151
|
Hou Q, Li S, Zhang B, Chu H, Ni C, Fei X, Zheng H. LncRNA Riken Attenuated Sevoflurane-Induced Neuroinflammation by Regulating the MicroRNA-101a/MKP-1/JNK Pathway. Neurotox Res 2021; 40:186-197. [PMID: 34826047 DOI: 10.1007/s12640-021-00443-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 12/28/2022]
Abstract
The induction of anesthesia in children makes its safety one of the most important global health problems. Neuroinflammation contributes to anesthesia-induced neurotoxicity in young individuals. However, the mechanisms underlying anesthesia-induced neurotoxicity have not been established. In this study, the level of interleukin (IL)-6 in the hippocampus of mice and N2A cells treated with sevoflurane was increased, and long noncoding RNA (LncRNA) Riken was sufficient to decrease sevoflurane-induced neurotoxicity, and the level of inflammatory cytokine IL-6. The RNA pull-down assay verified that miR-101a was bound to lncRNA Riken in N2A cells. In addition, miR-101a blocked the protective effect of lncRNA Riken on anesthesia-induced neuroinflammation. These data suggest that lncRNA Riken attenuated anesthesia-induced neuroinflammation by interacting with microRNA-101a. Finally, we also demonstrated that MAPK phosphatase 1 (MKP-1) was a downstream target of miR-101a, and lncRNA Riken can regulate the expression of MKP-1; the JNK signal transduction pathway has been implicated in sevoflurane-induced IL-6 secretion. Our findings demonstrated that lncRNA Riken alleviated the sevoflurane-induced neurotoxic effects, and the lncRNA Riken/miR-101a/MKP-1/JNK axis plays an important role in the cognitive disorder.
Collapse
Affiliation(s)
- Qi Hou
- 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, 100021, China
| | - Shuai Li
- 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, 100021, China
| | - Bo Zhang
- 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, 100021, China
| | - Huaqing Chu
- 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, 100021, 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, 100021, China
| | - Xuejie Fei
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, 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, 100021, China.
| |
Collapse
|
152
|
Elevated neuron-specific enolase level is associated with postoperative delirium and detection of phosphorylated neurofilament heavy subunit: A prospective observational study. PLoS One 2021; 16:e0259217. [PMID: 34797829 PMCID: PMC8604326 DOI: 10.1371/journal.pone.0259217] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/14/2021] [Indexed: 01/21/2023] Open
Abstract
Background Delirium is the most common central nervous system complication after surgery. Detection of phosphorylated neurofilament heavy subunit in the serum reflects axonal damage within the central cervous system and is associated with the severity of postoperative delirium. Neuron-specific enolase and S100 calcium-binding protein β have been identified as possible serum biomarkers of postoperative delirium. This study examined the association of the levels of these markers with incidence of postoperative delirium and detection of phosphorylated neurofilament heavy subunit. Methods This study represents a post hoc analysis of 117 patients who participated in a prospective observational study of postoperative delirium in patients undergoing cancer surgery. Patients were clinically assessed for development of postoperative delirium within the first five days of surgery. Serum levels of phosphorylated neurofilament heavy subunit, neuron-specific enolase, and S100 calcium-binding protein β levels were measured on postoperative day 3. Results Forty-one patients (35%) were clinically diagnosed with postoperative delirium. Neuron-specific enolase level (P < 0.0001) and the proportion of patients positive for phosphorylated neurofilament heavy subunit (P < 0.0001) were significantly higher in the group of patients with postoperative delirium. Neuron-specific enolase level discriminated between patients with and without clinically diagnosed postoperative delirium with significantly high accuracy (area under the curve [AUC], 0.87; 95% confidence interval [CI], 0.79–0.95; P < 0.0001). Neuron-specific enolase level was associated with incidence of postoperative delirium independently of age (adjusted odds ratio, 8.291; 95% Cl, 3.506−33.286; P < 0.0001). The AUC for the serum neuron-specific enolase level in detecting phosphorylated neurofilament heavy subunit was significant (AUC, 0.78; 95% CI, 0.66–0.90; P < 0.0001). Conclusion Elevated serum neuron-specific enolase was associated with postoperative delirium independent of age as well as detection of phosphorylated neurofilament heavy subunit in serum. Serum neuron-specific enolase and phosphorylated neurofilament heavy subunit might be useful as biomarkers of postoperative delirium. Trial registration University Medical Information Network (UMIN) trial ID: UMIN000010329; https://clinicaltrials.gov/.
Collapse
|
153
|
Gut microbiota modulates the inflammatory response and cognitive impairment induced by sleep deprivation. Mol Psychiatry 2021; 26:6277-6292. [PMID: 33963281 DOI: 10.1038/s41380-021-01113-1] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 02/03/2023]
Abstract
Sleep deprivation (SD) is increasingly common in modern society, which can lead to the dysregulation of inflammatory responses and cognitive impairment, but the mechanisms remain unclear. Emerging evidence suggests that gut microbiota plays a critical role in the pathogenesis and development of inflammatory and psychiatric diseases, possibly via gut microbiota-brain interactions and neuroinflammation. The present study investigated the impact of SD on gut microbiota composition and explored whether alterations of the gut microbiota play a causal role in chronic inflammatory states and cognitive impairment that are induced by SD. We found that SD-induced gut dysbiosis, inflammatory responses, and cognitive impairment in humans. Moreover, the absence of the gut microbiota suppressed inflammatory response and cognitive impairment induced by SD in germ-free (GF) mice. Transplantation of the "SD microbiota" into GF mice activated the Toll-like receptor 4/nuclear factor-κB signaling pathway and impaired cognitive function in the recipient mice. Mice that harbored "SD microbiota" also exhibited increases in neuroinflammation and microglial activity in the hippocampus and medial prefrontal cortex. These findings indicate that gut dysbiosis contributes to both peripheral and central inflammatory processes and cognitive deficits that are induced by SD, which may open avenues for potential interventions that can relieve the detrimental consequences of sleep loss.
Collapse
|
154
|
Yang S, Zhang S, Tang W, Fang S, Zhang H, Zheng J, Liu X, Zhang Y, Zhao L, Huang L, Li B. Enriched Environment Prevents Surgery-Induced Persistent Neural Inhibition and Cognitive Dysfunction. Front Aging Neurosci 2021; 13:744719. [PMID: 34658844 PMCID: PMC8517535 DOI: 10.3389/fnagi.2021.744719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/03/2021] [Indexed: 11/21/2022] Open
Abstract
Perioperative neurocognitive disorders (PND) encompass short-term delirium and long-term cognitive dysfunction. Aging increases the susceptibility to PND, yet the neural mechanism is not known. In this study, we monitored the dynamic changes of neuronal activity in the prelimbic cortex before and after surgery. We found that anesthesia combined with surgery, but not anesthesia alone, induced a prolonged decrease in neuronal activity during the post-operation period in the aged mice, but not in the adult mice. The prolonged decrease in neuronal activity was accompanied by surgery-induced microglial activation and proinflammatory cytokines expression. Importantly, we found that the enriched environment (EE) completely prevented both the prolonged neural inhibition and neuroinflammation, and improved cognitive function in the aged mice. These results indicate that the prolonged neural inhibition correlated to PND and that EE before the surgery could effectively alleviate the surgery- induced cognitive dysfunction.
Collapse
Affiliation(s)
- Shana Yang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Song Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenting Tang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shunchang Fang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongyang Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieyan Zheng
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xia Liu
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Zhang
- Department of Spine Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Liang Zhao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lianyan Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Boxing Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
155
|
Hovens IB, van Leeuwen BL, Falcao-Salles J, de Haan JJ, Schoemaker RG. Enteral enriched nutrition to prevent cognitive dysfunction after surgery; a study in rats. Brain Behav Immun Health 2021; 16:100305. [PMID: 34589797 PMCID: PMC8474614 DOI: 10.1016/j.bbih.2021.100305] [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: 05/07/2021] [Revised: 06/17/2021] [Accepted: 07/24/2021] [Indexed: 11/26/2022] Open
Abstract
Background Inflammation plays an important role in postoperative cognitive dysfunction (POCD), particularly in elderly patients. Enteral enriched nutrition was shown to inhibit the response on inflammatory stimuli. Aim of the present study was to explore the therapeutic potential of enteral enriched nutrition in our rat model for POCD. The anticipated mechanism of action was examined in young rats, while responses in the target group of elderly patients were evaluated in old rats. Methods Male 3 and 23 months old Wistar rats received a bolus of enteral fat/protein-enriched nutrition 2 h and 30 min before surgery. The inflammatory response was evaluated by systemic inflammation markers and brain microglia activity. Additionally, in old rats, the role of the gut-brain axis was studied by microbiome analyses of faecal samples. Days 9–14 after surgery, rats were subjected to cognitive testing. Day 16, rats were sacrificed and brains were collected for immunohistochemistry. Results In young rats, enriched nutrition improved long-term spatial learning and memory in the Morris Water Maze, reduced plasma IL1-β and VEGF levels, but left microglia activity and neurogenesis unaffected. In contrast, in old rats, enriched nutrition improved short-term memory in the novel object- and novel location recognition tests, but impaired development of long-term memory in the Morris Water Maze. Systemic inflammation was not affected, but microglia activity seemed even increased. Gut integrity and microbiome were not affected. Conclusion Enteral enriched nutrition before surgery in young rats indeed reduced systemic inflammation and improved cognitive performance after surgery, whereas old rats showed a mixed favorable/unfavorable cognitive response, without effect on systemic inflammation. Anti-inflammatory effects of enriched nutrition were not reflected in decreased microglia activity. Neither was an important role for the gut-brain axis observed. Since the relatively straight forward effects of enriched nutrition in young rats could not be shown in old rats, as indicated by a mixed beneficial/detrimental cognitive outcome in the latter, caution is advised by translating effects seen in younger patients to older ones. Enriched nutrition reduced inflammation after surgery in young rats. Enriched nutrition improved postoperative cognitive outcome in young rats. Enteral enriched nutrition did not inhibit neuroinflammation. Effects in young rats do not predict effects in old rats. Enteral enriched nutrition caused mixed improved/declined cognition in old rats.
Collapse
Affiliation(s)
- Iris B Hovens
- Department of Neurobiology, GELIFES, University of Groningen, Netherlands
| | | | - Joana Falcao-Salles
- Department of Microbial Ecology, GELIFES, University of Groningen, Netherlands
| | - Jacco J de Haan
- Department of Medical Oncology, University Medical Center Groningen, Netherlands
| | | |
Collapse
|
156
|
Song P, Yi Z, Fu Y, Song D, Chen K, Zheng J, Sun Y, Diao Y. Reversing Postcardiopulmonary Bypass Associated Cognitive Dysfunction Using k-Opioid Receptor Agonists to Regulate Microglial Polarization via the NLRP3/Caspase-1 Pathway. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:3048383. [PMID: 34630980 PMCID: PMC8500742 DOI: 10.1155/2021/3048383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/14/2021] [Indexed: 12/21/2022]
Abstract
Cardiopulmonary bypass (CPB) is mainly used during cardiac surgeries that treat ischemic, valvular, or congenital heart disease and aortic dissections. The disorders of central nervous system (CNS) that occur after cardiopulmonary bypass are attracting considerable interest. Postoperative neurocognitive disorders (PND) have been reported as the leading cause of patients' disability and death following CPB. The k-opioid receptor (KOR) agonists (U50488H) have been suggested to be vital in the treatment of surgically induced CNS neuroinflammatory responses. In this article, the transitions between the M1 and M2 microglial polarization state phenotypes were hypothesized to significantly affect the regulatory mechanisms of KOR agonists on postcardiopulmonary bypass (post-CPB) neuroinflammation. We investigated the effects of U50488H on neuroinflammation and microglia polarization in rats exposed to CPB and explored the method of the NLRP3/caspase-1 pathway. Thirty SD rats were randomly divided into three groups: sham operation group, cardiopulmonary bypass model group, and CPB+ k-opioid receptor agonist (U50488H) group, with ten rats in each group. The Morris water maze was used to evaluate the changes in the cognitive function of CPB rats. Hematoxylin and eosin (HE) staining and TUNEL were performed to assess the rats' hippocampal damage. Enzyme-Linked Immunosorbent Assay (ELISA) was used to detect changes in brain injury markers and inflammatory factors. Furthermore, immunofluorescence was used to observe the expression of microglia polarization and NLRP3 followed by Western blots to detect the expression of the NLRP3/caspase-1 pathway and microglia polarization-related proteins. Rat microglia were cultured in vitro, with LPS stimulation, and treated with U50488H and a caspase-1 antagonist to evaluate the effects and mechanism of action of U50488H. KORs alleviated hippocampal damage caused by CPB and improved PND. CPB activated the NLRP3 inflammasome and upregulated pro-caspase-1 expression which promoted the expression of pro-IL-lβ and pro-IL-18 and resulted in increased inflammation. However, KORs also inhibited NLRP3 and transformed microglia from the M1 to the M2 state. Caspase-1 inhibitor treatment reduced the microglial polarization induced by KORs. The κ-opioid receptor agonists inhibited the inflammation mediated by microglia and improved PND through the NLRP3/caspase-1 signaling pathway.
Collapse
Affiliation(s)
- Pei Song
- Department of Anesthesia, Postgraduate Training Base of Jinzhou Medical University in the General Hospital of Northern Theater Command, Shenyang 110016, China
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Zhuo Yi
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Yiji Fu
- Department of Anesthesiology, Anshan Central Hospital, Anshan 114002, Liaoning, China
| | - Dandan Song
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Keyan Chen
- Department of Laboratory Animal Science, China Medical University, Shenyang 110000, Liaoning, China
| | - Jingjing Zheng
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Yingjie Sun
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Yugang Diao
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| |
Collapse
|
157
|
Heng L, Wang M, Wang M, Li L, Zhu S. Thoracic Paravertebral Block Ameliorates Postoperative Delirium in Geriatric Patients. Thorac Cardiovasc Surg 2021; 70:439-444. [PMID: 34521135 DOI: 10.1055/s-0041-1731788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Thoracic surgery often causes postoperative delirium (POD) in geriatric patients. This study aimed to explore the effect of ultrasound-guided continuous thoracic paravertebral block (UG-TPVB) on POD in geriatric patients undergoing pulmonary resection. METHODS Total 128 patients who underwent pulmonary resection were randomly allocated to either the conventional patient-controlled analgesia (PCA) group or the UG-TPVB group (n = 64 per group). The consumption of opioid agents (propofol and remifentanil), postoperative hospital stay, postoperative pulmonary atelectasis, postoperative nausea/vomiting, and postoperative itchiness were recorded. The diagnosis of delirium was dependent on the Nursing Delirium Screening Scale. The postoperative pain was assessed by visual analogue scale (VAS) score. The serum levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α were used to evaluate the postoperative neuroinflammation. RESULTS The consumption of propofol and remifentanil, postoperative hospital stay, postoperative pulmonary atelectasis, postoperative nausea/vomiting, and postoperative itchiness in the UG-TPVB group were lower than that in the PCA group. Compared with the PCA group, the prevalence of POD was decreased in the UG-TPVB group. In addition, use of UG-TPVB not only reduced postoperative pain (VAS score) but also decreased postoperative neuroinflammation compared with PCA in geriatric patients undergoing pulmonary resection. CONCLUSIONS This study determined the benefits of UG-TPVB over PCA, providing an effectiveness approach to alleviate POD in geriatric patients undergoing pulmonary resection.
Collapse
Affiliation(s)
- Lei Heng
- Department of Anesthesia, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu Province, China
| | - Mingyu Wang
- Department of Anesthesia, Xuzhou Maternity and Child Health Care Hospital, Xuzhou City, Jiangsu Province, China
| | - Mingquan Wang
- Department of Anesthesia, The People's Hospital of Jiawang District of Xuzhou, Xuzhou City, Jiangsu Province, China
| | - Li Li
- Intensive Care Unit, Xuzhou No. 1 People's Hospital, College Road, Xuzhou City, Jiangsu Province, China
| | - Shanshan Zhu
- Department of Anesthesia, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu Province, China
| |
Collapse
|
158
|
Lai Z, Min J, Li J, Shan W, Yu W, Zuo Z. Surgery Trauma Severity but not Anesthesia Length Contributes to Postoperative Cognitive Dysfunction in Mice. J Alzheimers Dis 2021; 80:245-257. [PMID: 33523008 DOI: 10.3233/jad-201232] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Perioperative, modifiable factors contributing to perioperative neurocognitive disorders (PND) have not been clearly defined. OBJECTIVE To determine the contribution of anesthesia lengths and the degrees of surgical trauma to PND and neuroinflammation, a critical process for PND. METHODS Three-month-old C57BL/6J mice were subjected to 2 h or 6 h isoflurane anesthesia plus a 5 min or 15 min left common carotid artery exposure (surgery) in a factorial design (two factors: anesthesia with two levels and surgery with three levels). Their learning and memory were tested by Barnes maze and novel object recognition paradigms. Blood, spleen, and hippocampus were harvested for measuring interleukin (IL)-6 and IL-1β. Eighteen-month-old C57BL/6J mice (old mice) were subjected to 6 h isoflurane anesthesia or 2 h isoflurane anesthesia plus 15 min surgery and then had learning and memory tested. RESULTS Three-month-old mice with 15 min surgery (long surgery) under 2 h or 6 h anesthesia performed poorly in the learning and memory tests compared with controls. Anesthesia alone or anesthesia plus 5 min surgery did not affect mouse performance in these tests. Similarly, only mice with long surgery but not mice with other experimental conditions had increased IL-6 and IL-1β in the blood, spleen, and hippocampus and decreased spleen weights. Splenocytes were found in the hippocampus after surgery. Similarly, old mice with long surgery but not the mice with isoflurane anesthesia alone had poor performance in the Barnes maze and novel object recognition tests. CONCLUSION Surgical trauma, but not anesthesia, contributes to the development of PND and neuroinflammation. Splenocytes may modulate these processes.
Collapse
Affiliation(s)
- Zhongmeng Lai
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA.,Department of Anesthesiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jia Min
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA.,Department of Anesthesiology, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Jun Li
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Weiran Shan
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
159
|
Transcutaneous electrical acupoint stimulation applied in lower limbs decreases the incidence of paralytic ileus after colorectal surgery: A multicenter randomized controlled trial. Surgery 2021; 170:1618-1626. [PMID: 34497027 DOI: 10.1016/j.surg.2021.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Postoperative paralytic ileus prolongs hospitalization duration, increases medical expenses, and is even associated with postoperative mortality; however, effective prevention of postoperative paralytic ileus is not yet available. This trial aimed to assess the preventative effectiveness of transcutaneous electrical acupoint stimulation applied in the lower limbs on postoperative paralytic ileus incidence after colorectal surgery. METHODS After ethics approval and written informed consent, 610 patients from 10 hospitals who were scheduled for colorectal surgery between May 2018 and September 2019 were enrolled. Patients were randomly allocated into the transcutaneous electrical acupoint stimulation (stimulated on bilateral Zusanli, Shangjuxu, and Sanyinjiao acupoints in lower limbs for 30 minutes each time, total 4 times) or sham (without currents delivered) group with 1:1 ratio. The primary outcome was postoperative paralytic ileus incidence, defined as no flatus for >72 hours after surgery. RESULTS Compared to the sham treatment, transcutaneous electrical acupoint stimulation lowered the postoperative paralytic ileus incidence by 8.7% (32.3% vs 41.0%, P = .026) and decreased the risk of postoperative paralytic ileus by 32% (OR, 0.68; P = .029). Transcutaneous electrical acupoint stimulation also shortened the recovery time to flatus, defecation, normal diet, and bowel sounds. Transcutaneous electrical acupoint stimulation treatment significantly increased median serum acetylcholine by 55% (P = .007) and interleukin-10 by 88% (P < .001), but decreased interleukin-6 by 47% (P < .001) and inducible nitric oxide synthase by 42% (P = .002) at 72 hours postoperatively. CONCLUSION Transcutaneous electrical acupoint stimulation attenuated the postoperative paralytic ileus incidence and enhanced gastrointestinal functional recovery, which may be associated with increasing parasympathetic nerve tone and its anti-inflammatory actions.
Collapse
|
160
|
Han FF, Wang HX, Wu JJ, Yao W, Hao CF, Pei JJ. Depressive symptoms and cognitive impairment: A 10-year follow-up study from the Survey of Health, Ageing and Retirement in Europe. Eur Psychiatry 2021; 64:e55. [PMID: 34446123 PMCID: PMC8446071 DOI: 10.1192/j.eurpsy.2021.2230] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Depressive symptoms and cognitive impairment often coexisted in the elderly. This study investigates the effect of late-life depressive symptoms on risk of mild cognitive impairment (MCI). Methods A total of 14,231 dementia- and MCI free participants aged 60+ from the Survey of Health, Ageing, and Retirement in Europe were followed-up for 10 years to detect incident MCI. MCI was defined as 1.5 standard deviation (SD) below the mean of the standardized global cognition score. Depressive symptoms were assessed by a 12-item Europe-depression scale (EURO-D). Severity of depressive symptoms was grouped as: no/minimal (score 0–3), moderate (score 4–5), and severe (score 6–12). Significant depressive symptoms (SDSs) were defined as EURO-D score ≥ 4. Results During an average of 8.2 (SD = 2.4)-year follow-up, 1,352 (9.50%) incident MCI cases were identified. SDSs were related to higher MCI risk (hazard ratio [HR] = 1.26, 95% confidence intervals [CI]: 1.10–1.44) in total population, individuals aged 70+ (HR = 1.35, 95% CI: 1.14–1.61) and women (HR = 1.28, 95% CI: 1.08–1.51) in Cox proportional hazard model adjusting for confounders. In addition, there was a dose–response association between the severity of depressive symptoms and MCI incidence in total population, people aged ≥70 years and women (p-trend <0.001). Conclusions Significant depressive symptoms were associated with higher incidence of MCI in a dose–response fashion, especially among people aged 70+ years and women. Treating depressive symptoms targeting older population and women may be effective in preventing MCI.
Collapse
Affiliation(s)
- Fei-Fei Han
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Hui-Xin Wang
- Stress Research Institute, Department of Psychology, Stockholm University, Stockholm, Sweden
| | - Jia-Jia Wu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Wu Yao
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Chang-Fu Hao
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jin-Jing Pei
- Stress Research Institute, Department of Psychology, Stockholm University, Stockholm, Sweden
| |
Collapse
|
161
|
Muscat SM, Barrientos RM. The Perfect Cytokine Storm: How Peripheral Immune Challenges Impact Brain Plasticity & Memory Function in Aging. Brain Plast 2021; 7:47-60. [PMID: 34631420 PMCID: PMC8461734 DOI: 10.3233/bpl-210127] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Precipitous declines in cognitive function can occur in older individuals following a variety of peripheral immune insults, such as surgery, infection, injury, and unhealthy diet. Aging is associated with numerous changes to the immune system that shed some light on why this abrupt cognitive deterioration may occur. Normally, peripheral-to-brain immune signaling is tightly regulated and advantageous; communication between the two systems is bi-directional, via either humoral or neural routes. Following an immune challenge, production, secretion, and translocation of cytokines into the brain is critical to the development of adaptive sickness behaviors. However, aging is normally associated with neuroinflammatory priming, notably microglial sensitization. Microglia are the brain's innate immune cells and become sensitized with advanced age, such that upon immune stimulation they will mount more exaggerated neuroimmune responses. The resultant elevation of pro-inflammatory cytokine expression, namely IL-1β, has profound effects on synaptic plasticity and, consequentially, cognition. In this review, we (1) investigate the processes which lead to aberrantly elevated inflammatory cytokine expression in the aged brain and (2) examine the impact of the pro-inflammatory cytokine IL-1β on brain plasticity mechanisms, including its effects on BDNF, AMPA and NMDA receptor-mediated long-term potentiation.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
162
|
Rahman R, Rahman S, Ghani H, Lozada-Martinez ID, Al-Salihi MM, Rahman MM. Letter: Presurgical Identification of Patients With Glioblastoma at Risk for Cognitive Impairment at 3-Month Follow-up. Neurosurgery 2021; 89:E270-E271. [PMID: 34318895 DOI: 10.1093/neuros/nyab282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/22/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Raphia Rahman
- Rowan School of Osteopathic Medicine Stratford, New Jersey, USA
| | - Sabrina Rahman
- Department of Public Health Independent University-Bangladesh Dhaka, Bangladesh
| | - Hira Ghani
- NYIT College of Osteopathic Medicine Old Westbury, New York, USA
| | | | | | - Md Moshiur Rahman
- Department of Neurosurgery Holy Family Red Crescent Medical College Dhaka, Bangladesh
| |
Collapse
|
163
|
Mao M, Zhou Z, Sun M, Wang C, Sun J. The dysfunction of parvalbumin interneurons mediated by microglia contributes to cognitive impairment induced by lipopolysaccharide challenge. Neurosci Lett 2021; 762:136133. [PMID: 34311051 DOI: 10.1016/j.neulet.2021.136133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND The mechanisms underlying cognitive impairments induced by systemic inflammation remain unclear. Increasing evidence has suggested that parvalbumin (PV) interneurons play an important role in regulating cognitive behaviors and its dysfunction is implicated in many neurological disorders. Thus, the present study was aimed to detect whether the destruction of PV interneurons mediates cognitive impairment associated with systemic inflammation. METHODS Male wild-type C57BL/6J mice (12-14 weeks old) received lipopolysaccharide (LPS 2 mg/kg i.p.) injection to establish the systemic inflammation model. For the suppression of microglial activation, minocycline (50 mg/kg i.p.) was applied. Animal behavior tests were conducted on day 3 post-LPS injection including the open field test, fear conditioning test and Y maze test. The PV expression in hippocampus was detected by Western blot and immunofluorescence. The number of perisomatic boutons around the NeuN-positive cells and microglia in hippocampus was detected by immunofluorescence. RESULTS LPS induced hippocampus-dependent memory and working memory impairment, coinciding with decreased PV expression, reduced perisomatic boutons around the NeuN-positive cells and activated microglia in the hippocampus. Notably, the treatment of minocycline suppressed the microglial activation and rescued the PV expression as well as the perisomatic boutons around the NeuN-positive cells in the hippocampus, contributing to improved cognitive function. CONCLUSION Our study suggests that the dysfunction of parvalbumin interneurons mediated by microglia plays a key role in LPS-induced cognitive impairments, which may serve a therapeutic strategy for cognitive disorders associated with systemic inflammation.
Collapse
Affiliation(s)
- Meng Mao
- Department of Anesthesiology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhenhui Zhou
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Menghan Sun
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Chaoran Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Sun
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
164
|
Goldstein MR, Cheslock M. On the prevention and treatment of Alzheimer's disease: Control the promoters and look beyond the brain. Med Hypotheses 2021; 154:110645. [PMID: 34315048 DOI: 10.1016/j.mehy.2021.110645] [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: 02/25/2021] [Revised: 06/05/2021] [Accepted: 07/13/2021] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) is a progressive incurable neurodegenerative disease of the brain afflicting a third of the population aged 85 and older. Pathologic hallmarks include extracellular plaques of amyloid-beta (Aß), intraneuronal neurofibrillary tangles of hyperphosphorylated tau protein, synaptic destruction, neuronal death, and brain atrophy. Neuroinflammation, mediated by microglia, is a central component of the disease, and is intricately connected with peripheral inflammation. The clinical manifestations include progressive memory loss and eventual death. The present treatment of AD is largely ineffective. Nearly all AD is late-onset and presents age 65 or older, and the most common genetic risk factor is carriage of an apolipoprotein (APO) E4 allele, seen in about 25% of the general population. Individuals carrying an APOE4 allele produce more Aß and clear it less efficiently from the brain throughout life. There has been accumulating pathologic and clinical evidence that microbes, particularly the herpes simplex virus (HSV), is a causative factor for AD, most notable in carriers of the APOE4 allele. Eighty percent of the adult population harbors HSV and it resides in the trigeminal ganglion in latent state throughout life, but periodically reactivates, traveling antegrade resulting in herpes labialis and traveling retrograde into the brain leading to neuroinflammation. Functioning as an antimicrobial peptide, Aß inactivates HSV and the recurring process culminates in a buildup of Aß plaque and other hallmarks of AD over time. Periodontal disease exists in 20-50% of the adult population and is also a causative factor for AD. Accordingly, bacteria causing periodontal disease and their byproducts can enter the brain directly via the trigeminal nerve or indirectly through the bloodstream, resulting in AD pathology over time. There are many other promoters of AD, particularly inflammatory conditions outside of the brain, that can be mitigated. Small trials are finally in progress testing antimicrobial drugs for the prevention and treatment of AD. In the meantime, a more proactive approach to the prevention and treatment of AD is posited, with an emphasis on prevention, since the pathologic underpinnings of the disease start decades before the clinical manifestations. Individuals can be stratified in risk categories using family history, periodontal disease presence, APOE4 carriage, and HSV IgG positivity. Moderate- and high-risk individuals can be treated safely with various preventive measures and appropriate antimicrobial agents as discussed. Importantly, the proposed treatments are concordant with the accepted practice of medicine, and if utilized, could significantly decrease AD prevalence.
Collapse
Affiliation(s)
| | - Megan Cheslock
- Harvard Medical School Multi-Campus Geriatric Fellowship, Boston, Massachusetts, USA.
| |
Collapse
|
165
|
Oberman K, Hovens I, de Haan J, Falcao-Salles J, van Leeuwen B, Schoemaker R. Acute pre-operative ibuprofen improves cognition in a rat model for postoperative cognitive dysfunction. J Neuroinflammation 2021; 18:156. [PMID: 34238316 PMCID: PMC8265047 DOI: 10.1186/s12974-021-02206-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/23/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Inflammation is considered a key factor in the development of postoperative cognitive dysfunction (POCD). Therefore, we hypothesized that pre-operative anti-inflammatory treatment with ibuprofen would inhibit POCD in our rat-model. METHODS Male Wistar rats of 3 or 23 months old received a single injection of ibuprofen (15 mg/kg i.p.) or were control handled before abdominal surgery. Timed blood and fecal samples were collected for analyses of inflammation markers and gut microbiome changes. Behavioral testing was performed from 9 to 14 days after surgery, in the open field, novel object- and novel location-recognition tests and Morris water maze. Neuroinflammation and neurogenesis were assessed by immune histochemistry after sacrifice on postoperative day 14. RESULTS Ibuprofen improved short-term spatial memory in the novel location recognition test, and increased hippocampal neurogenesis. However, these effects were associated with increased hippocampal microglia activity. Whereas plasma cytokine levels (IL1-β, IL6, IL10, and TNFα) were not significantly affected, VEGF levels increased and IFABP levels decreased after ibuprofen. Long-term memory in the Morris water maze was not significantly improved by ibuprofen. The gut microbiome was neither significantly affected by surgery nor by ibuprofen treatment. In general, effects in aged rats appeared similar to those in young rats, though less pronounced. CONCLUSION A single injection of ibuprofen before surgery improved hippocampus-associated short-term memory after surgery and increased neurogenesis. However, this favorable outcome seemed not attributable to inhibition of (neuro)inflammation. Potential contributions of intestinal and blood-brain barrier integrity need further investigation. Although less pronounced compared to young rats, effects in aged rats indicate that even elderly individuals could benefit from ibuprofen treatment.
Collapse
Affiliation(s)
- Klaske Oberman
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747, AG, Groningen, The Netherlands
| | - Iris Hovens
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747, AG, Groningen, The Netherlands
| | - Jacco de Haan
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Joana Falcao-Salles
- Department of Microbial Ecology, GELIFES, University of Groningen, Groningen, The Netherlands
| | - Barbara van Leeuwen
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Regien Schoemaker
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747, AG, Groningen, The Netherlands.
| |
Collapse
|
166
|
Huang CM, Cai JJ, Jin SW, Lin QC, Fang QJ, Nan K, Han Y, Ge WW, Liu Y, Tao YX, Cao H, Li J. Class IIa HDAC Downregulation Contributes to Surgery-Induced Cognitive Impairment Through HMGB1-Mediated Inflammatory Response in the Hippocampi of Aged Mice. J Inflamm Res 2021; 14:2301-2315. [PMID: 34103963 PMCID: PMC8180279 DOI: 10.2147/jir.s304060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/18/2021] [Indexed: 02/01/2023] Open
Abstract
Objective Perioperative neurocognitive disorders (PND) are a common complication in the elderly. Histone deacetylases (HDACs) are a class of enzymes that control the acetylation status of intracellular proteins. Thus, we explored whether HDACs trigger the release of high mobility group box 1 (HMGB1) through altering the acetylation status in the hippocampi of aged mice. Materials and Methods The effect of the Class IIa HDAC in PND was explored using an in vivo form of splenectomy. Sixteen-month-old healthy male C57BL/6J mice were randomly divided into five groups: control, anesthesia plus sham surgery, anesthesia plus splenectomy, LMK235 treatment, and PBS treatment. The hippocampi were harvested on either first, third, or seventh postoperative day. Cognitive function was assessed via a Morris water maze (MWM) test. Quantitative RT-PCR, Western blots and ELISAs were carried out to assess the targeted gene expression at transcriptional and translational levels. Results Splenectomy led to a significant deficiency in spatial memory acquisition, marked decreases in mRNA and protein levels of HDAC4 and HDAC5 in the hippocampus, and increases in the levels of total HMGB1 and acetylated HMGB1. In a similar fashion to splenectomy, treatment with the HDAC4/5 inhibitor LMK235 produced impaired spatial memory and an increase in the expression of HMGB1 and its acetylated counterpart in the hippocampus. Conclusion These results suggest that surgery leads to PND through class IIa HDAC downregulation-triggered HMGB1 release in hippocampus of aged mice. HDACs may be a potential therapeutic target for postoperative cognitive dysfunction.
Collapse
Affiliation(s)
- Chen-Miao Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Jia-Jing Cai
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Shao-Wu Jin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Qi-Cheng Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Qian-Juan Fang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Ke Nan
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Yuan Han
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Wen-Wei Ge
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Yuan-Xiang Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Hong Cao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| | - Jun Li
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China.,Zhejiang Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, People's Republic of China
| |
Collapse
|
167
|
Lv XC, Lin Y, Wu QS, Wang L, Hou YT, Dong Y, Chen LW. Plasma interleukin-6 is a potential predictive biomarker for postoperative delirium among acute type a aortic dissection patients treated with open surgical repair. J Cardiothorac Surg 2021; 16:146. [PMID: 34044881 PMCID: PMC8161913 DOI: 10.1186/s13019-021-01529-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES The relationship between inflammatory cytokines and postoperative delirium (POD) remains to be further investigated, especially in patients undergoing acute type A aortic dissection (AAD). Interleukin-6 (IL-6) is involved in the inflammatory process and has recently been identified as a biomarker of cerebral dysfunction. We explored the hypothesis that IL-6 was one of the critical causes of POD after surgical repair of AAD. METHODS Plasma IL-6 was measured using electrochemiluminescence technology in patients preoperatively and 24 h, 48 h, and 72 h after surgical repair of acute type A aortic dissection. After the first three postoperative days, delirium was evaluated twice daily using the Confusion Assessment Method. ROC curves were used to evaluate the ability of IL-6 measurements to distinguish POD. RESULTS The incidence of POD was 14.03% (31 of 221 patients). The patients in the POD group were significantly older than the patients in the non-POD group (56.48 ± 11.68 years vs 52.22 ± 10.50 years, P = 0.040). Plasma IL-6 concentrations were significantly higher in the POD group than in the non-POD group at three time points: preoperatively, after 24 h, and after 48 h. The AUC values corresponding to IL-6 preoperatively and 24 h after surgery were 0.73 and 0.72, respectively. CONCLUSIONS Cerebral dysfunction after the surgical repair of AAD shows elevated stress levels and inflammatory responses. Plasma IL-6 is a potential biomarker to predict the onset of POD in acute type A aortic dissection patients following surgical repair.
Collapse
Affiliation(s)
- Xiao-Chai Lv
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Xinquan Road No. 29, Fuzhou, 350001, Fujian, China.,Department of Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fujian, China.,Department of Fujian Provincial Special Reserve Talents Laboratory, Fujian, China.,Department of Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fujian, China
| | - Yong Lin
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Xinquan Road No. 29, Fuzhou, 350001, Fujian, China.,Department of Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fujian, China.,Department of Fujian Provincial Special Reserve Talents Laboratory, Fujian, China.,Department of Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fujian, China
| | - Qing-Song Wu
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Xinquan Road No. 29, Fuzhou, 350001, Fujian, China.,Department of Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fujian, China.,Department of Fujian Provincial Special Reserve Talents Laboratory, Fujian, China.,Department of Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fujian, China
| | - Lei Wang
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Xinquan Road No. 29, Fuzhou, 350001, Fujian, China.,Department of Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fujian, China.,Department of Fujian Provincial Special Reserve Talents Laboratory, Fujian, China.,Department of Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fujian, China
| | - Yan-Ting Hou
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Xinquan Road No. 29, Fuzhou, 350001, Fujian, China.,Department of Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fujian, China.,Department of Fujian Provincial Special Reserve Talents Laboratory, Fujian, China.,Department of Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fujian, China
| | - Yi Dong
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Xinquan Road No. 29, Fuzhou, 350001, Fujian, China.,Department of Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fujian, China.,Department of Fujian Provincial Special Reserve Talents Laboratory, Fujian, China.,Department of Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fujian, China
| | - Liang-Wan Chen
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Xinquan Road No. 29, Fuzhou, 350001, Fujian, China. .,Department of Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fujian, China. .,Department of Fujian Provincial Special Reserve Talents Laboratory, Fujian, China. .,Department of Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fujian, China.
| |
Collapse
|
168
|
Yamanashi T, Saito T, Yu T, Alario A, Comp K, Crutchley KJ, Sullivan EJ, Anderson ZEM, Marra PS, Chang G, Wahba NE, Jellison SS, Meyer AA, Mathur S, Pandharipande P, Yoshino A, Kaneko K, Lee S, Toda H, Iwata M, Shinozaki G. DNA methylation in the TNF-alpha gene decreases along with aging among delirium inpatients. Neurobiol Aging 2021; 105:310-317. [PMID: 34192631 DOI: 10.1016/j.neurobiolaging.2021.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/25/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
It has been suggested that aging and inflammation play key roles in the development of delirium. In the present study, we investigated the differences of the DNAm patterns in the TNF gene between patients with delirium and without. The data and samples derived from previous and ongoing cohort studies were analyzed. DNAm levels of the TNF gene were analyzed using the Illumina EPIC array genome-wide method and pyrosequencing method. Correlations between age and DNAm levels of each CpG were calculated. Several CpG in the TNF gene in blood showed negative correlation between their DNAm and age in delirium cases both with the EPIC array and by the pyrosequencing method. However, there was no CpG that had significant correlation between their DNAm and age regardless of delirium status among buccal samples. On the other hand, among peripheral blood mononuclear cells samples, it was found that several CpG showed negative correlation between their DNAm and age in delirium cases. The evidence of DNAm change in the TNF gene among delirious subjects was demonstrated.
Collapse
Affiliation(s)
- Takehiko Yamanashi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA; Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Neuropsychiatry, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Taku Saito
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Psychiatry, National Defense Medical College School of Medicine, Tokorozawa, Saitama, Japan
| | - Tong Yu
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Alexandra Alario
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Katie Comp
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Kaitlyn J Crutchley
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Eleanor J Sullivan
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Zoe-Ella M Anderson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Pedro S Marra
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Gloria Chang
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Nadia E Wahba
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sydney S Jellison
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Alissa A Meyer
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Srishti Mathur
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Pratik Pandharipande
- Department of anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aihide Yoshino
- Department of Psychiatry, National Defense Medical College School of Medicine, Tokorozawa, Saitama, Japan
| | - Koichi Kaneko
- Department of Neuropsychiatry, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Sangil Lee
- Department of Emergency Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Hiroyuki Toda
- Department of Psychiatry, National Defense Medical College School of Medicine, Tokorozawa, Saitama, Japan
| | - Masaaki Iwata
- Department of Neuropsychiatry, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Gen Shinozaki
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA; Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
169
|
Zhang L, Xiao F, Zhang J, Wang X, Ying J, Wei G, Chen S, Huang X, Yu W, Liu X, Zheng Q, Xu G, Yu S, Hua F. Dexmedetomidine Mitigated NLRP3-Mediated Neuroinflammation via the Ubiquitin-Autophagy Pathway to Improve Perioperative Neurocognitive Disorder in Mice. Front Pharmacol 2021; 12:646265. [PMID: 34079457 PMCID: PMC8165564 DOI: 10.3389/fphar.2021.646265] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/27/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Surgery and anesthesia-induced perioperative neurocognitive disorder (PND) are closely related to NOD-like receptors (NLR) family, pyrin domain containing 3 (NLRP3) inflammasome microglia inflammatory response. Inhibiting the occurrence of neuroinflammation is an important treatment method to improve postoperative delirium. Fewer NLRP3-targeting molecules are currently available in the clinic to reduce the incidence of postoperative delirium. Dexmedetomidine (DEX), an α2 adrenergic receptor agonist has been shown to have antioxidant and anti-inflammatory activities. The present study showed that DEX reduced the production of cleaved caspase1 (CASP1) and destroyed the NLRP3–PYD And CARD Domain Containing (PYCARD)–CASP1 complex assembly, thereby reducing the secretion of IL-1β interleukin beta (IL-1β). DEX promoted the autophagy process of microglia and reduced NLRP3 expression. More interestingly, it promoted the ubiquitination and degradation of NLRP3. Thus, this study demonstrated that DEX reduced NLRP3-mediated inflammation through the activation of the ubiquitin-autophagy pathway. This study provided a new mechanism for treating PND using DEX. Methods: C57BL/6 mice were pre-administered DEX 3 days in advance, and an abdominal exploration model was used to establish a perioperative neurocognitive disorder model. The anti-inflammatory effect of DEX was explored in vivo by detecting NLRP3-CASP1/IL-1β protein expression and behavioral testing. Primary microglia were stimulated with lipopolysaccharide (LPS) and adenosine triphosphate (ATP) in vitro, the expression of CASP1 and IL-1β was detected in the supernatant of cells, and the expression of autophagy-related proteins microtubule-associated protein 1 light chain 3 beta (MAP1LC3B) and sequestosome 1 (SQSTM1) was examined in the cytoplasm. Meanwhile, Co-immunoprecipitation (Co-IP) was used to detect NLRP3 protein ubiquitination so as to clarify the new mechanism underlying the anti-inflammatory effect of DEX. Results: Pre-administration of DEX reduced the protein expression of NLRP3, CASP1, and IL-1β in the hippocampus of mice induced by surgery and also improved the impairment of learning and memory ability. At the same time, DEX also effectively relieved the decrease in spine density of the hippocampal brain induced by surgery. DEX decreased the cleaved CASP1 expression, blocked the assembly of NLRP3–PYCARD–CASP1 complex, and also reduced the secretion of mature IL-1β in vitro. Mechanically, it accelerated the degradation of NLRP3 inflammasome via the autophagy–ubiquitin pathway and reduced the green fluorescent protein/red fluorescent protein MAP1LC3B ratio, which was comparable to the effect when using the autophagy activator rapamycin (Rapa). Furthermore, it increased the ubiquitination of NLRP3 after LPS plus ATP stimulated microglia. Conclusion: DEX attenuated the hippocampal brain inflammation by promoting NLRP3 inflammasome degradation via the autophagy–ubiquitin pathway, thus improving cognitive impairment in mice.
Collapse
Affiliation(s)
- Lieliang Zhang
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fan Xiao
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hosptial of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Gen Wei
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Shoulin Chen
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xiangfei Huang
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Wen Yu
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xing Liu
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Shuchun Yu
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hosptial of Nanchang Univerisity, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| |
Collapse
|
170
|
Perioperative Vascular Biomarker Profiling in Elective Surgery Patients Developing Postoperative Delirium: A Prospective Cohort Study. Biomedicines 2021; 9:biomedicines9050553. [PMID: 34063403 PMCID: PMC8155907 DOI: 10.3390/biomedicines9050553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Postoperative delirium (POD) ranks among the most common complications in surgical patients. Blood-based biomarkers might help identify the patient at risk. This study aimed to assess how serum biomarkers with specificity for vascular and endothelial function and for inflammation are altered, prior to or following surgery in patients who subsequently develop POD. Methods: This was a study on a subcohort of consecutively recruited elective non-cardiac as well as cardiac surgery patients (age > 60 years) of the single-center PROPDESC trial at a German tertiary care hospital. Serum was sampled prior to and following surgery, and the samples were subjected to bead-based multiplex analysis of 17 serum proteins (IL-3, IL-8, IL-10, Cripto, CCL2, RAGE, Resistin, ANGPT2, TIE2, Thrombomodulin, Syndecan-1, E-Selectin, VCAM-1, ICAM-1, CXCL5, NSE, and uPAR). Development of POD was assessed during the first five days after surgery, using the Confusion Assessment Method for ICU (CAM-ICU), the CAM, the 4-‘A’s test (4AT), and the Delirium Observation Scale (DOS). Patients were considered positive if POD was detected at least once during the visitation period by any of the applied methods. Non-parametric testing, as well as propensity score matching were used for statistical analysis. Results: A total of 118 patients were included in the final analysis; 69% underwent non-cardiac surgery, median overall patient age was 71 years, and 59% of patients were male. In the whole cohort, incidence of POD was 28%. The male gender was significantly associated with the development of POD (p = 0.0004), as well as a higher ASA status III (p = 0.04). Incidence of POD was furthermore significantly increased in cardiac surgery patients (p = 0.002). Surgery induced highly significant changes in serum levels of almost all biomarkers except uPAR. In preoperative serum samples, none of the analyzed parameters was significantly altered in subsequent POD patients. In postoperative samples, CCL2 was significantly increased by a factor of 1.75 in POD patients (p = 0.03), as compared to the no-POD cohort. Following propensity score matching, CCL2 remained the only biomarker that showed significant differences in postoperative values (p = 0.01). In cardiac surgery patients, postoperative CCL2 serum levels were more than 3.5 times higher than those following non-cardiac surgery (p < 0.0001). Moreover, after cardiac surgery, Syndecan-1 serum levels were significantly increased in POD patients, as compared to no-POD cardiac surgery patients (p = 0.04). Conclusions: In a mixed cohort of elective non-cardiac as well as cardiac surgery patients, preoperative serum biomarker profiling with specificity for vascular dysfunction and for systemic inflammation was not indicative of subsequent POD development. Surgery-induced systemic inflammation—as evidenced by the significant increase in CCL2 release—was associated with POD, particularly following cardiac surgery. In those patients, postoperative glycocalyx injury might furthermore contribute to POD development.
Collapse
|
171
|
Shen Z, Xu H, Song W, Hu C, Guo M, Li J, Li J. Galectin-1 ameliorates perioperative neurocognitive disorders in aged mice. CNS Neurosci Ther 2021; 27:842-856. [PMID: 33942523 PMCID: PMC8193703 DOI: 10.1111/cns.13645] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/23/2022] Open
Abstract
Introduction The incidence of perioperative neurocognitive disorders (PND) is higher in the elderly patients undergoing surgery. Microglia activation‐mediated neuroinflammation is one of the hallmarks of PND. Galectin‐1 has been identified as a pivotal modulator in the central nervous system (CNS), while the role of galectin‐1 in PND induced by microglia‐mediated neuroinflammation is still undetermined. Methods An exploratory laparotomy model anesthetized with isoflurane was employed to investigate the role of galectin‐1 on PND in aged mice. Open field test and Morris water maze were used to test the cognitive function 3‐ or 7‐days post‐surgery. The activation of microglia in the hippocampus of aged mice was tested by immunohistochemistry. Western blot, enzyme‐linked immunosorbent assay (ELISA), and quantitative real‐time polymerase chain reaction (qRT‐PCR) were employed to elucidate the underlying mechanisms. Results Galectin‐1 attenuated the cognitive dysfunction induced by surgery in aged mice and inhibited microglial activity. Moreover, galectin‐1 decreased the expression level of inflammatory proteins (interleukin‐1β, interleukin‐6, and tumor necrosis factor‐α), and prevented neuronal loss in the hippocampus. Galectin‐1 inhibited the inflammation of BV2 microglial cells induced by lipopolysaccharide via decreasing the translocation of NF‐κB p65 and c‐Jun, while this kind of inhibition was rescued when overexpressing IRAK1. Conclusion Our findings provide evidence that galectin‐1 may inhibit IRAK1 expression, thus suppressing inflammatory response, inhibiting neuroinflammation, and improving ensuing cognitive dysfunction. Collectively, these findings unveil that galectin‐1 may elicit protective effects on surgery‐induced neuroinflammation and neurocognitive disorders.
Collapse
Affiliation(s)
- Zhiwen Shen
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Xu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wen Song
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuwen Hu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mingyan Guo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinfeng Li
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junhua Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
172
|
Grigolashvili MA, Mustafina RM. [The role of the inflammatory process in the development of post-stroke cognitive impairment]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:16-21. [PMID: 33908227 DOI: 10.17116/jnevro202112103216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Post-stroke cognitive impairment (PCI) is a common complication of stroke. PCI in most cases is associated with an increased risk of progression to dementia, with a progression rate of 8-15% per year. When post-stroke cognitive impairment reaches dementia, patients lose independence, professional and social maladjustment occurs, which, in turn, significantly worsen the quality of life and reduce the rehabilitation potential. According to many experimental and clinical studies, the inflammatory process has an important role in the development of PCI. Several previous studies have looked at the association between inflammatory markers and PCI, with some results conflicting with specific biomarkers. Based on the results of studies, inflammatory markers such as IL-8, IL-12 and ESR were closely associated with PCI, high ESR values are associated with worse cognitive impairment, especially memory. The relationship was not confirmed between the markers IFN-gamma, TNF-α and PCI. With regard to IL-1β, IL-6, IL-10, CRP, the results obtained are not unambiguous. Thus, the inflammatory process in the development of PCI has an important role, including a series of complex reactions, the combined effect of which induces neuronal damage and loss of synapses that ultimately leads to cognitive impairment.
Collapse
Affiliation(s)
| | - R M Mustafina
- Medical University of Karaganda, Karaganda, Kazakhstan
| |
Collapse
|
173
|
Burfeind KG, Tirado Navales AA, Togioka BM, Schenning K. Prevention of postoperative delirium through the avoidance of potentially inappropriate medications in a geriatric surgical patient. BMJ Case Rep 2021; 14:14/4/e240403. [PMID: 33875501 PMCID: PMC8057549 DOI: 10.1136/bcr-2020-240403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We demonstrate the utility of risk stratification for postoperative delirium in geriatric patients and show that postoperative delirium can be prevented in high-risk patients when potentially inappropriate medications (PIMs) (medications that are best avoided in older adults) are avoided. In this case, a 65-year-old woman underwent two debridement procedures with similar presurgical risk for postoperative delirium. There was no risk stratification or preoperative cognitive assessment in the first procedure, she received PIMs and developed postoperative delirium. In the second procedure, PIMs were intentionally avoided and postoperative delirium did not occur. This case supports recent recommendations from the European Society of Anaesthesiology, the American Society of Anesthesiologists and the American Geriatrics Society that providers assess a patient's cognitive function and delirium risk profile preoperatively to appropriately guide perioperative management.
Collapse
Affiliation(s)
- Kevin G Burfeind
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA.,Medical Scientist Training Program, Oregon Health & Science University, Portland, Oregon, USA
| | - Andrés A Tirado Navales
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Brandon Michael Togioka
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Katie Schenning
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
174
|
Zhao Q, Gao R, Liu C, Chen H, Zhang X, Guan J, Xie X, Qiu Y, Cheng X, Lv P, Zhu T, Chen C. Dynamic Change of Lymphocyte-to-Monocyte Is Associated With the Occurrence of POCD After Cardiovascular Surgery: A Prospective Observational Study. Front Behav Neurosci 2021; 15:646528. [PMID: 33927600 PMCID: PMC8076514 DOI: 10.3389/fnbeh.2021.646528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/16/2021] [Indexed: 02/05/2023] Open
Abstract
Objective: Postoperative cognitive dysfunction (POCD) is a common and severe complication of cardiovascular surgery. Lymphocyte-to-monocyte ratio (LMR) has been reported to be an independent predictor of lots of diseases associated with inflammation, but the association between the LMR and POCD is not clear. The present study aimed to investigate the potential value of LMR level to predict POCD in patients undergoing cardiovascular surgery. Methods: A prospective observational study was performed on the patients diagnosed with heart diseases undergoing cardiovascular surgeries with cardiopulmonary bypass. The leukocyte counts were measured by blood routine examination preoperatively. Then we calculated the LMR by dividing the lymphocyte count by the monocyte count. Neurocognitive functions were assessed 1 day before and 7 days after surgery. Perioperative factors were recorded to explore the relationship between LMR and POCD. Results: In total, 75 patients finished the whole study, while 34 patients developed POCD. The preoperative LMR level in the POCD group was higher than that in the non-POCD group. A cutoff value of 4.855 was identified to predict POCD occurrence according to ROC curve. The perioperative dynamic change of LMR level in the POCD group was higher than those in the non-POCD group. A cutoff value of 2.255 was identified to predict POCD occurrence according to ROC curve and the dynamic LMR change had similar varying trend with preoperative LMR level. Conclusions: The dynamic change of LMR level in the peripheral blood is associated with occurrence of POCD, and preoperative LMR level seems to be a prognostic biomarker of postoperative cognitive dysfunction in patients after cardiovascular surgery.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Gao
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Changliang Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hai Chen
- Targeted Tracer Research and Development Laboratory, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xueying Zhang
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Guan
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Xie
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanhua Qiu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Cheng
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Peilin Lv
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chan Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
175
|
Dong L, Li J, Zhang C, Liu D. Gut microbiota: a new player in the pathogenesis of perioperative neurocognitive disorder? IBRAIN 2021; 7:37-43. [PMID: 37786871 PMCID: PMC10529199 DOI: 10.1002/j.2769-2795.2021.tb00063.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/21/2021] [Accepted: 03/07/2021] [Indexed: 10/04/2023]
Abstract
Perioperative neurocognitive disorder (PND), including postoperative delirium and postoperative cognitive dysfunction (POCD), is a common postoperative complication in elderly patients, who represent an expanding segment of our population. PND is a multifactorial disease resulting in higher morbidity and mortality. The precise mechanism of PND is yet to be fully delineated. Identifying the modifiable risk factors and mechanisms for PND would be an important step forward in preventing such adverse events and thus improving patients' outcomes. It is increasingly recognized that gut microbiota also manifest effects in the central nervous system via the microbiota-gut-brain axis, which has emerged as an important player in shaping aspects of behavior and cognitive function. Recent studies have found that patients with cognitive dysfunction after surgery and anesthesia have obvious gut microbiome 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. Here, we present a broad topical review of the literature supporting the role of gut microbiota in PND. We provide an overview of the mechanisms underlying the pathogenesis of PND from pre-clinical and human studies. Therefore, gut microbiota could be a putative therapeutic target for PND in the future.
Collapse
Affiliation(s)
| | - Juan Li
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Chao Zhang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - De‐Xing Liu
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
176
|
A Prediction Model for Cognitive Impairment Risk in Colorectal Cancer after Chemotherapy Treatment. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6666453. [PMID: 33688501 PMCID: PMC7914097 DOI: 10.1155/2021/6666453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/01/2021] [Accepted: 02/10/2021] [Indexed: 11/17/2022]
Abstract
Background A prediction model can be developed to predict the risk of cancer-related cognitive impairment in colorectal cancer patients after chemotherapy. Methods A regression analysis was performed on 386 colorectal cancer patients who had undergone chemotherapy. Three prediction models (random forest, logistic regression, and support vector machine models) were constructed using collected clinical and pathological data of the patients. Calibration and ROC curves and C-indexes were used to evaluate the selected models. A decision curve analysis (DCA) was used to determine the clinical utility of the line graph. Results Three prediction models including a random forest, a logistic regression, and a support vector machine were constructed. The logistic regression model had the strongest predictive power with an area under the curve (AUC) of 0.799. Age, BMI, colostomy, complications, CRA, depression, diabetes, QLQ-C30 score, exercise, hypercholesterolemia, diet, marital status, education level, and pathological stage were included in the nomogram. The C-index (0.826) and calibration curve showed that the nomogram had good predictive ability and the DCA curves indicated that the model had strong clinical utility. Conclusions A prediction model with good predictive ability and practical clinical value can be developed for predicting the risk of cognitive impairment in colorectal cancer after chemotherapy.
Collapse
|
177
|
Tachi K, Fukuda T, Tanaka M. Olanzapine attenuates postoperative cognitive dysfunction in adult rats. Heliyon 2021; 7:e06218. [PMID: 33659744 PMCID: PMC7890212 DOI: 10.1016/j.heliyon.2021.e06218] [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: 06/04/2020] [Revised: 01/01/2021] [Accepted: 02/04/2021] [Indexed: 11/27/2022] Open
Abstract
Background Postoperative cognitive dysfunction (POCD) is associated with poor quality of life and difficulty working. Its impact may be greater in middle-aged patients than in elderly patients. Neuroinflammation is reported to be a main cause of POCD. Olanzapine has been reported to improve learning and memory functions. We therefore investigated olanzapine's effectiveness and mechanisms in an adult rat POCD model. Methods Six-month-old rats underwent laparotomy and lipopolysaccharide (LPS group) or LPS + olanzapine (OLA group) intraperitoneal injection or anesthesia alone (CON group) 1 week after a Barnes maze training session. A Barnes maze test trial was then conducted the day after surgery or anesthesia. The microglial activity in the hippocampus and cytokine levels were measured by Iba1 staining and enzyme-linked immunosorbent assay, respectively. Results The OLA group had significantly higher success rates of Barnes maze trial than the LPS group. The success rate in time of the OLA group was inferior to that of the CON group. On the other hand, the success rate in distance of the OLA group was similar to that of the CON group. Iba1 staining areas in the LPS and OLA groups were larger than that in the CON group; however, the staining area in the OLA group was smaller than that of the LPS group. Plasma interleukin-1β concentration in the LPS and OLA groups was significantly higher than that in the CON group; however, there was no significant difference between the LPS and OLA groups. Conclusion Olanzapine attenuated both spatial cognitive dysfunction and microglial activity of the hippocampus, which were induced by surgery and LPS injection. These effects were unrelated to inflammatory cytokine concentrations in plasma and hippocampus.
Collapse
Affiliation(s)
- Keitaro Tachi
- Department of Anesthesiology, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| | - Taeko Fukuda
- Department of Anesthesiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Anesthesiology, Kasumigaura Medical Center, Tsuchiura Center for Medical Education and Training, University of Tsukuba Hospital, Tsuchiura, Ibaraki, Japan
| | - Makoto Tanaka
- Department of Anesthesiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
178
|
Wang X, Chen L, Xu Y, Wang W, Wang Y, Zhang Z, Zheng J, Bao H. Gastrodin alleviates perioperative neurocognitive dysfunction of aged mice by suppressing neuroinflammation. Eur J Pharmacol 2021; 892:173734. [PMID: 33220272 DOI: 10.1016/j.ejphar.2020.173734] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/03/2020] [Accepted: 11/13/2020] [Indexed: 01/20/2023]
Abstract
Perioperative neurocognitive disorder (PND) is a common complication of elderly patients after surgery and lacks effective prevention and treatment measures. We investigated the effect and mechanism of gastrodin (GAS), a natural plant ingredient, on postoperative cognition induced by laparotomy in aged mice. Male aged (18 months) mice were subjected to laparotomy and orally treated with GAS (25, 50, and 100 mg/kg) 3 weeks before surgery and 1 week after surgery. In addition, some male aged (18 months) mice were subjected to viral vector or GSK-3β expression virus injection followed by laparotomy with or without 100 mg/kg GAS treatment. GAS improved learning and memory in aged mice after surgery. Surgery increased the levels of pro-inflammatory factors (TNF-α, IL-1β and IL-6) and decreased the level of an anti-inflammatory factor (IL-10) in the mouse hippocampus, and these changes were reversed by GAS treatment. GAS also suppressed the activation of microglia. GAS inhibited the phosphorylation of GSK-3β and Tau. Furthermore, surgery induced more serious cognitive dysfunction, inflammatory factors, activation of microglia, and phosphorylation of GSK-3β and Tau in GSK-3β overexpressing aged mice. The improvement of learning and memory, the reduction of inflammation and microglia activation, and the suppression of GSK-3β and Tau phosphorylation by GAS were prevented when GSK-3β was overexpressed in aged mice subjected to surgery. Our finding suggested that GAS exerts neuroprotective effects in aged mice subjected to laparotomy by suppressing neuroinflammation and GSK-3β and Tau phosphorylation. Thus, these findings suggest that GAS may be a promising agent for PND.
Collapse
Affiliation(s)
- Xiaoliang Wang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lihai Chen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yajie Xu
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wanling Wang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Youran Wang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhiyuan Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Jing Zheng
- Department of Neurosurgery, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China.
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
179
|
Granger KT, Barnett JH. Postoperative cognitive dysfunction: an acute approach for the development of novel treatments for neuroinflammation. Drug Discov Today 2021; 26:1111-1114. [PMID: 33497828 DOI: 10.1016/j.drudis.2021.01.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
Inflammation within the central nervous system (CNS; neuroinflammation) is a major contributor to lasting symptoms of traumatic brain injury and stroke, and likely has a casual role Alzheimer's disease (AD) and other neurodegenerative conditions. Therapeutic modulation of the immune processes that initiate and maintain neuroinflammation is of growing scientific interest but neuroinflammatory drug development is hampered by limited reliability and availability of neuroimaging or other biomarkers in humans. Better means of establishing drug efficacy on human neuroinflammation would have great value in accelerating the development of neuroinflammatory compounds for many clinical indications. Here, we discuss the use of postoperative cognitive decline (POCD), which is hypothesised to have a neuroinflammatory basis, as an acute indication to demonstrate the efficacy of novel neuroinflammatory drugs.
Collapse
Affiliation(s)
- Kiri T Granger
- Monument Therapeutics, Cambridge, UK; Cambridge Cognition, Cambridge, UK; School of Psychology, University of Nottingham, Nottingham, UK.
| | - Jennifer H Barnett
- Monument Therapeutics, Cambridge, UK; Cambridge Cognition, Cambridge, UK; Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
180
|
Yu L, Wen G, Zhu S, Hu X, Huang C, Yang Y. Abnormal phosphorylation of tau protein and neuroinflammation induced by laparotomy in an animal model of postoperative delirium. Exp Brain Res 2021; 239:867-880. [PMID: 33409674 DOI: 10.1007/s00221-020-06007-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Postoperative delirium (POD) is an acute neuropsychological disturbance after surgery, whose prevalence is related with advancing age. Neuroinflammation and abnormal tau phosphorylation that commonly presenting in Alzheimer's disease (AD) may contribute to the progression and duration of POD. To study the acute influence of surgery on cognitive function, wild type male C57BL/6 N mice were randomly divided into three groups: Control (CON), Laparotomy at 4 h and 24 h (LAP-4 h, LAP-24 h), then subjected to laparotomy under sevoflurane anaesthesia. The cognitive performance, peripheral and central inflammatory responses and tau phosphorylation levels were evaluated at 4 h and 24 h postoperatively. When LAP4-hrs displayed anxiety behaviors with high mRNA levels of inflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, IL-8, TNF-α and MCP-1 in the liver, and IL-8 in the hippocampus, results at 24 h were different. In the liver, only IL-10 protein was obviously elevated, but in the hippocampus, both pro- and anti-inflammatory cytokines were significantly decreased whilst the elimination of anxiety. The activity of major related kinases and phosphatases was remarkably changed which may contribute to the dephosphorylated tau protein. With tremendous neuropathological changes and significant numbers of activated microglias and astrocytes observed in the sub-regions of hippocampus, the memory impairment existed at both 4 h and 24 h. Since the association of dephosphorylated tau with POD, these findings may supply novel implications for the understanding of tauopathies and as a theoretical basis for preventions from the postoperative cognitive dysfunction (POCD).
Collapse
Affiliation(s)
- Le Yu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China.,Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Guanghua Wen
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China
| | - Shoufeng Zhu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Xianwen Hu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China.,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China
| | - Chunxia Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, People's Republic of China. .,Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, People's Republic of China.
| | - Yan Yang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230001, People's Republic of China.
| |
Collapse
|
181
|
Komleva YK, Lopatina OL, Gorina YV, Chernykh AI, Trufanova LV, Vais EF, Kharitonova EV, Zhukov EL, Vahtina LY, Medvedeva NN, Salmina AB. Expression of NLRP3 Inflammasomes in Neurogenic Niche Contributes to the Effect of Spatial Learning in Physiological Conditions but Not in Alzheimer's Type Neurodegeneration. Cell Mol Neurobiol 2021; 42:1355-1371. [PMID: 33392919 DOI: 10.1007/s10571-020-01021-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/27/2020] [Indexed: 12/27/2022]
Abstract
A common feature of neurodegenerative disorders, in particular Alzheimer's disease (AD), is a chronic neuroinflammation associated with aberrant neuroplasticity. Development of neuroinflammation affects efficacy of stem and progenitor cells proliferation, differentiation, migration, and integration of newborn cells into neural circuitry. However, precise mechanisms of neurogenesis alterations in neuroinflammation are not clear yet. It is well established that expression of NLRP3 inflammasomes in glial cells marks neuroinflammatory events, but less is known about contribution of NLRP3 to deregulation of neurogenesis within neurogenic niches and whether neural stem cells (NSCs), neural progenitor cells (NPCs) or immature neuroblasts may express inflammasomes in (patho)physiological conditions. Thus, we studied alterations of neurogenesis in rats with the AD model (intra-hippocampal injection of Aβ1-42). We found that in Aβ-affected brain, number of CD133+ cells was elevated after spatial training in the Morris water maze. The number of PSA-NCAM+ neuroblasts diminished by Aβ injection was completely restored by subsequent spatial learning. Spatial training leads to elevated expression of NLRP3 inflammasomes in the SGZ (subgranular zones): CD133+ and PSA-NCAM+ cells started to express NLRP3 in sham-operated, but not AD rats. Taken together, our data suggest that expression of NLRP3 inflammasomes in CD133+ and PSA-NCAM+ cells may contribute to stimulation of adult neurogenesis in physiological conditions, whereas Alzheimer's type neurodegeneration abolishes stimuli-induced overexpression of NLRP3 within the SGZ neurogenic niche.
Collapse
Affiliation(s)
- Yulia K Komleva
- The Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia. .,Research Institute of Molecular Medicine and Pathobiochemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia.
| | - O L Lopatina
- The Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia.,Research Institute of Molecular Medicine and Pathobiochemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Ya V Gorina
- The Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia.,Research Institute of Molecular Medicine and Pathobiochemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - A I Chernykh
- Research Institute of Molecular Medicine and Pathobiochemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - L V Trufanova
- The Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - E F Vais
- The Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - E V Kharitonova
- The Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - E L Zhukov
- Department of Pathological Anatomy Named After Prof. P.G. Podzolkov, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - L Yu Vahtina
- Department of Human Anatomy, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - N N Medvedeva
- Department of Human Anatomy, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - A B Salmina
- The Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia.,Research Institute of Molecular Medicine and Pathobiochemistry, Ministry of Health of the Russian Federation, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| |
Collapse
|
182
|
Wang Z, Liu T, Yin C, Li Y, Gao F, Yu L, Wang Q. Electroacupuncture Pretreatment Ameliorates Anesthesia and Surgery-Induced Cognitive Dysfunction via Activation of an α7-nAChR Signal in Aged Rats. Neuropsychiatr Dis Treat 2021; 17:2599-2611. [PMID: 34413646 PMCID: PMC8370114 DOI: 10.2147/ndt.s322047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Postoperative cognitive dysfunction (POCD) after anesthesia and surgery (AS) is a common complication in the elderly population. A cholinergic-dependent signal, the alpha7-nicotinic acetylcholine receptor (α7-nAChR), has been suggested to regulate cognitive processes in a variety of neurologic diseases. In the current study, we determined whether electroacupuncture (EA) pretreatment ameliorates AS-induced POCD in aged rats, as well as the underlying mechanism. METHODS Male Sprague-Dawley rats (20 months old) were randomly assigned to the following 5 groups (n=12): vehicle; POCD (tibial fracture surgery); EA plus POCD; EA plus POCD and alpha-bungarotoxin (α-BGT); and POCD plus α-BGT groups. Alpha-bungarotoxin (1 μg/kg), a selective antagonist of α7-nAChR, was administrated via intraperitoneal injection before EA. Thirty days post-AS, the Morris water maze and a novel objective recognition test were used to evaluate cognitive function. Neuronal amount, apoptosis, microglial activation, percentage of high mobility group box 1 (HMGB1)- and nuclear factor-κB (NF-κB)-positive microglia, and levels of HMGB-1 downstream factors, including NF-κB, interleukin-6 (IL-6), and IL-1β, were detected by Nissl staining, immunofluorescence, and Western blot assays. RESULTS EA pretreatment significantly increased crossing platform times and elevated the time with a novel object, restored the quantity of neurons, decreased TUNEL-positive neurons, alleviated activation of microglia, downregulated expression of HMGB1 and NF-κB in the microglia, and reduced levels of phosphor-NF-κB, IL-6, and IL-1β 35 days after AS, while α-BGT partially reversed these changes. CONCLUSION EA pretreatment improved AS-induced POCD in aged rats, and the underlying mechanism may be associated with inhibition of HMGB1-NF-κB via an α7-nAChR signal in the microglia.
Collapse
Affiliation(s)
- Zhigang Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China.,Department of Anesthesiology, Handan Central Hospital, Handan, Hebei, People's Republic of China
| | - Tianlin Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China
| | - Chunping Yin
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China
| | - Yanan Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China
| | - Fang Gao
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China
| | - Lili Yu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China
| | - Qiujun Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China
| |
Collapse
|
183
|
Resveratrol Mitigates Hippocampal Tau Acetylation and Cognitive Deficit by Activation SIRT1 in Aged Rats following Anesthesia and Surgery. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4635163. [PMID: 33381265 PMCID: PMC7758127 DOI: 10.1155/2020/4635163] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/09/2020] [Accepted: 11/28/2020] [Indexed: 12/14/2022]
Abstract
Postoperative cognitive dysfunction (POCD) is a sever postsurgical neurological complication in the elderly population. As the global acceleration of population ageing, POCD is proved to be a great challenge to the present labor market and healthcare system. In the present study, our findings showed that tau acetylation mediated by SIRT1 deficiency resulted in tau hyperphosphorylation in the hippocampus of the aged POCD model and consequently contributed to cognitive impairment. Interestingly, pretreatment with resveratrol almost restored the expression of SIRT1, reduced the levels of acetylated tau and hyperphosphorylated tau in the hippocampus, and improved the cognitive performance in the behavioral tests. What is more, we observed that microglia-derived neuroinflammation resulting from SIRT1 inhibition in microglia probably aggravated the tau acetylation in cultured neurons in vitro. Our findings supported the notion that activation SIRT1 provided dually beneficial effect in the aged POCD model. Taken together, our findings provided the initial evidence that tau acetylation was associated with cognitive impairment in the aged POCD model and paved a promising avenue to prevent POCD by inhibiting tau acetylation in a SIRT1-dependent manner.
Collapse
|
184
|
Shen Y, Zhang Y, Chen L, Du J, Bao H, Xing Y, Cai M, Si Y. Chemokine CXCL13 acts via CXCR5-ERK signaling in hippocampus to induce perioperative neurocognitive disorders in surgically treated mice. J Neuroinflammation 2020; 17:335. [PMID: 33161894 PMCID: PMC7648984 DOI: 10.1186/s12974-020-02013-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
Background Perioperative neurocognitive disorders (PNDs) occur frequently after surgery and worsen patient outcome. How C-X-C motif chemokine (CXCL) 13 and its sole receptor CXCR5 contribute to PNDs remains poorly understood. Methods A PND model was created in adult male C57BL/6J and CXCR5−/− mice by exploratory laparotomy. Mice were pretreated via intracerebroventricular injection with recombinant CXCL13, short hairpin RNA against CXCL13 or a scrambled control RNA, or ERK inhibitor PD98059. Then surgery was performed to induce PNDs, and animals were assessed in the Barnes maze trial followed by a fear-conditioning test. Expression of CXCL13, CXCR5, and ERK in hippocampus was examined using Western blot, quantitative PCR, and immunohistochemistry. Levels of interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) in hippocampus were assessed by Western blot. Results Surgery impaired learning and memory, and it increased expression of CXCL13 and CXCR5 in the hippocampus. CXCL13 knockdown partially reversed the effects of surgery on CXCR5 and cognitive dysfunction. CXCR5 knockout led to similar cognitive outcomes as CXCL13 knockdown, and it repressed surgery-induced activation of ERK and production of IL-1β and TNF-α in hippocampus. Recombinant CXCL13 induced cognitive deficits and increased the expression of phospho-ERK as well as IL-1β and TNF-α in hippocampus of wild-type mice, but not CXCR5−/− mice. PD98059 partially blocked CXCL13-induced cognitive dysfunction as well as production of IL-1β and TNF-α. Conclusions CXCL13-induced activation of CXCR5 may contribute to PNDs by triggering ERK-mediated production of pro-inflammatory cytokines in hippocampus.
Collapse
Affiliation(s)
- Yanan Shen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yuan Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Lihai Chen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Jiayue Du
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yan Xing
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 211118, People's Republic of China
| | - Mengmeng Cai
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yanna Si
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China.
| |
Collapse
|
185
|
Muscat SM, Barrientos RM. Lifestyle modifications with anti-neuroinflammatory benefits in the aging population. Exp Gerontol 2020; 142:111144. [PMID: 33152515 DOI: 10.1016/j.exger.2020.111144] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/18/2020] [Accepted: 10/28/2020] [Indexed: 01/03/2023]
Abstract
Aging-associated microglial priming results in the potential for an exaggerated neuroinflammatory response to a subsequent inflammatory challenge in regions of the brain known to support learning and memory. This excessive neuroinflammation in the aging brain is known to occur following a variety of peripheral insults, including infection and surgery, where it has been associated with precipitous declines in cognition and memory. As the average lifespan increases worldwide, identifying interventions to prevent and treat aging-associated excessive neuroinflammation and ensuing cognitive impairments is of critical importance. Lifestyle has emerged as a potential non-pharmacological target in this endeavor. Here, we review important and recent preclinical and clinical literature demonstrating the anti-inflammatory effects of lifestyle modifications such as exercise, diet, and environmental enrichment in the context of aging and memory. Importantly, we focus on research indicating that these lifestyle modifications do not need to be lifelong, suggesting that such interventions may be efficacious in the prevention and treatment of aging- and neuroinflammation-associated cognitive impairment, even when initiated in older age.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
186
|
Ye C, Zhang Y, Luo S, Cao Y, Gao F, Wang E. Correlation of Serum BACE1 With Emergence Delirium in Postoperative Patients: A Preliminary Study. Front Aging Neurosci 2020; 12:555594. [PMID: 33192455 PMCID: PMC7655534 DOI: 10.3389/fnagi.2020.555594] [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: 06/18/2020] [Accepted: 09/28/2020] [Indexed: 11/24/2022] Open
Abstract
Background: The mechanism underlying delirium, a common acute fluctuating mental state, may be related to the activation of a neuroinflammatory response. In this study, we attempted to investigate whether plasma inflammatory response markers, vascular and cerebrovascular injury-related markers, and neurodegeneration-associated markers were associated with emergence delirium (ED). Methods: Patients aged 50 years or above who underwent elective laparoscopic surgery under general anesthesia were included in this study. Delirium was assessed postoperatively with the Richmond Agitation Sedation Scale (RASS) and the Confusion Assessment Method for the Intensive Care Unit (CAM-ICU) scale. Plasma samples were collected from ED patients and non-ED patients to test concentrations of inflammation markers, including interleukin 6 (IL-6), chitinase 3-like 1 (CHI3L1), S100 calcium-binding protein B (S100B), lipoprotein-associated phospholipase-A2 (Lp-PLA2), and macrophage migration inhibitory factor (MIF); vascular and cerebrovascular injury-related markers, including intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule (VCAM-1); and neurodegeneration-associated markers, including alpha-synuclein (α-Syn) and β-secretase 1 (BACE1). Binary logistic regression analysis was performed to analyze the relationship between biomarkers and ED, and receiver operating characteristic (ROC) curves were used to analyze the diagnostic value of biomarkers. Results: A total of 104 patients were included in this study, with an average age of 63.69 ± 7.21. IL-6 (OR = 2.73, 95% CI: 1.66–6.44, P = 0.022), S100B (OR = 4.74, 95% CI: 1.88–11.95, P = 0.001), and BACE1 (OR = 6.54, 95% CI: 2.57–16.65, P < 0.000) were independent biological indicators for the occurrence of ED.CHI3L1, Lp-PLA2, MIF, ICAM-1, VCAM-1, and α-Syn were unrelated to ED. Plasma BACE1 level had a possible diagnostic value for ED [area under curve (AUC) = 0.75, 95% CI: 0.66–0.85], whereas plasma IL-6 (AUC = 0.62, 95% CI: 0.51–0.73) and S100B (AUC = 0.65, 95% CI: 0.54–0.76) levels had little diagnostic value for distinguishing ED vs. non-ED. Conclusion: Higher levels of systemic inflammation marker IL-6, cerebral inflammation marker S100B, and neurodegeneration-associated marker BACE1 are related to ED. Plasma BACE1 may be a potential diagnostic biomarker for ED.
Collapse
Affiliation(s)
- Chunyan Ye
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Yanrong Zhang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Sumei Luo
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Yanan Cao
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Feng Gao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
| |
Collapse
|
187
|
Yu X, Xie Y. Effect of dexmedetomidine combined with etomidate on IL-17A and S-100β expression levels in rats with postoperative cognitive dysfunction. Exp Ther Med 2020; 20:275. [PMID: 33200000 DOI: 10.3892/etm.2020.9405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 08/03/2020] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to explore the effects of dexmedetomidine combined with etomidate on the expression levels of interleukin (IL)-17A and S-100β in rats with postoperative cognitive dysfunction (POCD). A total of 50 SD rats were randomly allocated into the control group, model group, etomidate group (Eto group), dexmedetomidine group (Dex group) and dexmedetomidine combined with etomidate group (Dex-Eto group). Inhalation anesthesia was used in all five groups. Apart from the control group, partial lobectomy was performed to construct a rat model of cognitive dysfunction. The rats of the model group received no intravenous anesthesia, except general anesthesia with intubation. Morris water maze test was performed before injection (T0), at the 1st day (T1), the 3rd day (T2) and the 5th day (T3) after operation to assess the memory ability of the rats. At the end of T3, the expression levels of IL-17A, S-100β, TNF-α, IL-6 and IL-1β in serum were detected by ELISA and the expression of NF-κB p65 by western blot analysis. Compared with the control group, the model group showed an increased escape latency and swimming distance, decreased number of times of crossing the platform and target quadrant residence time, and increased expression levels of IL-17A, S-100β, TNF-α, IL-6, IL-1β and NF-κB p65. Compared with the model group, the escape latency and swimming distance in the Dex, Eto and Dex-Eto groups were reduced, whereas the number of times of crossing the platform and the target quadrant residence time were increased. In addition, the expression levels of IL-17A, S-100β, TNF-α, IL-6, IL-1β and NF-κB p65 were decreased in the Dex, Eto and Dex-Eto groups, compared with the model group. Among the Dex, Eto and Dex-Eto groups, the escape latency and swimming distance in the Dex-Eto group were the shortest, the number of times of crossing the platform and the target quadrant residence time were the highest, and IL-17A, S-100β, TNF-α, IL-6, IL-1β and NF-κB p65 expression levels were the lowest. In conclusion, dexmedetomidine combined with etomidate can effectively improve POCD.
Collapse
Affiliation(s)
- Xiaoli Yu
- Department of Anesthesiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yunfang Xie
- Department of Anesthesiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
188
|
Jin Z, Hu J, Ma D. Postoperative delirium: perioperative assessment, risk reduction, and management. Br J Anaesth 2020; 125:492-504. [DOI: 10.1016/j.bja.2020.06.063] [Citation(s) in RCA: 238] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/22/2020] [Accepted: 06/20/2020] [Indexed: 12/20/2022] Open
|
189
|
Lan F, Lin G, Cao G, Li Z, Ma D, Liu F, Duan M, Fu H, Xiao W, Qi Z, Wang T. Altered Intrinsic Brain Activity and Functional Connectivity Before and After Knee Arthroplasty in the Elderly: A Resting-State fMRI Study. Front Neurol 2020; 11:556028. [PMID: 33133006 PMCID: PMC7550714 DOI: 10.3389/fneur.2020.556028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/18/2020] [Indexed: 12/03/2022] Open
Abstract
Objective: This study aimed to investigate the brain functional alterations with resting-state functional magnetic resonance imaging (rs-fMRI) in older patients with knee osteoarthritis (KOA) before and after total knee arthroplasty (TKA) and to assess the causal relationship of the brain function and neuropsychological changes. Methods: We performed rs-fMRI to investigate brain function of 23 patients aged ≥65 with KOA and 23 healthy matched controls. Of the KOA patients, 15 completed postoperative rs-fMRI examinations. Analyzes of the amplitude of low-frequency fluctuation (ALFF) and functional connectivity (FC) were used to estimate differences in brain functional parameters between KOA patients, postoperative patients, and the controls. The relationship between changes of pre- and post-surgical status in ALFF and neuropsychological test results was analyzed. Results: Compared with the controls, all patients with KOA exhibited decreased ALFF in the default mode network (bilateral angular gyrus, precuneus gyrus, medial superior frontal gyrus) and increased ALFF in the bilateral amygdala and cerebellum posterior lobe before surgery (P < 0.001). Altered ALFF persisted in the same brain regions 1 week postoperatively. The decreased ALFF in the left precuneus gyrus and middle temporal gyrus was found after surgery when compared with preoperative data (P < 0.01). Preoperatively, the KOA patients exhibited increased FC between the left precuneus gyrus and the right supplementary motor area compared to the controls (P < 0.001), but this connectivity became no significant difference after TKA. The left Cerebelum_9 was found to have decreased FC with the right precuneus gyrus postoperatively (P < 0.001) although this was not significantly different before surgery. The significantly altered ALFF values were not correlated with changes in cognitive assessment scores. Conclusion: In older patients with end-stage KOA, functional alterations in important brain regions were detected with the persistence and further changes observed at an early stage after knee replacement. Our data further our understanding of brain functional abnormalities and cognitive impairment in older patients following knee replacement, which may provide therapeutic targets for preventive/treatment strategy to be developed. Trial registration: Clinical Trial Registration: http://www.chictr.org.cn/index.aspx, ChiCTR1800016437; Registered June 1, 2018.
Collapse
Affiliation(s)
- Fei Lan
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing Institute for Brain Disorders, Beijing, China
| | - Guanwen Lin
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing Institute for Brain Disorders, Beijing, China.,Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Guanglei Cao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zheng Li
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Daqing Ma
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Anaesthesia Research of the Section of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital, London, United Kingdom
| | - Fangyan Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing Institute for Brain Disorders, Beijing, China
| | - Mei Duan
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing Institute for Brain Disorders, Beijing, China
| | - Huiqun Fu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing Institute for Brain Disorders, Beijing, China
| | - Wei Xiao
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhigang Qi
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tianlong Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
190
|
Amiri AA, Karvandian K, Ramezani N, Amiri AA. Short-term memory impairment in patients undergoing general anesthesia and its contributing factors. Saudi J Anaesth 2020; 14:454-458. [PMID: 33447186 PMCID: PMC7796728 DOI: 10.4103/sja.sja_651_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/29/2019] [Indexed: 11/25/2022] Open
Abstract
Background: Short-term memory disorder following surgery and anesthesia is a common complication of anesthesia and a common complaint of the patients. Aims: This study was designed to assess memory impairment in patients undergoing elective surgery, investigate the effect of general anesthesia (GA) on memory, and identify the factors contributing to it, as well as the specific effect of anesthesia on each of the memory domains. Setting and Design: This cross-sectional study was performed in a university hospital. Methods and Materials: Patients with the American Society of Anesthesiologists (ASA) Class I, II, and III who were candidates for elective abdominal surgery were enrolled. Patients answered several questions based on the Wechsler Memory Scale–Revised V (WMS-R-V), a standardized questionnaire, minutes before entering the operating room (OR) and again after 24 h postoperation, and the differences were recorded. Statistical Analysis: Analysis was performed using T-independent and Chi-square tests with Pearson's coefficient and Fischer's exact test and Man–Whitney test. Data were analyzed using the Statistical Package for the Social Sciences (SPSS) software. Results: Four hundred patients (198 females and 202 males) with a mean age of 50.75 years were enrolled in our study. Our study results showed that short-term memory after GA was significantly decreased compared with preanesthesia (P < 0.05). There was no significant relationship between memory disorder following GA and gender (P = 0.18) or comorbidities (P = 0.138). However, older age was found to be a contributing factor to memory loss following GA (P < 0.001). The highest and lowest effect of GA were found on the number repeat (45.2%) and personal information (16.2%) domain of the memory. Conclusion: GA significantly reduces the patient's short-term memory after the surgery.
Collapse
Affiliation(s)
- Ali Ahmadzadeh Amiri
- Department of Anesthesiology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Kasra Karvandian
- Department of Anesthesiology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Ramezani
- Department of Anesthesiology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ahmadzadeh Amiri
- Department of Anesthesiology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
191
|
Dabrowski W, Siwicka-Gieroba D, Gasinska-Blotniak M, Zaid S, Jezierska M, Pakulski C, Williams Roberson S, Wesley Ely E, Kotfis K. Pathomechanisms of Non-Traumatic Acute Brain Injury in Critically Ill Patients. ACTA ACUST UNITED AC 2020; 56:medicina56090469. [PMID: 32933176 PMCID: PMC7560040 DOI: 10.3390/medicina56090469] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/27/2022]
Abstract
Delirium, an acute alteration in mental status characterized by confusion, inattention and a fluctuating level of arousal, is a common problem in critically ill patients. Delirium prolongs hospital stay and is associated with higher mortality. The pathophysiology of delirium has not been fully elucidated. Neuroinflammation and neurotransmitter imbalance seem to be the most important factors for delirium development. In this review, we present the most important pathomechanisms of delirium in critically ill patients, such as neuroinflammation, neurotransmitter imbalance, hypoxia and hyperoxia, tryptophan pathway disorders, and gut microbiota imbalance. A thorough understanding of delirium pathomechanisms is essential for effective prevention and treatment of this underestimated pathology in critically ill patients.
Collapse
Affiliation(s)
- Wojciech Dabrowski
- Department of Anaesthesiology and Intensive Care, Medical University of Lublin, 20-954 Lublin, Poland; (D.S.-G.); (M.G.-B.); (M.J.)
- Correspondence: or (W.D.); (K.K.)
| | - Dorota Siwicka-Gieroba
- Department of Anaesthesiology and Intensive Care, Medical University of Lublin, 20-954 Lublin, Poland; (D.S.-G.); (M.G.-B.); (M.J.)
| | - Malgorzata Gasinska-Blotniak
- Department of Anaesthesiology and Intensive Care, Medical University of Lublin, 20-954 Lublin, Poland; (D.S.-G.); (M.G.-B.); (M.J.)
| | - Sami Zaid
- Department of Anaesthesia, Al-Emadi-Hospital Doha, P.O. Box 5804 Doha, Qatar;
| | - Maja Jezierska
- Department of Anaesthesiology and Intensive Care, Medical University of Lublin, 20-954 Lublin, Poland; (D.S.-G.); (M.G.-B.); (M.J.)
| | - Cezary Pakulski
- Department of Anaesthesiology, Intensive Therapy and Emergency Medicine, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland;
| | - Shawniqua Williams Roberson
- Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Vanderbilt University Medical Center, 1211, Nashville, TN 37232, USA; (S.W.R.); (E.W.E.)
- Department of Neurology, Vanderbilt University Medical Center, 1211, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, 1211, Nashville, TN 37232, USA
| | - Eugene Wesley Ely
- Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Vanderbilt University Medical Center, 1211, Nashville, TN 37232, USA; (S.W.R.); (E.W.E.)
- Geriatric Research, Education and Clinical Center (GRECC), Tennessee Valley Veterans Affairs Healthcare System, 1310, Nashville, TN 37212, USA
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, 1211, Nashville, TN 37232, USA
| | - Katarzyna Kotfis
- Department of Anaesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University, 70-111 Szczecin, Poland
- Correspondence: or (W.D.); (K.K.)
| |
Collapse
|
192
|
Li M, Su S, Cai W, Cao J, Miao X, Zang W, Gao S, Xu Y, Yang J, Tao YX, Ai Y. Differentially Expressed Genes in the Brain of Aging Mice With Cognitive Alteration and Depression- and Anxiety-Like Behaviors. Front Cell Dev Biol 2020; 8:814. [PMID: 33015035 PMCID: PMC7493670 DOI: 10.3389/fcell.2020.00814] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/31/2020] [Indexed: 01/21/2023] Open
Abstract
Despite the great increase in human lifespan with improved medical care, the physiological and pathological changes such as memory and cognitive disorders and associated anxiety and depression are major concern with aging. Molecular mechanisms underlying these changes are little known. The present study examined the differentially expressed genes (DEGs) and the genes with differentially expressed isoforms in three brain regions, anterior cingulate cortex (ACC), amygdala and hippocampus, throughout the lifespan of mice. Compared to 2-month old mice, both 12- and 24-month old mice displayed memory and cognitive impairments in the Morris water maze, Y-maze, and novel object recognition tests and depression- and anxiety-like behaviors in the tail suspension, forced swimming, open field, and elevated plus maze tests. RNA sequencing analysis identified 634 and 1078 DEGs in ACC, 453 and 1015 DEGs in the amygdala and 884 and 1054 DEGs in hippocampus in the 12- and 24-month old mice, respectively. Similarly, many genes with differentially expressed isoforms were also identified in these three brain regions in the 12- and 24-month old mice. Further functional analysis revealed that many DEGs and the genes with differentially expressed isoforms in the ACC and amygdala were mapped to depression- and anxiety-related genes, respectively and that a lot of DEGs and the genes with differentially expressed isoforms in hippocampus were mapped to cognitive dysfunction-related genes from both 12- and 24-month old mice. All of these mapped DEGs and the genes with differentially expressed isoforms were closely related to neuroinflammation. Our findings indicate that these neuroinflammation-related DEGs and the genes with differentially expressed isoforms are likely new targets in the management of memory/cognitive impairment and emotional disorders during the aging.
Collapse
Affiliation(s)
- Mengqi Li
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, China
| | - Songxue Su
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, China.,Department of Anatomy, College of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Weihua Cai
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, China.,Department of Anatomy, College of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Jing Cao
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, China.,Department of Anatomy, College of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Xuerong Miao
- Department of Anesthesiology and Intensive Care, Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Weidong Zang
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, China.,Department of Anatomy, College of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Shichao Gao
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York, Buffalo, NY, United States
| | - Ying Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York, Buffalo, NY, United States
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Yanqiu Ai
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, China
| |
Collapse
|
193
|
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: 3.0] [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
|
194
|
Tasbihgou SR, Absalom AR. Postoperative neurocognitive disorders. Korean J Anesthesiol 2020; 74:15-22. [PMID: 32623846 PMCID: PMC7862941 DOI: 10.4097/kja.20294] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022] Open
Abstract
A decline in cognitive function is a frequent complication of major surgery. Postoperative cognitive impairments have generally been divided into short- (postoperative delirium) and long-term disturbances (postoperative cognitive dysfunction [POCD]). Long-term impairments are often subtle and overlooked. They need to be objectively assessed using neuropsychological tests to be diagnosed. Although POCD has been the subject of considerable research over the past decades, it remains uncertain why some patients do not return to preoperative levels of cognitive function. Surgery and anesthesia have both been implicated to play a role in POCD development, and certain patient-related factors, such as advanced age and low preoperative baseline cognitive function, have consistently been found to predict postoperative cognitive decline. This article will present an overview of POCD and its etiology and provide advice on possible strategies on its prevention.
Collapse
Affiliation(s)
- Setayesh Reza Tasbihgou
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anthony Ray Absalom
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
195
|
Saito J, Ma D. Can dexmedetomidine protect against surgical stress response? Clin Transl Med 2020; 10:e96. [PMID: 32594657 PMCID: PMC7403666 DOI: 10.1002/ctm2.96] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 05/23/2020] [Indexed: 01/01/2023] Open
Affiliation(s)
- Junichi Saito
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
196
|
Zhou HS, Cao SM, Liao HS, Huo HY. Hirsutanol A exhibits neuroprotective activities against sevoflurane-induced neurotoxicity in aged rats. Anat Rec (Hoboken) 2020; 304:591-601. [PMID: 32536020 DOI: 10.1002/ar.24473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/26/2020] [Accepted: 04/03/2020] [Indexed: 01/07/2023]
Abstract
The neurotoxicity of the inhaled anesthetic, sevoflurane, has been documented in a number of studies. In this study, we conducted experiments to investigate whether Hirsutanol A (HA), a sesquiterpene compound from the fungus, Chondrostereum sp., can provide protection from sevoflurane-induced neurological toxicity in aged rats, and analyzed the underlying mechanisms. The cognitive dysfunction of rats following sevoflurane exposure was evaluated by behavioral tests. The neuronal cell survival was determined by Nissl staining. In addition, human neuroblastoma H4 cells were exposed to sevoflurane to establish an in vitro model. Apoptotic marker expression in hippocampal tissue was determined by western blotting. Cell apoptosis in vitro was also examined by TUNEL assay and flow cytometry. The expression and translocation of Nrf2 were examined by both western blot and immunofluorescence staining. Our results show that HA significantly attenuated sevoflurane-induced cognitive impairment in aged rats. In addition, HA treatment decreased sevoflurane-induced neuronal apoptosis in the hippocampus and alleviated Aβ accumulation. Our results also show that the neuroprotective effect of HA is associated with the activation of Nrf2 signaling. Human neuroblastoma H4 cells were used as a model to examine the protective activity of HA against sevoflurane-induced neurotoxicity. In addition, our results show that the inhibition of Nrf2 by a specific inhibitor or targeting siRNA significantly compromises the attenuating effect of HA on sevoflurane-induced cell apoptosis and Aβ accumulation. Our results suggest that HA may function as a neuroprotective agent against sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Hong-She Zhou
- Department of Anesthesiology, Xi'an High Tech Hospital, Xi'an, Shaanxi, China
| | - Shu-Mei Cao
- Department of Anesthesiology, Xi'an High Tech Hospital, Xi'an, Shaanxi, China
| | - Hua-Shan Liao
- Department of Anesthesiology, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Hong-Yan Huo
- Department of Anesthesiology, Xi'an High Tech Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
197
|
Yang N, Li Z, Han D, Mi X, Tian M, Liu T, Li Y, He J, Kuang C, Cao Y, Li L, Ni C, Wang JQ, Guo X. Autophagy prevents hippocampal α-synuclein oligomerization and early cognitive dysfunction after anesthesia/surgery in aged rats. Aging (Albany NY) 2020; 12:7262-7281. [PMID: 32335546 PMCID: PMC7202547 DOI: 10.18632/aging.103074] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 03/29/2020] [Indexed: 12/13/2022]
Abstract
Stress-induced α-synuclein aggregation, especially the most toxic species (oligomers), may precede synaptic and cognitive dysfunction. Under pathological conditions, α-synuclein is degraded primarily through the autophagic/lysosomal pathway. We assessed the involvement of autophagy in α-synuclein aggregation and cognitive impairment following general anesthesia and surgical stress. Autophagy was found to be suppressed in the aged rat hippocampus after either 4-h propofol anesthesia alone or 2-h propofol anesthesia during a laparotomy surgery. This inhibition of autophagy was accompanied by profound α-synuclein oligomer aggregation and neurotransmitter imbalances in the hippocampus, along with hippocampus-dependent cognitive deficits. These events were not observed 18 weeks after propofol exposure with or without surgical stress. The pharmacological induction of autophagy using rapamycin markedly suppressed α-synuclein oligomerization, restored neurotransmitter equilibrium, and improved cognitive behavior after prolonged anesthesia or anesthesia combined with surgery. Thus, both prolonged propofol anesthesia alone and propofol anesthesia during surgery impaired autophagy, which may have induced abnormal hippocampal α-synuclein aggregation and neurobehavioral deficits in aged rats. These findings suggest that the activation of autophagy and the clearance of pathological α-synuclein oligomers may be novel strategies to ameliorate the common occurrence of postoperative cognitive dysfunction.
Collapse
Affiliation(s)
- Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Miao Tian
- Chinese Traditional and Herbal Drugs Editorial Office, Tianjin Institute of Pharmaceutical Research, Tianjin 300193, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Jindan He
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Chongshen Kuang
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Yiyun Cao
- Department of Anesthesiology, Shanghai Sixth People's Hospital East Affiliated with Shanghai University of Medicine and Health Sciences, Shanghai 200233, China
| | - Lunxu Li
- Department of Anesthesiology, Peking University International Hospital, Beijing 102200, China
| | - Cheng Ni
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - John Q Wang
- Department of Anesthesiology, University of Missouri Kansas City, School of Medicine, Kansas, MO 64110, USA
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
198
|
Le K, Wu S, Chibaatar E, Ali AI, Guo Y. Alarmin HMGB1 Plays a Detrimental Role in Hippocampal Dysfunction Caused by Hypoxia-Ischemia Insult in Neonatal Mice: Evidence from the Application of the HMGB1 Inhibitor Glycyrrhizin. ACS Chem Neurosci 2020; 11:979-993. [PMID: 32073822 DOI: 10.1021/acschemneuro.0c00084] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hippocampal dysfunction related to cognitive impairment and emotional disorders in young children and adolescents caused by neonatal hypoxic-ischemic brain injury (HIBI) has attracted increasing attention in recent years. Crosstalk between the nervous and immune systems organized by hypoxia-ischemia (HI) insult may contribute to hippocampal dysfunction after HIBI. Extracellular HMGB1 functions as a damage-associated molecular pattern to instigate and amplify inflammatory responses, but whether this molecule is correlated with hippocampal dysfunction after HIBI is largely unknown. Therefore, this study examined hippocampal function after HMGB1 inhibition in an experimental HIBI model to verify the hypothesis that HMGB1 is a key mediator of hippocampal neuropathology in neonatal HIBI. By administering different doses of the HMGB1-specific inhibitor glycyrrhizin (GLY), we first found that GLY reversed the HI insult-induced loss of neurons and myelin in the hippocampal region and neurobehavioral impairments, partially in a dose-dependent manner, and based on this, we determined the optimal drug concentration to be 50 mg/kg. Subsequent analysis found that this neuroprotective effect was achieved through the inhibition of HMGB1 expression and nucleocytoplasmic translocation, a reduction in the abnormal expression of proteins associated with the downstream signaling pathway of HMGB1, a decrease in the inflammatory response, the suppression of increases in microglia/astrocytes, and the inhibition of hippocampal cell apoptosis. Collectively, our discoveries contribute to the rising appreciation of the role of HMGB1 in the neuropathology of hippocampal dysfunction and related behavioral outcomes following HIBI.
Collapse
Affiliation(s)
- Kai Le
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province 210009, China
- School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Shanshan Wu
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province 210009, China
- School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Enkhmurun Chibaatar
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province 210009, China
- School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Abdoulaye Idriss Ali
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province 210009, China
- School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, China
| | - Yijing Guo
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province 210009, China
| |
Collapse
|
199
|
Yuan Y, Li Z, Yang N, Han Y, Ji X, Han D, Wang X, Li Y, Liu T, Yuan F, He J, Liu Y, Ni C, Zou P, Wang G, Guo X, Zhou Y. Exosome α-Synuclein Release in Plasma May be Associated With Postoperative Delirium in Hip Fracture Patients. Front Aging Neurosci 2020; 12:67. [PMID: 32231560 PMCID: PMC7082759 DOI: 10.3389/fnagi.2020.00067] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/25/2020] [Indexed: 01/24/2023] Open
Abstract
Background: Little is known about the underlying mechanisms of the similarities in the core features of postoperative delirium (POD) and α-synuclein (α-syn)-related cognitive disorders. We herein investigated associations between fluctuated levels of exosomal α-syn in the plasma and POD presentation in geriatric hip fracture patients. Methods: We conducted an observational, prospective, and 1:1 matched (on age older than 65, hip fracture diagnosis, American Society of Anesthesiologist’ (ASA) physical status, duration of surgery, and intraoperative bleeding) case-control study: POD cases and non-POD controls were selected from the overall cohort by using Confusion Assessment Method (CAM). Delirium severity was measured by the Memorial Delirium Assessment Scale (MDAS). Plasma exosome levels of α-syn were examined preoperatively and at the time that POD was diagnosed, by using an established immunocapture technology based on a putative brain-cell-specific marker. Circulating concentrations of interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were also determined. The relationship between α-syn levels and POD risk, as well as the association between α-syn and MDAS scores and plasma cytokines, were assessed. Results: POD incidence was 8.4% (17/202). Postoperative α-syn were either elevated or lowered. As primary outcome variables, the change of α-syn in POD patients was significantly higher than non-POD ones (21.0 ± 29.3 pg.ml−1 vs.1.9 ± 20.0, P = 0.047). The α-syn alteration was positively correlated to MDAS (r = 0.436, P = 0.010) and the change of IL-6 (r = 0.383, P = 0.025). Conclusions: Exosome α-syn release in plasma may be associated with the POD development which might be due to systemic inflammation. Clinical Trial Registration: www.clinicaltrials.gov, identifier ChiCTR-IPR-17012301. Prior Presentation: The abstract of this work has been selected for presentation in the 2019 ANESTHESIOLOGY Journal Symposium “What’s New with the old,” and it has been present in the ASA 2019 annual meeting October 21st, 2019 in Florida.
Collapse
Affiliation(s)
- Yi Yuan
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China.,Department of Anesthesiology, Beijing Jishuitan Hospital, Beijing, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yongzheng Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiaojuan Ji
- Department of Cadre Health Care, Beijing Jishuitan Hospital, Beijing, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiaoxiao Wang
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Feng Yuan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Jindan He
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yajie Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Cheng Ni
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Peng Zou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Geng Wang
- Department of Anesthesiology, Beijing Jishuitan Hospital, Beijing, China
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yang Zhou
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
200
|
Borchert T, Hess A, Lukačević M, Ross TL, Bengel FM, Thackeray JT. Angiotensin-converting enzyme inhibitor treatment early after myocardial infarction attenuates acute cardiac and neuroinflammation without effect on chronic neuroinflammation. Eur J Nucl Med Mol Imaging 2020; 47:1757-1768. [PMID: 32125488 PMCID: PMC7248052 DOI: 10.1007/s00259-020-04736-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Purpose Myocardial infarction (MI) triggers a local inflammatory response which orchestrates cardiac repair and contributes to concurrent neuroinflammation. Angiotensin-converting enzyme (ACE) inhibitor therapy not only attenuates cardiac remodeling by interfering with the neurohumoral system, but also influences acute leukocyte mobilization from hematopoietic reservoirs. Here, we seek to dissect the anti-inflammatory and anti-remodeling contributions of ACE inhibitors to the benefit of heart and brain outcomes after MI. Methods C57BL/6 mice underwent permanent coronary artery ligation (n = 41) or sham surgery (n = 9). Subgroups received ACE inhibitor enalapril (20 mg/kg, oral) either early (anti-inflammatory strategy; 10 days treatment beginning 3 days prior to surgery; n = 9) or delayed (anti-remodeling; continuous from 7 days post-MI; n = 16), or no therapy (n = 16). Cardiac and neuroinflammation were serially investigated using whole-body macrophage- and microglia-targeted translocator protein (TSPO) PET at 3 days, 7 days, and 8 weeks. In vivo PET signal was validated by autoradiography and histopathology. Results Myocardial infarction evoked higher TSPO signal in the infarct region at 3 days and 7 days compared with sham (p < 0.001), with concurrent elevation in brain TSPO signal (+ 18%, p = 0.005). At 8 weeks after MI, remote myocardium TSPO signal was increased, consistent with mitochondrial stress, and corresponding to recurrent neuroinflammation. Early enalapril treatment lowered the acute TSPO signal in the heart and brain by 55% (p < 0.001) and 14% (p = 0.045), respectively. The acute infarct signal predicted late functional outcome (r = 0.418, p = 0.038). Delayed enalapril treatment reduced chronic myocardial TSPO signal, consistent with alleviated mitochondrial stress. Early enalapril therapy tended to lower TSPO signal in the failing myocardium at 8 weeks after MI (p = 0.090) without an effect on chronic neuroinflammation. Conclusions Whole-body TSPO PET identifies myocardial macrophage infiltration and neuroinflammation after MI, and altered cardiomyocyte mitochondrial density in chronic heart failure. Improved chronic cardiac outcome by enalapril treatment derives partially from acute anti-inflammatory activity with complementary benefits in later stages. Whereas early ACE inhibitor therapy lowers acute neuroinflammation, chronic alleviation is not achieved by early or delayed ACE inhibitor therapy, suggesting a more complex mechanism underlying recurrent neuroinflammation in ischemic heart failure. Electronic supplementary material The online version of this article (10.1007/s00259-020-04736-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tobias Borchert
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Annika Hess
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Mario Lukačević
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany.
| |
Collapse
|