1
|
Zou K, Deng Q, Zhang H, Huang C. Glymphatic system: a gateway for neuroinflammation. Neural Regen Res 2024; 19:2661-2672. [PMID: 38595285 PMCID: PMC11168510 DOI: 10.4103/1673-5374.391312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/15/2023] [Accepted: 11/09/2023] [Indexed: 04/11/2024] Open
Abstract
The glymphatic system is a relatively recently identified fluid exchange and transport system in the brain. Accumulating evidence indicates that glymphatic function is impaired not only in central nervous system disorders but also in systemic diseases. Systemic diseases can trigger the inflammatory responses in the central nervous system, occasionally leading to sustained inflammation and functional disturbance of the central nervous system. This review summarizes the current knowledge on the association between glymphatic dysfunction and central nervous system inflammation. In addition, we discuss the hypothesis that disease conditions initially associated with peripheral inflammation overwhelm the performance of the glymphatic system, thereby triggering central nervous system dysfunction, chronic neuroinflammation, and neurodegeneration. Future research investigating the role of the glymphatic system in neuroinflammation may offer innovative therapeutic approaches for central nervous system disorders.
Collapse
Affiliation(s)
- Kailu Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qingwei Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hong Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Changsheng Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
2
|
Tastan B, Heneka MT. The impact of neuroinflammation on neuronal integrity. Immunol Rev 2024; 327:8-32. [PMID: 39470038 DOI: 10.1111/imr.13419] [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] [Indexed: 10/30/2024]
Abstract
Neuroinflammation, characterized by a complex interplay among innate and adaptive immune responses within the central nervous system (CNS), is crucial in responding to infections, injuries, and disease pathologies. However, the dysregulation of the neuroinflammatory response could significantly affect neurons in terms of function and structure, leading to profound health implications. Although tremendous progress has been made in understanding the relationship between neuroinflammatory processes and alterations in neuronal integrity, the specific implications concerning both structure and function have not been extensively covered, with the exception of perspectives on glial activation and neurodegeneration. Thus, this review aims to provide a comprehensive overview of the multifaceted interactions among neurons and key inflammatory players, exploring mechanisms through which inflammation influences neuronal functionality and structural integrity in the CNS. Further, it will discuss how these inflammatory mechanisms lead to impairment in neuronal functions and architecture and highlight the consequences caused by dysregulated neuronal functions, such as cognitive dysfunction and mood disorders. By integrating insights from recent research findings, this review will enhance our understanding of the neuroinflammatory landscape and set the stage for future interventions that could transform current approaches to preserve neuronal integrity and function in CNS-related inflammatory conditions.
Collapse
Affiliation(s)
- Bora Tastan
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, Massachusetts, USA
| |
Collapse
|
3
|
Li S, Liu H, Lv P, Yao Y, Peng L, Xia T, Yan C, Ma Z, Chen ZP, Zhao C, Gu X. Microglia mediate memory dysfunction via excitatory synaptic elimination in a fracture surgery mouse model. J Neuroinflammation 2024; 21:227. [PMID: 39285282 PMCID: PMC11406843 DOI: 10.1186/s12974-024-03216-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024] Open
Abstract
Cognitive impairment is a common issue among human patients undergoing surgery, yet the neural mechanism causing this impairment remains unidentified. Surgical procedures often lead to glial cell activation and neuronal hypoexcitability, both of which are known to contribute to postoperative cognitive dysfunction (POCD). However, the role of neuron-glia crosstalk in the pathology of POCD is still unclear. Through integrated transcriptomics and proteomics analyses, we found that the complement cascades and microglial phagocytotic signaling pathways are activated in a mouse model of POCD. Following surgery, there is a significant increase in the presence of complement C3, but not C1q, in conjunction with presynaptic elements. This triggers a reduction in excitatory synapses, a decline in excitatory synaptic transmission, and subsequent memory deficits in the mouse model. By genetically knockout out C3ar1 or inhibiting p-STAT3 signaling, we successfully prevented neuronal hypoexcitability and alleviated cognitive impairment in the mouse model. Therefore, targeting the C3aR and downstream p-STAT3 signaling pathways could serve as potential therapeutic approaches for mitigating POCD.
Collapse
Affiliation(s)
- Shuming Li
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Huan Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Pin Lv
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yu Yao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Liangyu Peng
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tianjiao Xia
- Medical School, Nanjing University, Nanjing, China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhang-Peng Chen
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
4
|
Li Y, Li YJ, Fang X, Chen DQ, Yu WQ, Zhu ZQ. Peripheral inflammation as a potential mechanism and preventive strategy for perioperative neurocognitive disorder under general anesthesia and surgery. Front Cell Neurosci 2024; 18:1365448. [PMID: 39022312 PMCID: PMC11252726 DOI: 10.3389/fncel.2024.1365448] [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: 01/04/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
General anesthesia, as a commonly used medical intervention, has been widely applied during surgical procedures to ensure rapid loss of consciousness and pain relief for patients. However, recent research suggests that general anesthesia may be associated with the occurrence of perioperative neurocognitive disorder (PND). PND is characterized by a decline in cognitive function after surgery, including impairments in attention, memory, learning, and executive functions. With the increasing trend of population aging, the burden of PND on patients and society's health and economy is becoming more evident. Currently, the clinical consensus tends to believe that peripheral inflammation is involved in the pathogenesis of PND, providing strong support for further investigating the mechanisms and prevention of PND.
Collapse
Affiliation(s)
- Yuan Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| | - Ying-Jie Li
- Department of General Surgery, Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| | - Xu Fang
- Department of Anesthesiology, Nanchong Central Hospital, The Second Clinical Medical School of North Sichuan Medical College, Zunyi, China
| | - Dong-Qin Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wan-Qiu Yu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhao-Qiong Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Early Clinical Research Ward of Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
5
|
Xu D, Liu J, Meng S, Sun M, Chen Y, Hong Y. Isoflurane-induced neuroinflammation and NKCC1/KCC2 dysregulation result in long-term cognitive disorder in neonatal mice. BMC Anesthesiol 2024; 24:200. [PMID: 38840092 PMCID: PMC11151488 DOI: 10.1186/s12871-024-02587-6] [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: 03/12/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND The inhalational anesthetic isoflurane is commonly utilized in clinical practice, particularly in the field of pediatric anesthesia. Research has demonstrated its capacity to induce neuroinflammation and long-term behavioral disorders; however, the underlying mechanism remains unclear [1]. The cation-chloride cotransporters Na+-K+-2Cl--1 (NKCC1) and K+-2Cl--2 (KCC2) play a pivotal role in regulating neuronal responses to gamma-aminobutyric acid (GABA) [2]. Imbalances in NKCC1/KCC2 can disrupt GABA neurotransmission, potentially leading to neural circuit hyperexcitability and reduced inhibition following neonatal exposure to anesthesia [3]. Therefore, this study postulates that anesthetics have the potential to dysregulate NKCC1 and/or KCC2 during brain development. METHODS We administered 1.5% isoflurane anesthesia to neonatal rats for a duration of 4 h at postnatal day 7 (PND7). Anxiety levels were assessed using the open field test at PND28, while cognitive function was evaluated using the Morris water maze test between PND31 and PND34. Protein levels of NKCC1, KCC2, BDNF, and phosphorylated ERK (P-ERK) in the hippocampus were measured through Western blotting analysis. Pro-inflammatory cytokines IL-1β, IL-6, and TNF-α were quantified using ELISA. RESULTS We observed a decrease in locomotion trajectories within the central region and a significantly shorter total distance in the ISO group compared to CON pups, indicating that isoflurane induces anxiety-like behavior. In the Morris water maze (MWM) test, rats exposed to isoflurane exhibited prolonged escape latency onto the platform. Additionally, isoflurane administration resulted in reduced time spent crossing in the MWM experiment at PND34, suggesting long-term impairment of memory function. Furthermore, we found that isoflurane triggered activation of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α; downregulated KCC2/BDNF/P-ERK expression; and increased the NKCC1/KCC2 ratio in the hippocampus of PND7 rats. Bumetadine (NKCC1 specific inhibitors) reversed cognitive damage and effective disorder induced by isoflurane in neonatal rats by inhibiting TNF-α activation, normalizing IL-6 and IL-1β levels, restoring KCC2 expression levels as well as BDNF and ERK signaling pathways. Based on these findings, it can be speculated that BDNF, P-ERK, IL-1β, IL-6 and TNF - α may act downstream of the NKCC1/KCC2 pathway. CONCLUSIONS Our findings provide evidence that isoflurane administration in neonatal rats leads to persistent cognitive deficits through dysregulation of the Cation-Chloride Cotransporters NKCC1 and KCC2, BDNF, p-ERK proteins, as well as neuroinflammatory processes.
Collapse
Affiliation(s)
- Dongni Xu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Jiayi Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Shiyu Meng
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Meixian Sun
- The Eighth People's Hospital of Qingdao, Qingdao, Shandong Province, China
| | - Yuqing Chen
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China.
| | - Yu Hong
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China.
| |
Collapse
|
6
|
Mei B, Xu X, Weng J, Yang Y, Wang P, Qiu G, Zhang C, Zhang Q, Lu Y, Liu X. Activating astrocytic α2A adrenoceptors in hippocampus reduces glutamate toxicity to attenuate sepsis-associated encephalopathy in mice. Brain Behav Immun 2024; 117:376-398. [PMID: 38320682 DOI: 10.1016/j.bbi.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/09/2023] [Accepted: 02/02/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Glutamate metabolism disorder is an important mechanism of sepsis-associated encephalopathy (SAE). Astrocytes regulate glutamate metabolism. In septic mice, α2A adrenoceptor (α2A-AR) activation in the central nervous system provides neuroprotection. α2A-ARs are expressed abundantly in hippocampal astrocytes. This study was performed to determine whether hippocampal astrocytic α2A-AR activation confers neuroprotection against SAE and whether this protective effect is astrocyte specific and achieved by the modulation of glutamate metabolism. METHODS Male C57BL/6 mice with and without α2A-AR knockdown were subjected to cecal ligation and puncture (CLP). They were treated with intrahippocampal guanfacine (an α2A-AR agonist) or intraperitoneal dexmedetomidine in the presence or absence of dihydrokainic acid [DHK; a glutamate transporter 1 (GLT-1) antagonist] and/or UCPH-101 [a glutamate/aspartate transporter (GLAST) antagonist]. Hippocampal tissue was collected for the measurement of astrocyte reactivity, GLT-1 and GLAST expression, and glutamate receptor subunit 2B (GluN2B) phosphorylation. In vivo real-time extracellular glutamate concentrations in the hippocampus were measured by ultra-performance liquid chromatography tandem mass spectrometry combined with microdialysis, and in vivo real-time hippocampal glutamatergic neuron excitability was assessed by calcium imaging. The mice were subjected to the Barnes maze and fear conditioning tests to assess their learning and memory. Golgi staining was performed to assess changes in the hippocampal synaptic structure. In vitro, primary astrocytes with and without α2A-AR knockdown were stimulated with lipopolysaccharide (LPS) and treated with guanfacine or dexmedetomidine in the presence or absence of 8-bromo- cyclic adenosine monophosphate (8-Br-cAMP, a cAMP analog). LPS-treated primary and BV2 microglia were also treated with guanfacine or dexmedetomidine. Astrocyte reactivity, PKA catalytic subunit, GLT-1 an GLAST expression were determined in primary astrocytes. Interleukin-1β, interleukin-6 and tumor necrosis factor-alpha in the medium of microglia culture were measured. RESULTS CLP induced synaptic injury, impaired neurocognitive function, increased astrocyte reactivity and reduced GLT-1 and GLAST expression in the hippocampus of mice. The extracellular glutamate concentration, phosphorylation of GluN2B at Tyr-1472 and glutamatergic neuron excitability in the hippocampus were increased in the hippocampus of septic mice. Intraperitoneal dexmedetomidine or intrahippocampal guanfacine administration attenuated these effects. Hippocampal astrocytes expressed abundant α2A-ARs; expression was also detected in neurons but not microglia. Specific knockdown of α2A-ARs in hippocampal astrocytes and simultaneous intrahippocampal DHK and UCPH-101 administration blocked the neuroprotective effects of dexmedetomidine and guanfacine. Intrahippocampal administration of DHK or UCPH-101 alone had no such effect. In vitro, guanfacine or dexmedetomidine inhibited astrocyte reactivity, reduced PKA catalytic subunit expression, and increased GLT-1 and GLAST expression in primary astrocytes but not in primary astrocytes that received α2A-AR knockdown or were treated with 8-Br-cAMP. Guanfacine or dexmedetomidine inhibited microglial reactivity in BV2 but not primary microglia. CONCLUSIONS Our results suggest that neurocognitive protection against SAE after hippocampal α2A-AR activation is astrocyte specific. This protection may involve the inhibition of astrocyte reactivity and alleviation of glutamate neurotoxicity, thereby reducing synaptic injury. The cAMP/protein kinase A (PKA) signaling pathway is a potential cellular mechanism by which activating α2A-AR modulates astrocytic function.
Collapse
Affiliation(s)
- Bin Mei
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China.
| | - Xiaoxia Xu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Juntao Weng
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Yueyue Yang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Peng Wang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Gaolin Qiu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Chi Zhang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, 230001, China
| | - Qunlin Zhang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, 230001, China
| | - Yao Lu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China
| | - Xuesheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui Province, 230022, China.
| |
Collapse
|
7
|
Aldecoa C, Bettelli G, Bilotta F, Sanders RD, Aceto P, Audisio R, Cherubini A, Cunningham C, Dabrowski W, Forookhi A, Gitti N, Immonen K, Kehlet H, Koch S, Kotfis K, Latronico N, MacLullich AMJ, Mevorach L, Mueller A, Neuner B, Piva S, Radtke F, Blaser AR, Renzi S, Romagnoli S, Schubert M, Slooter AJC, Tommasino C, Vasiljewa L, Weiss B, Yuerek F, Spies CD. Update of the European Society of Anaesthesiology and Intensive Care Medicine evidence-based and consensus-based guideline on postoperative delirium in adult patients. Eur J Anaesthesiol 2024; 41:81-108. [PMID: 37599617 PMCID: PMC10763721 DOI: 10.1097/eja.0000000000001876] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Postoperative delirium (POD) remains a common, dangerous and resource-consuming adverse event but is often preventable. The whole peri-operative team can play a key role in its management. This update to the 2017 ESAIC Guideline on the prevention of POD is evidence-based and consensus-based and considers the literature between 01 April 2015, and 28 February 2022. The search terms of the broad literature search were identical to those used in the first version of the guideline published in 2017. POD was defined in accordance with the DSM-5 criteria. POD had to be measured with a validated POD screening tool, at least once per day for at least 3 days starting in the recovery room or postanaesthesia care unit on the day of surgery or, at latest, on postoperative day 1. Recent literature confirmed the pathogenic role of surgery-induced inflammation, and this concept reinforces the positive role of multicomponent strategies aimed to reduce the surgical stress response. Although some putative precipitating risk factors are not modifiable (length of surgery, surgical site), others (such as depth of anaesthesia, appropriate analgesia and haemodynamic stability) are under the control of the anaesthesiologists. Multicomponent preoperative, intra-operative and postoperative preventive measures showed potential to reduce the incidence and duration of POD, confirming the pivotal role of a comprehensive and team-based approach to improve patients' clinical and functional status.
Collapse
Affiliation(s)
- César Aldecoa
- From the Department of Anaesthesia and Postoperative Critical Care, Hospital Universitario Rio Hortega, Valladolid, Spain (CA), Department of Biomedical Studies, University of the Republic of San Marino, San Marino (GB), Department of Anesthesiology, Critical Care and Pain Medicine, 'Sapienza' University of Rome, Rome, Italy (FB, AF, LM), Specialty of Anaesthetics & NHMRC Clinical Trials Centre, University of Sydney & Department of Anaesthetics and Institute of Academic Surgery, Royal Prince Alfred Hospital (RDS), Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt Universität zu Berlin, Campus Charité Mitte, and Campus Virchow Klinikum (CDS, SK, AM, BN, LV, BW, FY), Dipartimento di Scienze dell'Emergenza, Anestesiologiche e della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (PA), Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy (PA), Department of Surgery, Institute of Clinical Sciences, Sahlgrenska University Hospital, Göteborg, Sweden (RA), Geriatria, Accettazione Geriatrica e Centro di ricerca per l'invecchiamento, IRCCS INRCA, Ancona, Italy (AC), School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland (CC), First Department of Anaesthesiology and Intensive Care Medical University of Lublin, Poland (WD), Research Unit of Nursing Science and Health Management, University of Oulu, Oulu, Finland (KI), Section of Surgical Pathophysiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (HK), Department of Anesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University in Szczecin, Poland (KK), Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia (NG, NL, SP, SR), Department of Anesthesia, Critical Care and Emergency, Spedali Civili University Hospital, Brescia, Italy (NL, SP), Edinburgh Delirium Research Group, Ageing and Health, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (AMJM), Department of Anaesthesia and Intensive Care, Nykoebing Hospital; University of Southern Denmark, SDU (SK, FR), Department of Anaesthesiology and Intensive Care, University of Tartu, Tartu, Estonia (ARB), Center for Intensive Care Medicine, Luzerner Kantonsspital, Lucerne, Switzerland (ARB), Department of Health Science, Section of Anesthesiology, University of Florence (SR), Department of Anaesthesia and Critical Care, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy (SR), School of Health Sciences, Institute of Nursing, ZHAW Zurich University of Applied Science, Winterthur, Switzerland (MS), Departments of Psychiatry and Intensive Care Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands (AJCS), Department of Neurology, UZ Brussel and Vrije Universiteit Brussel, Brussels, Belgium (AJCS) and Dental Anesthesia and Intensive Care Unit, Polo Universitario Ospedale San Paolo, Department of Biomedical, Surgical and Odontoiatric Sciences, University of Milano, Milan, Italy (CT)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Meng H, Wei JH, Yu PZ, Ren JX, Tang MY, Sun JY, Yan XY, Su J. Insights into Advanced Neurological Dysfunction Mechanisms Following DBS Surgery in Parkinson's Patients: Neuroinflammation and Pyroptosis. Curr Issues Mol Biol 2023; 45:4480-4494. [PMID: 37232753 DOI: 10.3390/cimb45050284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023] Open
Abstract
Parkinson's disease is a severe neurodegenerative disorder. Currently, deep brain electrical stimulation (DBS) is the first line of surgical treatment. However, serious neurological impairments such as speech disorders, disturbances of consciousness, and depression after surgery limit the efficacy of treatment. In this review, we summarize the recent experimental and clinical studies that have explored the possible causes of neurological deficits after DBS. Furthermore, we tried to identify clues from oxidative stress and pathological changes in patients that could lead to the activation of microglia and astrocytes in DBS surgical injury. Notably, reliable evidence supports the idea that neuroinflammation is caused by microglia and astrocytes, which may contribute to caspase-1 pathway-mediated neuronal pyroptosis. Finally, existing drugs and treatments may partially ameliorate the loss of neurological function in patients following DBS surgery by exerting neuroprotective effects.
Collapse
Affiliation(s)
- Hao Meng
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Jia-Hang Wei
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Peng-Zheng Yu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Jia-Xin Ren
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Meng-Yao Tang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Jun-Yi Sun
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Xiao-Yu Yan
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Jing Su
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| |
Collapse
|
9
|
Wang LF, Liang WD, Wang BY, Guo ML, Zhou JS, Chen L, Zhong ML, Ye JM. Transcutaneous electrical acupoint stimulation for reducing cognitive dysfunction in lumbar spine surgery: A randomized, controlled trail. Front Aging Neurosci 2022; 14:1034998. [PMID: 36545028 PMCID: PMC9760873 DOI: 10.3389/fnagi.2022.1034998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/21/2022] [Indexed: 12/07/2022] Open
Abstract
Objective This study aimed to evaluate the effect of perioperative transcutaneous electrical acupoint stimulation (TEAS) on postoperative cognitive dysfunction (POCD) in older patients with lumbar spine surgery. Methods Older patients (aged 60-80 years old) receiving lumbar spine surgery under general anesthesia were randomly divided into group A, 3-day intervention group; group B, 7-day intervention group; control group C, sham TEAS group, selected "Baihui" (GV 20) and "Dazhui" (GV 14) point was intervened once 30 min before operation with "HANS" transcutaneous electrical stimulation device, and then once a day after operation for 30 min each time. The primary outcome was the incidence of postoperative cognitive impairment assessed by the use of the Mini Mental Rating Scale (MMSE), patients developed POCD according to the Z score method. The secondary outcome was serum interleukin-6 (IL-6), tumor Necrosis factor α (TNF-α), neuron-specific enolase (NSE), and S100β protein levels. Results Three days after surgery, the incidence of POCD in groups A((22.4%)) and B ((18.3%)) were lower than those in group C ((42.9%)) (P < 0.05). There was no significant difference between groups A and B (P > 0.05). Seven days after surgery, the incidence of POCD in group B (18.3%) was lower than that in groups A (26.5%) and B (42.9%), and the comparison between groups B and C was statistically significant (P < 0.05). On the 3rd and 7th days after surgery, the levels of IL-6, TNF-α, NSE, and S100β in the two TEAS groups were lower than those in the sham TEAS group (P < 0.01), but higher than the preoperative levels in the three groups (P < 0.01). Conclusion It seems that Perioperative TEAS intervention could reduce the level of inflammatory factors IL-6, TNF-α in the blood of older patients with lumbar spine surgery, and reduce the incidence of POCD. Clinical trial registration www.chictr.org.cn, identifier ChiCTR2200063030.
Collapse
Affiliation(s)
- Li-feng Wang
- Department of Anesthesiology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China,Department of Anesthesiology, First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Wei-dong Liang
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Bing-yu Wang
- Department of Anesthesiology, Gannan Medical College, Ganzhou, Jiangxi, China
| | - Ming-ling Guo
- Department of Anesthesiology, Gannan Medical College, Ganzhou, Jiangxi, China
| | - Jian-shun Zhou
- Department of Anesthesiology, Gannan Medical College, Ganzhou, Jiangxi, China
| | - Li Chen
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Mao-lin Zhong
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Jun-ming Ye
- Department of Anesthesiology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China,Department of Anesthesiology, Gannan Medical College, Ganzhou, Jiangxi, China,*Correspondence: Jun-ming Ye,
| |
Collapse
|
10
|
Zhang J, Liu Y, Li H, Hu Y, Yu S, Liu Q, Chen Y. Stellate Ganglion Block Improves Postoperative Cognitive Dysfunction in aged rats by SIRT1-mediated White Matter Lesion Repair. Neurochem Res 2022; 47:3838-3853. [DOI: 10.1007/s11064-022-03800-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/13/2022] [Accepted: 10/16/2022] [Indexed: 12/04/2022]
|
11
|
Huang R, Han S, Qiu Y, Zhou T, Wu Y, Du H, Xu J, Wei X. Glucocorticoid regulation of lactate release from spinal astrocytes contributes to the induction of spinal LTP of C-fiber-evoked field potentials and the development of mechanical allodynia. Neuropharmacology 2022; 219:109253. [PMID: 36108796 DOI: 10.1016/j.neuropharm.2022.109253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/28/2022] [Accepted: 09/06/2022] [Indexed: 10/31/2022]
Abstract
High-frequency stimulation (HFS) of the sciatic nerve leads to long-term potentiation (LTP) at C-fiber synapse and long-lasting pain hypersensitivity. The underlying mechanisms, however, are still unclear. In the present study, we investigated the involvement of astrocytes derived l-lactate in the spinal dorsal horn subsequent to glucocorticoid (GC) secretion into the plasma in this process using Sprague-Dawley rats and Aldh1L1-CreERT2 mice of either sex. We found that HFS increased l-lactate and monocarboxylate transporters 1/2 (MCT1/2) in the spinal dorsal horn. Inhibition of glycogenolysis or blocking lactate transport prevented the induction of spinal LTP following HFS. Furthermore, Chemogenetical inhibition of dorsal horn astrocytes, which were activated by HFS, prevented spinal LTP, alleviated the mechanical allodynia and the decreased the level l-lactate and GFAP expression in the dorsal horn following HFS. In contrast, Chemogenetics activation of dorsal horn astrocytes in naïve rats induced spinal LTP as well as mechanical allodynia, and increased GFAP expression and l-lactate. Application of l-lactate directly to the spinal cord of naïve rats induced spinal LTP, mechanical allodynia, and increased spinal expression of p-ERK. Importantly, HFS increased GC in the plasma and glucocorticoid receptor (GR) expression in spinal astrocytes, adrenalectomy or knocking down of GR in astrocytes by using Cre-Loxp system blocked the mechanical allodynia, prevented the spinal LTP and the enhancement of lactate after HFS. These results show that lactate released from spinal astrocytes following glucocorticoid release into the plasma enhance synaptic transmission at the C-fiber synapse and underlie pain chronicity.
Collapse
Affiliation(s)
- Ruizhen Huang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuang Han
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuxin Qiu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Taihe Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuning Wu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hongchun Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Jing Xu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China; Center for Laboratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xuhong Wei
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
12
|
He W, Xie X, Li C, Ding H, Ye J. Adenosine A2A Receptor Antagonist Improves Cognitive Impairment by Inhibiting Neuroinflammation and Excitatory Neurotoxicity in Chronic Periodontitis Mice. Molecules 2022; 27:molecules27196267. [PMID: 36234803 PMCID: PMC9571030 DOI: 10.3390/molecules27196267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022] Open
Abstract
The adenosine A2A receptor antagonist SCH58261 has been reported to have anti-inflammatory effects. However, its role in chronic periodontitis (CP)-induced cognitive impairment, which is associated with Porphyromonas gingivalis lipopolysaccharide (P. gingivalis LPS), remains unclear. This study investigated the role of SCH58261 in mice with CP-induced cognitive impairment. C57BL/6J mice were used to develop CP model by injecting 0.5 mg/kg P. gingivalis LPS into the palatal gingival sulcus of maxillary first molars twice a week for four weeks. The mice were divided into control, P. gingivalis LPS (P-LPS), P-LPS + SCH58261, and SCH58261 groups. The passive avoidance test (PAT) and Morris water maze (MWM) were used to assess cognition in mice. Furthermore, CD73/adenosine, neuroinflammation, glutamate transporters, and glutamate were assessed. Compared with the P-LPS group, 0.1 and 0.5 mg/kg SCH58261 increased latency and decreased error times in PAT, but increased platform crossing number in MWM. SCH58261 inhibited microglial activation, and decreased pro-inflammatory cytokines and glutamate levels, but increased GLT-1 and PSD95 expression in the hippocampus. This was the first report of SCH58261 treatment for CP-induced cognitive impairment, which may be related to its anti-inflammatory activities and anti-glutamate excitatory neurotoxicity. This suggests that SCH58261 can be used as a novel agent to treat cognitive impairment.
Collapse
Affiliation(s)
- Wendan He
- Department of Stomatology, The Affiliated Hospital of Wuhan Traditional Chinese and Western Medicine, Tongji Medical College of HUST, Wuhan 430022, China
- Correspondence: ; Tel.: +86-027-8586-0666
| | - Xianlong Xie
- Department of General Practice, The Affiliated Hospital of Wuhan Traditional Chinese and Western Medicine, Tongji Medical College of HUST, Wuhan 430022, China
| | - Chenxi Li
- Laboratory for Tumor Genetics and Regenerative Medicine, Department of Oral and Maxillofacial Surgery, The Head and Neurocenter, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Huang Ding
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan 430060, China
| | - Jishi Ye
- Department of Pain, Renmin Hospital, Wuhan University, Wuhan 430060, China
| |
Collapse
|
13
|
Zhang G, Wang F, Ran Y, Liu D. Applications of the ultrasound-guided nerve block technique for nonanalgesic effects. IBRAIN 2022; 8:389-400. [PMID: 37786735 PMCID: PMC10528970 DOI: 10.1002/ibra.12061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/28/2022] [Accepted: 07/31/2022] [Indexed: 10/04/2023]
Abstract
The nerve block technique guided by ultrasound has been able to accurately block tiny nerves throughout the body in recent years. It has been increasingly used to treat multisystem diseases or analgesia in surgical patients, but the latter accounted for the vast majority of cases. The nonanalgesic effect of nerve blocks is also in wide demand. After searching ultrasound-guided nerve block works on the PubMed database, we systematically summarized the current clinical application of the nerve block technique and the unique role and related mechanism of nerve block in the prevention and treatment of multi-system diseases or symptoms, including disorders of the circulatory and respiratory systems, postoperative cognitive dysfunction, immune function, posttraumatic stress disorder, and postoperative digestive system, to put forward the potential prospective application in future and serve as a reference for future research of nerve block therapy in these diseases mentioned.
Collapse
Affiliation(s)
- Guang‐Ting Zhang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunYiGuizhouChina
| | - Feng‐Lin Wang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunYiGuizhouChina
| | - Ying Ran
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunYiGuizhouChina
| | - De‐Xing Liu
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunYiGuizhouChina
| |
Collapse
|
14
|
Suo Z, Yang J, Zhou B, Qu Y, Xu W, Li M, Xiao T, Zheng H, Ni C. Whole-transcriptome sequencing identifies neuroinflammation, metabolism and blood-brain barrier related processes in the hippocampus of aged mice during perioperative period. CNS Neurosci Ther 2022; 28:1576-1595. [PMID: 35899365 PMCID: PMC9437242 DOI: 10.1111/cns.13901] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 11/28/2022] Open
Abstract
AIM Perioperative neurocognitive disorders (PND) occur frequently after surgery and anesthesia, especially in aged patients. Previous studies have shown multiple PND related mechanisms in the hippocampus; however, their relationships remain unclear. Meanwhile, the perioperative neuropathological processes are sophisticated and changeable, single period study could not reveal the accurate mechanisms. Thus, multiperiod whole-transcriptome study is necessary to elucidate the gene expression patterns during perioperative period. METHODS Aged C57BL/6 mice were subjected to exploratory laparotomy under sevoflurane anesthesia. Whole-transcriptome sequencing (RNA-seq analysis) was performed on the hippocampi from control condition (Con), 30 min (Day0), 2 days (Day2), and 7 days (Day7) after surgery. Gene Ontology/Kyoto Encyclopedia of Genes and Genomes analyses, quantitative real-time PCR, immunofluorescence, and fear conditioning test were also performed to elucidate the pathological processes and modulation networks during the period. RESULTS Through RNA-seq analysis, 328, 3597, and 4179 differentially expressed genes (DEGs) were screened out in intraoperative period (Day0 vs. Con), early postoperative period (Day2 vs. Day0), and late postoperative period (Day7 vs. Day2). The involved GO biological processes were divided into 9 categories, and positive-regulated processes were more than negative-regulated ones. Seventy-four transcription factors were highlighted. The potential synaptic and neuroinflammatory pathways were constructed for Neurotransmitter, Synapse and Neuronal alteration categories with 9 genes (Htr1a, Rims1, and Ezh2, etc.). The metabolic and mitochondrial pathways were constructed for metabolism, oxidative stress, and biological rhythm categories with 9 genes (Gpld1, Sirt1, and Cry2, etc.). The blood-brain barrier and neurotoxicity related pathways were constructed for blood-brain barrier, neurotoxicity, and cognitive function categories with 10 genes (Mmp2, Itpr1, and Nrf1, etc.). CONCLUSION The results revealed gene expression patterns and modulation networks in the aged hippocampus during perioperative period, which provide insights into overall mechanisms and potential therapeutic targets for prevention and treatment of perioperative central nervous system diseases, such as PND, from the genetic level.
Collapse
Affiliation(s)
- Zizheng Suo
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Bowen Zhou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinyin Qu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Wenjie Xu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Ting Xiao
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Ni
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Chen X, Qiao WH, Cao H, Shi JW, Du XL, Dong NG. Role of Neuroimmune Interactions in COVID-19-related Cardiovascular Damage. Curr Med Sci 2022; 42:555-560. [PMID: 35678914 PMCID: PMC9178934 DOI: 10.1007/s11596-022-2529-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/07/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has caused a global pandemic impacting over 200 countries/regions and more than 200 million patients worldwide. Among the infected patients, there is a high prevalence of COVID-19-related cardiovascular injuries. However, the specific mechanisms linking cardiovascular damage and COVID-19 remain unclear. The COVID-19 pandemic also has exacerbated the mental health burden of humans. Considering the close association between neuroimmune interactions and cardiovascular disease, this review assessed the complex pathophysiological mechanisms connecting neuroimmune interactions and cardiovascular disease. It was revealed that the mental health burden might be a pivotal accomplice causing COVID-19-associated cardiovascular damage. Specifically, the proinflammatory status of patients with a terrible mood state is closely related to overdrive of the hypothalamus-pituitary-adrenal (HPA) axis, sympathovagal imbalance, and endothelial dysfunction, which lead to an increased risk of developing cardiovascular injury during COVID-19. Therefore, during the prevention and treatment of cardiovascular complications in COVID-19 patients, particular attention should be given to relieve the mental health burden of these patients.
Collapse
Affiliation(s)
- Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Wei-hua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Hong Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Jia-wei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Xin-ling Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Nian-guo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| |
Collapse
|
16
|
Miller-Rhodes P, Li H, Velagapudi R, Chiang W, Terrando N, Gelbard HA. URMC-099 prophylaxis prevents hippocampal vascular vulnerability and synaptic damage in an orthopedic model of delirium superimposed on dementia. FASEB J 2022; 36:e22343. [PMID: 35535564 PMCID: PMC9175136 DOI: 10.1096/fj.202200184rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/11/2022]
Abstract
Systemic perturbations can drive a neuroimmune cascade after surgical trauma, including affecting the blood-brain barrier (BBB), activating microglia, and contributing to cognitive deficits such as delirium. Delirium superimposed on dementia (DSD) is a particularly debilitating complication that renders the brain further vulnerable to neuroinflammation and neurodegeneration, albeit these molecular mechanisms remain poorly understood. Here, we have used an orthopedic model of tibial fracture/fixation in APPSwDI/mNos2-/- AD (CVN-AD) mice to investigate relevant pathogenetic mechanisms underlying DSD. We conducted the present study in 6-month-old CVN-AD mice, an age at which we speculated amyloid-β pathology had not saturated BBB and neuroimmune functioning. We found that URMC-099, our brain-penetrant anti-inflammatory neuroprotective drug, prevented inflammatory endothelial activation, breakdown of the BBB, synapse loss, and microglial activation in our DSD model. Taken together, our data link post-surgical endothelial activation, microglial MafB immunoreactivity, and synapse loss as key substrates for DSD, all of which can be prevented by URMC-099.
Collapse
Affiliation(s)
- Patrick Miller-Rhodes
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA.,Neuroscience Graduate Program, University of Rochester Medical Center, Rochester, New York, USA
| | - Herman Li
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Ravikanth Velagapudi
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Wesley Chiang
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, USA
| | - Niccolò Terrando
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, North Carolina, USA.,Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Harris A Gelbard
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA.,Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA.,Department of Neuroscience, University of Rochester Medical Center, Rochester, New York, USA.,Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA.,Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
17
|
Connexin 43 gap junction-mediated astrocytic network reconstruction attenuates isoflurane-induced cognitive dysfunction in mice. J Neuroinflammation 2022; 19:64. [PMID: 35255943 PMCID: PMC8903726 DOI: 10.1186/s12974-022-02424-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/23/2022] [Indexed: 01/18/2023] Open
Abstract
Background Postoperative cognitive dysfunction (POCD) is a common complication following anesthesia and surgery. General anesthetic isoflurane has potential neurotoxicity and induces cognitive impairments, but the exact mechanism remains unclear. Astrocytes form interconnected networks in the adult brain through gap junctions (GJs), which primarily comprise connexin 43 (Cx43), and play important roles in brain homeostasis and functions such as memory. However, the role of the GJ-Cx43-mediated astrocytic network in isoflurane-induced cognitive dysfunction has not been defined. Methods 4-month-old male C57BL/6 mice were exposure to long-term isoflurane to induce cognitive impairment. To simulate an in vitro isoflurane-induced cognitive dysfunction‐like condition, primary mouse astrocytes were subjected to long-term isoflurane exposure. Cognitive function was assessed by Y-maze and fear conditioning tests. Western blot was used to determine the expression levels of different functional configurations of Cx43. The morphology of the GJs-Cx43 was evaluated by immunofluorescence staining. Levels of IL-1β and IL-6 were examined by ELISA. The ability of GJs-Cx43-mediated intercellular communication was examined by lucifer yellow dye transfer assay. Ethidium bromide uptake assays were used to measure the activity of Cx43 hemichannels. The ultrastructural morphology of astrocyte gap junctions and tripartite synapse were observed by transmission electron microscopy. Results After long-term isoflurane anesthesia, the GJs formed by Cx43 in the mouse hippocampus and primary mouse astrocytes were significantly reduced, GJs function was impaired, hemichannel activity was enhanced, the levels of IL-1β and IL-6 were increased, and mice showed significant cognitive impairment. After treatment with the novel GJ-Cx43 enhancer ZP1609, GJ-Cx43-mediated astrocytic network function was enhanced, neuroinflammation was alleviated, and ameliorated cognition dysfunction induced by long-term isoflurane exposure. However, ZP1609 enhances the astrocytic network by promoting Cx43 to form GJs without affecting hemichannel activity. Additionally, our data showed that long-term isoflurane exposure does not alter the structure of tripartite synapse. Conclusion Our results reveal a novel mechanism of the GJ-Cx43-mediated astrocytic network involved in isoflurane-induced neuroinflammation and cognitive impairments, which provides new mechanistic insight into the pathogenesis of POCD and identifies potential targets for its treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02424-y.
Collapse
|
18
|
Rosén A, Gennser M, Oscarsson N, Kvarnström A, Sandström G, Seeman-Lodding H, Simrén J, Zetterberg H. Protein tau concentration in blood increases after SCUBA diving: an observational study. Eur J Appl Physiol 2022; 122:993-1005. [PMID: 35142945 PMCID: PMC8926952 DOI: 10.1007/s00421-022-04892-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/05/2022] [Indexed: 11/27/2022]
Abstract
Purpose It is speculated that diving might be harmful to the nervous system. The aim of this study was to determine if established markers of neuronal injury were increased in the blood after diving. Methods Thirty-two divers performed two identical dives, 48 h apart, in a water-filled hyperbaric chamber pressurized to an equivalent of 42 m of sea water for 10 min. After one of the two dives, normobaric oxygen was breathed for 30 min, with air breathed after the other. Blood samples were obtained before and at 30–45 and 120 min after diving. Concentrations of glial fibrillary acidic, neurofilament light, and tau proteins were measured using single molecule array technology. Doppler ultrasound was used to detect venous gas emboli. Results Tau was significantly increased at 30–45 min after the second dive (p < 0.0098) and at 120 min after both dives (p < 0.0008/p < 0.0041). Comparison of matching samples showed that oxygen breathing after diving did not influence tau results. There was no correlation between tau concentrations and the presence of venous gas emboli. Glial fibrillary acidic protein was decreased 30–45 min after the first dive but at no other point. Neurofilament light concentrations did not change. Conclusions Tau seems to be a promising marker of dive-related neuronal stress, which is independent of the presence of venous gas emboli. Future studies could validate these results and determine if there is a quantitative relationship between dive exposure and change in tau blood concentration. Supplementary Information The online version contains supplementary material available at 10.1007/s00421-022-04892-9.
Collapse
Affiliation(s)
- Anders Rosén
- Department of Anaesthesia and Intensive Care Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Anaesthesia and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Mikael Gennser
- Swedish Aerospace Physiology Centre, Division of Environmental Physiology, Department of Biomedical Engineering and Health Systems, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology, KTH, Stockholm, Sweden
| | - Nicklas Oscarsson
- Department of Anaesthesia and Intensive Care Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Anaesthesia and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Kvarnström
- Department of Anaesthesia and Intensive Care Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Anaesthesia and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Göran Sandström
- Department of Anaesthesia and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Swedish Armed Forces, Center for Defence Medicine, Gothenburg, Sweden
| | - Helen Seeman-Lodding
- Department of Anaesthesia and Intensive Care Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Anaesthesia and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Simrén
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| |
Collapse
|
19
|
Mkrtchian S, Ebberyd A, Veerman RE, Méndez-Lago M, Gabrielsson S, Eriksson LI, Gómez-Galán M. Surgical Trauma in Mice Modifies the Content of Circulating Extracellular Vesicles. Front Immunol 2022; 12:824696. [PMID: 35116043 PMCID: PMC8804340 DOI: 10.3389/fimmu.2021.824696] [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: 11/29/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Surgical interventions rapidly trigger a cascade of molecular, cellular, and neural signaling responses that ultimately reach remote organs, including the brain. Using a mouse model of orthopedic surgery, we have previously demonstrated hippocampal metabolic, structural, and functional changes associated with cognitive impairment. However, the nature of the underlying signals responsible for such periphery-to-brain communication remains hitherto elusive. Here we present the first exploratory study that tests the hypothesis of extracellular vesicles (EVs) as potential mediators carrying information from the injured tissue to the distal organs including the brain. The primary goal was to investigate whether the cargo of circulating EVs after surgery can undergo quantitative changes that could potentially trigger phenotypic modifications in the target tissues. EVs were isolated from the serum of the mice subjected to a tibia surgery after 6, 24, and 72 h, and the proteome and miRNAome were investigated using mass spectrometry and RNA-seq approaches. We found substantial differential expression of proteins and miRNAs starting at 6 h post-surgery and peaking at 24 h. Interestingly, one of the up-regulated proteins at 24 h was α-synuclein, a pathogenic hallmark of certain neurodegenerative syndromes. Analysis of miRNA target mRNA and corresponding biological pathways indicate the potential of post-surgery EVs to modify the extracellular matrix of the recipient cells and regulate metabolic processes including fatty acid metabolism. We conclude that surgery alters the cargo of circulating EVs in the blood, and our results suggest EVs as potential systemic signal carriers mediating remote effects of surgery on the brain.
Collapse
Affiliation(s)
- Souren Mkrtchian
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anette Ebberyd
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rosanne E. Veerman
- Department of Clinical Immunology and Transfusion Medicine and Division of Immunology and Allergy, Department of Medicine, Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - María Méndez-Lago
- Genomics Core Facility, Institute of Molecular Biology gGmbH (IMB), Mainz, Germany
| | - Susanne Gabrielsson
- Department of Clinical Immunology and Transfusion Medicine and Division of Immunology and Allergy, Department of Medicine, Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Lars I. Eriksson
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
- Function Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Marta Gómez-Galán
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Marta Gómez-Galán,
| |
Collapse
|
20
|
Morcuende A, García-Gutiérrez MS, Tambaro S, Nieto E, Manzanares J, Femenia T. Immunomodulatory Role of CB2 Receptors in Emotional and Cognitive Disorders. Front Psychiatry 2022; 13:866052. [PMID: 35492718 PMCID: PMC9051035 DOI: 10.3389/fpsyt.2022.866052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Emotional behavior, memory, and learning have been associated with alterations in the immune system in neuropsychiatric and neurodegenerative diseases. In recent years, several studies pointed out the involvement of the cannabinoid receptor 2 (CB2r) in the immune system and the regulation of inflammation. This receptor is widely distributed in different tissues and organs with higher expression in spleen and immune system cells. However, CB2r has also been detected in several brain areas and different brain cell types, such as neurons and glia. These findings suggest that CB2r may closely relate the immune system and the brain circuits regulating inflammation, mood, and cognitive functions. Therefore, we review the studies that may help elucidate the molecular bases of CB2r in regulating inflammation in different brain cells and its role in the pathophysiology of psychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Alvaro Morcuende
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (CSIC), Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (CSIC), Alicante, Spain.,Redes de Investigación Cooperativa Orientada a Resultados en Salud, Red de Investigación en Atención Primaria de Adicciones, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (MICINN) and Fondo Europeo de Desarrollo Regional (FEDER), Madrid, Spain.,Instituto de Investigación Sanitaria y Biomédica de Alicante, Alicante, Spain
| | - Simone Tambaro
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Elena Nieto
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (CSIC), Alicante, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (CSIC), Alicante, Spain.,Redes de Investigación Cooperativa Orientada a Resultados en Salud, Red de Investigación en Atención Primaria de Adicciones, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (MICINN) and Fondo Europeo de Desarrollo Regional (FEDER), Madrid, Spain.,Instituto de Investigación Sanitaria y Biomédica de Alicante, Alicante, Spain
| | - Teresa Femenia
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (CSIC), Alicante, Spain.,Redes de Investigación Cooperativa Orientada a Resultados en Salud, Red de Investigación en Atención Primaria de Adicciones, Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación (MICINN) and Fondo Europeo de Desarrollo Regional (FEDER), Madrid, Spain
| |
Collapse
|
21
|
Chen L, Yang N, Li Y, Li Y, Hong J, Wang Q, Liu K, Han D, Han Y, Mi X, Shi C, Zhou Y, Li Z, Liu T, Guo X. Cholecystokinin octapeptide improves hippocampal glutamatergic synaptogenesis and postoperative cognition by inhibiting induction of A1 reactive astrocytes in aged mice. CNS Neurosci Ther 2021; 27:1374-1384. [PMID: 34402181 PMCID: PMC8504528 DOI: 10.1111/cns.13718] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 11/28/2022] Open
Abstract
Aims Delayed neurocognitive recovery (dNCR) is a common postoperative complication in geriatric surgical patients for which there is no efficacious therapy. Cholecystokinin octapeptide (CCK‐8), an immunomodulatory peptide, regulates memory and learning. Here, we explored the effects and mechanism of action of CCK‐8 on dNCR. Methods We applied laparotomy to establish a model of dNCR in aged mice. Morris water maze and fear conditioning tests were used to evaluate cognition. Immunofluorescence was used to detect the density of CCK‐8, A1 reactive astrocytes, glutamatergic synapses, and activation of microglia in the hippocampus. Quantitative PCR was performed to determine mRNA levels of synapse‐associated factors. A1 reactive astrocytes, activated microglia, and glutamatergic synapse‐associated protein levels in the hippocampus were assessed by western blotting. Results Administration of CCK‐8 suppressed the activation of microglia, the induction of A1 reactive astrocytes, and the expression of tumor necrosis factor alpha, complement 1q, and interleukin 1 alpha in the hippocampus. Furthermore, it promoted glutamatergic synaptogenesis and neurocognitive recovery in aged dNCR model mice. Conclusion Our findings indicated that CCK‐8 alleviated cognitive impairment and promoted glutamatergic synaptogenesis by inhibiting the induction of A1 reactive astrocytes and the activation of microglia. CCK‐8 is, therefore, a potential therapeutic target for dNCR.
Collapse
Affiliation(s)
- Lei Chen
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yitong Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jingshu Hong
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Qian Wang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Kaixi Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yongzheng Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Chengmei Shi
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Ying Zhou
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
22
|
Liang Z, Xu X, Qi X, Zhang F. Efficacy and safety of ulinastatin on cognitive dysfunction after general anesthesia in elderly patients: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e24814. [PMID: 33787575 PMCID: PMC8021364 DOI: 10.1097/md.0000000000024814] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND With the aging of society, the incidence of diseases increases. And along with the increase of surgery rate, the number of elderly patients with postoperative cognitive dysfunction (POCD) is also increasing. POCD seriously affects the mental state and quality of life of patients and their families. Clinical studies have shown that POCD is closely related to inflammatory reaction, and Ulinastatin can inhibit the inflammatory reaction and reduce the incidence of POCD in elderly patients under general anesthesia. However. the effect of Ulinastatin on POCD in elderly patients under general anesthesia has not been systematically evaluated. OBJECTIVE Meta analysis will be used to evaluate the efficacy and safety of Ulinastatin in elderly patients with general anesthesia POCD during perioperative period. METHODS We will search China Science and Technology Journal Database Chinese database, China National Knowledge Infrastructure, Wanfang, China biomedical database, PubMed, EMBASE, Cochrane Library and web of science for randomized controlled trials of the effect of Ulinastatin on POCD of elderly patients with general anesthesia from the establishment of the database to November 2020. The 2 researchers will independently screen the literature and conducted quality assessment and data extraction for the included studies, Revman5.3 software will be used for risk assessment and meta analysis. RESULTS In this study, the efficacy and safety of Ulinastatin in elderly patients with general anesthesia POCD will be evaluated by the incidence of postoperative cognitive impairment, mini mental state examination (Mini-Mental State Examination [MMSE]), visual regeneration, associative memory score, S100 β protein, tumor necrosis factor α (TNF- α), interleukin 6 (IL-6), IL- 10 inflammatory factors and the incidence of adverse reactions. CONCLUSION The use of Ulinastatin in perioperative period can significantly reduce the inflammatory level of elderly patients after general anesthesia, effectively prevent the occurrence of POCD and reduce its incidence. ETHICS AND DISSEMINATION The private information from individuals will not be published. This systematic review also will not involve endangering participant rights. Ethical approval is not required. The results may be published in a peer-reviewed journal or disseminated in relevant conferences. OSF REGISTRATION NUMBER DOI 10.17605/OSF.IO/GY3V7.
Collapse
|
23
|
Chen SM, Li M, Xie J, Li S, Xiang SS, Liu HY, Chen Z, Zhang P, Kuang X, Tang XQ. Hydrogen sulfide attenuates postoperative cognitive dysfunction through promoting the pathway of Warburg effect-synaptic plasticity in hippocampus. Toxicol Appl Pharmacol 2020; 409:115286. [PMID: 33068621 DOI: 10.1016/j.taap.2020.115286] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/01/2020] [Accepted: 10/12/2020] [Indexed: 01/03/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is deemed to a severe surgical complication without effective treatment. Previous work has confirmed the important modulatory role of hydrogen sulfide (H2S) in cognitive function. This study was proposed to explore whether H2S relieves POCD and the possible mechanisms. We demonstrated that NaHS (a donor of H2S) reversed the inhibited endogenous H2S generation in the hippocampus of postoperative rats. NaHS attenuated the cognitive impairment of postoperative rats in the Y-maze, Novel object recognition, and Morris water maze tests. NaHS enhanced the expressions of synaptic plasticity-related proteins, synapsin-1 and PSD-95, increased the synaptic density, and decreased the destruction of synaptic structures in the hippocampus of postoperative rats. Moreover, NaHS promoted Warburg effect in the hippocampus of postoperative rats, as reflected by increases in the expressions of hexokinase 2, pyruvate kinase M2, lactate dehydrogenase A, and pyruvate dehydrogenase kinase 1, an enhancement in the content of lactate, and a reduction in the expression of pyruvate dehydrogenase. The inhibitor of Warburg effect, 2-Deoxy-D-glucose (2-DG), not only reversed NaHS-enhanced Warburg effect in the hippocampus of postoperative rats, but also significantly abolished NaHS-exerted protective effect on cognitive function. Furthermore, 2-DG reversed NaHS-exerted enhancement in the expressions of synapsin-1 and PSD-95, increase in the synaptic density, and decrease in the destruction of synaptic structures in the hippocampus of postoperative rats. Collectively, these results indicate that H2S alleviates POCD through enhancing hippocampal Warburg effect, which subsequently improves synaptic plasticity in the hippocampus.
Collapse
Affiliation(s)
- Si-Min Chen
- Institute of Neurology, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, PR China; Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang 421001, Hunan, PR China
| | - Min Li
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang 421001, Hunan, PR China; Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang 421001, Hunan, PR China
| | - Juan Xie
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang 421001, Hunan, PR China; Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang 421001, Hunan, PR China
| | - Sha Li
- Institute of Neurology, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, PR China; Department of Anesthesiology, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, PR China
| | - Shi-Shi Xiang
- Institute of Neurology, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, PR China; Department of Anesthesiology, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, PR China
| | - Hai-Yao Liu
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang 421001, Hunan, PR China; Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang 421001, Hunan, PR China
| | - Zhuo Chen
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang 421001, Hunan, PR China; Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang 421001, Hunan, PR China
| | - Ping Zhang
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang 421001, Hunan, PR China; Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang 421001, Hunan, PR China.
| | - Xin Kuang
- Institute of Neurology, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, PR China; Department of Anesthesiology, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, PR China
| | - Xiao-Qing Tang
- Institute of Neurology, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, PR China; Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang 421001, Hunan, PR China.
| |
Collapse
|
24
|
lncRNAs Are Involved in Sevoflurane Anesthesia-Related Brain Function Modulation through Affecting Mitochondrial Function and Aging Process. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8841511. [PMID: 33354572 PMCID: PMC7735847 DOI: 10.1155/2020/8841511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 02/01/2023]
Abstract
Long noncoding RNAs (lncRNAs) play important roles in brain function modulation and neurodegenerative diseases. However, whether lncRNA regulations are involved in the mechanisms of perioperative neurocognitive disorders, especially in anesthesia-related brain dysfunction, remain unknown. Therefore, we explored the expression and regulation pattern profiles of lncRNAs in the hippocampus of aged rats after sevoflurane anesthesia. Three lncRNAs and 772 protein-coding genes were identified by microarray analysis and evidenced by in vitro and in vivo experiments as differentially expressed. Functional annotation and differentially expressed- (DE-) lncRNA-mRNA coexpression networks reveal that DE-lncRNAs are associated with mitochondrial dysfunction and oxidative stress, aging-related metabolism alterations, DNA damage, and apoptosis, as well as neurodegenerative features during sevoflurane anesthesia. These results suggest that lncRNAs play roles in general anesthesia-related brain function modulation during the perioperative context and provide insights into the lncRNA-related modulation mechanisms and targets.
Collapse
|
25
|
Biomarkers of neuronal damage in saturation diving-a controlled observational study. Eur J Appl Physiol 2020; 120:2773-2784. [PMID: 32975632 PMCID: PMC7674315 DOI: 10.1007/s00421-020-04499-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/10/2020] [Indexed: 11/17/2022]
Abstract
Purpose A prospective and controlled observational study was performed to determine if the central nervous system injury markers glial fibrillary acidic protein (GFAp), neurofilament light (NfL) and tau concentrations changed in response to a saturation dive. Methods The intervention group consisted of 14 submariners compressed to 401 kPa in a dry hyperbaric chamber. They remained pressurized for 36 h and were then decompressed over 70 h. A control group of 12 individuals was used. Blood samples were obtained from both groups before, during and after hyperbaric exposure, and from the intervention group after a further 25–26 h. Results There were no statistically significant changes in the concentrations of GFAp, NfL and tau in the intervention group. During hyperbaric exposure, GFAp decreased in the control group (mean/median − 15.1/ − 8.9 pg·mL−1, p < 0.01) and there was a significant difference in absolute change of GFAp and NfL between the groups (17.7 pg·mL−1, p = 0.02 and 2.34 pg·mL−1, p = 0.02, respectively). Albumin decreased in the control group (mean/median − 2.74 g/L/ − 0.95 g/L, p = 0.02), but there was no statistically significant difference in albumin levels between the groups. In the intervention group, haematocrit and mean haemoglobin values were slightly increased after hyperbaric exposure (mean/median 2.3%/1.5%, p = 0.02 and 4.9 g/L, p = 0.06, respectively). Conclusion Hyperbaric exposure to 401 kPa for 36 h was not associated with significant increases in GFAp, NfL or tau concentrations. Albumin levels, changes in hydration or diurnal variation were unlikely to have confounded the results. Saturation exposure to 401 kPa seems to be a procedure not harmful to the central nervous system. Trial registration ClinicalTrials.gov NCT03192930.
Collapse
|
26
|
Mkrtchian S, Kåhlin J, Gómez-Galán M, Ebberyd A, Yoshitake T, Schmidt S, Kehr J, Hildenborg M, Jonsson Fagerlund M, Erlandsson Harris H, Eriksson LI. The impact of damage-associated molecular patterns on the neurotransmitter release and gene expression in the ex vivo rat carotid body. Exp Physiol 2020; 105:1634-1647. [PMID: 32652583 DOI: 10.1113/ep088705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022]
Abstract
NEW FINDINGS What is the central question of this study? Are carotid bodies (CBs) modulated by the damage-associated molecular patterns (DAMPs) and humoral factors of aseptic tissue injury? What are the main findings and their importance? DAMPs (HMGB1, S100 A8/A9) and blood plasma from rats subjected to tibia surgery, a model of aseptic injury, stimulate the release of neurotransmitters (ATP, dopamine) and TNF-α from ex vivo rat CBs. All-thiol HMGB1 mediates upregulation of immune-related biological pathways. These data suggest regulation of CB function by endogenous mediators of innate immunity. ABSTRACT The glomus cells of carotid bodies (CBs) are the primary sensors of arterial partial O2 and CO2 tensions and moreover serve as multimodal receptors responding also to other stimuli, such as pathogen-associated molecular patterns (PAMPs) produced by acute infection. Modulation of CB function by excessive amounts of these immunomodulators is suggested to be associated with a detrimental hyperinflammatory state. We have hypothesized that yet another class of immunomodulators, endogenous danger-associated molecular patterns (DAMPs), released upon aseptic tissue injury and recognized by the same pathogen recognition receptors as PAMPs, might modulate the CB activity in a fashion similar to PAMPs. We have tested this hypothesis by exposing rat CBs to various DAMPs, such as HMGB1 (all-thiol and disulfide forms) and S100 A8/A9 in a series of ex vivo experiments that demonstrated the release of dopamine and ATP, neurotransmitters known to mediate CB homeostatic responses. We observed a similar response after incubating CBs with conditioned blood plasma obtained from the rats subjected to tibia surgery, a model of aseptic injury. In addition, we have investigated global gene expression in the rat CB using an RNA sequencing approach. Differential gene expression analysis showed all-thiol HMGB1-driven upregulation of a number of prominent pro-inflammatory markers including Il1α and Il1β. Interestingly, conditioned plasma had a more profound effect on the CB transcriptome resulting in inhibition rather than activation of the immune-related pathways. These data are the first to suggest potential modulation of CB function by endogenous mediators of innate immunity.
Collapse
Affiliation(s)
- Souren Mkrtchian
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jessica Kåhlin
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.,Function Perioperative Medicine and Intensive Care, Karolinska University Hospital and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marta Gómez-Galán
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anette Ebberyd
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Takashi Yoshitake
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Kehr
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Pronexus Analytical AB, Bromma, Sweden
| | - Malin Hildenborg
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.,Function Perioperative Medicine and Intensive Care, Karolinska University Hospital and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Malin Jonsson Fagerlund
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.,Function Perioperative Medicine and Intensive Care, Karolinska University Hospital and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Helena Erlandsson Harris
- Department of Medicine Solna, Section for Rheumatology, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars I Eriksson
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.,Function Perioperative Medicine and Intensive Care, Karolinska University Hospital and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Katsumi Y, Racine AM, Torrado-Carvajal A, Loggia ML, Hooker JM, Greve DN, Hightower BG, Catana C, Cavallari M, Arnold SE, Fong TG, Vasunilashorn SM, Marcantonio ER, Schmitt EM, Xu G, Libermann TA, Barrett LF, Inouye SK, Dickerson BC, Touroutoglou A, Collins JA. The Role of Inflammation after Surgery for Elders (RISE) study: Examination of [ 11C]PBR28 binding and exploration of its link to post-operative delirium. Neuroimage Clin 2020; 27:102346. [PMID: 32712451 PMCID: PMC7390821 DOI: 10.1016/j.nicl.2020.102346] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/11/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Major surgery is associated with a systemic inflammatory cascade that is thought, in some cases, to contribute to transient and/or sustained cognitive decline, possibly through neuroinflammatory mechanisms. However, the relationship between surgery, peripheral and central nervous system inflammation, and post-operative cognitive outcomes remains unclear in humans, primarily owing to limitations of in vivo biomarkers of neuroinflammation which vary in sensitivity, specificity, validity, and reliability. In the present study, [11C]PBR28 positron emission tomography, cerebrospinal fluid (CSF), and blood plasma biomarkers of inflammation were assessed pre-operatively and 1-month post-operatively in a cohort of patients (N = 36; 30 females; ≥70 years old) undergoing major orthopedic surgery under spinal anesthesia. Delirium incidence and severity were evaluated daily during hospitalization. Whole-brain voxel-wise and regions-of-interest analyses were performed to determine the magnitude and spatial extent of changes in [11C]PBR28 uptake following surgery. Results demonstrated that, compared with pre-operative baseline, [11C]PBR28 binding in the brain was globally downregulated at 1 month following major orthopedic surgery, possibly suggesting downregulation of the immune system of the brain. No significant relationship was identified between post-operative delirium and [11C]PBR28 binding, possibly due to a small number (n = 6) of delirium cases in the sample. Additionally, no significant relationships were identified between [11C]PBR28 binding and CSF/plasma biomarkers of inflammation. Collectively, these results contribute to the literature by demonstrating in a sizeable sample the effect of major surgery on neuroimmune activation and preliminary evidence identifying no apparent associations between [11C]PBR28 binding and fluid inflammatory markers or post-operative delirium.
Collapse
Affiliation(s)
- Yuta Katsumi
- Department of Psychology, Northeastern University, Boston, MA, United States; Japan Society for the Promotion of Science, Tokyo, Japan; Harvard Medical School, Boston, MA, United States
| | - Annie M Racine
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States
| | - Angel Torrado-Carvajal
- Harvard Medical School, Boston, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States; Medical Image Analysis and Biometry Laboratory, Universidad Rey Juan Carlos, Madrid, Spain
| | - Marco L Loggia
- Harvard Medical School, Boston, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Jacob M Hooker
- Harvard Medical School, Boston, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Douglas N Greve
- Harvard Medical School, Boston, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Baileigh G Hightower
- Harvard Medical School, Boston, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Ciprian Catana
- Harvard Medical School, Boston, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Michele Cavallari
- Harvard Medical School, Boston, MA, United States; Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States
| | - Steven E Arnold
- Harvard Medical School, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Tamara G Fong
- Harvard Medical School, Boston, MA, United States; Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States; Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Sarinnapha M Vasunilashorn
- Harvard Medical School, Boston, MA, United States; Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Edward R Marcantonio
- Harvard Medical School, Boston, MA, United States; Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Eva M Schmitt
- Harvard Medical School, Boston, MA, United States; Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States
| | - Guoquan Xu
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States
| | - Towia A Libermann
- Harvard Medical School, Boston, MA, United States; Genomics, Proteomics, Bioinformatics and Systems Biology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Lisa Feldman Barrett
- Department of Psychology, Northeastern University, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Sharon K Inouye
- Harvard Medical School, Boston, MA, United States; Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Bradford C Dickerson
- Harvard Medical School, Boston, MA, United States; Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Alexandra Touroutoglou
- Harvard Medical School, Boston, MA, United States; Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Jessica A Collins
- Harvard Medical School, Boston, MA, United States; Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
28
|
Zhang W, Xiong BR, Zhang LQ, Huang X, Zhou WC, Zou Q, Manyande A, Wang J, Tian XB, Tian YK. Disruption of the GABAergic system contributes to the development of perioperative neurocognitive disorders after anesthesia and surgery in aged mice. CNS Neurosci Ther 2020; 26:913-924. [PMID: 32488976 PMCID: PMC7415208 DOI: 10.1111/cns.13388] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Aims Perioperative neurocognitive disorders (PND) are associated with cognitive impairment in the preoperative or postoperative period, and neuroinflammation is thought to be the most important mechanisms especially during the postoperative period. The GABAergic system is easily disrupted by neuroinflammation. This study investigated the impact of the GABAergic system on PND after anesthesia and surgery. Methods An animal model of laparotomy with inhalation anesthesia in 16‐month‐old mice was addressed. Effects of the GABAergic system were assessed using biochemical analysis. Pharmacological blocking of α5GABAARs or P38 mitogen‐activated protein kinase (MAPK) were applied to investigate the effects of the GABAergic system. Results After laparotomy, the hippocampus‐dependent memory and long‐term potentiation were impaired, the levels of IL‐6, IL‐1β and TNF‐α up‐regulated in the hippocampus, the concentration of GABA decreased, and the protein levels of the surface α5GABAARs up‐regulated. Pharmacological blocking of α5GABAARs with L655,708 alleviated laparotomy induced cognitive deficits. Further studies found that the P38 MAPK signaling pathway was involved and pharmacological blocking with SB203,580 alleviated memory dysfunctions. Conclusions Anesthesia and surgery caused neuroinflammation in the hippocampus, which consequently disrupted the GABAergic system, increased the expressions of surface α5GABAARs especially through the P38 MAPK signaling pathway, and eventually led to hippocampus‐dependent memory dysfunctions.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing-Rui Xiong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Long-Qing Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian Huang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Chang Zhou
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Zou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, United Kingdom
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Xue-Bi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Ke Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Hippocampal complement C3 might contribute to cognitive impairment induced by anesthesia and surgery. Neuroreport 2020; 31:507-514. [PMID: 32168099 DOI: 10.1097/wnr.0000000000001422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Postoperative cognitive dysfunction is a well-recognized complication after major surgery in the elderly, but its pathophysiological mechanism is not fully understood. In the present study, we used liquid chromatography-tandem mass spectrometry combined with tandem mass tags to identify differentially expressed proteins and perform further functional studies on protein of interest. Here, we showed that hippocampal complement C3 was significantly upregulated after surgery, which was accompanied by marked decreases in synaptic related proteins and density. In aged patients undergoing gastrointestinal surgery, we also found significantly increased plasma level of C3b postoperatively and were negatively associated with cognitive performance. Notably, selective inhibition of complement C3 by compstatin was able to rescue synaptic and cognitive impairments induced by surgery in aged mice. Collectively, our study confirms that surgery can induce cognitive impairments, and the possible mechanisms might be related to abnormal complement signaling and synaptic disruption.
Collapse
|
30
|
Novel Insight into Neuroimmune Regulatory Mechanisms and Biomarkers Linking Major Depression and Vascular Diseases: The Dilemma Continues. Int J Mol Sci 2020; 21:ijms21072317. [PMID: 32230840 PMCID: PMC7177743 DOI: 10.3390/ijms21072317] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
Abstract
Major depressive disorder (MDD) represents a serious health problem estimated to affect 350 million people globally. Importantly, MDD has repeatedly emerged as an etiological or prognostic factor in cardiovascular disease (CVD) development, including vascular pathology. Several linking pathomechanisms between MDD and CVD involve abnormal autonomic regulation, inflammation, and endothelial dysfunction as an early preclinical stage of atherosclerosis. However, the cause of accelerated atherosclerosis in MDD patients remains unclear. Recently, the causal relationships between MDD and mediator (e.g., inflammation and/or endothelial dysfunction), as well as the causal pathways from the mediator to atherosclerosis, were discussed. Specifically, MDD is accompanied by immune dysregulation, resulting in increased production of proinflammatory cytokines (e.g., interleukin (IL)-6 and tumor necrosis factor (TNF)-α), which could lead to depression-linked abnormalities in brain function. Further, MDD has an adverse effect on endothelial function; for example, circulating markers of endothelial dysfunction (e.g., soluble adhesion molecules, von Willebrand factor) have been linked with depression. Additionally, MDD-linked autonomic dysregulation, which is characterized by disrupted sympathovagal balance associated with excessive circulating catecholamines, can contribute to CVD. Taken together, activated inflammatory response, endothelial dysfunction, and autonomic dysregulation could affect gradual atherosclerosis progression, resulting in a higher risk of developing CVD in MDD. This review focused on the pathomechanisms linking MDD and CVD with respect to neuroimmune regulation, and the description of promising biomarkers, which is important for the early diagnosis and personalized prevention of CVD in major depression.
Collapse
|
31
|
Qiu LL, Pan W, Luo D, Zhang GF, Zhou ZQ, Sun XY, Yang JJ, Ji MH. Dysregulation of BDNF/TrkB signaling mediated by NMDAR/Ca 2+/calpain might contribute to postoperative cognitive dysfunction in aging mice. J Neuroinflammation 2020; 17:23. [PMID: 31948437 PMCID: PMC6966800 DOI: 10.1186/s12974-019-1695-x] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023] Open
Abstract
Background Postoperative cognitive decline (POCD) is a recognized clinical phenomenon characterized by cognitive impairments in patients following anesthesia and surgery, yet its underlying mechanism remains unclear. Brain-derived neurotrophic factor (BDNF) plays an important role in neuronal plasticity, learning, and memory via activation of TrkB-full length (TrkB-FL) receptors. It has been reported that an abnormal truncation of TrkB mediated by calpain results in dysregulation of BDNF/TrkB signaling and is associated with cognitive impairments in several neurodegenerative disorders. Calpains are Ca2+-dependent proteases, and overactivation of calpain is linked to neuronal death. Since one source of intracellular Ca2+ is N-methyl-d-aspartate receptors (NMDARs) related and the function of NMDARs can be regulated by neuroinflammation, we therefore hypothesized that dysregulation of BDNF/TrkB signaling mediated by NMDAR/Ca2+/calpain might be involved in the pathogenesis of POCD. Methods In the present study, 16-month-old C57BL/6 mice were subjected to exploratory laparotomy with isoflurane anesthesia to establish the POCD animal model. For the interventional study, mice were treated with either NMDAR antagonist memantine or calpain inhibitor MDL-28170. Behavioral tests were performed by open field, Y maze, and fear conditioning tests from 5 to 8 days post-surgery. The levels of Iba-1, GFAP, interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α), NMDARs, calpain, BDNF, TrkB, bax, bcl-2, caspase-3, and dendritic spine density were determined in the hippocampus. Results Anesthesia and surgery-induced neuroinflammation overactivated NMDARs and then triggered overactivation of calpain, which subsequently led to the truncation of TrkB-FL, BDNF/TrkB signaling dysregulation, dendritic spine loss, and cell apoptosis, contributing to cognitive impairments in aging mice. These abnormities were prevented by memantine or MDL-28170 treatment. Conclusion Collectively, our study supports the notion that NMDAR/Ca2+/calpain is mechanistically involved in anesthesia and surgery-induced BDNF/TrkB signaling disruption and cognitive impairments in aging mice, which provides one possible therapeutic target for POCD.
Collapse
Affiliation(s)
- Li-Li Qiu
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Wei Pan
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dan Luo
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Guang-Fen Zhang
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Zhi-Qiang Zhou
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xiao-Yun Sun
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jian-Jun Yang
- Department of Anesthesiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Mu-Huo Ji
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Nanjing, 210009, China.
| |
Collapse
|
32
|
Abstract
Neuroinflammation has become a key hallmark of neurological complications including perioperative pathologies such as postoperative delirium and longer-lasting postoperative cognitive dysfunction. Dysregulated inflammation and neuronal injury are emerging from clinical studies as key features of perioperative neurocognitive disorders. These findings are paralleled by a growing body of preclinical investigations aimed at better understanding how surgery and anesthesia affect the central nervous system and possibly contribute to cognitive decline. Herein, we review the role of postoperative neuroinflammation and underlying mechanisms in immune-to-brain signaling after peripheral surgery.
Collapse
Affiliation(s)
- Saraswathi Subramaniyan
- From the Center for Translational Pain Medicine, Department of Anesthe siology, Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
33
|
Lin X, Chen Y, Zhang P, Chen G, Zhou Y, Yu X. The potential mechanism of postoperative cognitive dysfunction in older people. Exp Gerontol 2019; 130:110791. [PMID: 31765741 DOI: 10.1016/j.exger.2019.110791] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/18/2019] [Accepted: 11/21/2019] [Indexed: 02/08/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a common disorder following surgery, which seriously threatens the quality of patients' life, especially the older people. Accumulating attention has been paid to POCD worldwide in pace with the popularization of anesthesia/surgery. The development of medical humanities and rehabilitation medicine sets higher demands on accurate diagnosis and safe treatment system of POCD. Although the research on POCD is in full swing, underlying pathogenesis is still inconclusive due to these conflicting results and controversial evidence. Generally, POCD is closely related to neuropsychiatric diseases such as dementia, depression and Alzheimer's disease in molecular pathways. Researchers have come up with various hypotheses to reveal the mechanisms of POCD, including neuroinflammation, oxidative stress, autophagy disorder, impaired synaptic function, lacking neurotrophic support, etc. Recent work focused on molecular mechanism of POCD in older people has been thoroughly reviewed and summed up here, concerning the changes of peripheral circulation, pathological pathways of central nervous system (CNS), the microbiota-gut-brain axis and the related brain regions. Accordingly, this article provides a better perspective to understand the development situation of POCD in older people, which is conductive to uncover the pathological mechanism and exploit reasonable treatment strategy of POCD.
Collapse
Affiliation(s)
- Xianyi Lin
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Piao Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China.
| | - Youfa Zhou
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Xin Yu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
34
|
Sargolzaei S, Cai Y, Walker MJ, Hovda DA, Harris NG, Giza CC. Craniectomy Effects on Resting State Functional Connectivity and Cognitive Performance in Immature Rats. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2019; 2018:5414-5417. [PMID: 30441561 DOI: 10.1109/embc.2018.8513500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Experimental models have been proven to be valuable tools to understand downstream cellular mechanisms of Traumatic Brain Injury (TBI). The models allow for reduction of confounding variables and tighter control of varying parameters. It has been recently reported that craniectomy induces pro-inflammatory responses, which therefore needs to be properly addressed given the fact that craniectomy is often considered a control procedure for experimental TBI models. The current study aims to determine whether a craniectomy induces alterations in Resting State Network (RSN) in a developmental rodent model. Functional Magnetic Resonance Imaging (fMRI) data-driven RSN show clusters of peak differences (left caudate putamen, somatosensory cortex, amygdala and piriform cortex) between craniectomy and control group, four days post-craniectomy. In addition, the Novel Object Recognition (NOR) task revealed impaired working memory in the craniectomy group. This evidence supports craniectomy-induced neurological changes which need to be carefully addressed, considering the frequent use of craniectomy as a control procedure for experimental models of TBI.
Collapse
|
35
|
Zhou B, Chen L, Liao P, Huang L, Chen Z, Liao D, Yang L, Wang J, Yu G, Wang L, Zhang J, Zuo Y, Liu J, Jiang R. Astroglial dysfunctions drive aberrant synaptogenesis and social behavioral deficits in mice with neonatal exposure to lengthy general anesthesia. PLoS Biol 2019; 17:e3000086. [PMID: 31433818 PMCID: PMC6719896 DOI: 10.1371/journal.pbio.3000086] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 09/03/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
Lengthy use of general anesthetics (GAs) causes neurobehavioral deficits in the developing brain, which has raised significant clinical concerns such that the United States Food and Drug Administration (FDA) is warning on the use of GAs in children younger than 3 years. However, the molecular and cellular mechanisms for GAs-induced neurotoxicity remain largely unknown. Here, we report that sevoflurane (Sevo), a commonly used GA in pediatrics, caused compromised astrocyte morphogenesis spatiotemporally correlated to synaptic overgrowth, with reduced synaptic function in developing cortex in a regional-, exposure-length-, and age-specific manner. Sevo disrupted astrocyte Ca2+ homeostasis both acutely and chronically, which led to the down-regulation of Ezrin, an actin-binding membrane-bound protein, which we found was critically involved in astrocyte morphogenesis in vivo. Importantly, overexpression of astrocyte Ezrin rescued astrocytic and neuronal dysfunctions and fully corrected deficits in social behaviors in developing mice with lengthy Sevo exposure. Our data uncover that, in addition to neurons, astrocytes may represent important targets for GAs to exert toxic effects and that astrocyte morphological integrity is crucial for synaptogenesis and neurological behaviors. The extended use of general anesthetics can cause neurobehavioral deficits in the developing brain, leading to clinical concerns regarding their use in children younger than 3 years. This study shows that general anesthetics target glial cells to disrupt neural circuit formation in the developing brain, an effect that may underlie the observed learning, cognitive, or emotional deficits.
Collapse
Affiliation(s)
- Bin Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingmin Chen
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Huang
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhuo Chen
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Daqing Liao
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Wang
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guoqiang Yu
- Bradley Department of Electrical & Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, Virginia, United States of America
| | - Li Wang
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jianguo Zhang
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yunxia Zuo
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruotian Jiang
- Laboratory of Anesthesia and Critical Care Medicine, Sichuan University, Chengdu, Sichuan, China
- Translational Neuroscience Center, Sichuan University, Chengdu, Sichuan, China
- Department of Anesthesiology of West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
36
|
Preoperative Sleep Disturbance Exaggerates Surgery-Induced Neuroinflammation and Neuronal Damage in Aged Mice. Mediators Inflamm 2019; 2019:8301725. [PMID: 31011286 PMCID: PMC6442479 DOI: 10.1155/2019/8301725] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/23/2018] [Indexed: 12/20/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is defined as new cognitive impairment (memory impairment and impaired performance) after surgery, especially in aged patients. Sleep disturbance is a common phenomenon before surgery that has been increasingly thought to affect patient recovery. However, little is known about the functional impact of preoperative sleep disturbance on POCD. Here, we showed that tibial fracture surgery induced cognitive deficit and production of proinflammatory cytokines interleukin-6 (IL-6) and IL-1β, along with microglia and astrocyte activation, neuronal damage, and blood-brain barrier (BBB) disruption. Preoperative sleep disturbance enhanced the surgery-induced neuroinflammation, neuronal damage, BBB disruption, and memory impairment 24 h after surgery. Taken together, these results demonstrated that preoperative sleep disturbance aggravated postoperative cognitive function in aged mice and the mechanism may be related to central nervous system (CNS) inflammation and neuronal damage.
Collapse
|
37
|
Xiang X, Yu Y, Tang X, Chen M, Zheng Y, Zhu S. Transcriptome Profile in Hippocampus During Acute Inflammatory Response to Surgery: Toward Early Stage of PND. Front Immunol 2019; 10:149. [PMID: 30804943 PMCID: PMC6370675 DOI: 10.3389/fimmu.2019.00149] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/17/2019] [Indexed: 01/08/2023] Open
Abstract
Perioperative neurocognitive disorders (PND) are common complications observed in surgical patients, but there are no effective treatments and the detailed mechanisms remain largely unknown. In this study, transcriptome analysis was performed to investigate the hippocampal changes after surgery and underlying molecular mechanisms of PND. Tibial fracture surgery was performed in 3–4 months old C57BL/6J mice to mimic human orthopedic surgery. We demonstrated that memory consolidation of the hippocampal-dependent trace-fear conditioning task was significantly impaired. By using ELISA, a significant elevated IL-6 was observed both in circulating system and central nervous system and peaked at 6 h post-surgery, but transiently returned to baseline thereafter. Hippocampus were collected at 6 h post-surgery then processed for RNA-Seq. A total of 268 genes were screened differentially expressed between the Surgery and Control group, including 170 up-regulated genes and 98 down-regulated genes. By functional enrichment analysis of differently expressed genes, several KEGG pathways involved in inflammatory mediator regulation of TRP channels, neuroactive ligand-receptor interaction and cholinergic synapse were overrepresented. Quantitative real-time PCR confirmed 15 dysregulated genes of interest. These results provide a comprehensive insight into global gene expression changes during the acute presence of hippocampal inflammation and a better understanding on early stage of PND.
Collapse
Affiliation(s)
- Xuwu Xiang
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Yu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodong Tang
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Manli Chen
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yueying Zheng
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
38
|
Leukocyte expression profiles reveal gene sets with prognostic value for seizure-free outcome following stereotactic laser amygdalohippocampotomy. Sci Rep 2019; 9:1086. [PMID: 30705324 PMCID: PMC6355811 DOI: 10.1038/s41598-018-37763-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/13/2018] [Indexed: 02/08/2023] Open
Abstract
Among patients with intractable epilepsy, the most commonly performed surgical procedure is craniotomy for amygdalohippocampectomy (AH). Stereotactic laser amygdalohippocampotomy (SLAH) has also been recently employed as a minimally invasive treatment for intractable temporal lobe epilepsy (TLE). Among patients treated with AH and SLAH approximately 65% and 54% of patients become seizure-free, respectively. Therefore, selection criteria for surgical candidates with improved prognostic value for post-operative seizure-free outcome are greatly needed. In this study, we perform RNA sequencing (RNA-Seq) on whole blood leukocyte samples taken from 16 patients with intractable TLE prior to SLAH to test the hypothesis that pre-operative leukocyte RNA expression profiles are prognostic for post-operative seizure outcome. Multidimensional scaling analysis of the RNA expression data indicated separate clustering of patients with seizure free (SF) and non-seizure-free (NSF) outcomes. Differential expression (DE) analysis performed on SF versus NSF groups revealed 24 significantly differentially expressed genes (≥2.0-fold change, p-value < 0.05, FDR <0.05). Network and pathway analyses identified differential activation of pathways involved in lipid metabolism, morphology of oligodendrocytes, inflammatory response, and development of astrocytes. These results suggest that pre-operative leukocyte expression profiles have prognostic value for seizure outcome following SLAH.
Collapse
|
39
|
Li Z, Luo T, Ning X, Xiong C, Wu A. Neurokinin-1 receptor antagonism improves postoperative neurocognitive disorder in mice. Neurosci Lett 2018; 687:189-195. [DOI: 10.1016/j.neulet.2018.09.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/01/2018] [Accepted: 09/27/2018] [Indexed: 11/29/2022]
|
40
|
Xiong C, Liu J, Lin D, Zhang J, Terrando N, Wu A. Complement activation contributes to perioperative neurocognitive disorders in mice. J Neuroinflammation 2018; 15:254. [PMID: 30180861 PMCID: PMC6123969 DOI: 10.1186/s12974-018-1292-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/26/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The complement system plays an important role in many neurological disorders. Complement modulation, including C3/C3a receptor signaling, shows promising therapeutic effects on cognition and neurodegeneration. Yet, the implications for this pathway in perioperative neurocognitive disorders (PND) are not well established. Here, we evaluated the possible role for C3/C3a receptor signaling after orthopedic surgery using an established mouse model of PND. METHODS A stabilized tibial fracture surgery was performed in adult male C57BL/6 mice under general anesthesia and analgesia to induce PND-like behavior. Complement activation was assessed in the hippocampus and choroid plexus. Changes in hippocampal neuroinflammation, synapse numbers, choroidal blood-cerebrospinal fluid barrier (BCSFB) integrity, and hippocampal-dependent memory function were evaluated after surgery and treatment with a C3a receptor blocker. RESULTS C3 levels and C3a receptor expression were specifically increased in hippocampal astrocytes and microglia after surgery. Surgery-induced neuroinflammation and synapse loss in the hippocampus were attenuated by C3a receptor blockade. Choroidal BCSFB dysfunction occurred 1 day after surgery and was attenuated by C3a receptor blockade. Administration of exogenous C3a exacerbated cognitive decline after surgery, whereas C3a receptor blockade improved hippocampal-dependent memory function. CONCLUSIONS Orthopedic surgery activates complement signaling. C3a receptor blockade may be therapeutically beneficial to attenuate neuroinflammation and PND.
Collapse
Affiliation(s)
- Chao Xiong
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Jinhu Liu
- Department of Anesthesiology, Beijing First Hospital of Integrated Chinese and Western Medicine, Beijing, 100021 China
| | - Dandan Lin
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Juxia Zhang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Niccolò Terrando
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710 USA
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| |
Collapse
|