1
|
Gargiulo S, Albanese S, Megna R, Gramanzini M, Marsella G, Vecchiarelli L. Veterinary medical care in rodent models of stroke: Pitfalls and refinements to balance quality of science and animal welfare. Neuroscience 2025; 572:269-302. [PMID: 39894435 DOI: 10.1016/j.neuroscience.2025.01.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/25/2024] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
Rodent models of cerebral ischemia provide a valuable contribution to a better understanding of stroke pathophysiology, to validate diagnostic methods, and to enable testing of new treatments for ischemia-reperfusion damage and comorbidities. However, ethical concerns have led to increased attention to the welfare aspects of such models. Supportive therapies are an essential part of the overall animal care and use program and should be tailored to the experimental model being studied, the regulatory requirements, and research objectives to achieve high-quality preclinical studies and ethical research practices. On the other hand, the use of veterinary medical treatments in preclinical models of stroke must balance the needs of animal care and potential sources of bias in experimental results. This report provides a systematic review of the scientific literature covering the relevant period from years 1988 to September 2024, with the aim to investigating veterinary medical interventions useful to minimize suffering in rodent models of stroke without producing experimental bias. The research findings, consolidated from 181 selected studies, published from 1991 to 2023, indicate the feasibility of implementing personalized protocols of anesthesia, analgesics, antibiotics, and other supportive therapies in rodent models of stroke, while avoiding scientific interferences. These data fill a gap in current knowledge and could be of interest for an interdisciplinary audience working with rodent models of stroke, stimulating further refinements to safeguard both animal welfare and the validity of experimental findings, and may promote the culture of ethical conduct in various research fields and disciplines.
Collapse
Affiliation(s)
- Sara Gargiulo
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy.
| | - Sandra Albanese
- Institute of Biostructures and Bioimaging, National Research Council, 80131 Naples, Italy.
| | - Rosario Megna
- Institute of Biostructures and Bioimaging, National Research Council, 80131 Naples, Italy.
| | - Matteo Gramanzini
- Institute of Chemical Sciences and Technologies "Giulio Natta", National Research Council, L.go F. Vito, 00168 Rome, Italy.
| | - Gerardo Marsella
- Animal Care Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Lidovina Vecchiarelli
- Animal Welfare at Animal and Plant Health Agency, Department for Environment Food and Rural Affairs, Midlands, UK.
| |
Collapse
|
2
|
Yi S, Cao H, Zheng W, Wang Y, Li P, Wang S, Zhou Z. Targeting the opioid remifentanil: Protective effects and molecular mechanisms against organ ischemia-reperfusion injury. Biomed Pharmacother 2023; 167:115472. [PMID: 37716122 DOI: 10.1016/j.biopha.2023.115472] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/18/2023] Open
Abstract
Opioids are widely used in clinical practice by activating opioid receptors (OPRs), but their clinical application is limited by a series of side effects. Researchers have been making tremendous efforts to promote the development and application of opioids. Fortunately, recent studies have identified the additional effects of opioids in addition to anesthesia and analgesia, particularly in terms of organ protection against ischemia-reperfusion (I/R) injury, with unique advantages. I/R injury in vital organs not only leads to cell dysfunction and structural damage but also induces acute and chronic organ failure, even death. Early prevention and appropriate therapeutic targets for I/R injury are crucial for organ protection. Opioids have shown cardioprotective effects for over 20 years, especially remifentanil, a derivative of fentanyl, which is a new ultra-short-acting opioid analgesic widely used in clinical anesthesia induction and maintenance. In this review, we provide current knowledge about the physiological effects related to OPR-mediated organ protection, focusing on the protective effect and mechanism of remifentanil on I/R injury in the heart and other vital organs. Herein, we also explored the potential application of remifentanil in clinical I/R injury. These findings provide a theoretical basis for the use of remifentanil to inhibit or alleviate organ I/R injury during the perioperative period and provide insights for opioid-induced human organ protection and drug development.
Collapse
Affiliation(s)
- Shuyuan Yi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China; Department of Anaesthesiology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China; School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Hong Cao
- Department of Anaesthesiology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China
| | - Weilei Zheng
- Department of Anaesthesiology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Shoushi Wang
- Department of Anaesthesiology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China.
| | - Zhixia Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
3
|
Han C, Ji H, Guo Y, Fei Y, Wang C, Yuan Y, Ruan Z, Ma T. Effect of Subanesthetic Dose of Esketamine on Perioperative Neurocognitive Disorders in Elderly Undergoing Gastrointestinal Surgery: A Randomized Controlled Trial. Drug Des Devel Ther 2023; 17:863-873. [PMID: 36974331 PMCID: PMC10039635 DOI: 10.2147/dddt.s401161] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Perioperative neurocognitive disorders (PND), including delayed neurocognitive recovery (dNCR) and postoperative cognitive dysfunction (POCD), are common postoperative complications in elderly patients and adversely affect their prognosis. The study was designed to explore the effects of esketamine on postoperative cognitive function in elderly patients who underwent gastrointestinal surgery under general anesthesia and its potential mechanism. METHODS Eighty-four patients aged 65 and above undergoing gastrointestinal surgery were randomly divided into 2 groups: the esketamine group (group S) and the control group (group C). Group S received intravenous sub-anesthetic doses of esketamine (0.15 mg/kg) 5 minutes before the initiation of surgery, while group C received the same volume of saline. A battery of neuropsychological tests was used to assess cognitive function before surgery, 7 days, and 3 months after surgery. The primary outcome was the incidence of dNCR at 7 days postoperatively and POCD at 3 months postoperatively in both groups. The secondary outcome measures included changes in the levels of serum interleukin-6 (IL-6) and calcium-binding protein β (S100β) before and 1 day after surgery. RESULTS The incidence of dNCR in group S was lower than that of group C (18.15% vs 38.24% P=0.033). Contrarily, there was no difference in both groups regarding POCD 3 months postoperatively (6.06% vs 14.37% P=0.247). Plasma IL-6 and S100β levels were significantly elevated in both groups on postoperative day 1 (p<0.05), but esketamine pretreatment reduced these levels to some extent compared with group C (p<0.05). CONCLUSION Sub-anesthetic doses of esketamine might reduce the incidence of dNCR and improve early postoperative cognitive function in elderly patients undergoing gastrointestinal surgery, which might be related to the anti-neuroinflammation effects of esketamine.
Collapse
Affiliation(s)
- Chao Han
- Department of Anesthesiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, 214200, People’s Republic of China
- Yixing Clinical College, Medical College of Yangzhou University, Yixing, Jiangsu, 214200, People’s Republic of China
| | - Haiyan Ji
- Department of Anesthesiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, 214200, People’s Republic of China
| | - Yaxin Guo
- Department of Anesthesiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, 214200, People’s Republic of China
| | - Yuanhui Fei
- Department of Anesthesiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, 214200, People’s Republic of China
| | - Chunhui Wang
- Department of Anesthesiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, 214200, People’s Republic of China
| | - Yanbo Yuan
- Department of Anesthesiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, 214200, People’s Republic of China
| | - Zhihui Ruan
- Department of Anesthesiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, 214200, People’s Republic of China
| | - Tieliang Ma
- Department of Anesthesiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, 214200, People’s Republic of China
- Yixing Clinical College, Medical College of Yangzhou University, Yixing, Jiangsu, 214200, People’s Republic of China
| |
Collapse
|
4
|
Remifentanil alleviates hypoxic-ischemic brain damage-induced cognitive impairment via BACH1. Neurosci Lett 2022; 786:136802. [PMID: 35853564 DOI: 10.1016/j.neulet.2022.136802] [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: 04/08/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022]
Abstract
Hypoxia-ischemia (HI) is among the most frequent causes of death and disability in neonates. We aimed here to examine the neuroprotective effects of Remifentanil (RE) and the underlying mechanisms in a rat model of hypoxic-ischemic brain damage (HIBD). We found that RE improved the learning memory ability, reduced neuronal cell damage and apoptosis, reduced inflammation induced by suppressing the expression of BTB domain and CNC homolog 1 (BACH1) in rats with HIBD. BACH1 attenuated the alleviating effect of RE on cognitive impairment in HIBD rats. Moreover, RE inhibited TRAF3 expression by downregulating BACH1, and TRAF3 attenuated the therapeutic effect of RE on cognitive impairment by activating the NF-κB signaling. In conclusion, our findings demonstrated that RE inhibits the expression of BACH1, which in turn inhibits the NF-κB signaling pathway by suppressing TRAF3. RE may be a promising therapeutic agent to attenuate HIBD-induced cognitive impairment.
Collapse
|
5
|
Gong WY, Xu B, Liu L, Li ST. Dezocine relieves the postoperative hyperalgesia in rats through suppressing the hyper-action of Akt1/GSK-3β pathway. Exp Brain Res 2022; 240:1435-1444. [PMID: 35333956 DOI: 10.1007/s00221-022-06341-7] [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: 08/17/2021] [Accepted: 02/24/2022] [Indexed: 11/28/2022]
Abstract
The relieving role of dezocine in pain after surgery was previously reported, while the potential mechanism was not completely clear. Therefore, the current research probed into the regulatory mechanism of dezocine in pain after surgery. A postoperative pain model was established by performing plantar incision surgery on the juvenile Sprague-Dawley rats. After the rats were treated with dezocine or SC79 (Akt1 activator), the paw withdrawal threshold and paw withdrawal latency of rats were detected to evaluate the mechanical allodynia and thermal hyperalgesia. After the plantar tissue, dorsal root ganglions, and spinal cord of rats were collected, the expressions of Akt1, p-Akt1, GSK-3β, and p-GSK-3β in the tissues were determined by western blot to evaluate the activation state of the Akt1/GSK-3β pathway. After surgery, the paw withdrawal threshold and paw withdrawal latency of rats were lessened, whereas the ratios of p-Akt1/Akt1 and p-GSK-3β/GSK-3β were augmented in rat plantar tissue, dorsal root ganglions, and spinal cord. After treatment with dezocine alone, the paw withdrawal threshold and paw withdrawal latency of postoperative rats were elevated, but ratios of p-Akt1/Akt1 and p-GSK-3β/GSK-3β were reduced. After co-treatment with dezocine and SC79, SC79 reversed the effects of dezocine on elevating the paw withdrawal threshold and paw withdrawal latency, and reducing the ratios of p-Akt1/Akt1 and p-GSK-3β/GSK-3β in postoperative rats. Dezocine ameliorated the postoperative hyperalgesia in rats via repressing the hyper-action of Akt1/GSK-3β pathway.
Collapse
Affiliation(s)
- Wen-Yi Gong
- Department of Anesthesiology, Shanghai General Hospital of Nanjing Medical University, No.100, Haining Road, Hongkou District, Shanghai, 200080, People's Republic of China.,Department of Anesthesiology, Wusong Hospital, No.101, North Tongtai Road, Baoshan District, Shanghai, 200940, People's Republic of China
| | - Bing Xu
- Department of Anesthesiology, Wusong Hospital, No.101, North Tongtai Road, Baoshan District, Shanghai, 200940, People's Republic of China
| | - Li Liu
- Department of Anesthesiology, Wusong Hospital, No.101, North Tongtai Road, Baoshan District, Shanghai, 200940, People's Republic of China
| | - Shi-Tong Li
- Department of Anesthesiology, Shanghai General Hospital of Nanjing Medical University, No.100, Haining Road, Hongkou District, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
6
|
Liu W, Wang X, Du L, Sun Y. Remifentanil Suppresses Oxidative Stress and Inflammation Induced by Glutamate via Activation of PPARγ/HO-1 Signaling Pathway in Hippocampal Neuronal Cells. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Excitotoxicity caused by glutamate severely damages the central nervous system, contributing to the progress of neurodegenerative diseases. Remifentanil is an ultra-short acting synthetic α-opioid receptor agonist and it protects the body against oxidative stress. Oxidative
stress is a causative factor for neuronal cell death, contributing to the pathogenesis of neurological diseases. More importantly, remifentanil has been confirmed to have neuroprotective effects on cerebral ischemia. Hence, the aim of the present study was to investigate the molecular mechanism
underlying the effect of remifentanil on glutamate (Glu)-induced oxidative stress and inflammation in hippocampal cells. In present study, the cell viability was detected via CCk-8 assay. The cell apoptosis was evaluated by tunel assay. Western blot was performed for measurement of protein
expression level. Generation of ROS level was detected by the ROS Activity Assay Kit (KA3842, Abnova) and DCF-DA staining method. MDA and SOD levels were detected by corresponding kits. The results from the present study suggested that remifentanil enhanced cell viability, reduced cell apoptosis
rate and prevented oxidative stress in glutamate-induced HT22 cells. The PPARγ/HO-1 pathway was activated by remifentanil. After inhibition of PPARγ/HO-1 pathway, the anti-apoptosis and anti-oxidative stress effects of remifentanil were abolished. In conclusion,
remifentanil has anti-apoptosis and anti-oxidative stress effects on glutamate-induced HT22 Cells via PPARγ/HO-1 pathway. Hence, remifentanil is a promising agent for attenuation of cytotoxicity induced by glutamate, providing a new strategy for treatment of excitotoxicity
caused by glutamate in the central nervous system.
Collapse
Affiliation(s)
- Weihua Liu
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University (Qingyuan People’s Hospital), Guangzhou City, Guangdong Province, 511518, China
| | - Xinli Wang
- Department of Anesthesiology, Capital Health Care Hospital for Children and Women, Beijing City, 100069, China
| | - Liangqin Du
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University (Qingyuan People’s Hospital), Guangzhou City, Guangdong Province, 511518, China
| | - Yanlin Sun
- Department of Anesthesiology, Qingdao Lianchi Maternity and Infant Hospital, Qingdao City, Shandong Province, 266000, China
| |
Collapse
|
7
|
Orum MH, Kalenderoglu A. Acute opioid use may cause choroidal thinning and retinal nerve fiber layer increase. J Addict Dis 2021; 39:322-330. [PMID: 33555234 DOI: 10.1080/10550887.2021.1874816] [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] [Indexed: 10/22/2022]
Abstract
The number of optical coherence tomography (OCT) examinations in substance use disorders is gradually increasing. However, OCT findings in opioid use disorder (OUD) have not yet been investigated. In this study, we compared the retinal nerve fiber layer (RNFL), the ganglion cell layer (GCL), the inner plexiform layer (IPL), and choroid thickness (CT) of OUD and control groups. We included 43 male patients and 43 healthy male controls of similar age (p = 0.296) in the study, prospectively. On the day of OCT application, urine toxic screening test results of all OUD patients were positive for opioid use. There was a significant difference between OUD and control groups in terms of CT (p < 0.05), nasal superior (NS), and nasal (N) sectors of the RNFL (p < 0.05) values of both eyes. According to the binary logistic regression analysis, the sensitivity of mean NS (p = 0.001) and mean CT (p = 0.007) related to the diagnosis of OUD was 72.1 percent, and the specificity was 65.1 percent. Receiver operating characteristic (ROC) analysis revealed that the sensitivity and specificity of mean CT for the diagnosis of OUD were 18.6% and 97.7%, respectively. This is the first study to investigate the OCT findings in OUD. Our findings are important in terms of showing thinning in the choroidal layer and an increase in the volume of the NS and N sectors of RNFL while detecting opioids in the body/urine. Further studies are needed to clarify whether these differences are due to the acute and/or chronic effects of opioids.
Collapse
Affiliation(s)
- Mehmet H Orum
- Psychiatry Outpatient Clinic, Kahta State Hospital, Adiyaman, Turkey
| | - Aysun Kalenderoglu
- Department of Psychiatry, Adiyaman University, Faculty of Medicine, Adiyaman, Turkey
| |
Collapse
|
8
|
Kong C, Miao F, Wu Y, Wang T. Oxycodone suppresses the apoptosis of hippocampal neurons induced by oxygen-glucose deprivation/recovery through caspase-dependent and caspase-independent pathways via κ- and δ-opioid receptors in rats. Brain Res 2019; 1721:146319. [PMID: 31276638 DOI: 10.1016/j.brainres.2019.146319] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/21/2019] [Accepted: 07/01/2019] [Indexed: 02/05/2023]
Abstract
Cerebral ischemia/reperfusion injury (CIRI) can lead to perioperative neurocognitive disorders (PND) during clinical recanalization procedures in cerebral vessels, principally due to neuronal apoptosis in the hippocampus. Oxycodone appears to be a multiple opioid receptor agonist and exerts intrinsic antinociception activity via κ-opioid receptor (KOR). Recent evidence has revealed that activation of both δ-opioid receptor (DOR) and KOR can provide neuroprotection against CIRI in vivo and in vitro. In our study, we established an oxygen-glucose deprivation/recovery (OGD/R) model with fetal hippocampal neurons and found that oxycodone could induce CIRI tolerance in these neurons, primarily through KOR and DOR. Possible mechanisms might involve the regulatory effect of oxycodone on the MAPK-Bcl2/Bax-caspase-9-caspase-3 pathway, as well as its inhibitory effect on cellular reactive oxygen species (ROS) production and mitochondrial membrane potential activation. Taken together, our findings may indicate a potential method for the prevention and treatment of PND associated with CIRI.
Collapse
Affiliation(s)
- Cuicui Kong
- Department of Anesthesiology, Capital Medical University Xuanwu Hospital, Beijing 100053, China
| | - Fangfang Miao
- Department of Anesthesiology, Capital Medical University Xuanwu Hospital, Beijing 100053, China
| | - Yan Wu
- Department of Anatomy, Beijing Institute for Brain Disorders, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China
| | - Tianlong Wang
- Department of Anesthesiology, Capital Medical University Xuanwu Hospital, Beijing 100053, China.
| |
Collapse
|
9
|
Ahn JH, Kim D, Chung IS, Lee JJ, Lee EK, Jeong JS. Pre-administration of remifentanil in target-controlled propofol and remifentanil anesthesia prolongs anesthesia induction in neurosurgical patients: A double-blind randomized controlled trial. Medicine (Baltimore) 2019; 98:e14144. [PMID: 30653147 PMCID: PMC6370149 DOI: 10.1097/md.0000000000014144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Pre- and co-administration of remifentanil in target-controlled propofol and remifentanil anesthesia are the most common methods in clinical practice. However, anesthesia induction time by timing remifentanil administration was not identified. Therefore, we investigated the induction time of anesthesia based on type of remifentanil administration in target-controlled anesthesia. METHODS A total of 60 patients were randomly assigned to 1 of 2 groups: Pre-administered with remifentanil before propofol infusion (Group R, n = 30) and co-administered with remifentanil with propofol (Group N, n = 30). The primary outcome was total induction time based on the order of remifentanil administration. Secondary outcomes were from start of the propofol infusion time to loss of consciousness (LOC), rocuronium onset time, time to Bispectral index (BIS) 60, and hemodynamic variables. RESULTS The mean ± SD of total induction time was 180.5 ± 49.0 s in Group N and 246.3 ± 64.7 s in Group R (mean difference: 65.8 seconds; 95% CI: 35.0-96.5 s, P < .01). Time to BIS 60 and rocuronium onset time were longer in the Group R (P < .01 and P < .01, respectively). The Δheart rate and Δcardiac output values were lower in the Group R (P = .02 and P = .04, respectively). Injection pain was reported by 11 of 28 (39%) in the Group N and in 2 of 28 (7%) in the Group R (difference in proportion: 32%, 95% CI: 10-51%, P = .01). CONCLUSION Pre-administration of remifentanil in target-controlled propofol and remifentanil anesthesia prolongs total induction time about 35% compared to co-administration of remifentanil and propofol by decreased CO.
Collapse
Affiliation(s)
- Jin Hee Ahn
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Anesthesiology and Pain Medicine, College of Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Doyeon Kim
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Ik Soo Chung
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Jeong Jin Lee
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Eun Kyung Lee
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Ji Seon Jeong
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| |
Collapse
|
10
|
Prolonged DADLE exposure epigenetically promotes Bcl-2 expression and elicits neuroprotection in primary rat cortical neurons via the PI3K/Akt/NF-κB pathway. Acta Pharmacol Sin 2018; 39:1582-1589. [PMID: 29795362 DOI: 10.1038/aps.2018.7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/02/2018] [Indexed: 01/09/2023]
Abstract
Both in vivo and in vitro studies have shown the beneficial effects of the delta-opioid receptor (DOR) on neurodegeneration in hypoxia/ischemia. We previously reported that DOR stimulation with [(D-Ala2, D-Leu5) enkephalin] (DADLE), a potent DOR agonist, for both a short (minutes) and long (days) time has notable protective effects against sodium azide (NaN3)-induced cell injury in primary cultured rat cortical neurons. We further demonstrated that short-term DADLE stimulation increased neuronal survival through the PKC-mitochondrial ERK pathway. However, the mechanisms underlying long-term neuroprotection by DADLE remain unclear. Here, we showed that DOR stimulation with DADLE (0.1 μmol/L) for 2 d selectively activates the PI3K/Akt/NF-κB pathway in NaN3-treated neurons; this activation increased Bcl-2 expression, attenuated Cyto c release and promoted neuronal survival. Further investigation revealed that sustained DADLE stimulation increased Bcl-2 expression by enhancing NF-κB binding to the Bcl-2 promoter and upregulating the histone acetylation levels of the Bcl-2 promoter. Our results demonstrate that prolonged DADLE exposure epigenetically promotes Bcl-2 expression and elicits neuroprotective effects in the NaN3 model via the PI3K/Akt/NF-κB pathway.
Collapse
|
11
|
The neuroprotective role of the brain opioid system in stroke injury. Drug Discov Today 2018; 23:1385-1395. [DOI: 10.1016/j.drudis.2018.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/30/2018] [Accepted: 02/26/2018] [Indexed: 11/18/2022]
|
12
|
Niu RN, Shang XP, Teng JF. Overexpression of Egr2 and Egr4 protects rat brains against ischemic stroke by downregulating JNK signaling pathway. Biochimie 2018; 149:62-70. [PMID: 29580816 DOI: 10.1016/j.biochi.2018.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/20/2018] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate the effect of Egr2 and Egr4 upregulation on ischemic stroke recovery of rats. METHODS In this study, Sprague Dawley (SD) rats assigned at random into control, sham and MCAO (middle cerebral artery occlusion) group were treated accordingly to build MCAO models. The neurological severity scores (NSS) test was applied to assess rats' behavior. Triphenyltetrazolium chloride (TTC) staining reflected infarct areas while Nissl staining revealed the number of neurons. Levels of pro-inflammatory cytokines (interleukin [IL]-1β, IL-6 and tumor necrosis factor [TNF]-α) were judged by enzyme-linked immunosorbent assay (ELISA) in brain and serum tissues. We applied western blot to check the expression of Egr2, Egr4 and JNK/c-JUN (c-Jun N-terminal kinase) pathway. Further grouping of rats were based on various transfection, requiring control, sham, MCAO, MCAO + Egr2 cDNA (complementary DNA), MCAO + Egr4 cDNA, MCAO + Egr2 cDNA + Egr4 cDNA group to observe difference in MCAO recovery and JNK/c-JUN-pathway-related protein expression. RESULTS Under successful modeling of MCAO, western blot results suggested down-regulation of Egr2 and Egr4 and overexpression of pro-inflammatory cytokines. The JNK/c-JUN pathway was activated. On upregulation of Egr2 and Egr4 in infarct areas, neurological function of SD rats recovered along with repressed JNK/c-JUN pathway activation and increased neuron number. CONCLUSION Upregulation of Egr2 and Egr4 could demote the activation of JNK/c-JUN pathway and the expression of pro-inflammatory cytokines in MCAO rats, so that Egr2 and Egr4 might be potential targets for ischemic stroke in future.
Collapse
Affiliation(s)
- Rui-Na Niu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiao-Ping Shang
- Department of Medical Records, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jun-Fang Teng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
13
|
Smith JA, Donepudi RV, Argoti PS, Giezentanner AL, Jain R, Boring N, Garcia E, Moise KJ. Exploring the Pharmacokinetic Profile of Remifentanil in Mid-Trimester Gestations Undergoing Fetal Intervention Procedures. Front Pharmacol 2017; 8:11. [PMID: 28174536 PMCID: PMC5260009 DOI: 10.3389/fphar.2017.00011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/05/2017] [Indexed: 11/13/2022] Open
Abstract
Background: Indications for surgery during pregnancy have increased. Specifically fetal interventions have increased from conditions that were considered lethal like twin-twin transfusion syndrome and severe fetal anemia to non-lethal conditions like myelomeningocele. The optimal anesthetic agent for in utero surgery is yet to be determined. Success of the procedure is often dictated by the efficacy of the anesthetic to immobilize the fetus without over-sedating mom. Remifentanil is used as preferred agent due to its short half-life however pharmacokinetics in pregnancy is unknown. Objective: To determine the pharmacokinetic parameters of remifentanil in a mid-trimester pregnant patient population undergoing fetal intervention. Study Design: A validated liquid chromatography assay with ultraviolet absorbance was employed to estimate maternal serum remifentanil levels. Blood samples were obtained at baseline and at selected time points: 5, 15, 30, 45, 60 min after the beginning of the remifentanil infusion and at 15, 30, and 60 min post end of infusion. Results: Ten pregnant patients were enrolled in the study however only eight patients had sampling obtained at all time points. The mean gestational age was 22.2 (±2.7) weeks, maternal age was 27.8 (±5.1) years and body mass index was 29.6 (±6.3). After receiving a continuous infusion of remifentanil, mean total dose was 975.3 μg, Cmin was 2.0 ng/mL and Cmax was 8.4 ng/mL. A two-compartment model best described the plasma remifentanil data. Mean pharmacokinetic parameters were: volume of distribution (Vdc) = 124.6 L (16.2–530.8 L), maternal remifentanil total clearance (Clt) = 170.7 L/h (17.7–486.9 L/h), and half-life (t½) = 0.6 h (0.2–0.9 h). The maternal remifentanil area under the curve (AUC) ranged from 2.7 to 21.7 μg/L*h. The mean alpha-acidic glycoprotein was 124.8 mg/dL (81.3–149.8). Conclusion: The pharmacokinetic profile of remifentanil in pregnant women is similar to previously reported general population profiles. This data did provide potential rationale for the clinical observations why when remifentanil is dosed based on non-pregnant guidelines, it did not uniformly provide adequate fetal immobilization as per anecdotal perception of operating fetal surgeons. These findings are important for the development of further clinical studies to optimize dosing for surgery during pregnancy including the estimation of placental transfer and total fetal exposure.
Collapse
Affiliation(s)
- Judith A Smith
- Department of Obstetrics, Gynecology and Reproductive Medicine, UTHealth McGovern Medical SchoolHouston, TX, USA; Department of Pharmacy, Memorial Hermann Hospital-Texas Medical Centre (TMC)Houston, TX, USA
| | - Roopali V Donepudi
- Department of Obstetrics, Gynecology and Reproductive Medicine, UTHealth McGovern Medical School Houston, TX, USA
| | - Pedro S Argoti
- Department of Obstetrics, Gynecology and Reproductive Medicine, UTHealth McGovern Medical School Houston, TX, USA
| | - Anita L Giezentanner
- Department of Pharmacy, Memorial Hermann Hospital-Texas Medical Centre (TMC) Houston, TX, USA
| | - Ranu Jain
- The Fetal Center, Children's Memorial Hermann HospitalHouston, TX, USA; Department of Anesthesiology, UTHealth McGovern Medical SchoolHouston, TX, USA
| | - Noemi Boring
- Department of Obstetrics, Gynecology and Reproductive Medicine, UTHealth McGovern Medical School Houston, TX, USA
| | - Elisa Garcia
- Department of Obstetrics, Gynecology and Reproductive Medicine, UTHealth McGovern Medical School Houston, TX, USA
| | - Kenneth J Moise
- Department of Obstetrics, Gynecology and Reproductive Medicine, UTHealth McGovern Medical School Houston, TX, USA
| |
Collapse
|
14
|
Kim YS, Kim WY, Kim YH, Yoo JW, Min TJ. The protective effect of hydromorphone to ischemia in rat glial cells. SPRINGERPLUS 2016; 5:610. [PMID: 27247906 PMCID: PMC4864736 DOI: 10.1186/s40064-016-2281-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/05/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Ischemic insults during operation can cause ischemic-reperfusion injuries in brain as well as memory impairments. Total intravenous anesthesia (TIVA) is the preferred anesthetic method in brain surgery, as it utilizes motor evoked potential monitoring. And the use of opioids is common in TIVA. However there are few studies about ischemic protective effect of opioids to glial cells. METHODS We used mixed cultures of rat glial cells, which were harvested from the brain of 1-day old rat. We divided the experimental groups according to their hydromorphone conditioning period: (a) pre-culture, (b) per-culture, or (c) pre- and per-culture. We measured the levels of the reactive oxygen species (ROS) induced by tert-butyl hydroperoxide (TBH) using flow cytometry. The ROS levels in the glial cells were also measured after the administration of 100 nM hydromorphone and selective opioid receptor antagonists. RESULTS The ROS levels were reduced in the hydromorphone-treated group, as compared to the control group (only TBH treated). There were no differences between pre-conditioned and per-conditioned groups. However, the ROS levels were more reduced in pre- and per-conditioned group compared to pre-conditioned or per-conditioned only groups. Furthermore, selective antagonists for the delta, kappa, or mu opioid receptor partially negated the hydromorphone effect. CONCLUSION This study demonstrated that hydromorphone can have additional protective effects on oxidative stress when pre- and per-conditioning is combined. Furthermore we proved that μ, δ, κ opioid receptors participate in protective mechanism of hydromorphone to glial cells.
Collapse
Affiliation(s)
- Young Sung Kim
- />Department of Anesthesiology, Korea University College of Medicine, Seoul, Korea
| | - Woon Young Kim
- />Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Korea
| | - Yeon-hwa Kim
- />Institute of Medical Science, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Ji Won Yoo
- />Department of Internal Medicine and Institute of Gerontology, University of Nevada, School of Medicine, Las Vegas, NV USA
| | - Too Jae Min
- />Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Korea
| |
Collapse
|
15
|
Shen JT, Li YS, Xia ZQ, Wen SH, Yao X, Yang WJ, Li C, Liu KX. Remifentanil preconditioning protects the small intestine against ischemia/reperfusion injury via intestinal δ- and μ-opioid receptors. Surgery 2016; 159:548-559. [PMID: 26410664 DOI: 10.1016/j.surg.2015.07.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 07/02/2015] [Accepted: 07/31/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) injury can cause a high rate of mortality in the perioperative period. Remifentanil has been reported to provide protection for organs against I/R injury. We hypothesized that remifentanil preconditioning would attenuate the small intestinal injury induced by intestinal I/R. METHODS We used both an in vivo rat model of intestinal I/R injury and a cell culture model using IEC-6 cells (the rat intestinal epithelial cell line) subjected to oxygen and glucose deprivation (OGD). Remifentanil was administered before ischemia or OGD, and 3 specific opioid receptors antagonists, naltrindole (a δ-OR selective antagonist), nor-binaltorphimine (nor-BNI, a κ-OR selective antagonist), and CTOP (a μ-OR selective antagonist), were administered before preconditioning to determine the role of opioid receptors in the intestinal protection mediated by remifentanil. RESULTS In the in vivo rat model, intestinal I/R induced obvious intestinal injury as evidenced by increases in the Chiu score, serum diamine oxidase activity, the apoptosis index, and the level of cleaved caspase-3 protein expression, whereas remifentanil preconditioning significantly improved these changes in vivo. In the in vitro cell culture exposed to OGD, cell viability (MTT, ie, (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay and flow cytometric analysis showed that remifentanil preconditioning enhanced IEC-6 cell viability and decreased apoptosis. In both in vitro and in vivo models, the aforementioned protective effects of remifentanil preconditioning were abolished completely by previous administration of the δ- or μ-opioid markedly attentuated but not the κ-opioid receptor antagonist. CONCLUSION Remifentanil preconditioning appears to act via δ- and μ-opioid receptors to protect the small intestine from intestinal I/R injury by attenuating apoptosis of the intestinal mucosal epithelial cells.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Biomarkers/metabolism
- Cell Line
- Cell Survival/drug effects
- Intestinal Mucosa/blood supply
- Intestinal Mucosa/drug effects
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Intestine, Small/blood supply
- Intestine, Small/drug effects
- Intestine, Small/metabolism
- Intestine, Small/pathology
- Ischemic Preconditioning/methods
- Male
- Piperidines/pharmacology
- Piperidines/therapeutic use
- Protective Agents/pharmacology
- Protective Agents/therapeutic use
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Remifentanil
- Reperfusion Injury/prevention & control
Collapse
Affiliation(s)
- Jian-Tong Shen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yun-Sheng Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Qiu Xia
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shi-Hong Wen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Yao
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wen-Jing Yang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cai Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
16
|
Lee CH, Park YC, Kim JH, Kim WY, Lee YS, Kim YH, Min TJ. The effects of hydromorphone on astrocytic responses in cerebral ischemia. Anesth Pain Med (Seoul) 2016. [DOI: 10.17085/apm.2016.11.1.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Chang Hyung Lee
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Young Cheol Park
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Jae Hwan Kim
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Woon Young Kim
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Yoon Sook Lee
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Yun Hee Kim
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Too Jae Min
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| |
Collapse
|
17
|
|
18
|
Mandeville JB, Liu CH, Vanduffel W, Marota JJA, Jenkins BG. Data collection and analysis strategies for phMRI. Neuropharmacology 2014; 84:65-78. [PMID: 24613447 PMCID: PMC4058391 DOI: 10.1016/j.neuropharm.2014.02.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 02/07/2014] [Accepted: 02/25/2014] [Indexed: 12/24/2022]
Abstract
Although functional MRI traditionally has been applied mainly to study changes in task-induced brain function, evolving acquisition methodologies and improved knowledge of signal mechanisms have increased the utility of this method for studying responses to pharmacological stimuli, a technique often dubbed "phMRI". The proliferation of higher magnetic field strengths and the use of exogenous contrast agent have boosted detection power, a critical factor for successful phMRI due to the restricted ability to average multiple stimuli within subjects. Receptor-based models of neurovascular coupling, including explicit pharmacological models incorporating receptor densities and affinities and data-driven models that incorporate weak biophysical constraints, have demonstrated compelling descriptions of phMRI signal induced by dopaminergic stimuli. This report describes phMRI acquisition and analysis methodologies, with an emphasis on data-driven analyses. As an example application, statistically efficient data-driven regressors were used to describe the biphasic response to the mu-opioid agonist remifentanil, and antagonism using dopaminergic and GABAergic ligands revealed modulation of the mesolimbic pathway. Results illustrate the power of phMRI as well as our incomplete understanding of mechanisms underlying the signal. Future directions are discussed for phMRI acquisitions in human studies, for evolving analysis methodologies, and for interpretative studies using the new generation of simultaneous PET/MRI scanners. This article is part of the Special Issue Section entitled 'Neuroimaging in Neuropharmacology'.
Collapse
Affiliation(s)
- Joseph B Mandeville
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | - Christina H Liu
- National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD 20817, USA
| | - Wim Vanduffel
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - John J A Marota
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Bruce G Jenkins
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
19
|
Eroglu A. The effect of intravenous anesthetics on ischemia-reperfusion injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:821513. [PMID: 24527458 PMCID: PMC3914339 DOI: 10.1155/2014/821513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 12/03/2013] [Indexed: 02/07/2023]
Abstract
The effects of intravenous anesthetics on ischemia-reperfusion injury (IRI) have been investigated in both animals and clinical studies. The protective effects and the dosages of the intravenous anesthetics on IRI were discussed in this paper. The prevention of the tissue injury after the IRI was demonstrated with intravenous anesthetics in some studies. In the future, the studies should be focused on the dosage of the anesthetics related to diminishing the tissue injuries. Further studies might be required in order to investigate the effects of the anesthetics on molecular levels.
Collapse
Affiliation(s)
- Ahmet Eroglu
- Karadeniz Technical University, Anesthesiology and Intensive Care Medicine, 61000 Trabzon, Turkey
- *Ahmet Eroglu:
| |
Collapse
|
20
|
Abstract
This paper is the thirty-fifth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2012 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
21
|
Ban K, Peng Z, Kozar RA. Inhibition of ERK1/2 worsens intestinal ischemia/reperfusion injury. PLoS One 2013; 8:e76790. [PMID: 24073294 PMCID: PMC3779170 DOI: 10.1371/journal.pone.0076790] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 09/03/2013] [Indexed: 01/09/2023] Open
Abstract
Background The role of extracellular signal-regulated protein kinase (ERK) in intestinal ischemia/reperfusion (I/R) injury has not been well investigated. The aim of the current study was to examine the effect of inhibition of the ERK pathway in an in vitro and in vivo model of intestinal I/R injury. Methods ERK1/2 activity was inhibited using the specific inhibitor, U0126, in intestinal epithelial cells under hypoxia/reoxygenation conditions and in mice subjected to 1 hour of intestinal ischemia followed by 6 hours reperfusion. In vitro, cell proliferation was assessed by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay, apoptosis by DNA fragmentation, and migration using an in vitro model of intestinal wound healing. Cells were also transfected with a p70S6K plasmid and the effects of overexpression similarly analyzed. In vivo, the effects of U0126 on intestinal cell proliferation and apoptosis, intestinal permeability, lung and intestinal neutrophil infiltration and injury, and plasma cytokine levels were measured. Survival was also assessed after U0126. Activity of p70S6 kinase (p70S6K) was measured by Western blot. Results In vitro, inhibition of ERK1/2 by U0126 significantly decreased cell proliferation and migration but enhanced cell apoptosis. Overexpression of p70S6K promoted cell proliferation and decreased cell apoptosis. In vivo, U0126 significantly increased cell apoptosis and decreased cell proliferation in the intestine, increased intestinal permeability, intestinal and lung neutrophil infiltration, and injury, as well as systemic pro-inflammatory cytokines, TNF-α, IL-6 and IL-1β. Mortality was also significantly increased by U0126. Inhibition of ERK1/2 by U0126 also abolished activity of p70S6K both in vitro and in vivo models. Conclusion Pharmacologic inhibition of ERK1/2 by U0126 worsens intestinal IR injury. The detrimental effects are mediated, at least in part, by inhibition of p70S6K, the major effector of mammalian target of rapamycin pathway.
Collapse
Affiliation(s)
- Kechen Ban
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail:
| | - Zhanglong Peng
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Rosemary A. Kozar
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
22
|
Remifentanil postconditioning improves global cerebral ischemia-induced spatial learning and memory deficit in rats via inhibition of neuronal apoptosis through the PI3K signaling pathway. Neurol Sci 2013; 34:1955-62. [DOI: 10.1007/s10072-013-1419-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/20/2013] [Indexed: 02/06/2023]
|
23
|
|
24
|
Liu X, An C, Jin P, Liu X, Wang L. Protective effects of cationic bovine serum albumin-conjugated PEGylated tanshinone IIA nanoparticles on cerebral ischemia. Biomaterials 2013; 34:817-30. [DOI: 10.1016/j.biomaterials.2012.10.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 10/08/2012] [Indexed: 11/26/2022]
|
25
|
Wang Z, Ma N, Riley J, Armstead WM, Liu R. Salvinorin A administration after global cerebral hypoxia/ischemia preserves cerebrovascular autoregulation via kappa opioid receptor in piglets. PLoS One 2012; 7:e41724. [PMID: 22911847 PMCID: PMC3404042 DOI: 10.1371/journal.pone.0041724] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/27/2012] [Indexed: 11/29/2022] Open
Abstract
Background Cerebral hypoxia/ischemia (HI) is not uncommon during the perinatal period. If occurring, it can result in severe neurologic disabilities that persist throughout life. Salvinorin A, a non-opioid Kappa opioid receptors (KOR) selective agonist, has the potential to address this devastating situation. We have demonstrated that salvinorin A administration before HI, preserves pial artery autoregulative function through both the KOR and extracellular signal-regulated kinases (ERK) pathways. In the present study, we tested the hypothesis that administration of salvinorin A after HI could preserve cerebral autoregulation via KOR and ERK pathway. Methodology/Principal Findings The response of the pial artery to hypercapnia, hypotension and isoproterenol were monitored before and 1 hour after HI in piglets equipped with a cranial window. Four groups of drug administration were performed after HI. The control group had DMSO (1 µl/kg, i.v.) administrated immediately after HI. Two salvinorin A treated groups had salvinorin A (10 µg/kg, i.v.) administrated 0 and 30 min after HI, respectively. The 4th group had salvinorin A and the KOR antagonist norbinaltorphimine (Nor-BIN, 1 µM topical) co-administrated 0 min after HI (n = 5). The dilation responses of the pial artery to hypercapnia and hypotension were impaired after global HI and were preserved with salvinorin A administration immediately or 30 min after HI. The preservation of autoregulation was abolished when nor-BIN was administered. Levels of phosphor-ERK(pERK)/ERK in the cerebrospinal fluid (CSF) were measured before and 1 hour after HI. After HI, the pERK/ERK levels significantly increased in both DMSO control group and salvinorin A and nor-BIN co-administration group. The elevated levels of pERK/ERK were not observed with salvinorin A only groups. Conclusions Salvinorin A administration 0 and 30 min after HI preserves autoregulation of pial artery to hypercapnia and hypotension via kappa opioid receptor and ERK pathway.
Collapse
MESH Headings
- Administration, Intravenous
- Animals
- Animals, Newborn
- Cerebral Arteries/drug effects
- Cerebral Arteries/enzymology
- Cerebral Arteries/pathology
- Cerebral Arteries/physiopathology
- Cerebrovascular Circulation/drug effects
- Diterpenes, Clerodane/administration & dosage
- Diterpenes, Clerodane/pharmacology
- Diterpenes, Clerodane/therapeutic use
- Extracellular Signal-Regulated MAP Kinases/cerebrospinal fluid
- Homeostasis/drug effects
- Hypercapnia/complications
- Hypercapnia/pathology
- Hypercapnia/physiopathology
- Hypotension/complications
- Hypotension/drug therapy
- Hypotension/pathology
- Hypotension/physiopathology
- Hypoxia-Ischemia, Brain/complications
- Hypoxia-Ischemia, Brain/drug therapy
- Hypoxia-Ischemia, Brain/physiopathology
- Isoproterenol/pharmacology
- MAP Kinase Signaling System/drug effects
- Pia Mater/blood supply
- Pia Mater/drug effects
- Pia Mater/physiopathology
- Receptors, Opioid, kappa/metabolism
- Sus scrofa
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Zhenhong Wang
- Department of Anesthesiology and Critical Care, Hospital of University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Anesthesiology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Nan Ma
- Department of Anesthesiology and Critical Care, Hospital of University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John Riley
- Department of Anesthesiology and Critical Care, Hospital of University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - William M. Armstead
- Department of Anesthesiology and Critical Care, Hospital of University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (RL); (WMA)
| | - Renyu Liu
- Department of Anesthesiology and Critical Care, Hospital of University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (RL); (WMA)
| |
Collapse
|
26
|
Wu L, Miao S, Zou LB, Wu P, Hao H, Tang K, Zeng P, Xiong J, Li HH, Wu Q, Cai L, Ye DY. Lipoxin A4 inhibits 5-lipoxygenase translocation and leukotrienes biosynthesis to exert a neuroprotective effect in cerebral ischemia/reperfusion injury. J Mol Neurosci 2012; 48:185-200. [PMID: 22661361 DOI: 10.1007/s12031-012-9807-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 05/07/2012] [Indexed: 12/31/2022]
Abstract
Lipoxin A(4) (LXA(4)), a biologically active eicosanoid with anti-inflammatory and pro-resolution properties, was recently found to have neuroprotective effects in brain ischemia. As 5-lipoxygenase (5-LOX) and leukotrienes are generally considered to aggravate cerebral ischemia/reperfusion (I/R) injury, we investigated their effects on LXA(4)-mediated neuroprotection by studying middle cerebral artery occlusion (MCAO)/reperfusion in rats and oxygen-glucose deprivation (OGD)/recovery in neonatal rat astrocyte primary cultures. LXA(4) effectively reduced infarct volumes and brain edema, and improved neurological scores in the MCAO/reperfusion experiments; this effect was partially blocked by butoxycarbonyl-Phe-Leu-Phe-Leu-Phe (Boc2), a specific antagonist of the LXA(4) receptor (ALXR). Total 5-LOX expression did not change, regardless of treatment, but LXA(4) could inhibit nuclear translocation induced by MCAO or OGD. We also found that LXA(4) inhibits the upregulation of both leukotriene B(4) (LTB(4)) and leukotriene C(4) (LTC(4)) and the phosphorylation of extracellular signal-regulated kinase (ERK) induced by MCAO or OGD. The phosphorylation of the 38-kDa protein kinase (p38) and c-Jun N-terminal kinase (JNK) was not altered throughout the experiment. These results suggest that the neuroprotective effects of LXA(4) are probably achieved by anti-inflammatory mechanisms that are partly mediated by ALXR and through an ERK signal transduction pathway.
Collapse
Affiliation(s)
- Le Wu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|