1
|
Yu MH, Yang Q, Zhang YP, Wang JH, Zhang RJZ, Liu ZG, Liu XC. Cannabinoid Receptor Agonist WIN55, 212-2 Attenuates Injury in the Hippocampus of Rats after Deep Hypothermic Circulatory Arrest. Brain Sci 2023; 13:brainsci13030525. [PMID: 36979335 PMCID: PMC10046860 DOI: 10.3390/brainsci13030525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
OBJECTIVES Postoperative neurological deficits remain a challenge in cardiac surgery employing deep hypothermic circulatory arrest (DHCA). This study aimed to investigate the effect of WIN55, 212-2, a cannabinoid agonist, on brain injury in a rat model of DHCA. METHODS Twenty-four male Sprague Dawley rats were randomly divided into three groups: a control group (which underwent cardiopulmonary bypass (CPB) only), a DHCA group (CPB with DHCA), and a WIN group (WIN55, 212-2 pretreatment before CPB with DHCA). Histopathological changes in the brain were evaluated by hematoxylin-eosin staining. Plasma levels of superoxide dismutase (SOD) and proinflammatory cytokines including interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-a) were determined using an enzyme-linked immunosorbent assay (ELISA). The expression of SOD in the hippocampus was detected by Western blot and immunofluorescence staining. Levels of apoptotic-related protein caspase-3 and type 1 cannabinoid receptor (CB1R) in the hippocampus were evaluated by Western blot. RESULTS WIN55, 212-2 administration attenuated histopathological injury of the hippocampus in rats undergoing DHCA, associated with lowered levels of IL-1β, IL-6, and TNF-α (p < 0.05, p < 0.001, and p < 0.01, vs. DHCA, respectively) and an increased level of SOD (p < 0.05 vs. DHCA). WIN55, 212-2 treatment also increased the content of SOD in the hippocampus. The protein expression of caspase-3 was downregulated and the expression of CB1R was upregulated in the hippocampus by WIN55, 212-2. CONCLUSIONS the administration of WIN55, 212-2 alleviates hippocampal injury induced by DHCA in rats by regulating intrinsic inflammatory and oxidative stress responses through a CB1R-dependent mechanism.
Collapse
Affiliation(s)
- Ming-Huan Yu
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, 61 Third Avenue, TEDA, Tianjin 300456, China
| | - Qin Yang
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, 61 Third Avenue, TEDA, Tianjin 300456, China
| | - You-Peng Zhang
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, 61 Third Avenue, TEDA, Tianjin 300456, China
| | - Jia-Hui Wang
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, 61 Third Avenue, TEDA, Tianjin 300456, China
| | - Ren-Jian-Zhi Zhang
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, 61 Third Avenue, TEDA, Tianjin 300456, China
| | - Zhi-Gang Liu
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, 61 Third Avenue, TEDA, Tianjin 300456, China
| | - Xiao-Cheng Liu
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, 61 Third Avenue, TEDA, Tianjin 300456, China
| |
Collapse
|
2
|
Jufar AH, Lankadeva YR, May CN, Cochrane AD, Marino B, Bellomo R, Evans RG. Renal and Cerebral Hypoxia and Inflammation During Cardiopulmonary Bypass. Compr Physiol 2021; 12:2799-2834. [PMID: 34964119 DOI: 10.1002/cphy.c210019] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiac surgery-associated acute kidney injury and brain injury remain common despite ongoing efforts to improve both the equipment and procedures deployed during cardiopulmonary bypass (CPB). The pathophysiology of injury of the kidney and brain during CPB is not completely understood. Nevertheless, renal (particularly in the medulla) and cerebral hypoxia and inflammation likely play critical roles. Multiple practical factors, including depth and mode of anesthesia, hemodilution, pump flow, and arterial pressure can influence oxygenation of the brain and kidney during CPB. Critically, these factors may have differential effects on these two vital organs. Systemic inflammatory pathways are activated during CPB through activation of the complement system, coagulation pathways, leukocytes, and the release of inflammatory cytokines. Local inflammation in the brain and kidney may be aggravated by ischemia (and thus hypoxia) and reperfusion (and thus oxidative stress) and activation of resident and infiltrating inflammatory cells. Various strategies, including manipulating perfusion conditions and administration of pharmacotherapies, could potentially be deployed to avoid or attenuate hypoxia and inflammation during CPB. Regarding manipulating perfusion conditions, based on experimental and clinical data, increasing standard pump flow and arterial pressure during CPB appears to offer the best hope to avoid hypoxia and injury, at least in the kidney. Pharmacological approaches, including use of anti-inflammatory agents such as dexmedetomidine and erythropoietin, have shown promise in preclinical models but have not been adequately tested in human trials. However, evidence for beneficial effects of corticosteroids on renal and neurological outcomes is lacking. © 2021 American Physiological Society. Compr Physiol 11:1-36, 2021.
Collapse
Affiliation(s)
- Alemayehu H Jufar
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia.,Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Yugeesh R Lankadeva
- Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia.,Department of Critical Care, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria, Australia
| | - Clive N May
- Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia.,Department of Critical Care, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria, Australia
| | - Andrew D Cochrane
- Department of Cardiothoracic Surgery, Monash Health and Department of Surgery (School of Clinical Sciences at Monash Health), Monash University, Melbourne, Victoria, Australia
| | - Bruno Marino
- Cellsaving and Perfusion Resources, Melbourne, Victoria, Australia
| | - Rinaldo Bellomo
- Department of Critical Care, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria, Australia.,Department of Intensive Care, Austin Health, Heidelberg, Victoria, Australia
| | - Roger G Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia.,Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Chen Q, Lei YQ, Liu JF, Wang ZC, Cao H. Beneficial effects of chlorogenic acid treatment on neuroinflammation after deep hypothermic circulatory arrest may be mediated through CYLD/NF-κB signaling. Brain Res 2021; 1767:147572. [PMID: 34216581 DOI: 10.1016/j.brainres.2021.147572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/30/2021] [Accepted: 06/25/2021] [Indexed: 10/21/2022]
Abstract
Deep hypothermic circulatory arrest (DHCA) during heart surgery may induce neuroinflammation leading to neurocognitive dysfunction. Chlorogenic acid (CA) is a common phytochemical, which can attenuate neuroinflammation. Nevertheless, the underlying mechanism involved in the anti-inflammatory effect of CA after DHCA is unknown. The present study therefore characterized the anti-inflammatory functions of CA after DHCA using in vivo and in vitro DHCA models. The activation of microglia, inflammatory cytokine levels, and the NF-κB pathway were measured. The results showed that CA treatment ameliorated neurocognitive function and reduced the inflammatory cytokine levels in the brain and circulation. Furthermore, the microglial and NF-κB activations were suppressed after DHCA. CA exerted the same anti-inflammatory effect in hypothermia OGD microglial cells as the in vivo study. Additional studies indicated that the regulation of ubiquitin ligase activity of TRAF6 and RIP1 by CYLD was related to the mechanism involving inhibition of CA in the NF-κB pathway. Together, the results showed that CA may attenuate neuroinflammation after DHCA by modulating the signaling of CYLD/NF-κB.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China.
| | - Yu-Qing Lei
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Jian-Feng Liu
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Zeng-Chun Wang
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Hua Cao
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| |
Collapse
|
4
|
Chen Q, Lei YQ, Liu JF, Wang ZC, Cao H. Triptolide improves neurobehavioral functions, inflammation, and oxidative stress in rats under deep hypothermic circulatory arrest. Aging (Albany NY) 2021; 13:3031-3044. [PMID: 33465048 PMCID: PMC7880355 DOI: 10.18632/aging.202460] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/10/2020] [Indexed: 11/25/2022]
Abstract
This study investigated the neuroprotective effects of triptolide (TPL) in a rat model of cardiopulmonary bypass with deep hypothermia circulatory arrest (DHCA). Rats were randomly divided into six groups: control, sham, DHCA, and DHCA + TPL (100, 200, 300 μg/kg). Neurobehavioral functions were measured using the elevated plus-maze, Y-maze, and Morris water maze tests. Levels of inflammatory cytokines, oxidative stress indices, and brain neurotrophins were measured by ELISA. Microglial activation and cell death was measured by immunofluorescence staining and TUNEL assay, respectively. Finally, activation of the Nrf2 pathway and NF-κB were detected by western blot. The elevated plus-maze, Y-maze, and Morris water maze tests all showed that TPL mitigated anxiety-like behavior, working memory, spatial learning, and memory in DHCA rats. TPL inhibited inflammatory responses and oxidative stress, as well as increased brain neurotrophin levels in DHCA rats. Moreover, TPL attenuated microglia activation and cell death in DHCA rats. Finally, TPL activated the Nrf2 pathway and inhibited NF-κB activity in DHCA rats. These results demonstrated that TPL improved neurobehavioral functions, neuroinflammation, and oxidative stress in DHCA rats, which may be associated with the Nrf2 and NF-κB pathways.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, P. R. China
| | - Yu-Qing Lei
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, P. R. China
| | - Jian-Feng Liu
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, P. R. China
| | - Zeng-Chun Wang
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, P. R. China
| | - Hua Cao
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, P. R. China
| |
Collapse
|
5
|
Wang ZC, Chen Q, Wang J, Yu LS, Chen LW. Sulforaphane mitigates LPS-induced neuroinflammation through modulation of Cezanne/NF-κB signalling. Life Sci 2020; 262:118519. [PMID: 33010279 DOI: 10.1016/j.lfs.2020.118519] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/13/2020] [Accepted: 09/25/2020] [Indexed: 10/23/2022]
Abstract
AIM Neuroinflammation is a potent pathological process of various neurodegenerative diseases. Sulforaphane (SFN) is a natural product and acts as a neuroprotective agent to suppress inflammatory response in brain. The present study investigated the protective effect of Sulforaphane (SFN) on lipopolysaccharide (LPS)-induced neuroinflammation. MATERIALS AND METHODS Rats were divided into three groups: control group, LPS group and LPS + SFN group. Morris water maze test was carried out to evaluate the spatial memory and learning function of rats. The inflammatory cytokines levels in hippocampal tissues, plasma were measured by ELISA. The western blot was used to detect Cezanne/NF-κB signalling. For in vitro study, the Cezanne siRNA and scrambled control were transfected into BV2 cells, and then treated with or without 20 μM SFN before exposed to LPS. The inflammatory cytokines levels and Cezanne/NF-κB signalling were detected by ELISA and western blot, respectively. Co-IP assay were applied to investigate the regulation of Cezanne on ubiquitination of TRAF6 and RIP1. KEY FINDINGS SFN improved LPS-induced neurocognitive dysfunction in rats. It inhibited the neuroinflammation and activation of NF-κB pathway induced by LPS. The modulation of TRAF6 and RIP1 ubiquitination by Cezanne was playing a pivotal role in relation to the mechanism of SFN inhibiting NF-κB pathway. SIGNIFICANCE The results of our study demonstrated that SFN could attenuate LPS-induced neuroinflammation through the modulation of Cezanne/NF-κB signalling.
Collapse
Affiliation(s)
- Zeng-Chun Wang
- Department of Cardiovascular Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China; Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, China.
| | - Qiang Chen
- Department of Cardiovascular Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China; Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, China
| | - Jing Wang
- Department of Cardiovascular Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Ling-Shan Yu
- Department of Cardiovascular Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Liang-Wan Chen
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
6
|
Chen Q, Sun KP, Huang JS, Wang ZC, Hong ZN. Resveratrol attenuates neuroinflammation after deep hypothermia with circulatory arrest in rats. Brain Res Bull 2020; 155:145-154. [DOI: 10.1016/j.brainresbull.2019.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/24/2019] [Accepted: 12/12/2019] [Indexed: 11/29/2022]
|
7
|
Almahozi A, Radhi M, Alzayer S, Kamal A. Effects of Memantine in a Mouse Model of Postoperative Cognitive Dysfunction. Behav Sci (Basel) 2019; 9:bs9030024. [PMID: 30845688 PMCID: PMC6466583 DOI: 10.3390/bs9030024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/18/2019] [Accepted: 02/27/2019] [Indexed: 02/08/2023] Open
Abstract
Persistent impairment in cognitive functioning postoperatively is reported by clinical and animal studies, and is labeled as postoperative cognitive dysfunction (POCD). Evidence points to an exaggerated neuroinflammatory response resulting from peripheral systemic inflammation after surgery, with subsequent cytokine-induced glutamatergic excitotoxicity and synaptic impairment. These immunological changes, among many others, are also observed in Alzheimer’s disease. Memantine is an N-methyl-D-aspartate receptor (NMDAR) antagonist commonly used to treat Alzheimer’s disease. Surprisingly, little research exists on the role of memantine in preventing POCD. The purpose of this study is to investigate the effects of memantine on a spectrum of cognitive functions postoperatively. Mice were divided into 3 groups and each received treatment for 4 weeks. Placebo groups received a placebo then underwent either a sham procedure or a laparotomy procedure. The memantine group received memantine hydrochloride then underwent a laparotomy procedure. Cognitive tests were performed on postoperative days (POD) 1 and 7. Compared to sham-operated mice, placebo groups that underwent a laparotomy procedure showed impaired memory in the Morris water maze test, higher anxiety-like behavior in the open field and the elevated plus maze tests, increased depression-like behavior in the tail suspension test, and lack of preference for social novelty in the three-chamber test. On the other hand, memantine-treated mice that underwent a laparotomy procedure showed enhanced memory on POD7, improved depression-like behavior on POD1 and POD7, enhanced preference for social novelty on POD1, and no improvement in anxiety-like behavior. These findings suggest a potential protective effect of memantine in mice postoperatively on memory, depression-like behavior, and preference for social novelty.
Collapse
Affiliation(s)
- Ahmad Almahozi
- Physiology Department, College of Medicine and Medical Sciences, Arabian Gulf University, P.O. Box 26671, Manama 1111, Bahrain.
| | - Mohamed Radhi
- Physiology Department, College of Medicine and Medical Sciences, Arabian Gulf University, P.O. Box 26671, Manama 1111, Bahrain.
| | - Suja Alzayer
- Physiology Department, College of Medicine and Medical Sciences, Arabian Gulf University, P.O. Box 26671, Manama 1111, Bahrain.
| | - Amer Kamal
- Physiology Department, College of Medicine and Medical Sciences, Arabian Gulf University, P.O. Box 26671, Manama 1111, Bahrain.
| |
Collapse
|
8
|
Influence of sex and hormonal status on initial impact and neurocognitive outcome after subarachnoid haemorrhage in rats. Behav Brain Res 2019; 363:13-22. [PMID: 30703399 DOI: 10.1016/j.bbr.2019.01.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/24/2019] [Accepted: 01/26/2019] [Indexed: 12/23/2022]
Abstract
The aim of this study was to detect differences in functional outcome after experimental subarachnoid haemorrhage (SAH) in rodents with different hormonal status. For this purpose, the endovascular perforation model was applied to four groups of Sprague-Dawley-Rats: male intact, male neutered, female intact and female neutered animals. Initial impact was measured by ICP, CPP and cerebral blood flow in the first hour after SAH. From day 4-14, the modified hole board test was applied to assess functional and neuro-cognitive outcome. Histological outcome was examined in the motor cortex and hippocampus of each hemisphere. Mortality was highest in the female intact group albeit not statistically significant. Physiologic parameters did not differ significantly between groups either. In the modified hole board test, male intact animals showed a greater impairment of declarative memory than the female intact and neutered groups. However, male intact animals showed greater avoidance behaviour and male animals revealed higher anxiety levels independent of hormonal status. No differences in histological damage of hippocampus and motor cortex between groups could be shown. We therefore speculate that the marginal deficits in cognitive performance that are shown by the male intact group in the modified hole board test are mostly caused by higher anxiety levels and cannot be interpreted as pure cognitive impairment.
Collapse
|
9
|
ZHAO FY, ZHANG ZY, ZHAO YX, YAN HX, HONG YF, XIA XJ, XU H. The effect of electroacupuncture preconditioning on cognitive impairments following knee replacement among elderly: A randomized controlled trial. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2018. [DOI: 10.1016/j.wjam.2018.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
10
|
Zhang Z, Ma Q, Shah B, Mackensen GB, Lo DC, Mathew JP, Podgoreanu MV, Terrando N. Neuroprotective Effects of Annexin A1 Tripeptide after Deep Hypothermic Circulatory Arrest in Rats. Front Immunol 2017; 8:1050. [PMID: 28912778 PMCID: PMC5582068 DOI: 10.3389/fimmu.2017.01050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/14/2017] [Indexed: 01/01/2023] Open
Abstract
Resolution agonists, including lipid mediators and peptides such as annexin A1 (ANXA1), are providing novel approaches to treat inflammatory conditions. Surgical trauma exerts a significant burden on the immune system that can affect and impair multiple organs. Perioperative cerebral injury after cardiac surgery is associated with significant adverse neurological outcomes such as delirium and postoperative cognitive dysfunction. Using a clinically relevant rat model of cardiopulmonary bypass (CPB) with deep hypothermic circulatory arrest (DHCA), we tested the pro-resolving effects of a novel bioactive ANXA1 tripeptide (ANXA1sp) on neuroinflammation and cognition. Male rats underwent 2 h CPB with 1 h DHCA at 18°C, and received vehicle or ANXA1sp followed by timed reperfusion up to postoperative day 7. Immortalized murine microglial cell line BV2 were treated with vehicle or ANXA1sp and subjected to 2 h oxygen-glucose deprivation followed by timed reoxygenation. Microglial activation, cell death, neuroinflammation, and NF-κB activation were assessed in tissue samples and cell cultures. Rats exposed to CPB and DHCA had evident neuroinflammation in various brain areas. However, in ANXA1sp-treated rats, microglial activation and cell death (apoptosis and necrosis) were reduced at 24 h and 7 days after surgery. This was associated with a reduction in key pro-inflammatory cytokines due to inhibition of NF-κB activation in the brain and systemically. Treated rats also had improved neurologic scores and shorter latency in the Morris water maze. In BV2 cells treated with ANXA1sp, similar protective effects were observed including decreased pro-inflammatory cytokines and cell death. Notably, we also found increased expression of ANXA1, which binds to NF-κB p65 and thereby inhibits its transcriptional activity. Our findings provide evidence that treatment with a novel pro-resolving ANXA1 tripeptide is neuroprotective after cardiac surgery in rats by attenuating neuroinflammation and may prevent postoperative neurologic complications.
Collapse
Affiliation(s)
- Zhiquan Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Qing Ma
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Bijal Shah
- Center for Drug Discovery, Department of Neurobiology, Duke University Medical Center, Durham, NC, United States
| | - G. Burkhard Mackensen
- Department of Anesthesiology & Pain Medicine, University of Washington Medical Center, Seattle, WA, United States
| | - Donald C. Lo
- Center for Drug Discovery, Department of Neurobiology, Duke University Medical Center, Durham, NC, United States
| | - Joseph P. Mathew
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Mihai V. Podgoreanu
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Niccolò Terrando
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
- Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
11
|
Influence of the postoperative inflammatory response on cognitive decline in elderly patients undergoing on-pump cardiac surgery: a controlled, prospective observational study. BMC Anesthesiol 2017; 17:113. [PMID: 28851286 PMCID: PMC5576316 DOI: 10.1186/s12871-017-0408-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/22/2017] [Indexed: 02/03/2023] Open
Abstract
Background The role of non-infective inflammatory response (IR) in the aetiology of postoperative cognitive dysfunction (POCD) is still controversial. The aim of this controlled, prospective observational study was to assess the possible relationship between the grade of IR, defined by procalcitonin (PCT) changes, and development of POCD related to cardiac surgery. Methods Forty-two patients, who were ≥ 60 years of age and scheduled for elective cardiac surgery, were separated into the low inflammatory (LIR) and high inflammatory (HIR) response groups based on their PCT levels measured on the first postoperative day. A matched normative control group of 32 subjects was recruited from primary care practice. The PCT and C-reactive protein (CRP) levels were monitored daily during the first five postoperative days. The cognitive function and mood state were preoperatively tested with a set of five neurocognitive tests and two mood inventories and at the seventh postoperative day. The Reliable Change Index modified for practice (RCIp) using data from normative controls was applied to determine the significant decline in test performance. Results The LIR (n = 20) and HIR (n = 22) groups differed significantly in the PCT (p < 0.001) but not in the CRP time courses. The incidence of POCD at the first postoperative week was 35.7% in the cohort. The LIR and HIR groups did not vary in the RCIp Z scores of neurocognitive tests and frequencies of POCD (7 vs 8 cases, respectively, p > 0.05). Additionally, there was no difference in the mood states, anxiety levels and perioperative parameters known to influence the development of POCD. Conclusions In this study, the magnitude of the non-infective inflammatory response generated by on-pump cardiac surgery did not influence the development of POCD in the early postoperative period in elderly patients.
Collapse
|
12
|
Pappa M, Theodosiadis N, Tsounis A, Sarafis P. Pathogenesis and treatment of post-operative cognitive dysfunction. Electron Physician 2017; 9:3768-3775. [PMID: 28465805 PMCID: PMC5410904 DOI: 10.19082/3768] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 01/18/2017] [Indexed: 12/19/2022] Open
Abstract
Cognitive disorders common in the post-operative period, are the post-operative delirium (POD) and the post-operative cognitive dysfunction (POCD). The Diagnostic and Statistical Manual of Mental Disorders (DSM) does not mention POCD as a separate disease entity, and thus little is known about the pathogenesis of this disorder. The aim of this study was to review, detect and highlight the most important data cited, regarding pathogenesis mechanisms and treatment of the post-operative cognitive dysfunction (POCD). The study was carried out from March 2015 to June 2015. Literature review was achieved by searching a number of bibliographic databases including PubMed, Google Scholar and SCOPUS, surveying published articles from 1955 to 2014. As far as the selection criteria, the material consists of scientific articles published mainly over the last fifteen years, while material published before 2000 was selected because it was considered to be important. This review showed that deficits are observed in one or more discrete areas of the patient's mental state, such as attention, concentration, memory, psychomotor speed and more. This condition is usually developed over a period of more than a week or month after surgery and is more common in elderly patients. Mechanisms that have been proposed to explain this phenomenon are hyperventilation, hypotension, cerebral microemboli and inflammatory mechanisms. Its differential diagnosis will be made mainly from delirium. POCD treatment will first include the exclusion of any other serious diseases that can cause organic psychosyndrome and then focus on the actual symptoms.
Collapse
Affiliation(s)
- Maria Pappa
- 414 Military Hospital of Special Diseases, Athens, Greece
| | | | - Andreas Tsounis
- Centers for the Prevention of Addictions and Promoting Psychosocial Health of Municipality of Thessaloniki, Thessaloniki, Greece
| | - Pavlos Sarafis
- Nursing Department, Cyprus University of Technology, Limassol, Cyprus
| |
Collapse
|
13
|
He S, Lin K, Ma R, Xu R, Xiao Y. Effect of the urinary tryptin inhibitor ulinastatin on cardiopulmonary bypass-related inflammatory response and clinical outcomes: a meta-analysis of randomized controlled trials. Clin Ther 2015; 37:643-53. [PMID: 25660078 DOI: 10.1016/j.clinthera.2014.12.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 11/19/2014] [Accepted: 12/14/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Cardiopulmonary bypass (CPB) can cause systemic inflammatory responses and a series of subsequent complications that may harm patients. The aim of this study was to explore the effects of ulinastatin on inflammatory responses and clinical outcomes of CPB via a meta-analysis of published randomized controlled trials. METHODS A literature search was conducted, both manually and by using the PubMed, EMBASE, Cochrane Library, and Web of Knowledge databases from inception to February 2013, to identify randomized controlled trials. The abstracted efficacy measures included changes in the plasma levels of cytokines (interleukin-6 [IL-6], IL-8, and tumor necrosis factor-α [TNF-α]) measured during the perioperative period and clinical indicators of efficacy, including the duration of mechanical ventilation and the length of intensive care unit stay. Ten ulinastatin-related randomized controlled trials related to cardiac surgeries involving CPB were selected. FINDINGS In terms of cytokine concentrations, there were no significant differences between patients who received ulinastatin and those who received placebo before CPB. However, as the surgeries progressed, cytokine concentrations were all significantly lower in the ulinastatin group (P < 0.05 at 1 hour; P < 0.0001 at 6 hours), and the respective plasma concentrations returned to baseline values 24 hours after CPB. In terms of the clinical outcome indices, the length of intensive care unit stay was not significantly different, but the duration of mechanical ventilation (95% CI, -6.75 to -0.39; P = 0.03) was significantly shorter in the ulinastatin group. IMPLICATIONS This meta-analysis found that changes in inflammatory cytokines occurred in a time-dependent manner and that the use of ulinastatin resulted in decreased duration of mechanical ventilation with CPB compared with placebo.
Collapse
Affiliation(s)
- Siyi He
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Cardiovascular Surgery, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Kailong Lin
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Ruiyan Ma
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Rufu Xu
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yingbin Xiao
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
14
|
Liguori GR, Kanas AF, Moreira LFP. Managing the inflammatory response after cardiopulmonary bypass: review of the studies in animal models. Braz J Cardiovasc Surg 2014; 29:93-102. [PMID: 24896169 PMCID: PMC4389477 DOI: 10.5935/1678-9741.20140017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/24/2013] [Indexed: 11/28/2022] Open
Abstract
Objective To review studies performed in animal models that evaluated therapeutic
interventions to inflammatory response and microcirculatory changes after
cardiopulmonary bypass. Methods It was used the search strategy ("Cardiopulmonary Bypass" (MeSH)) and
("Microcirculation" (MeSH) or "Inflammation" (MeSH) or
"Inflammation Mediators" (MeSH)). Repeated results, human studies,
non-English language articles, reviews and studies without control were
excluded. Results Blood filters, system miniaturization, specific primers regional perfusion,
adequate flow and temperature and pharmacological therapies with anticoagulants,
vasoactive drugs and anti-inflammatories reduced changes in microcirculation and
inflammatory response. Conclusion Demonstrated efficacy in animal models establishes a perspective for evaluating
these interventions in clinical practice.
Collapse
Affiliation(s)
- Gabriel Romero Liguori
- Correspondence address: Gabriel Romero Liguori, Instituto do Coração
(InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo,
Av. Dr. Enéas de Carvalho Aguiar, 44 - 2º andar - bloco II - sala 13 - Cerqueira
César, São Paulo, SP, Brazil - Zip code: 05403-000. E-mail:
| | | | | |
Collapse
|
15
|
Drabek T, Wilson CD, Janata A, Stezoski JP, Janesko-Feldman K, Garman RH, Tisherman SA, Kochanek PM. Unique brain region-dependent cytokine signatures after prolonged hypothermic cardiac arrest in rats. Ther Hypothermia Temp Manag 2014; 5:26-39. [PMID: 25423415 DOI: 10.1089/ther.2014.0013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We previously showed that prolonged cardiac arrest (CA) produces neuronal death with microglial proliferation. Microglial proliferation, but not neuronal death, was attenuated by deeper hypothermia. Microglia are reportedly a major source of cytokines. In this study, we tested the hypotheses that (1) CA will result in highly specific regional and temporal increases in brain cytokines; and (2) these increases will be attenuated by deep hypothermia. Adult male Sprague-Dawley rats were subjected to rapid exsanguination. After 6 minutes of normothermic no-flow, different levels of hypothermia were induced by either ice-cold (IC) or room-temperature (RT) aortic flush. After 20 minutes CA, rats were resuscitated with cardiopulmonary bypass (CPB), and sacrificed at 6 or 24 hours. Rats subjected to CPB only (without CA) and shams (no CPB or CA) served as controls (n=6 per group). Cytokines were analyzed in cerebellum, cortex, hippocampus, and striatum. Immunofluorescence was used to identify cell types associated with individual cytokines. Intra-CA temperature was lower after IC versus RT flush (21°C vs. 28°C, p<0.05). At 6 hours, striatum showed a massive increase in interleukin (IL)-1α and tumor necrosis factor-alpha (TNF-α) (>100-fold higher than in hippocampus), which was attenuated by deeper hypothermia in the IC versus RT group. In contrast, IL-12 was 50-fold higher in hippocampus versus striatum. At 24 hours, cytokines decreased. In striatum, IL-1α colocalized with astrocytes while TNF-α colocalized with neurons. In hippocampus, IL-12 colocalized with hippocampal hilar neurons, the only region where neuronal degeneration was observed at 24 hours at both IC and RT groups. We report important temporo-spatial differences in the brain cytokine response to hypothermic CA, with a novel role of striatum. Astrocytes and neurons, but not microglia colocalized with individual cytokines. Hypothermia showed protective effects. These neuroinflammatory reactions precede neuronal death. New therapeutic strategies may need to target early regional neuroinflammation.
Collapse
Affiliation(s)
- Tomas Drabek
- 1 Safar Center for Resuscitation Research, University of Pittsburgh , Pittsburgh, Pennsylvania
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Inflammatory Response in Patients under Coronary Artery Bypass Grafting Surgery and Clinical Implications: A Review of the Relevance of Dexmedetomidine Use. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/905238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite the fact that coronary artery bypass grafting surgery (CABG) with cardiopulmonary bypass (CPB) prolongs life and reduces symptoms in patients with severe coronary artery diseases, these benefits are accompanied by increased risks. Morbidity associated with cardiopulmonary bypass can be attributed to the generalized inflammatory response induced by blood-xenosurfaces interactions during extracorporeal circulation and the ischemia/reperfusion implications, including exacerbated inflammatory response resembling the systemic inflammatory response syndrome (SIRS). The use of specific anesthetic agents with anti-inflammatory activity can modulate the deleterious inflammatory response. Consequently, anti-inflammatory anesthetics may accelerate postoperative recovery and better outcomes than classical anesthetics. It is known that the stress response to surgery can be attenuated by sympatholytic effects caused by activation of central (α-)2-adrenergic receptor, leading to reductions in blood pressure and heart rate, and more recently, that they can have anti-inflammatory properties. This paper discusses the clinical significance of the dexmedetomidine use, a selective (α-)2-adrenergic agonist, as a coadjuvant in general anesthesia. Actually, dexmedetomidine use is not in anesthetic routine, but this drug can be considered a particularly promising agent in perioperative multiple organ protection.
Collapse
|
17
|
Mishra MK, Beaty CA, Lesniak WG, Kambhampati SP, Zhang F, Wilson MA, Blue ME, Troncoso JC, Kannan S, Johnston MV, Baumgartner WA, Kannan RM. Dendrimer brain uptake and targeted therapy for brain injury in a large animal model of hypothermic circulatory arrest. ACS NANO 2014; 8:2134-47. [PMID: 24499315 PMCID: PMC4004292 DOI: 10.1021/nn404872e] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 02/05/2014] [Indexed: 05/20/2023]
Abstract
Treatment of brain injury following circulatory arrest is a challenging health issue with no viable therapeutic options. Based on studies in a clinically relevant large animal (canine) model of hypothermic circulatory arrest (HCA)-induced brain injury, neuroinflammation and excitotoxicity have been identified as key players in mediating the brain injury after HCA. Therapy with large doses of valproic acid (VPA) showed some neuroprotection but was associated with adverse side effects. For the first time in a large animal model, we explored whether systemically administered polyamidoamine (PAMAM) dendrimers could be effective in reaching target cells in the brain and deliver therapeutics. We showed that, upon systemic administration, hydroxyl-terminated PAMAM dendrimers are taken up in the brain of injured animals and selectively localize in the injured neurons and microglia in the brain. The biodistribution in other major organs was similar to that seen in small animal models. We studied systemic dendrimer-drug combination therapy with two clinically approved drugs, N-acetyl cysteine (NAC) (attenuating neuroinflammation) and valproic acid (attenuating excitotoxicity), building on positive outcomes in a rabbit model of perinatal brain injury. We prepared and characterized dendrimer-NAC (D-NAC) and dendrimer-VPA (D-VPA) conjugates in multigram quantities. A glutathione-sensitive linker to enable for fast intracellular release. In preliminary efficacy studies, combination therapy with D-NAC and D-VPA showed promise in this large animal model, producing 24 h neurological deficit score improvements comparable to high dose combination therapy with VPA and NAC, or free VPA, but at one-tenth the dose, while significantly reducing the adverse side effects. Since adverse side effects of drugs are exaggerated in HCA, the reduced side effects with dendrimer conjugates and suggestions of neuroprotection offer promise for these nanoscale drug delivery systems.
Collapse
Affiliation(s)
- Manoj K. Mishra
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Claude A. Beaty
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Wojciech G. Lesniak
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Siva P. Kambhampati
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Fan Zhang
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Mary A. Wilson
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, Maryland 21205, United States
| | - Mary E. Blue
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, Maryland 21205, United States
| | - Juan C. Troncoso
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Division of Neuropathology, Department of Anesthesiology and Critical Care Medicine, and Department of Pediatrics, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Sujatha Kannan
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, Maryland 21205, United States
- Division of Neuropathology, Department of Anesthesiology and Critical Care Medicine, and Department of Pediatrics, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Michael V. Johnston
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, Maryland 21205, United States
- Division of Neuropathology, Department of Anesthesiology and Critical Care Medicine, and Department of Pediatrics, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - William A. Baumgartner
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Address correspondence to ,
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
- Address correspondence to ,
| |
Collapse
|
18
|
Totally thoracoscopic repair of atrial septal defect reduces systemic inflammatory reaction and myocardial damage in initial patients. Eur J Med Res 2014; 19:13. [PMID: 24612760 PMCID: PMC3995714 DOI: 10.1186/2047-783x-19-13] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/21/2014] [Indexed: 11/24/2022] Open
Abstract
Background To compare the effect of totally thoracoscopic with conventional, open repair of atrial septal defect. Methods Forty atrial septal defect cases were divided into two groups by surgical approach: totally thoracoscopic approach (group A, n = 20) and conventional open approach (group B, n = 20). In group A, surgical procedures were performed through three portal incisions in the right lateral chest wall under thoracoscopic vision without the aid of a computerized robotic surgical system. Notably, all operations were completed by one surgeon who had just begun using this technique. In group B, the atrial septal defects were repaired in conventional open fashion. Clinical outcomes and serum levels of tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), interleukin-10 (IL-10), intercellular adhesion molecule 1 (ICAM-1), and creatine kinase isoenzyme-myocardial band (CK-MB) for the two groups were evaluated and compared. Results All operations were performed successfully without serious complications. Durations of cardiopulmonary bypass (CPB), CPB setup, aortic cross-clamping, and operative procedure were significantly longer in group A than in group B (P < 0.05). The recovery times for body temperature and laboratory values of leukocytes were significantly shorter for group A than for group B (P < 0.05). There were no differences in durations of postoperative assisted ventilation or intensive care unit and hospital stays, volumes of blood transfused intraoperatively or thoracic drainage, or medical costs between the two groups. Serum levels of inflammatory factors (TNF-α, IL-6, IL-10, and ICAM-1) and CK-MB increased significantly in both groups after surgery. However, 6 h and 12 h after surgery, levels of these inflammatory factors and CK-MB were significantly lower in group A than in group B (P < 0.05). Conclusions Thoracoscopic cardiac surgery is technically feasible and safe, with less trauma and quicker recovery even when done by a surgeon newly introduced to the technique.
Collapse
|
19
|
Drabek T, Janata A, Wilson CD, Stezoski J, Janesko-Feldman K, Tisherman SA, Foley LM, Verrier J, Kochanek PM. Minocycline attenuates brain tissue levels of TNF-α produced by neurons after prolonged hypothermic cardiac arrest in rats. Resuscitation 2014; 85:284-91. [PMID: 24513126 PMCID: PMC3952024 DOI: 10.1016/j.resuscitation.2013.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 09/24/2013] [Accepted: 10/15/2013] [Indexed: 12/14/2022]
Abstract
Neuro-cognitive disabilities are a well-recognized complication of hypothermic circulatory arrest. We and others have reported that prolonged cardiac arrest (CA) produces neuronal death and microglial proliferation and activation that are only partially mitigated by hypothermia. Microglia, and possibly other cells, are suggested to elaborate tumor necrosis factor alpha (TNF-α), which can trigger neuronal death cascades and exacerbate edema after CNS insults. Minocycline is neuroprotective in some brain ischemia models in part by blunting the microglial response. We tested the hypothesis that minocycline would attenuate neuroinflammation as reflected by brain tissue levels of TNF-α after hypothermic CA in rats. Rats were subjected to rapid exsanguination, followed by a 6 min normothermic CA. Hypothermia (30 °C) was then induced by an aortic saline flush. After a total of 20 min CA, resuscitation was achieved via cardiopulmonary bypass (CPB). After 5 min reperfusion, minocycline (90 mg kg−1; n = 6) or vehicle (PBS; n = 6) was given. Hypothermia (34 °C) was maintained for 6 h. Rats were sacrificed at 6 or 24 h. TNF-α was quantified (ELISA) in four brain regions (cerebellum, CEREB; cortex, CTX; hippocampus, HIP; striatum, STRI). Naïve rats (n = 6) and rats subjected to the same anesthesia and CPB but no CA served as controls (n = 6). Immunocytochemistry was used to localize TNF-α. Naïve rats and CPB controls had no detectable TNF-α in any brain region. CA markedly increased brain TNF-α. Regional differences were seen, with the highest TNF-α levels in striatum in CA groups (10-fold higher, P < 0.05 vs. all other brain regions). TNF-α was undetectable at 24 h. Minocycline attenuated TNF-α levels in CTX, HIP and STRI (P < 0.05). TNF-α showed unique co-localization with neurons. In conclusion, we report region-dependent early increases in brain TNF-α levels after prolonged hypothermic CA, with maximal increases in striatum. Surprisingly, TNF-α co-localized in neurons and not microglia. Minocycline attenuated TNF-α by approximately 50% but did not totally ablate its production. That minocycline decreased brain TNF-α levels suggests that it may represent a therapeutic adjunct to hypothermia in CA neuroprotection. University of Pittsburgh IACUC 0809278B-3.
Collapse
Affiliation(s)
- Tomas Drabek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andreas Janata
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Caleb D. Wilson
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason Stezoski
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Samuel A. Tisherman
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lesley M. Foley
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Jonathan Verrier
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW To summarize recent studies of neurocognitive dysfunction after cardiac surgery, as well as to outline efforts and approaches toward advancing the field. RECENT FINDINGS Observational studies have improved our understanding of the incidence and the trajectory of cognitive decline after cardiac surgery; however, the magnitude of this neurocognitive change remains controversial because of the inconsistent definitions and the lack of a gold-standard diagnostic modality. Nonetheless, physicians commonly see patients with functional and cognitive impairments after cardiac surgery, which utilize healthcare resources and impact quality of life. Novel approaches have utilized advanced neuroimaging techniques as well as innovative monitoring modalities to improve the efficiency of neuroprotective strategies during cardiac surgery. SUMMARY Adverse cognitive and neurologic outcomes following cardiac surgery range from discrete neurocognitive deficits to severe neurologic injury such as stroke and even death. The elderly are at higher risk of suffering these outcomes and the public health dimension of this problem is expected to accelerate. Future studies should combine advanced neuroimaging with genomic, transcriptional, proteomic, and metabolomic profiling to improve our understanding of the pathophysiologic mechanisms and optimize the diagnosis, prevention, and treatment of neurocognitive injury.
Collapse
|
21
|
Li W, Zheng B, Xu H, Deng Y, Wang S, Wang X, Su D. Isoflurane Prevents Neurocognitive Dysfunction After Cardiopulmonary Bypass in Rats. J Cardiothorac Vasc Anesth 2013; 27:502-9. [DOI: 10.1053/j.jvca.2012.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Indexed: 11/11/2022]
|
22
|
Wang X, Xue Q, Yan F, Li L, Liu J, Li S, Hu S. Ulinastatin as a neuroprotective and anti-inflammatory agent in infant piglets model undergoing surgery on hypothermic low-flow cardiopulmonary bypass. Paediatr Anaesth 2013; 23:209-16. [PMID: 23384299 DOI: 10.1111/pan.12073] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Infants are potentially more susceptible to brain injury mediated via cell death attributed to cardiopulmonary bypass (CPB) especially with prolonged hypothermic low flow (HLF). We hypothesized that a human urinary protease inhibitor (ulinastatin), by its anti-inflammatory effect, would reduce central nervous system (CNS) injury during HLF. METHODS Fifteen general-type infant piglets were randomized to ulinastatin group (group U, n = 5), control group (group C, n = 5), and sham operation group (group S, n = 5). Routine CPB was established after median thoracotomy in group U and C under anesthesia. When the temperature of infant piglets dropped down to 25 °C, low-flow CPB (50 ml·kg(-1) ·min(-1) ) was instituted. After 120 min of aortic cross-clamping and 20- to 30-min rewarming, the aortic cross-clamp was removed and finally the piglet was weaned from CPB. Five thousand units per killogram of ulinastatin and equivalently normal saline were, respectively, given at the beginning of and at aortic declamping in group U and group C. group S just received sham median thoracotomy. Venous blood samples were taken immediately after anesthesia induction in all three groups, 5- and 120-min post CPB in both group U and C, respectively; plasma markers of inflammation and CNS injury were compared. Pathology results of hippocampus were observed by light microscopy. RESULTS Statistically significant differences between group C and U were noted in the expression of inflammatory markers such as IL-10, TNF-α and neuron-specific enolase at 120-min post CPB. Brain injuries were observed in both groups (index cases and controls) and were milder in group U. CONCLUSIONS In our study, HLF CPB on infant piglets resulted in brain injury, and ulinastatin might reduce the extent of such injury.
Collapse
Affiliation(s)
- Xiaocou Wang
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
23
|
van Harten AE, Scheeren TWL, Absalom AR. A review of postoperative cognitive dysfunction and neuroinflammation associated with cardiac surgery and anaesthesia. Anaesthesia 2012; 67:280-93. [DOI: 10.1111/j.1365-2044.2011.07008.x] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
24
|
Abstract
Postoperative cognitive dysfunction (POCD) refers to a postoperative decline in cognitive function compared with preoperative cognitive function. Diagnosis requires pre- and postoperative testing, the latter of which is usually performed both 7 days and 3 months postoperatively. Although several risk factors for POCD have been described, age is the only consistently reported risk factor. Postoperative cognitive dysfunction is often transient. It may last several months, and is associated with leaving the labor market prematurely and increased mortality. As the pathophysiology of POCD is still a matter of debate and is likely to be multifactorial, there are no widely accepted prophylactic and therapeutic interventions. In this article, we discuss POCD's definition, risk factors, long-term significance, and pathophysiology. We also present data on prophylactic interventions that have been investigated in clinical trials.
Collapse
Affiliation(s)
- Christoph S Burkhart
- Department of Anesthesia and Intensive Care Medicine, University Hospital Basel, Basel, Switzerland.
| | | |
Collapse
|
25
|
Kellermann K, Dertinger N, Blobner M, Kees F, Kochs EF, Jungwirth B. Perioperative moxifloxacin treatment in rats subjected to deep hypothermic circulatory arrest: Reduction in cerebral inflammation but without improvement in cognitive performance. J Thorac Cardiovasc Surg 2011; 141:796-802. [DOI: 10.1016/j.jtcvs.2010.04.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 03/19/2010] [Accepted: 04/16/2010] [Indexed: 11/29/2022]
|
26
|
Jungwirth B, de Lange F. Animal models of cardiopulmonary bypass: development, applications, and impact. Semin Cardiothorac Vasc Anesth 2010; 14:136-40. [PMID: 20478954 DOI: 10.1177/1089253210370491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Neurologic and neurocognitive complications after cardiac surgery have been reported repeatedly. To better understand its etiology and design protective strategies, small animal models have been developed. This study describes the development of a survival rat cardiopulmonary bypass (CPB) model, along with the introduction of an appropriately sized oxygenator. This model led the way for even more complicated models with CPB, facilitating full cardiac arrest with anterograde cardioplegia administration, air embolization, and deep hypothermic circulatory arrest. In addition, the results of several of those rat CPB studies are summarized and their preclinical relevance is pointed out.
Collapse
|
27
|
|