role in the development of long-lasting neuroinflammation after the initial sepsis episode. This review provides a comprehensive analysis of the recent literature on the molecular signaling pathways involved in microglial cell activation and interleukin-1β release. It also explores the physiological and pathophysiological role of IL-1β in cognitive function, with a particular focus on its contribution to long-lasting neuroinflammation after sepsis. The findings from this review may assist healthcare providers in developing novel interventions against SAE.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Wan
- Department of Medical Genetics/Prenatal Diagnostic Center Nursing and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhimin Liao
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Tian M, Zhan Y, Cao J, Gao J, Sun J, Zhang L. Targeting blood-brain barrier for sepsis-associated encephalopathy: Regulation of immune cells and ncRNAs. Brain Res Bull 2024; 209:110922. [PMID: 38458135 DOI: 10.1016/j.brainresbull.2024.110922] [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: 10/21/2023] [Revised: 02/14/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Sepsis causes significant morbidity and mortality worldwide, most surviving patients show acute or chronic mental disorders, which are known as sepsis-associated encephalopathy (SAE). SAE involves many pathological processes, including the blood-brain barrier (BBB) damage. The BBB is located at the interface between the central nervous system and the surrounding environment, which protects the central nervous system (CNS) from the invasion of exogenous molecules, harmful substances or microorganisms in the blood. Recently, a growing number of studies have indicated that the BBB destruction was involved in SAE and played an important role in SAE-induced brain injury. In the present review, we firstly reveal the pathological processes of SAE such as the neurotransmitter disorders, oxidative stress, immune dysfunction and BBB destruction. Moreover, we introduce the structure of BBB, and describe the immune cells including microglia and astrocytes that participate in the BBB destruction after SAE. Furthermore, in view of the current research on non-coding RNAs (ncRNAs), we explain the regulatory mechanism of ncRNAs including long noncoding RNAs (lncRNAs), microRNAs (miRNAs) and circular RNAs (circRNAs) on BBB in the processes of SAE. Finally, we propose some challenges and perspectives of regulating BBB functions in SAE. Hence, on the basis of these effects, both immune cells and ncRNAs may be developed as therapeutic targets to protect BBB for SAE patients.
Collapse
Affiliation(s)
- Mi Tian
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, China
| | - Yunliang Zhan
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jinyuan Cao
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, China
| | - Jinqi Gao
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, China
| | - Jie Sun
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, China.
| | - Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
13
|
Mayer MG, Fischer T. Microglia at the blood brain barrier in health and disease. Front Cell Neurosci 2024; 18:1360195. [PMID: 38550920 PMCID: PMC10976855 DOI: 10.3389/fncel.2024.1360195] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/23/2024] [Indexed: 01/24/2025] Open
Abstract
The blood brain barrier (BBB) plays a crucial role in maintaining brain homeostasis by selectively preventing the entry of substances from the peripheral blood into the central nervous system (CNS). Comprised of endothelial cells, pericytes, and astrocytes, this highly regulated barrier encompasses the majority of the brain's vasculature. In addition to its protective function, the BBB also engages in significant crosstalk with perivascular macrophages (MΦ) and microglia, the resident MΦ of the brain. These interactions play a pivotal role in modulating the activation state of cells comprising the BBB, as well as MΦs and microglia, themselves. Alterations in systemic metabolic and inflammatory states can promote endothelial cell dysfunction, reducing the integrity of the BBB and potentially allowing peripheral blood factors to leak into the CNS compartment. This may mediate activation of perivascular MΦs, microglia, and astrocytes, and initiate further immune responses within the brain parenchyma, suggesting neuroinflammation can be triggered by signaling from the periphery, without primary injury or disease originating within the CNS. The intricate interplay between the periphery and the CNS through the BBB highlights the importance of understanding the role of microglia in mediating responses to systemic challenges. Despite recent advancements, our understanding of the interactions between microglia and the BBB is still in its early stages, leaving a significant gap in knowledge. However, emerging research is shedding light on the involvement of microglia at the BBB in various conditions, including systemic infections, diabetes, and ischemic stroke. This review aims to provide a comprehensive overview of the current research investigating the intricate relationship between microglia and the BBB in health and disease. By exploring these connections, we hope to advance our understanding of the role of brain immune responses to systemic challenges and their impact on CNS health and pathology. Uncovering these interactions may hold promise for the development of novel therapeutic strategies for neurological conditions that involve immune and vascular mechanisms.
Collapse
Affiliation(s)
- Meredith G. Mayer
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Tracy Fischer
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
14
|
Radke J, Meinhardt J, Aschman T, Chua RL, Farztdinov V, Lukassen S, Ten FW, Friebel E, Ishaque N, Franz J, Huhle VH, Mothes R, Peters K, Thomas C, Schneeberger S, Schumann E, Kawelke L, Jünger J, Horst V, Streit S, von Manitius R, Körtvélyessy P, Vielhaber S, Reinhold D, Hauser AE, Osterloh A, Enghard P, Ihlow J, Elezkurtaj S, Horst D, Kurth F, Müller MA, Gassen NC, Melchert J, Jechow K, Timmermann B, Fernandez-Zapata C, Böttcher C, Stenzel W, Krüger E, Landthaler M, Wyler E, Corman V, Stadelmann C, Ralser M, Eils R, Heppner FL, Mülleder M, Conrad C, Radbruch H. Proteomic and transcriptomic profiling of brainstem, cerebellum and olfactory tissues in early- and late-phase COVID-19. Nat Neurosci 2024; 27:409-420. [PMID: 38366144 DOI: 10.1038/s41593-024-01573-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/08/2024] [Indexed: 02/18/2024]
Abstract
Neurological symptoms, including cognitive impairment and fatigue, can occur in both the acute infection phase of coronavirus disease 2019 (COVID-19) and at later stages, yet the mechanisms that contribute to this remain unclear. Here we profiled single-nucleus transcriptomes and proteomes of brainstem tissue from deceased individuals at various stages of COVID-19. We detected an inflammatory type I interferon response in acute COVID-19 cases, which resolves in the late disease phase. Integrating single-nucleus RNA sequencing and spatial transcriptomics, we could localize two patterns of reaction to severe systemic inflammation, one neuronal with a direct focus on cranial nerve nuclei and a separate diffuse pattern affecting the whole brainstem. The latter reflects a bystander effect of the respiratory infection that spreads throughout the vascular unit and alters the transcriptional state of mainly oligodendrocytes, microglia and astrocytes, while alterations of the brainstem nuclei could reflect the connection of the immune system and the central nervous system via, for example, the vagus nerve. Our results indicate that even without persistence of severe acute respiratory syndrome coronavirus 2 in the central nervous system, local immune reactions are prevailing, potentially causing functional disturbances that contribute to neurological complications of COVID-19.
Collapse
Affiliation(s)
- Josefine Radke
- Institute of Pathology, Universitätsmedizin Greifswald, Greifswald, Germany.
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Jenny Meinhardt
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tom Aschman
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Lorenz Chua
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Vadim Farztdinov
- Core Facility High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sören Lukassen
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Foo Wei Ten
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ekaterina Friebel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Naveed Ishaque
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jonas Franz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Valerie Helena Huhle
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ronja Mothes
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kristin Peters
- Institute of Pathology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Carolina Thomas
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Shirin Schneeberger
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elisa Schumann
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Leona Kawelke
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Jünger
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Viktor Horst
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simon Streit
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Regina von Manitius
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Péter Körtvélyessy
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Vielhaber
- Department of Neurology, Otto von Guerike University Magdeburg, Magdeburg, Germany
| | - Dirk Reinhold
- Institute of Molecular and Clinical Immunology, Otto von Guerike University Magdeburg, Magdeburg, Germany
| | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Berlin, Germany
| | - Anja Osterloh
- Department of Pathology, University Medical Center Ulm, Ulm, Germany
| | - Philipp Enghard
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jana Ihlow
- Department of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sefer Elezkurtaj
- Department of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David Horst
- Department of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel A Müller
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils C Gassen
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Julia Melchert
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Jechow
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Camila Fernandez-Zapata
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Chotima Böttcher
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Markus Landthaler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institut für Biologie, Humboldt Universität, Berlin, Germany
| | - Emanuel Wyler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Victor Corman
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Centre for Infection Research (DZIF), associated partner, Berlin, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Markus Ralser
- Core Facility High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Eils
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank L Heppner
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Cluster of Excellence NeuroCure, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Conrad
- Center of Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Helena Radbruch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
15
|
Tuckey AN, Brandon A, Eslaamizaad Y, Siddiqui W, Nawaz T, Clarke C, Sutherland E, Williams V, Spadafora D, Barrington RA, Alvarez DF, Mulekar MS, Simmons JD, Fouty BW, Audia JP. Amyloid-β and caspase-1 are indicators of sepsis and organ injury. ERJ Open Res 2024; 10:00572-2023. [PMID: 38410714 PMCID: PMC10895426 DOI: 10.1183/23120541.00572-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/04/2023] [Indexed: 02/28/2024] Open
Abstract
Background Sepsis is a life-threatening condition that results from a dysregulated host response to infection, leading to organ dysfunction. Despite the prevalence and associated socioeconomic costs, treatment of sepsis remains limited to antibiotics and supportive care, and a majority of intensive care unit (ICU) survivors develop long-term cognitive complications post-discharge. The present study identifies a novel regulatory relationship between amyloid-β (Aβ) and the inflammasome-caspase-1 axis as key innate immune mediators that define sepsis outcomes. Methods Medical ICU patients and healthy individuals were consented for blood and clinical data collection. Plasma cytokine, caspase-1 and Aβ levels were measured. Data were compared against indices of multiorgan injury and other clinical parameters. Additionally, recombinant proteins were tested in vitro to examine the effect of caspase-1 on a functional hallmark of Aβ, namely aggregation. Results Plasma caspase-1 levels displayed the best predictive value in discriminating ICU patients with sepsis from non-infected ICU patients (area under the receiver operating characteristic curve=0.7080). Plasma caspase-1 and the Aβ isoform Aβx-40 showed a significant positive correlation and Aβx-40 associated with organ injury. Additionally, Aβ plasma levels continued to rise from time of ICU admission to 7 days post-admission. In silico, Aβ harbours a predicted caspase-1 cleavage site, and in vitro studies demonstrated that caspase-1 cleaved Aβ to inhibit its auto-aggregation, suggesting a novel regulatory relationship. Conclusions Aβx-40 and caspase-1 are potentially useful early indicators of sepsis and its attendant organ injury. Additionally, Aβx-40 has emerged as a potential culprit in the ensuing development of post-ICU syndrome.
Collapse
Affiliation(s)
- Amanda N. Tuckey
- Department of Microbiology and Immunology, University of South Alabama College of Medicine
- Center for Lung Biology, University of South Alabama College of Medicine
| | - Arcole Brandon
- Center for Lung Biology, University of South Alabama College of Medicine
| | - Yasaman Eslaamizaad
- Department of Internal Medicine, University of South Alabama College of Medicine
- Division of Pulmonary and Critical Care Medicine, University of South Alabama College of Medicine
| | - Waqar Siddiqui
- Department of Internal Medicine, University of South Alabama College of Medicine
- Division of Pulmonary and Critical Care Medicine, University of South Alabama College of Medicine
| | - Talha Nawaz
- Department of Internal Medicine, University of South Alabama College of Medicine
- Division of Pulmonary and Critical Care Medicine, University of South Alabama College of Medicine
| | - Christopher Clarke
- Department of Internal Medicine, University of South Alabama College of Medicine
- Division of Pulmonary and Critical Care Medicine, University of South Alabama College of Medicine
| | - Erica Sutherland
- Department of Internal Medicine, University of South Alabama College of Medicine
| | - Veronica Williams
- Department of Laboratory Medicine, University of South Alabama University Hospital
| | - Domenico Spadafora
- Flow Cytometry Shared Resources Laboratory, University of South Alabama College of Medicine
| | - Robert A. Barrington
- Department of Microbiology and Immunology, University of South Alabama College of Medicine
- Center for Lung Biology, University of South Alabama College of Medicine
- Flow Cytometry Shared Resources Laboratory, University of South Alabama College of Medicine
| | - Diego F. Alvarez
- Center for Lung Biology, University of South Alabama College of Medicine
- Department of Internal Medicine, University of South Alabama College of Medicine
- Department of Pharmacology College of Medicine, University of South Alabama College of Medicine
| | - Madhuri S. Mulekar
- Department of Mathematics and Statistics, University of South Alabama College of Arts and Sciences
| | - Jon D. Simmons
- Center for Lung Biology, University of South Alabama College of Medicine
- Department of Pharmacology College of Medicine, University of South Alabama College of Medicine
- Department of Surgery, University of South Alabama College of Medicine
| | - Brian W. Fouty
- Center for Lung Biology, University of South Alabama College of Medicine
- Department of Internal Medicine, University of South Alabama College of Medicine
- Division of Pulmonary and Critical Care Medicine, University of South Alabama College of Medicine
- Department of Pharmacology College of Medicine, University of South Alabama College of Medicine
| | - Jonathon P. Audia
- Department of Microbiology and Immunology, University of South Alabama College of Medicine
- Center for Lung Biology, University of South Alabama College of Medicine
| |
Collapse
|
16
|
Hu J, Xie S, Zhang H, Wang X, Meng B, Zhang L. Microglial Activation: Key Players in Sepsis-Associated Encephalopathy. Brain Sci 2023; 13:1453. [PMID: 37891821 PMCID: PMC10605398 DOI: 10.3390/brainsci13101453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a common brain dysfunction, which results in severe cognitive and neurological sequelae and an increased mortality rate in patients with sepsis. Depending on the stimulus, microglia (resident macrophages in the brain that are involved in SAE pathology and physiology) can adopt two polarization states (M1/M2), corresponding to altered microglial morphology, gene expression, and function. We systematically described the pathogenesis, morphology, function, and phenotype of microglial activation in SAE and demonstrated that microglia are closely related to SAE occurrence and development, and concomitant cognitive impairment. Finally, some potential therapeutic approaches that can prime microglia and neuroinflammation toward the beneficial restorative microglial phenotype in SAE were outlined.
Collapse
Affiliation(s)
- Jiyun Hu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shucai Xie
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Haisong Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinrun Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Binbin Meng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
17
|
Sieminski M, Szaruta-Raflesz K, Szypenbejl J, Krzyzaniak K. Potential Neuroprotective Role of Melatonin in Sepsis-Associated Encephalopathy Due to Its Scavenging and Anti-Oxidative Properties. Antioxidants (Basel) 2023; 12:1786. [PMID: 37760089 PMCID: PMC10525116 DOI: 10.3390/antiox12091786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/08/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. The brain is one of the organs involved in sepsis, and sepsis-induced brain injury manifests as sepsis-associated encephalopathy (SAE). SAE may be present in up to 70% of septic patients. SAE has a very wide spectrum of clinical symptoms, ranging from mild behavioral changes through cognitive disorders to disorders of consciousness and coma. The presence of SAE increases mortality in the population of septic patients and may lead to chronic cognitive dysfunction in sepsis survivors. Therefore, therapeutic interventions with neuroprotective effects in sepsis are needed. Melatonin, a neurohormone responsible for the control of circadian rhythms, exerts many beneficial physiological effects. Its anti-inflammatory and antioxidant properties are well described. It is considered a potential therapeutic factor in sepsis, with positive results from studies on animal models and with encouraging results from the first human clinical trials. With its antioxidant and anti-inflammatory potential, it may also exert a neuroprotective effect in sepsis-associated encephalopathy. The review presents data on melatonin as a potential drug in SAE in the wider context of the pathophysiology of SAE and the specific actions of the pineal neurohormone.
Collapse
Affiliation(s)
- Mariusz Sieminski
- Department of Emergency Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland; (K.S.-R.); (K.K.)
| | | | - Jacek Szypenbejl
- Department of Emergency Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland; (K.S.-R.); (K.K.)
| | | |
Collapse
|
18
|
Kim HY, Ashim J, Park S, Kim W, Ji S, Lee SW, Jung YR, Jeong SW, Lee SG, Kim HC, Lee YJ, Kwon MK, Hwang JS, Shin JM, Lee SJ, Yu W, Park JK, Choi SK. A preliminary study about the potential risks of the UV-weathered microplastic: The proteome-level changes in the brain in response to polystyrene derived weathered microplastics. ENVIRONMENTAL RESEARCH 2023; 233:116411. [PMID: 37354929 DOI: 10.1016/j.envres.2023.116411] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/11/2023] [Indexed: 06/26/2023]
Abstract
The growing use of plastic materials has resulted in a constant increase in the risk associated with microplastics (MPs). Ultra-violet (UV) light and wind break down modify MPs in the environment into smaller particles known as weathered MPs (WMPs) and these processes increase the risk of MP toxicity. The neurotoxicity of weathered polystyrene-MPs remains unclear. Therefore, it is important to understand the risks posed by WMPs. We evaluated the chemical changes of WMPs generated under laboratory-synchronized environmentally mimetic conditions and compared them with virgin MPs (VMPs). We found that WMP had a rough surface, slight yellow color, reduced molecular weight, and structural alteration compared with those of VMP. Next, 2 μg of ∼100 μm in size of WMP and VMP were orally administered once a day for one week to C57BL/6 male mice. Proteomic analysis revealed that the WMP group had significantly increased activation of immune and neurodegeneration-related pathways compared with that of the VMP group. Consistently, in in vitro experiments, the human brain-derived microglial cell line (HMC-3) also exhibited a more severe inflammatory response to WMP than to VMP. These results show that WMP is a more profound inflammatory factor than VMP. In summary, our findings demonstrate the toxicity of WMPs and provide theoretical insights into their potential risks to biological systems and even humans in the ecosystem.
Collapse
Affiliation(s)
- Hee-Yeon Kim
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Janbolat Ashim
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Song Park
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Wansoo Kim
- School of Life Science, BK21 FOUR KNU Creative Bioresearch Group, Kyungpook National University, Daegu, Republic of Korea; Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Sangho Ji
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Seoung-Woo Lee
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Yi-Rang Jung
- Department of Companion Animal Health Management, Daegu Health College, Daegu, Republic of Korea
| | - Sang Won Jeong
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Se-Guen Lee
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Hyun-Chul Kim
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Young-Jae Lee
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Mi Kyung Kwon
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | | | - Jung Min Shin
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Sung-Jun Lee
- Division of Biotechnology, DGIST, Daegu, Republic of Korea.
| | - Wookyung Yu
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Department of Brain Sciences, DGIST, Daegu, Republic of Korea.
| | - Jin-Kyu Park
- College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Seong-Kyoon Choi
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea; Division of Biotechnology, DGIST, Daegu, Republic of Korea.
| |
Collapse
|
19
|
Stins MF, Mtaja A, Mulendele E, Mwimbe DW, Pinilla G, Mutengo M, Pardo CA, Chipeta J. Elevated brain derived neurotrophic factor in plasma and interleukin-6 levels in cerebrospinal fluid in meningitis compared to cerebral malaria. J Neurol Sci 2023; 450:120663. [PMID: 37182424 PMCID: PMC10330544 DOI: 10.1016/j.jns.2023.120663] [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: 10/21/2022] [Revised: 03/11/2023] [Accepted: 04/22/2023] [Indexed: 05/16/2023]
Abstract
Neurological infections, such as Cerebral malaria (CM) and meningitis are associated with high mortality and in survivors, particularly young children, persistent neurologic deficits often remain. As brain inflammation plays a role in the development of these neurological sequelae, multiplex assays were used to assess a select set of immune mediators in both plasma and cerebrospinal fluid (CSF) from Zambian children with neurological infections. Both CM and meningitis patients showed high levels of markers for vascular inflammation, such as soluble ICAM-1 and angiopoietins. Although high levels of angiopoietin 1 and angiopoietin 2 were found in the meningitis group, their levels in the CSF were low and did not differ. As expected, there were high levels of cytokines and notably a significantly elevated IL-6 level in the CSF of the meningitis group. Interestingly, although elevated levels BDNF were found, BDNF levels were significantly higher in plasma of the meningitis group but similar in the CSF. The striking differences in plasma BDNF and IL-6 levels in the CSF point to markedly different neuro-pathological processes. Therefore, further investigations in the role of both IL-6 and BDNF in the neurological outcomes are needed.
Collapse
Affiliation(s)
- Monique F Stins
- Johns Hopkins School of Public Health, Malaria Research Institute, 615N Wolfe Street, SPH E45141, Baltimore, MD 21205, United States of America.
| | - Agnes Mtaja
- University of Zambia School of Medicine, Department of Paediatrics and Child Health, The School of Medicine, University Teaching Hospital Malaria Research Unit (SMUTH-MRU), P.O. Box 50110, Lusaka, Zambia
| | - Evan Mulendele
- University of Zambia School of Medicine, Department of Paediatrics and Child Health, The School of Medicine, University Teaching Hospital Malaria Research Unit (SMUTH-MRU), P.O. Box 50110, Lusaka, Zambia
| | - Daniel W Mwimbe
- University of Zambia School of Medicine, Department of Paediatrics and Child Health, The School of Medicine, University Teaching Hospital Malaria Research Unit (SMUTH-MRU), P.O. Box 50110, Lusaka, Zambia
| | - Gabriel Pinilla
- Johns Hopkins School of Medicine, Department of Neurology, Division of Neuroimmunology and Neuroinfectious Diseases, 600 N Wolfe Street, Baltimore, MD 21285, United States of America; Icesi University, Department of Clinical Sciences, Calle 18 No. 122-135, Cali 760031, Colombia
| | - Mable Mutengo
- University of Zambia School of Medicine, Department of Paediatrics and Child Health, The School of Medicine, University Teaching Hospital Malaria Research Unit (SMUTH-MRU), P.O. Box 50110, Lusaka, Zambia
| | - Carlos A Pardo
- Johns Hopkins School of Medicine, Department of Neurology, Division of Neuroimmunology and Neuroinfectious Diseases, 600 N Wolfe Street, Baltimore, MD 21285, United States of America
| | - James Chipeta
- University of Zambia School of Medicine, Department of Paediatrics and Child Health, The School of Medicine, University Teaching Hospital Malaria Research Unit (SMUTH-MRU), P.O. Box 50110, Lusaka, Zambia
| |
Collapse
|
20
|
Pagliaro P, Penna C. Inhibitors of NLRP3 Inflammasome in Ischemic Heart Disease: Focus on Functional and Redox Aspects. Antioxidants (Basel) 2023; 12:1396. [PMID: 37507935 PMCID: PMC10376505 DOI: 10.3390/antiox12071396] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is caused by several mechanisms, including the production of reactive oxygen species (ROS), altered cellular osmolarity, and inflammatory response. Calcium overload, altered oxygen levels, and mitochondrial ROS are also involved in these MIRI processes, resulting in the irreversible opening of the mitochondrial permeability transition pore (mPTP). These mechanisms and processes are associated with NLRP3 inflammasome priming and activation, which can also induce cell death by pyroptosis through the up-regulation of the caspase-1 pathway and IL-18 release. In addition, endothelial dysfunction, both in the presence and absence of MIRI, is also accompanied by altered oxygen levels, decreased nitric oxide production, and ROS overproduction, resulting in the expression of adhesion molecules and leukocyte infiltration in which the NLRP3 inflammasome plays a central role, thus contributing, through endothelial dysfunction, to the alteration of coronary flow, typical of ischemic heart disease. Given the intricate interrelationship between ROS and NLRP3, ROS inhibitors can reduce NLRP3 inflammasome activation, while NLRP3 inhibitors can reduce oxidative stress and inflammation. NLRP3 inhibitors have been intensively studied as anti-inflammatory agents in basic cardiovascular sciences. In this review, we analyze the interrelation between ROS and NLRP3 in ischemic heart disease and the effects of some NLRP3 inhibitors as possible therapeutic agents in this disease condition. All compounds considered in this review need larger studies to confirm their appropriate use in clinical scenarios as anti-ischemic drugs.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, Turin University, Orbassano, 10043 Turin, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, Turin University, Orbassano, 10043 Turin, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
21
|
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
|
22
|
Edler MK, Munger EL, Maycon H, Hopkins WD, Hof PR, Sherwood CC, Raghanti MA. The association of astrogliosis and microglial activation with aging and Alzheimer's disease pathology in the chimpanzee brain. J Neurosci Res 2023; 101:881-900. [PMID: 36647571 DOI: 10.1002/jnr.25167] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/31/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023]
Abstract
Aging and neurodegenerative disorders, such as Alzheimer's disease (AD), trigger an immune response known as glial activation in the brain. Recent evidence indicates species differences in inflammatory responses to AD pathology, highlighting the need for additional comparative studies to further understand human-specific neuropathologies. In the present study, we report on the occurrence of astrogliosis, microglial activation, and their relationship with age and AD-like pathology in a cohort of male and female chimpanzees (Pan troglodytes). Chimpanzees with severe astrogliosis exhibited widespread upregulation of hypertrophic astrocytes immunoreactive for glial fibrillary acidic protein (GFAP) throughout all layers of the dorsolateral prefrontal cortex and a loss of the interlaminar palisade. In addition, extreme astrogliosis was associated with increased astrocyte density in the absence of significant microglial activation and AD lesions. A shift from decreased resting to increased phagocytotic microglia occurred with aging, although proliferation was absent and no changes in astrogliosis was observed. Vascular amyloid correlated with decreased astrocyte and microglia densities, while tau lesions were associated with morphological changes in microglia and greater total glia density and glia: neuron ratio. These results further our understanding of inflammatory processes within the chimpanzee brain and provide comparative data to improve our understanding of human aging and neuropathological processes.
Collapse
Affiliation(s)
- Melissa K Edler
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Emily L Munger
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Hannah Maycon
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - William D Hopkins
- Department of Comparative Medicine, University of Texas MD Anderson Cancer Center, Bastrop, Texas, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
| | - Mary Ann Raghanti
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| |
Collapse
|
23
|
Bircak-Kuchtova B, Chung HY, Wickel J, Ehler J, Geis C. Neurofilament light chains to assess sepsis-associated encephalopathy: Are we on the track toward clinical implementation? Crit Care 2023; 27:214. [PMID: 37259091 DOI: 10.1186/s13054-023-04497-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Sepsis is the most common cause of admission to intensive care units worldwide. Sepsis patients frequently suffer from sepsis-associated encephalopathy (SAE) reflecting acute brain dysfunction. SAE may result in increased mortality, extended length of hospital stay, and long-term cognitive dysfunction. The diagnosis of SAE is based on clinical assessments, but a valid biomarker to identify and confirm SAE and to assess SAE severity is missing. Several blood-based biomarkers indicating neuronal injury have been evaluated in sepsis and their potential role as early diagnosis and prognostic markers has been studied. Among those, the neuroaxonal injury marker neurofilament light chain (NfL) was identified to potentially serve as a prognostic biomarker for SAE and to predict long-term cognitive impairment. In this review, we summarize the current knowledge of biomarkers, especially NfL, in SAE and discuss a possible future clinical application considering existing limitations.
Collapse
Affiliation(s)
- Barbora Bircak-Kuchtova
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Ha-Yeun Chung
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany.
| | - Jonathan Wickel
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Johannes Ehler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747, Jena, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| |
Collapse
|
24
|
Chung HY, Wickel J, Hahn N, Mein N, Schwarzbrunn M, Koch P, Ceanga M, Haselmann H, Baade-Büttner C, von Stackelberg N, Hempel N, Schmidl L, Groth M, Andreas N, Götze J, Coldewey SM, Bauer M, Mawrin C, Dargvainiene J, Leypoldt F, Steinke S, Wang ZQ, Hust M, Geis C. Microglia mediate neurocognitive deficits by eliminating C1q-tagged synapses in sepsis-associated encephalopathy. SCIENCE ADVANCES 2023; 9:eabq7806. [PMID: 37235660 DOI: 10.1126/sciadv.abq7806] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 04/20/2023] [Indexed: 05/28/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is a severe and frequent complication of sepsis causing delirium, coma, and long-term cognitive dysfunction. We identified microglia and C1q complement activation in hippocampal autopsy tissue of patients with sepsis and increased C1q-mediated synaptic pruning in a murine polymicrobial sepsis model. Unbiased transcriptomics of hippocampal tissue and isolated microglia derived from septic mice revealed an involvement of the innate immune system, complement activation, and up-regulation of lysosomal pathways during SAE in parallel to neuronal and synaptic damage. Microglial engulfment of C1q-tagged synapses could be prevented by stereotactic intrahippocampal injection of a specific C1q-blocking antibody. Pharmacologically targeting microglia by PLX5622, a CSF1-R inhibitor, reduced C1q levels and the number of C1q-tagged synapses, protected from neuronal damage and synapse loss, and improved neurocognitive outcome. Thus, we identified complement-dependent synaptic pruning by microglia as a crucial pathomechanism for the development of neuronal defects during SAE.
Collapse
Affiliation(s)
- Ha-Yeun Chung
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
| | - Jonathan Wickel
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
| | - Nina Hahn
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
| | - Nils Mein
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Meike Schwarzbrunn
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Philipp Koch
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena 07745, Germany
| | - Mihai Ceanga
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Holger Haselmann
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Carolin Baade-Büttner
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Nikolai von Stackelberg
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Nina Hempel
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Lars Schmidl
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Marco Groth
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena 07745, Germany
| | - Nico Andreas
- Institute of Immunology, Jena University Hospital, Jena 07743, Germany
| | - Juliane Götze
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena 07747, Germany
- Septomics Research Center, Jena University Hospital, Jena 07745, Germany
| | - Sina M Coldewey
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena 07747, Germany
- Septomics Research Center, Jena University Hospital, Jena 07745, Germany
| | - Michael Bauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena 07747, Germany
| | - Christian Mawrin
- Department of Neuropathology, University of Magdeburg, Magdeburg, Germany
- Section of Pathology, Institute of Forensic Medicine, Jena University Hospital, Jena 07749, Germany
| | | | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry, UKSH, Kiel/Lübeck, Germany
- Department of Neurology, Christian-Albrechts University, Kiel 24105, Germany
| | - Stephan Steinke
- Department Medical Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena 07745, Germany
- Faculty of Biological Sciences, Friedrich Schiller University, Jena 07745, Germany
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Michael Hust
- Department Medical Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Christian Geis
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
| |
Collapse
|
25
|
Song YQ, Lin WJ, Hu HJ, Wu SH, Jing L, Lu Q, Zhu W. Sodium tanshinone IIA sulfonate attenuates sepsis-associated brain injury via inhibiting NOD-like receptor 3/caspase-1/gasdermin D-mediated pyroptosis. Int Immunopharmacol 2023; 118:110111. [PMID: 37028275 DOI: 10.1016/j.intimp.2023.110111] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/19/2023] [Accepted: 03/26/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Sodium tanshinone IIA sulfonate (STS) has been reported to protect organ function in sepsis. However, the attenuation of sepsis-associated brain injury and its underlying mechanisms by STS has not been established. METHODS C57BL/6 mice were used to establish the cecal ligation perforation (CLP) model, and STS was injected intraperitoneally 30 min before the surgery. The BV2 cells were stimulated by lipopolysaccharide after being pre-treated with STS for 4 h. The STS protective effects against brain injury and in vivo anti-neuroinflammatory effects were investigated using the 48-hour survival rate and body weight changes, brain water content, histopathological staining, immunohistochemistry, ELISA, RT-qPCR, and transmission electron microscopy. The pro-inflammatory cytokines of BV2 cells were detected by ELISA and RT-qPCR. At last, the levels of NOD-like receptor 3 (NLRP3) inflammasome activation and pyroptosis in brain tissues of the CLP model and BV2 cells were detected using western blotting. RESULTS STS increased the survival rate, decreased brain water content, and improved brain pathological damage in the CLP models. STS increased the expressions of tight junction proteins ZO-1 and Claudin5 while reducing the expressions of tumor necrosis factor α (TNF-α), interleukin-1β(IL-1β), and interleukin-18 (IL-18) in the brain tissues of the CLP models. Meanwhile, STS inhibited microglial activation and M1-type polarization in vitro and in vivo. The NLRP3/caspase-1/ gasdermin D (GSDMD)-mediated pyroptosis was activated in the brain tissues of the CLP models and lipopolysaccharide (LPS)-treated BV2 cells, which was significantly inhibited by STS. CONCLUSIONS The activation of NLRP3/caspase-1/GSDMD-mediated pyroptosis and subsequent secretion of proinflammatory cytokines may be the underlying mechanisms of STS against sepsis-associated brain injury and neuroinflammatory response.
Collapse
Affiliation(s)
- Ya-Qin Song
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei-Ji Lin
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Jie Hu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shu-Hui Wu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liang Jing
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Lu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Zhu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
26
|
Activation of glucagon-like peptide-1 receptor in microglia exerts protective effects against sepsis-induced encephalopathy via attenuating endoplasmic reticulum stress-associated inflammation and apoptosis in a mouse model of sepsis. Exp Neurol 2023; 363:114348. [PMID: 36813224 DOI: 10.1016/j.expneurol.2023.114348] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/18/2023] [Accepted: 02/04/2023] [Indexed: 02/22/2023]
Abstract
Sepsis-induced encephalopathy (SAE) is a detrimental complication in patients with severe sepsis, while there is still no effective treatment. Previous studies have elucidated the neuroprotective effects of glucagon-like peptide-1 receptor (GLP-1R) agonists. However, the role of GLP-1R agonists in the pathological process of SAE is unclear. Here, we found that GLP-1R was up-regulated in the microglia of septic mice. The activation of GLP-1R with Liraglutide could inhibit endoplasmic reticulum stress (ER stress) and associated inflammatory response as well as apoptosis triggered by LPS or tunicamycin (TM) in BV2 cells. In vivo experiments confirmed the benefits of Liraglutide in the regulation of microglial activation, ER stress, inflammation, and apoptosis in the hippocampus of septic mice. Additionally, the survival rate and cognitive dysfunction of septic mice were also improved after Liraglutide administration. Mechanically, cAMP/PKA/CREB signaling is involved in the protection of ER stress-induced inflammation and apoptosis in cultured microglial cells under LPS or TM stimulations. In conclusion, we speculated that GLP-1/GLP-1R activation in microglia might be a potential therapeutic target for the treatment of SAE.
Collapse
|
27
|
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
|
28
|
Kou Y, Li Z, Yang T, Shen X, Wang X, Li H, Zhou K, Li L, Xia Z, Zheng X, Zhao Y. Therapeutic potential of plant iridoids in depression: a review. PHARMACEUTICAL BIOLOGY 2022; 60:2167-2181. [PMID: 36300881 PMCID: PMC9621214 DOI: 10.1080/13880209.2022.2136206] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/23/2022] [Accepted: 09/25/2022] [Indexed: 05/29/2023]
Abstract
CONTEXT Depression is a mental disorder characterized by low mood, reduced interest, impaired cognitive function, and vegetative symptoms such as sleep disturbances or poor appetite. Iridoids are the active constituents in several Chinese classical antidepressant formulae such as Yueju Pill, Zhi-Zi-Hou-Po Decoction, Zhi-Zi-Chi Decoction, and Baihe Dihuang Decoction. Parallel to their wide usages, iridoids are considered potential lead compounds for the treatment of neurological diseases. OBJECTIVE The review summarizes the therapeutic potential and molecular mechanisms of iridoids in the prevention or treatment of depression and contributes to identifying research gaps in iridoids as potential antidepressant medication. METHODS The following key phrases were sought in PubMed, Google Scholar, Web of Science, and China National Knowledge Internet (CNKI) without time limitation to search all relevant articles with in vivo or in vitro experimental studies as comprehensively as possible: ('iridoid' or 'seciridoid' or 'depression'). This review extracted the experimental data on the therapeutic potential and molecular mechanism of plant-derived iridoids for depression. RESULTS Plant iridoids (i.e., catalpol, geniposide, loganin), and secoiridoids (i.e., morroniside, gentiopicroside, oleuropein, swertiamarin), all showed significant improvement effects on depression. DISCUSSION AND CONCLUSIONS Iridoids exert antidepressant effects by elevating monoamine neurotransmitters, reducing pro-inflammatory factors, inhibiting hypothalamic-pituitary-adrenal (HPA) axis hyperactivity, increasing brain-derived neurotrophic factor (BDNF) and its receptors, and elevating intestinal microbial abundance. Further detailed studies on the pharmacokinetics, bioavailability, and key molecular targets of iridoids are also required in future research, ultimately to provide improvements to current antidepressant medications.
Collapse
Affiliation(s)
- Yaoyao Kou
- Three level Scientific Research Laboratory of National Administration of Traditional Chinese Medicine, Northwest University, Xi’an, PR China
| | - Zhihao Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, PR China
| | - Tong Yang
- Three level Scientific Research Laboratory of National Administration of Traditional Chinese Medicine, Northwest University, Xi’an, PR China
| | - Xue Shen
- Three level Scientific Research Laboratory of National Administration of Traditional Chinese Medicine, Northwest University, Xi’an, PR China
| | - Xin Wang
- Three level Scientific Research Laboratory of National Administration of Traditional Chinese Medicine, Northwest University, Xi’an, PR China
| | - Haopeng Li
- Three level Scientific Research Laboratory of National Administration of Traditional Chinese Medicine, Northwest University, Xi’an, PR China
| | - Kun Zhou
- Three level Scientific Research Laboratory of National Administration of Traditional Chinese Medicine, Northwest University, Xi’an, PR China
| | - Luyao Li
- Three level Scientific Research Laboratory of National Administration of Traditional Chinese Medicine, Northwest University, Xi’an, PR China
| | - Zhaodi Xia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, PR China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, PR China
| | - Ye Zhao
- Three level Scientific Research Laboratory of National Administration of Traditional Chinese Medicine, Northwest University, Xi’an, PR China
| |
Collapse
|
29
|
Reactive Microgliosis in Sepsis-Associated and Acute Hepatic Encephalopathies: An Ultrastructural Study. Int J Mol Sci 2022; 23:ijms232214455. [PMID: 36430933 PMCID: PMC9696099 DOI: 10.3390/ijms232214455] [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: 10/12/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
Sepsis and acute liver failure are associated with severe endogenous intoxication. Microglia, which are the resident immune brain cells, play diverse roles in central nervous system development, surveillance, and defense, as well as contributing to neuroinflammatory reactions. In particular, microglia are fundamental to the pathophysiology of reactive toxic encephalopathies. We analyzed microglial ultrastructure, morphotypes, and phagocytosis in the sensorimotor cortex of cecal ligation and puncture (CLP) and acetaminophen-induced liver failure (AILF) Wistar rats. A CLP model induced a gradual shift of ~50% of surveillant microglia to amoeboid hypertrophic-like and gitter cell-like reactive phenotypes with active phagocytosis and frequent contacts with damaged neurons. In contrast, AILF microglia exhibited amoeboid, rod-like, and hypertrophic-like reactive morphotypes with minimal indications for efficient phagocytosis, and were mostly in contact with edematous astrocytes. Close interactions of reactive microglia with neurons, astrocytes, and blood-brain barrier components reflect an active contribution of these cells to the tissue adaptation and cellular remodeling to toxic brain damage. Partial disability of reactive microglia may affect the integrity and metabolism in all tissue compartments, leading to failure of the compensatory mechanisms in acute endogenous toxic encephalopathies.
Collapse
|
30
|
Martinez-Orengo N, Tahmazian S, Lai J, Wang Z, Sinharay S, Schreiber-Stainthorp W, Basuli F, Maric D, Reid W, Shah S, Hammoud DA. Assessing organ-level immunoreactivity in a rat model of sepsis using TSPO PET imaging. Front Immunol 2022; 13:1010263. [PMID: 36439175 PMCID: PMC9685400 DOI: 10.3389/fimmu.2022.1010263] [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: 08/02/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
There is current need for new approaches to assess/measure organ-level immunoreactivity and ensuing dysfunction in systemic inflammatory response syndrome (SIRS) and sepsis, in order to protect or recover organ function. Using a rat model of systemic sterile inflammatory shock (intravenous LPS administration), we performed PET imaging with a translocator protein (TSPO) tracer, [18F]DPA-714, as a biomarker for reactive immunoreactive changes in the brain and peripheral organs. In vivo dynamic PET/CT scans showed increased [18F]DPA-714 binding in the brain, lungs, liver and bone marrow, 4 hours after LPS injection. Post-LPS mean standard uptake values (SUVmean) at equilibrium were significantly higher in those organs compared to baseline. Changes in spleen [18F]DPA-714 binding were variable but generally decreased after LPS. SUVmean values in all organs, except the spleen, positively correlated with several serum cytokines/chemokines. In vitro measures of TSPO expression and immunofluorescent staining validated the imaging results. Noninvasive molecular imaging with [18F]DPA-714 PET in a rat model of systemic sterile inflammatory shock, along with in vitro measures of TSPO expression, showed brain, liver and lung inflammation, spleen monocytic efflux/lymphocytic activation and suggested increased bone marrow hematopoiesis. TSPO PET imaging can potentially be used to quantify SIRS and sepsis-associated organ-level immunoreactivity and assess the effectiveness of therapeutic and preventative approaches for associated organ failures, in vivo.
Collapse
Affiliation(s)
- Neysha Martinez-Orengo
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sarine Tahmazian
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jianhao Lai
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Zeping Wang
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sanhita Sinharay
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - William Schreiber-Stainthorp
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - William Reid
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Swati Shah
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Dima A. Hammoud,
| |
Collapse
|
31
|
SS-31 Improves Cognitive Function in Sepsis-Associated Encephalopathy by Inhibiting the Drp1-NLRP3 Inflammasome Activation. Neuromolecular Med 2022:10.1007/s12017-022-08730-1. [DOI: 10.1007/s12017-022-08730-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
32
|
Xu B, Li M, Cheng T, Xia J, Deng X, Hou J. Resolvin D1 protects against sepsis-associated encephalopathy in mice by inhibiting neuro-inflammation induced by microglia. Am J Transl Res 2022; 14:6737-6750. [PMID: 36247289 PMCID: PMC9556482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/29/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVES Neuro-inflammation induced by microglia is crucial in the pathogenesis of sepsis-associated encephalopathy (SAE). The endogenous lipid mediator, Resolvin D1 (RvD1), which is synthesized from docosahexaenoic acid, has been extensively reported to attenuate inflammation in various diseases by its anti-inflammation and pro-resolving functions. However, the effect of RvD1 on SAE remains unclear. In this study, we aimed to ex the function and mechanism of RvD1 on SAE mice. METHODS In our study, the SAE mice model was established by the method of cecal ligation and perforation (CLP). C57BL/6J mice were randomly divided into three groups: the Sham group, the CLP group and the CLP+RvD1 group. Cognitive impairment of the mice was assessed by Morris water maze. Iba1 immunohistochemistry was conducted to observe the activation of microglia in hippocampus of the mice from different groups. The production of cytokines, including TNF-α, IL-6 and IL-1β, and their mRNA levels were evaluated by ELISA and Q-PCR. The expression of the molecules from inflammatory signaling pathways was assessed by Western blot. RESULTS xaRvD1 treatment significantly improved the learning and cognitive ability of SAE mice. The activation of microglia and the production of inflammatory cytokines in hippocampal tissues were inhibited in CLP+RvD1 group. We also found that the inflammation of microglia was attenuated by RvD1 treatment both in vivo and in vitro. Moreover, the activation of NF-κB, MAPK and STAT signaling pathways were inhibited by RvD1 treatment, which partly explained the anti-inflammation function of RvD1 on SAE mice. CONCLUSIONS RvD1 could improve the learning and cognitive ability of SAE mice by inhibiting the systemic and local inflammation. It could attenuate the inflammation in microglia by inhibiting the activation of inflammatory signaling pathways and then decreasing the production of cytokines. These findings are helpful to better understand the pathophysiology of SAE, which also provide a novel therapeutic method in clinic.
Collapse
Affiliation(s)
- Bing Xu
- Department of Anesthesiology, Changhai Hospital, Naval Medical UniversityShanghai 200433, China
| | - Mi Li
- School of Anesthesiology, Naval Medical UniversityShanghai 200433, China
| | - Tingting Cheng
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of MedicineShanghai 200025, China
| | - Jun Xia
- Department of Anesthesiology, Changhai Hospital, Naval Medical UniversityShanghai 200433, China
| | - Xiaoming Deng
- Department of Anesthesiology, Changhai Hospital, Naval Medical UniversityShanghai 200433, China
| | - Jiong Hou
- Department of Anesthesiology, Changhai Hospital, Naval Medical UniversityShanghai 200433, China
| |
Collapse
|
33
|
Mudra Rakshasa-Loots A, Whalley HC, Vera JH, Cox SR. Neuroinflammation in HIV-associated depression: evidence and future perspectives. Mol Psychiatry 2022; 27:3619-3632. [PMID: 35618889 PMCID: PMC9708589 DOI: 10.1038/s41380-022-01619-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 02/08/2023]
Abstract
People living with HIV face a high risk of mental illness, especially depression. We do not yet know the precise neurobiological mechanisms underlying HIV-associated depression. Depression severity in the general population has been linked to acute and chronic markers of systemic inflammation. Given the associations between depression and peripheral inflammation, and since HIV infection in the brain elicits a neuroinflammatory response, it is possible that neuroinflammation contributes to the high prevalence of depression amongst people living with HIV. The purpose of this review was to synthesise existing evidence for associations between inflammation, depression, and HIV. While there is strong evidence for independent associations between these three conditions, few preclinical or clinical studies have attempted to characterise their interrelationship, representing a major gap in the literature. This review identifies key areas of debate in the field and offers perspectives for future investigations of the pathophysiology of HIV-associated depression. Reproducing findings across diverse populations will be crucial in obtaining robust and generalisable results to elucidate the precise role of neuroinflammation in this pathophysiology.
Collapse
Affiliation(s)
- Arish Mudra Rakshasa-Loots
- Edinburgh Neuroscience, School of Biomedical Sciences, The University of Edinburgh, Edinburgh, UK.
- Lothian Birth Cohorts Group, Department of Psychology, The University of Edinburgh, Edinburgh, UK.
| | - Heather C Whalley
- Division of Psychiatry, Centre for Clinical Brain Sciences, Royal Edinburgh Hospital, The University of Edinburgh, Edinburgh, UK
| | - Jaime H Vera
- Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Simon R Cox
- Lothian Birth Cohorts Group, Department of Psychology, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
34
|
Deus CM, Tavares H, Beatriz M, Mota S, Lopes C. Mitochondrial Damage-Associated Molecular Patterns Content in Extracellular Vesicles Promotes Early Inflammation in Neurodegenerative Disorders. Cells 2022; 11:2364. [PMID: 35954208 PMCID: PMC9367540 DOI: 10.3390/cells11152364] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is a common hallmark in different neurodegenerative conditions that share neuronal dysfunction and a progressive loss of a selectively vulnerable brain cell population. Alongside ageing and genetics, inflammation, oxidative stress and mitochondrial dysfunction are considered key risk factors. Microglia are considered immune sentinels of the central nervous system capable of initiating an innate and adaptive immune response. Nevertheless, the pathological mechanisms underlying the initiation and spread of inflammation in the brain are still poorly described. Recently, a new mechanism of intercellular signalling mediated by small extracellular vesicles (EVs) has been identified. EVs are nanosized particles (30-150 nm) with a bilipid membrane that carries cell-specific bioactive cargos that participate in physiological or pathological processes. Damage-associated molecular patterns (DAMPs) are cellular components recognised by the immune receptors of microglia, inducing or aggravating neuroinflammation in neurodegenerative disorders. Diverse evidence links mitochondrial dysfunction and inflammation mediated by mitochondrial-DAMPs (mtDAMPs) such as mitochondrial DNA, mitochondrial transcription factor A (TFAM) and cardiolipin, among others. Mitochondrial-derived vesicles (MDVs) are a subtype of EVs produced after mild damage to mitochondria and, upon fusion with multivesicular bodies are released as EVs to the extracellular space. MDVs are particularly enriched in mtDAMPs which can induce an immune response and the release of pro-inflammatory cytokines. Importantly, growing evidence supports the association between mitochondrial dysfunction, EV release and inflammation. Here, we describe the role of extracellular vesicles-associated mtDAMPS in physiological conditions and as neuroinflammation activators contributing to neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Sandra Mota
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, III-Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; (C.M.D.); (H.T.); (M.B.)
| | - Carla Lopes
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, III-Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; (C.M.D.); (H.T.); (M.B.)
| |
Collapse
|
35
|
Peng LS, Xu Y, Wang QS. YY1 PROMOTES MICROGLIA M2 POLARIZATION THROUGH THE MIR-130A-3P/TREM-2 AXIS TO ALLEVIATE SEPSIS-ASSOCIATED ENCEPHALOPATHY. Shock 2022; 58:128-136. [PMID: 35234205 DOI: 10.1097/shk.0000000000001914] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Purpose: Sepsis-associated encephalopathy (SAE) induces cognitive dysfunction via mechanisms that commonly involve neuroinflammation. Yin Yang 1 (YY1) is an important transcription factor that acts as a key role in sepsis and neuroepithelium development. However, the function of YY1 in SAE remains unclear. Our study aimed to probe the intrinsic and concrete molecular mechanism of YY1 in SAE. Methods: SAE cell model and SAE animal model were constructed by lipopolysaccharide (LPS) treatment and cecal ligation and puncture surgery, respectively. Behavioral tests were performed to analyze the cognitive function. The polarization state of mouse microglia (BV-2 cells) was assessed by flow cytometry assay. The mRNA and protein expressions were assessed by qRT-PCR and western blot. Finally, the binding relationships between YY1, miR-130a-3p, andTREM-2were verified by dual luciferase reporter gene assay and/or ChIP assay. Results: Here our results described that YY1 and TREM-2 were downregulated and miR-130a-3p was upregulated in SAE. YY1 overexpression could promote M2 polarization of microglia, and alleviate neuroinflammation and behavioral deficits in vitro and in vivo. YY1 could inhibit miR-130a-3p promoter activity. As expected, miR-130a-3p overexpression abolished the effects of YY1 overexpression on LPS-treated BV-2 cells. Besides, TREM-2 was identified as the target of miR-130a-3p. TREM-2 silencing could reverse the effects of miR-130a-3p inhibition on LPS-treated BV-2 cells. Conclusion: Taken together, YY1 promoted microglia M2 polarization via upregulating TREM-2 by interacting with miR-130a-3p promoter, suggesting YY1 overexpression might be a novel therapeutic strategy of SAE.
Collapse
Affiliation(s)
- Liang-Shan Peng
- The First Affiliated Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | | | | |
Collapse
|
36
|
Zhang LM, Xin Y, Wu ZY, Song RX, Miao HT, Zheng WC, Li Y, Zhang DX, Zhao XC. STING mediates neuroinflammatory response by activating NLRP3-related pyroptosis in severe traumatic brain injury. J Neurochem 2022; 162:444-462. [PMID: 35892155 DOI: 10.1111/jnc.15678] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/21/2022] [Accepted: 07/19/2022] [Indexed: 12/01/2022]
Abstract
Long-term neurological deficits after severe traumatic brain injury (TBI), including cognitive dysfunction and emotional impairments, can significantly impair rehabilitation. Glial activation induced by inflammatory response is involved in the neurological deficits post-TBI. This study aimed to investigate the role of the stimulator of interferon genes (STING)-nucleotide-binding oligomerization domain-like receptor pyrin domain-containing-3 (NLRP3) signaling in a rodent model of severe TBI. Severe TBI models were established using weight-drop plus blood loss-reinfusion. Selective STING agonist ADU-S100 or antagonist C-176 was given as a single dose after modeling. Further, NLRP3 inhibitor MCC950 or activator nigericin, or caspase-1 inhibitor VX765, was given as an intracerebroventricular injection 30 min before modeling. After that, a novel object recognition test, open field test, force swimming test, western blot, and immunofluorescence assays were used to assess behavioral and pathological changes in severe TBI. Administration of C-176 alleviated TBI-induced cognitive dysfunction and emotional impairments, neuronal loss, and inflammatory activation of glia cells. However, the administration of STING agonist ADU-S100 exacerbated TBI-induced behavioral and pathological changes. In addition, STING activation exacerbated pyroptosis-associated neuroinflammation via promoting glial activation, as evidenced by increased cleaved caspase-1 and GSDMD N-terminal expression. In contrast, the administration of C-176 showed anti-pyroptotic effects. The neuroprotective effects of C-176 were partially reversed by the NLRP3 activator, nigericin. Collectively, glial STING is responsible for neuroinflammation post-TBI. However, pharmacologic inhibition of STING led to a remarkable improvement of neuroinflammation partly through suppressing NLRP3 signaling. The STING-NLRP3 signaling is a potential therapeutic target in TBI-induced neurological dysfunction.
Collapse
Affiliation(s)
- Li-Min Zhang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.,Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing)
| | - Yue Xin
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Zhi-You Wu
- Department of Neurosurgery, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Rong-Xin Song
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Hui-Tao Miao
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Wei-Chao Zheng
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiao-Chun Zhao
- Department of Anesthesiology, School and Hospital of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
37
|
Abstract
Systemic inflammation elicited by sepsis can induce an acute cerebral dysfunction known as sepsis-associated encephalopathy (SAE). Recent evidence suggests that SAE is common but shows a dynamic trajectory over time. Half of all patients with sepsis develop SAE in the intensive care unit, and some survivors present with sustained cognitive impairments for several years after initial sepsis onset. It is not clear why some, but not all, patients develop SAE and also the factors that determine the persistence of SAE. Here, we first summarize the chronic pathology and the dynamic changes in cognitive functions seen after the onset of sepsis. We then outline the cerebral effects of sepsis, such as neuroinflammation, alterations in neuronal synapses and neurovascular changes. We discuss the key factors that might contribute to the development and persistence of SAE in older patients, including premorbid neurodegenerative pathology, side effects of sedatives, renal dysfunction and latent virus reactivation. Finally, we postulate that some of the mechanisms that underpin neuropathology in SAE may also be relevant to delirium and persisting cognitive impairments that are seen in patients with severe COVID-19.
Collapse
Affiliation(s)
- Tatsuya Manabe
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Bonn, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
38
|
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
|
39
|
Aschman T, Mothes R, Heppner FL, Radbruch H. What SARS-CoV-2 does to our brains. Immunity 2022; 55:1159-1172. [PMID: 35777361 PMCID: PMC9212726 DOI: 10.1016/j.immuni.2022.06.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/22/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022]
Abstract
Neurological symptoms in SARS-CoV-2-infected patients have been reported, but their cause remains unclear. In theory, the neurological symptoms observed after SARS-CoV-2 infection could be (1) directly caused by the virus infecting brain cells, (2) indirectly by our body’s local or systemic immune response toward the virus, (3) by coincidental phenomena, or (4) a combination of these factors. As indisputable evidence of intact and replicating SARS-CoV-2 particles in the central nervous system (CNS) is currently lacking, we suggest focusing on the host’s immune reaction when trying to understand the neurocognitive symptoms associated with SARS-CoV-2 infection. In this perspective, we discuss the possible immune-mediated mechanisms causing functional or structural CNS alterations during acute infection as well as in the post-infectious context. We also review the available literature on CNS affection in the context of COVID-19 infection, as well as observations from animal studies on the molecular pathways involved in sickness behavior.
Collapse
|
40
|
Giridharan VV, Generoso JS, Lence L, Candiotto G, Streck E, Petronilho F, Pillai A, Sharshar T, Dal-Pizzol F, Barichello T. A crosstalk between gut and brain in sepsis-induced cognitive decline. J Neuroinflammation 2022; 19:114. [PMID: 35606817 PMCID: PMC9125851 DOI: 10.1186/s12974-022-02472-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 05/11/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Sepsis is a potentially fatal disease characterized by acute organ failure that affects more than 30 million people worldwide. Inflammation is strongly associated with sepsis, and patients can experience impairments in memory, concentration, verbal fluency, and executive functioning after being discharged from the hospital. We hypothesize that sepsis disrupts the microbiota-gut-brain axis homeostasis triggering cognitive impairment. This immune activation persists during treatment, causing neurological dysfunction in sepsis survivors. METHODS To test our hypothesis, adult Wistar rats were subjected to cecal-ligation and perforation (CLP) or sham (non-CLP) surgeries. The animals were subjected to the [11C]PBR28 positron emission tomography (PET)/computed tomography (CT) imaging at 24 h and 10 days after CLP and non-CLP surgeries. At 24 h and 10 days after surgery, we evaluated the gut microbiome, bacterial metabolites, cytokines, microglia, and astrocyte markers. Ten days after sepsis induction, the animals were subjected to the novel object recognition (NOR) and the Morris water maze (MWM) test to assess their learning and memory. RESULTS Compared to the control group, the 24-h and 10-day CLP groups showed increased [11C]PBR28 uptake, glial cells count, and cytokine levels in the brain. Results show that sepsis modulates the gut villus length and crypt depth, alpha and beta microbial diversities, and fecal short-chain fatty acids (SCFAs). In addition, sepsis surviving animals showed a significant cognitive decline compared with the control group. CONCLUSIONS Since several pharmacological studies have failed to prevent cognitive impairment in sepsis survivors, a better understanding of the function of glial cells and gut microbiota can provide new avenues for treating sepsis patients.
Collapse
Affiliation(s)
- Vijayasree V Giridharan
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Jaqueline S Generoso
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Leonardo Lence
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gabriela Candiotto
- Laboratory of Neurometabolic Diseases, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Emílio Streck
- Laboratory of Neurometabolic Diseases, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Anilkumar Pillai
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Psychiatry and Health Behavior, Augusta University, Augusta, GA, USA
| | - Tarek Sharshar
- GHU Paris Psychiatrie et Neuroscience, Neurointensive Care and Neuroanesthesia Department, Paris, France
- Université de Paris Cité, Paris, France
- Institute of Psychiatry and Neurosciences of Paris, NSERM UMR 1266, Paris, France
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Tatiana Barichello
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil.
| |
Collapse
|
41
|
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
|
42
|
Zhao H, Lyu Y, Zhai R, Sun G, Ding X. Metformin Mitigates Sepsis-Related Neuroinflammation via Modulating Gut Microbiota and Metabolites. Front Immunol 2022; 13:797312. [PMID: 35572534 PMCID: PMC9102391 DOI: 10.3389/fimmu.2022.797312] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/05/2022] [Indexed: 12/04/2022] Open
Abstract
Gut microbiota affects the functions of brains. However, its mechanism in sepsis remains unclear. This study evaluated the effect of metformin on ameliorating sepsis-related neurodamage by regulating gut microbiota and metabolites in septic rats. Cecal ligation and puncture (CLP) was used to establish the sepsis-related neurodamage animal models. Metformin therapy by gavage at 1 h after CLP administration was followed by fecal microbiota transplantation (FMT) to ensure the efficacy and safety of metformin on the sepsis-related neurodamage by regulating gut microbiota. The gut microbiota and metabolites were conducted by 16S rRNA sequencing and liquid chromatography-tandem mass spectrometry metabolomic analysis. The brain tissue inflammation response was analyzed by histopathology and reverse transcription-polymerase chain reaction (RT-PCR). This study reported brain inflammatory response, hemorrhage in sepsis-related neurodamage rats compared with the control group (C group). Surprisingly, the abundance of gut microbiota slightly increased in sepsis-related neurodamage rats than C group. The ratio of Firmicutes/Bacteroidetes was significantly increased in the CLP group than the C group. However, no difference was observed between the CLP and the metformin-treated rats (MET group). Interestingly, the abundance of Escherichia_Shigella increased in the MET group than the C and CLP groups, while Lactobacillaceae abundance decreased. Furthermore, Prevotella_9, Muribaculaceae, and Alloprevotella related to short-chain fatty acids production increased in the sepsis-related neurodamage of metformin-treated rats. Additionally, Prevotella_9 and Muribaculaceae correlated positively to 29 metabolites that might affect the inflammatory factors in the brain. The FMT assay showed that metformin improved sepsis-related neurodamage by regulating the gut microbiota and metabolites in septic rats. The findings suggest that metformin improves the sepsis-related neurodamage through modulating the gut microbiota and metabolites in septic rats, which may be an effective therapy for patients with sepsis-related neurodamage.
Collapse
Affiliation(s)
- Huayan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanjun Lyu
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruiqing Zhai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Guiying Sun
- Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Xianfei Ding
- General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Xianfei Ding,
| |
Collapse
|
43
|
Polarization of Microglia and Its Therapeutic Potential in Sepsis. Int J Mol Sci 2022; 23:ijms23094925. [PMID: 35563317 PMCID: PMC9101892 DOI: 10.3390/ijms23094925] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection, leaving the inflammation process without a proper resolution, leading to tissue damage and possibly sequelae. The central nervous system (CNS) is one of the first regions affected by the peripheral inflammation caused by sepsis, exposing the neurons to an environment of oxidative stress, triggering neuronal dysfunction and apoptosis. Sepsis-associated encephalopathy (SAE) is the most frequent sepsis-associated organ dysfunction, with symptoms such as deliriums, seizures, and coma, linked to increased mortality, morbidity, and cognitive disability. However, the current therapy does not avoid those patients’ symptoms, evidencing the search for a more optimal approach. Herein we focus on microglia as a prominent therapeutic target due to its multiple functions maintaining CNS homeostasis and its polarizing capabilities, stimulating and resolving neuroinflammation depending on the stimuli. Microglia polarization is a target of multiple studies involving nerve cell preservation in diseases caused or aggravated by neuroinflammation, but in sepsis, its therapeutic potential is overlooked. We highlight the peroxisome proliferator-activated receptor gamma (PPARγ) neuroprotective properties, its role in microglia polarization and inflammation resolution, and the interaction with nuclear factor-κB (NF-κB) and mitogen-activated kinases (MAPK), making PPARγ a molecular target for sepsis-related studies to come.
Collapse
|
44
|
Fan W, Huang W, Chen J, Li N, Mao L, Hou S. Retinal microglia: Functions and diseases. Immunology 2022; 166:268-286. [PMID: 35403700 DOI: 10.1111/imm.13479] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/26/2022] [Accepted: 02/22/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Wei Fan
- The First Affiliated Hospital of Chongqing Medical University Chongqing China
- Chongqing Key Laboratory of Ophthalmology Chongqing China
- Chongqing Eye Institute Chongqing China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases Chongqing China
| | - Weidi Huang
- The First Affiliated Hospital of Chongqing Medical University Chongqing China
- Department of Ophthalmology, Second Xiangya Hospital Central South University Changsha Hunan China
| | - Jiayi Chen
- The First Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Na Li
- College of Basic Medicine Chongqing Medical University Chongqing China
| | - Liming Mao
- Department of Immunology School of Medicine, Nantong University, 19 Qixiu Road Nantong Jiangsu China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University Chongqing China
- Chongqing Key Laboratory of Ophthalmology Chongqing China
- Chongqing Eye Institute Chongqing China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases Chongqing China
| |
Collapse
|
45
|
Zhang Y, Chen S, Tian W, Zhu H, Li W, Dai W, Zhang X, Gu X, Su D. Emerging Trends and Hot Spots in Sepsis-Associated Encephalopathy Research From 2001 to 2021: A Bibliometric Analysis. Front Med (Lausanne) 2022; 9:817351. [PMID: 35295600 PMCID: PMC8918530 DOI: 10.3389/fmed.2022.817351] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/27/2022] [Indexed: 12/30/2022] Open
Abstract
Study Objectives To evaluate sepsis-associated encephalopathy (SAE) research and to quantitatively and qualitatively predict research hot spots using bibliometric analysis. Methods We extracted relevant publications from the Web of Science Core Collection on July 28, 2021. We investigated the retrieved data by bibliometric analysis (e.g. co-cited and cluster analysis, keyword co-occurrence) using the software CiteSpace and VOSviewer, the Online Analysis Platform of Literature Metrology (http://bibliometric.com/) and Bibliometrix to analyse and predict the trends and hot spots in this field. Main Results We identified 1,582 published articles and reviews on SAE from 2001 to 2021. During this period, the number of manuscripts on SAE increased steadily and peaked in 2021. The USA and China were the leading countries that had a critical impact on SAE research. Among all institutions, Vanderbilt University and Pittsburgh University held leading positions and became central in the collaboration network. Among all the journals, Critical Care Medicine published the maximum number of manuscripts in the field of SAE within 20 years. Dal-Pizzol Felipe was the most productive author (61 papers) and received the largest number of citations (930 citations). Co-citation cluster analysis revealed that the most popular terms on SAE in the manner of cluster labels were critical illness, sepsis-associated encephalopathy, polymicrobial sepsis, posterior reversible encephalopathy syndrome, rat brain, intensive care unit, prior sepsis, molecular hydrogen, inflammation drive, metabolic encephalopathies, delirium pathophysiology, and clinical neuroscience. Keyword burst detection indicated that neuroinflammation, blood-brain barrier (BBB) and mitochondria dysfunction were the current research hot spots. Conclusions Our study revealed that neuroinflammation, blood-brain barrier, and mitochondria dysfunction had been the research foci of SAE over the past 20 years. These have emerged as the basis for transformation from basic research to clinical application in finding effective methods for the prevention and treatment of SAE.
Collapse
|
46
|
Garland EF, Hartnell IJ, Boche D. Microglia and Astrocyte Function and Communication: What Do We Know in Humans? Front Neurosci 2022; 16:824888. [PMID: 35250459 PMCID: PMC8888691 DOI: 10.3389/fnins.2022.824888] [Citation(s) in RCA: 66] [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: 11/29/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia and astrocytes play essential roles in the central nervous system contributing to many functions including homeostasis, immune response, blood-brain barrier maintenance and synaptic support. Evidence has emerged from experimental models of glial communication that microglia and astrocytes influence and coordinate each other and their effects on the brain environment. However, due to the difference in glial cells between humans and rodents, it is essential to confirm the relevance of these findings in human brains. Here, we aim to review the current knowledge on microglia-astrocyte crosstalk in humans, exploring novel methodological techniques used in health and disease conditions. This will include an in-depth look at cell culture and iPSCs, post-mortem studies, imaging and fluid biomarkers, genetics and transcriptomic data. In this review, we will discuss the advantages and limitations of these methods, highlighting the understanding these methods have brought the field on these cells communicative abilities, and the knowledge gaps that remain.
Collapse
Affiliation(s)
| | | | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
47
|
Retroviral infection of human neurospheres and use of stem Cell EVs to repair cellular damage. Sci Rep 2022; 12:2019. [PMID: 35132117 PMCID: PMC8821538 DOI: 10.1038/s41598-022-05848-x] [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: 07/24/2020] [Accepted: 01/05/2022] [Indexed: 12/18/2022] Open
Abstract
HIV-1 remains an incurable infection that is associated with substantial economic and epidemiologic impacts. HIV-associated neurocognitive disorders (HAND) are commonly linked with HIV-1 infection; despite the development of combination antiretroviral therapy (cART), HAND is still reported to affect at least 50% of HIV-1 infected individuals. It is believed that the over-amplification of inflammatory pathways, along with release of toxic viral proteins from infected cells, are primarily responsible for the neurological damage that is observed in HAND; however, the underlying mechanisms are not well-defined. Therefore, there is an unmet need to develop more physiologically relevant and reliable platforms for studying these pathologies. In recent years, neurospheres derived from induced pluripotent stem cells (iPSCs) have been utilized to model the effects of different neurotropic viruses. Here, we report the generation of neurospheres from iPSC-derived neural progenitor cells (NPCs) and we show that these cultures are permissive to retroviral (e.g. HIV-1, HTLV-1) replication. In addition, we also examine the potential effects of stem cell derived extracellular vesicles (EVs) on HIV-1 damaged cells as there is abundant literature supporting the reparative and regenerative properties of stem cell EVs in the context of various CNS pathologies. Consistent with the literature, our data suggests that stem cell EVs may modulate neuroprotective and anti-inflammatory properties in damaged cells. Collectively, this study demonstrates the feasibility of NPC-derived neurospheres for modeling HIV-1 infection and, subsequently, highlights the potential of stem cell EVs for rescuing cellular damage induced by HIV-1 infection.
Collapse
|
48
|
Di Liberto G, Egervari K, Kreutzfeldt M, Schürch CM, Hewer E, Wagner I, Du Pasquier R, Merkler D. OUP accepted manuscript. Brain 2022; 145:2730-2741. [PMID: 35808999 PMCID: PMC9420019 DOI: 10.1093/brain/awac102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 11/14/2022] Open
Abstract
Glial cell activation is a hallmark of several neurodegenerative and neuroinflammatory diseases. During HIV infection, neuroinflammation is associated with cognitive impairment, even during sustained long-term suppressive antiretroviral therapy. However, the cellular subsets contributing to neuronal damage in the CNS during HIV infection remain unclear. Using post-mortem brain samples from eight HIV patients and eight non-neurological disease controls, we identify a subset of CNS phagocytes highly enriched in LGALS3, CTSB, GPNMB and HLA-DR, a signature identified in the context of ageing and neurodegeneration. In HIV patients, the presence of this phagocyte phenotype was associated with synaptic stripping, suggesting an involvement in the pathogenesis of HIV-associated neurocognitive disorder. Taken together, our findings elucidate some of the molecular signatures adopted by CNS phagocytes in HIV-positive patients and contribute to the understanding of how HIV might pave the way to other forms of cognitive decline in ageing HIV patient populations.
Collapse
Affiliation(s)
- Giovanni Di Liberto
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Department of Clinical Neurosciences, Service of Neurology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kristof Egervari
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Christian M Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Ekkehard Hewer
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Renaud Du Pasquier
- Department of Clinical Neurosciences, Service of Neurology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Doron Merkler
- Correspondence to: Doron Merkler Centre Médical Universitaire (CMU) 1, rue Michel Servet 1211 Geneva, Switzerland E-mail:
| |
Collapse
|
49
|
Chernyak BV, Lyamzaev KG, Mulkidjanian AY. Innate Immunity as an Executor of the Programmed Death of Individual Organisms for the Benefit of the Entire Population. Int J Mol Sci 2021; 22:ijms222413480. [PMID: 34948277 PMCID: PMC8704876 DOI: 10.3390/ijms222413480] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 11/19/2022] Open
Abstract
In humans, over-activation of innate immunity in response to viral or bacterial infections often causes severe illness and death. Furthermore, similar mechanisms related to innate immunity can cause pathogenesis and death in sepsis, massive trauma (including surgery and burns), ischemia/reperfusion, some toxic lesions, and viral infections including COVID-19. Based on the reviewed observations, we suggest that such severe outcomes may be manifestations of a controlled suicidal strategy protecting the entire population from the spread of pathogens and from dangerous pathologies rather than an aberrant hyperstimulation of defense responses. We argue that innate immunity may be involved in the implementation of an altruistic programmed death of an organism aimed at increasing the well-being of the whole community. We discuss possible ways to suppress this atavistic program by interfering with innate immunity and suggest that combating this program should be a major goal of future medicine.
Collapse
Affiliation(s)
- Boris V. Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
- Correspondence: (B.V.C.); (A.Y.M.)
| | - Konstantin G. Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
| | - Armen Y. Mulkidjanian
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Physics, Osnabrueck University, D-49069 Osnabrueck, Germany
- Correspondence: (B.V.C.); (A.Y.M.)
| |
Collapse
|
50
|
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
|