1
|
Manohara N, Ferrari A, Greenblatt A, Berardino A, Peixoto C, Duarte F, Moyiaeri Z, Robba C, Nascimento FA, Kreuzer M, Vacas S, Lobo FA. Electroencephalogram monitoring during anesthesia and critical care: a guide for the clinician. J Clin Monit Comput 2025; 39:315-348. [PMID: 39704777 DOI: 10.1007/s10877-024-01250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
Perioperative anesthetic, surgical and critical careinterventions can affect brain physiology and overall brain health. The clinical utility of electroencephalogram (EEG) monitoring in anesthesia and intensive care settings is multifaceted, offering critical insights into the level of consciousness and depth of anesthesia, facilitating the titration of anesthetic doses, and enabling the detection of ischemic events and epileptic activity. Additionally, EEG monitoring can aid in predicting perioperative neurocognitive disorders, assessing the impact of systemic insults on cerebral function, and informing neuroprognostication. This review provides a comprehensive overview of the fundamental principles of electroencephalography, including the foundations of processed and quantitative electroencephalography. It further explores the characteristic EEG signatures associated wtih anesthetic drugs, the interpretation of the EEG data during anesthesia, and the broader clinical benefits and applications of EEG monitoring in both anesthetic practice and intensive care environments.
Collapse
Affiliation(s)
- Nitin Manohara
- Division of Anesthesiology, Cleveland Clinic Abu Dhabi, Integrated Hospital Care Institute, Abu Dhabi, United Arab Emirates
| | | | - Adam Greenblatt
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Andrea Berardino
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | | | - Flávia Duarte
- Department of Anesthesiology, Hospital Garcia de Orta, Almada, Portugal
| | - Zahra Moyiaeri
- Division of Anesthesiology, Cleveland Clinic Abu Dhabi, Integrated Hospital Care Institute, Abu Dhabi, United Arab Emirates
| | | | - Fabio A Nascimento
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Matthias Kreuzer
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Susana Vacas
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco A Lobo
- Division of Anesthesiology, Cleveland Clinic Abu Dhabi, Integrated Hospital Care Institute, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
2
|
Faheem M, Khan AU, Shah FA. Pharmacological investigation of natural compounds for therapeutic potential in neuropathic pain. Nat Prod Res 2024:1-25. [PMID: 39623812 DOI: 10.1080/14786419.2024.2429116] [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: 03/20/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 03/25/2025]
Abstract
The present research is based on the investigation of post treatment naturally occurring compounds berbamine (BBM), bergapten (BRG) and carveol (CAR) in relation to its therapeutic effect in neuroinflammation and chronic constriction injury induced neuropathic pain (CCI-NP). The drug-likeness of the compounds was explored by SwissADME (http://www.swissadme.ch/). Docking was performed by Auto dock, PyRx and Discovery Studio Visualiser 2016 against cyclooxygenase-2 (COX-2) (PDB ID: ICX2), tumour necrosis factor-alpha TNF-α (PDB ID: 10T7) and nuclear factor-kappa B (NF-κb) (PDB ID: INFK). Molecular dynamic simulation was performed through Desmond software. In in-vivo protocols, sciatic nerve was ligated and treatment was initiated and maintained until the 14th day. Behavioural assays (paw deformation, thermal hyperalgesia, mechanical allodynia and cold allodynia) were performed and tissues were extracted for molecular investigation. Hydrogen bonds and binding affinities of ligand target complex were determined. Berbamine showed binding against NF-κB (7.9 kcal/mol). Treatment reversed paw deformation, reduced thermal hyperalgesia, mechanical allodynia and cold allodynia. Treatment also improves the level of protective GSH and GST levels in the sciatic nerve and spinal cord and lowering the detrimental oxidative stress markers iNOS and LPO. Based on the results the aforementioned compounds correct behavioural deficit, inhibit COX-2, TNF-, and NF-κB over expression, as evidenced by Enzyme-linked immunosorbant assay (ELISA) provide neuroprotection in chronic constriction damage. Hence berbamine can be considered as neuroprotective compound.
Collapse
Affiliation(s)
- Muhammad Faheem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Arif-Ullah Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Fawad Ali Shah
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
3
|
Tao Z, Li P, Zhao X. Progress on the Mechanisms and Neuroprotective Benefits of Dexmedetomidine in Brain Diseases. Brain Behav 2024; 14:e70116. [PMID: 39482839 PMCID: PMC11527817 DOI: 10.1002/brb3.70116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/27/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024] Open
Abstract
INTRODUCTION Dexmedetomidine, a highly specific α2 agonist, has been extensively utilized in clinical sedation and surgical anesthesia since its introduction in 2000 due to its excellent sympatholytic, sedative, and analgesic effects. This review aimed to identify new approaches for the treatment of patients with brain disorders by thoroughly describing the mechanism of action of dexmedetomidine and examining its neuroprotective effects from the standpoints of basic and clinical research. METHODS The PubMed and Web of Science databases were searched using the keywords dexmedetomidine and related brain diseases, although relevant articles from the last decade were included for detailed summarization and analysis. RESULTS Dexmedetomidine has shown strong neuroprotective effects, such as protection of the blood-brain barrier, decreased neuronal death, maintained hemodynamic stability, and reduced postoperative agitation and cognitive dysfunction. Furthermore, dexmedetomidine has been shown to exert various neuroprotective effects, including anti-inflammatory and antioxidative stress effects, modulation of autophagy, and reduction of apoptosis in cerebral diseases. CONCLUSIONS Dexmedetomidine acts as a neuroprotective agent against brain diseases during all phases of treatment. However, clinical trials with larger sample sizes are required to optimize dosage and dosing strategies.
Collapse
Affiliation(s)
- Zhenxing Tao
- Wuxi Medical SchoolJiangnan UniversityWuxiChina
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
| | - Pengpeng Li
- Wuxi Medical SchoolJiangnan UniversityWuxiChina
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
| | - Xudong Zhao
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
- Wuxi Neurosurgical InstituteWuxiChina
| |
Collapse
|
4
|
Zhou M, Liu L, Tan Y, Huang R, Yang Z. The mechanism of Taohong Siwu decoction in treating chemotherapy-induced peripheral neuropathy: a network pharmacology and molecular docking study. Transl Cancer Res 2024; 13:3842-3853. [PMID: 39145055 PMCID: PMC11319946 DOI: 10.21037/tcr-24-1019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/19/2024] [Indexed: 08/16/2024]
Abstract
Background Taohong Siwu decoction (THSWD) is a classic traditional Chinese medicine (TCM) formula known for its effects in promoting blood circulation, removing blood stasis, and rejuvenating energy. There have been clinical reports of THSWD treating chemotherapy-induced peripheral neuropathy (CIPN) caused by paclitaxel. We conducted a network pharmacology and molecular docking analysis to further clarify the molecular mechanisms by which THSWD exerts its protective effects against CIPN. Methods Chemical components of THSWD and their corresponding targets were obtained through the traditional Chinese medicine systems pharmacology database and analysis platform (TCMSP), and related targets of CIPN were searched in disease databases including Online Mendelian Inheritance in Man (OMIM), Therapeutic Target Database (TTD), GeneCards, and DrugBank. Common targets between THSWD and CIPN were identified using Venn diagrams. A protein-protein interaction (PPI) network was constructed using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), which was followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. AutoDock and PyMOL were used for the molecular docking validation of the key components of THSWD with core targets. Results At total of 69 chemical components of THSWD were identified, corresponding to 856 targets; 2,297 targets were associated with CIPN, with an intersection of 105 common targets. PPI analysis identified eight core targets: MYC, TNF, MAPK14, AKT1, ESR1, RELA, TP53, and HSP90AA1; KEGG enrichment analysis implicated signaling pathways such as PI3K-Akt, NF-κB, and HIF-1, etc. Molecular docking results indicated that the selected active components and their corresponding target proteins have good binding activity. Conclusions Through network pharmacology, this study found that THSWD has significant advantages in treating CIPN. By analyzing potential core targets, biological functions, and involved signaling pathways, we clarified the potential molecular biological mechanisms involved in THSWD's treatment effect. This study provides a theoretical basis for the clinical application of THSWD in treating CIPN.
Collapse
Affiliation(s)
- Meiyu Zhou
- Division of Hematology, The Comprehensive Cancer Center, Chongqing University Fuling Hospital, Chongqing, China
| | - Li Liu
- Division of Hematology, The Comprehensive Cancer Center, Chongqing University Fuling Hospital, Chongqing, China
| | - Yonghong Tan
- Division of Hematology, The Comprehensive Cancer Center, Chongqing University Fuling Hospital, Chongqing, China
| | - Rui Huang
- Division of Hematology, The Comprehensive Cancer Center, Chongqing University Fuling Hospital, Chongqing, China
| | - Zailiang Yang
- Division of Hematology, The Comprehensive Cancer Center, Chongqing University Fuling Hospital, Chongqing, China
| |
Collapse
|
5
|
Khan J, Ali G, Saeed A, Khurshid A, Ahmad S, Kashtoh H, Ataya FS, Bathiha GES, Ullah A, Khan A. Efficacy assessment of novel methanimine derivatives in chronic constriction injury-induced neuropathic model: An in-vivo, ex-vivo and In-Silico approach. Eur J Pharm Sci 2024; 198:106797. [PMID: 38735401 DOI: 10.1016/j.ejps.2024.106797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
The multicomponent etiology, complex clinical implications, dose-based side effect and degree of pain mitigation associated with the current pharmacological therapy is incapable in complete resolution of chronic neuropathic pain patients which necessitates the perpetual requirement of novel medication therapy. Therefore, this study explored the ameliorative aptitude of two novel methanimine imitative like (E)-N-(4-nitrobenzylidene)-4‑chloro-2-iodobenzamine (KB 09) and (E)-N-(4-methylbenzylidene)-4‑chloro-2-iodobenzamine (KB 10) in chronic constriction injury (CCI) of sciatic nerve induced neuropathic pain in rat model. Standard behavioral tests like dynamic and static allodynia, cold, thermal and mechanical hyperalgesia along with rotarod activity were performed at various experimental days like 0, 3, 7, 14 and 21. Enzyme linked immunosorbent assay (ELISA) on spinal tissue and antioxidant assays on sciatic nerve were executed accompanied by molecular docking and simulation studies. Prolonged ligation of sciatic nerve expressively induced hyperalgesia as well as allodynia in rats. KB 09 and KB 10 substantially attenuated the CCI elicited hyperalgesia and allodynia. They significantly reduced the biomarkers of pain and inflammation like Interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) in ELISA and while enhanced the GSH, SOD and CAT and diminished the MDA levels during antioxidant assays. KB 09 displayed -9.62 kcal/mol with TNF-α and -7.68 kcal/mol binding energy with IL-6 whereas KB 10 exhibited binding energy of -8.20 kcal/mol with IL-6 while -11.68 kcal/mol with TNF-α and hence both trial compounds ensured stable interaction with IL-6 and TNF-α during computational analysis. The results advocated that both methanimine derivatives might be novel candidates for attenuation of CCI-induced neuropathic pain prospects via anti-nociceptive, anti-inflammatory and antioxidant mechanisms.
Collapse
Affiliation(s)
- Jawad Khan
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan.
| | - Gowhar Ali
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan.
| | - Aamer Saeed
- Department of Chemistry, Quaid-I-Azam University Islamabad 45320, Pakistan
| | - Asma Khurshid
- Department of Chemistry, Quaid-I-Azam University Islamabad 45320, Pakistan.
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University Peshawar 25000, Pakistan
| | - Hamdy Kashtoh
- Department of Biotechnology, Yeungnam University, Gyeongbuk, Korea.
| | - Farid S Ataya
- Department of Biochemistry, College of Science, King Saud University, PO Box 2455, Riyadh 11451, Saudi Arabia
| | - Gaber El-Saber Bathiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheria, Egypt
| | - Aman Ullah
- Department of Pharmacy, Saba Medical Center, Abu Dhabi PO Box 20316, United Arab Emirates
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, P.O Box 33, Postal Code 616, Birkat Al Mauz, Nizwa, Sultanate of Oman
| |
Collapse
|
6
|
Lu C, Lin C, Lu Y, Tsai H, Lin C, Wu C. CDDO regulates central and peripheral sensitization to attenuate post-herpetic neuralgia by targeting TRPV1/PKC-δ/p-Akt signals. J Cell Mol Med 2024; 28:e18131. [PMID: 38426931 PMCID: PMC10906387 DOI: 10.1111/jcmm.18131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/17/2023] [Accepted: 12/29/2023] [Indexed: 03/02/2024] Open
Abstract
Postherpetic neuralgia (PHN) is a notorious neuropathic pain featuring persistent profound mechanical hyperalgesia with significant negative impact on patients' life quality. CDDO can regulate inflammatory response and programmed cell death. Its derivative also protects neurons from damages by modulating microglia activities. As a consequence of central and peripheral sensitization, applying neural blocks may benefit to minimize the risk of PHN. This study aimed to explore whether CDDO could generate analgesic action in a PHN-rats' model. The behavioural test was determined by calibrated forceps testing. The number of apoptotic neurons and degree of glial cell reaction were assessed by immunofluorescence assay. Activation of PKC-δ and the phosphorylation of Akt were measured by western blots. CDDO improved PHN by decreasing TRPV1-positive nociceptive neurons, the apoptotic neurons, and reversed glial cell reaction in adult rats. It also suppressed the enhanced PKC-δ and p-Akt signalling in the sciatic nerve, dorsal root ganglia (DRG) and spinal dorsal horn. Our research is the promising report demonstrating the analgesic and neuroprotective action of CDDO in a PHN-rat's model by regulating central and peripheral sensitization targeting TRPV1, PKC-δ and p-Akt. It also is the first study to elucidate the role of oligodendrocyte in PHN.
Collapse
Affiliation(s)
- Chun‐Ching Lu
- Department of Orthopaedics and TraumatologyNational Yang Ming Chiao Tung University HospitalYilanTaiwan
- Department of Orthopaedics, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Department of Orthopaedics and TraumatologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Chia‐Yang Lin
- Department of Nuclear MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Ying‐Yi Lu
- Department of DermatologyKaohsiung Veterans General HospitalKaohsiungTaiwan
- Department of Post‐Baccalaureate Medicine, School of Medicine, College of MedicineNational Sun Yat‐sen UniversityKaohsiungTaiwan
- Shu‐Zen Junior College of Medicine and ManagementKaohsiungTaiwan
| | - Hung‐Pei Tsai
- Division of Neurosurgery, Department of SurgeryKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Chih‐Lung Lin
- Division of Neurosurgery, Department of SurgeryKaohsiung Medical University HospitalKaohsiungTaiwan
- Department of Surgery, School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chieh‐Hsin Wu
- Division of Neurosurgery, Department of SurgeryKaohsiung Medical University HospitalKaohsiungTaiwan
- Department of Surgery, School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Center for Big Data ResearchKaohsiung Medical UniversityKaohsiungTaiwan
- Drug Development and Value Creation Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
7
|
HUANG H, WANG B, CHEN S, FANG J, WANG X, CHEN L, JIANG Y, ZHANG H, CHEN J, LIN Z. Chinese Tuina remodels the synaptic structure in neuropathic pain rats by downregulating the expression of N-methyl D-aspartate receptor subtype 2B and postsynaptic density protein-95 in the spinal cord dorsal horn. J TRADIT CHIN MED 2023; 43:715-724. [PMID: 37454256 PMCID: PMC10626369 DOI: 10.19852/j.cnki.jtcm.20221214.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/08/2022] [Indexed: 07/18/2023]
Abstract
OBJECTIVE To investigate whether the Chinese massage system, Tuina, exerts analgesic effects in a rat model of chronic constriction injury (CCI) by remodeling the synaptic structure in the spinal cord dorsal horn (SCDH). METHODS Sixty-nine male Sprague-Dawley rats were randomly and evenly divided into the normal group, sham group, CCI group, CCI + Tuina group, CCI + MK-801 [an -methyl D-aspartate receptor subtype 2B (NR2B) antagonist] group, and CCI + MK-801 + Tuina group. The neuropathic pain model was established using CCI with right sciatic nerve ligation. Tuina was administered 4 d after CCI surgery, using pressing manipulation for 10 min, once daily. Motor function was observed with the inclined plate test, and pain behaviors were observed by the Von Frey test and acetone spray test. At 19 d after surgery, the L3-L5 spinal cord segments were removed. Glutamate, interleukin 1β (IL-1β), and tumor necrosis factor-α (TNF-α) levels were detected by enzyme-linked immunosorbent assay. The protein expression levels of NR2B and postsynaptic density protein-95 (PSD-95) were detected by Western blot, and the synaptic structure was observed by transmission electron microscopy (TEM). RESULTS CCI reduced motor function and caused mechanical and cold allodynia in rats, increased glutamate concentration and TNF-α and IL-1β levels, and increased expression of synapse-related proteins NR2B and PSD-95 in the SCDH. TEM revealed that the synaptic structure of SCDH neurons was altered. Most of these disease-induced changes were reversed by Tuina and intrathecal injection of MK-801 ( < 0.05 or < 0.01). For the majority of experiments, no significant differences were found between the CCI + MK-801 and CCI + MK-801 + Tuina groups. CONCLUSIONS Chinese Tuina can alleviate pain by remodeling the synaptic structure, and NR2B and PSD-95 receptors in the SCDH may be among its targets.
Collapse
Affiliation(s)
- Hongye HUANG
- 1 College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Bingqian WANG
- 4 Peking University International Hospital, Beijing 102206, China
| | - Shuijin CHEN
- 2 Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou 350003, China
- 3 Fujian Provincial Key Laboratory of Rehabilitation Technology, Fuzhou 350003, China
| | - Jiayu FANG
- 1 College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Xiaohua WANG
- 1 College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Lechun CHEN
- 2 Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou 350003, China
- 3 Fujian Provincial Key Laboratory of Rehabilitation Technology, Fuzhou 350003, China
| | - Yu JIANG
- 2 Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou 350003, China
- 3 Fujian Provincial Key Laboratory of Rehabilitation Technology, Fuzhou 350003, China
| | - Huanzhen ZHANG
- 2 Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou 350003, China
- 3 Fujian Provincial Key Laboratory of Rehabilitation Technology, Fuzhou 350003, China
| | - Jincheng CHEN
- 2 Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou 350003, China
- 3 Fujian Provincial Key Laboratory of Rehabilitation Technology, Fuzhou 350003, China
| | - Zhigang LIN
- 2 Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou 350003, China
- 3 Fujian Provincial Key Laboratory of Rehabilitation Technology, Fuzhou 350003, China
| |
Collapse
|
8
|
Pang J, Zhang S, Kong Y, Wang Z, Pei R, Zhuang P, Wang X. The effect of dexmedetomidine on expression of neuronal nitric oxide synthase in spinal dorsal cord in a rat model with chronic neuropathic pain. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:233-239. [PMID: 37059432 PMCID: PMC10104752 DOI: 10.1055/s-0043-1761491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/10/2022] [Indexed: 04/16/2023]
Abstract
BACKGROUND Neuropathic pain typically refers to the pain caused by somatosensory system injury or diseases, which is usually characterized by ambulatory pain, allodynia, and hyperalgesia. Nitric oxide produced by neuronal nitric oxide synthase (nNOS) in the spinal dorsal cord might serve a predominant role in regulating the algesia of neuropathic pain. The high efficacy and safety, as well as the plausible ability in providing comfort, entitle dexmedetomidine (DEX) to an effective anesthetic adjuvant. The aim of this study was to investigate the effect of DEX on the expression of nNOS in spinal dorsal cord in a rat model with chronic neuropathic pain. METHODS Male Sprague Dawley (SD) rats were randomly assigned into three groups: sham operation group (sham), (of the sciatic nerve) operation (CCI) group, and dexmedetomidine (DEX) group. Chronic neuropathic pain models in the CCI and DEX groups were established by sciatic nerve ligation. The thermal withdrawal latency (TWL) was measured on day 1 before operation and on day 1, 3, 7 and 14 after operation. Six animals were sacrificed after TWL measurement on day 7, and 14 days after operation, in each group, the L4-6 segment of the spinal cords was extracted for determination of nNOS expression by immunohistochemistry. RESULTS Compared with the sham group, the TWL threshold was significantly decreased and the expression of nNOS was up-regulated after operation in the CCI and DEX groups. Compared with the CCI grou[, the TWL threshold was significantly increased and the expression of nNOS was significantly down-regulated on day 7 and 14 days after operation in the DEX group. CONCLUSION Down-regulated nNOS in the spinal dorsal cord is involved in the attenuation of neuropathic pain by DEX.
Collapse
Affiliation(s)
- Jun Pang
- Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Department of Anesthesiology, Taiyuan, People's Republic of China.
- Huazhong University of Science and Technology, Tongji Hospital, Tongji Medical College, Wuhan, People's Republic of China.
| | - Suming Zhang
- Xuzhou Medical University, The Affiliated Hospital of Xuzhou, Department of Critical Care Medicine, Xuzhou, People's Republic of China.
| | - Ying Kong
- Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Department of Anesthesiology, Taiyuan, People's Republic of China.
- Huazhong University of Science and Technology, Tongji Hospital, Tongji Medical College, Wuhan, People's Republic of China.
| | - Zhe Wang
- Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Department of Anesthesiology, Taiyuan, People's Republic of China.
- Huazhong University of Science and Technology, Tongji Hospital, Tongji Medical College, Wuhan, People's Republic of China.
| | - Ruomeng Pei
- Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Department of Anesthesiology, Taiyuan, People's Republic of China.
- Huazhong University of Science and Technology, Tongji Hospital, Tongji Medical College, Wuhan, People's Republic of China.
| | - Ping Zhuang
- Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Department of Anesthesiology, Taiyuan, People's Republic of China.
- Huazhong University of Science and Technology, Tongji Hospital, Tongji Medical College, Wuhan, People's Republic of China.
| | - Xiaopeng Wang
- Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Department of Anesthesiology, Taiyuan, People's Republic of China.
- Huazhong University of Science and Technology, Tongji Hospital, Tongji Medical College, Wuhan, People's Republic of China.
| |
Collapse
|
9
|
Biswas P, Shahnaz M, Akhter M, Ripa AP, Ali T, Rafiq K. Effects of Azadirachta indica on neuropathic pain induced by chronic constriction injury to sciatic nerve of Wistar rat. J Adv Vet Anim Res 2022; 9:359-368. [PMID: 36382046 PMCID: PMC9597922 DOI: 10.5455/javar.2022.i603] [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: 01/14/2022] [Revised: 07/13/2022] [Accepted: 07/13/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE The research was designed to assess the consequences of Azadirachta indica aqueous leaf extract (AILE) on neuropathic pain in Wister rats and the role of the ATP-dependent potassium channel (KATP) as an underlying mechanism. MATERIALS AND METHODS This experimental layout was conducted on Wistar rats (n = 120) having 150 to 200 gm of body weight. On the foundation of the experimental design, rats were divided into group I (normal saline, 5 ml/kg/body weight) and group II (sham surgery and treatment with NS), group III [chronic constriction injury (CCI) in the sciatic nerve; and treated with NS], group IV (CCI and treated with AILE 400 mg/kg body weight), Group V (CCI, pretreated with Glibenclamide 15 mg/kg followed by treated with AILE 400 mg/kg). All the treatments were given once daily for a consecutive 21 days via the oral route, except Glibenclamide. Glibenclamide was given once through the intraperitoneal route on the day of the experiment. RESULTS Based on the neuropathic pain evaluation test, all groups were again sub-divided into subgroup "a" (walking tract analysis), "b" (cold tail immersion test), "c" (Von Frey test), and "d" (hot plate test). AILE showed a significantly higher sciatic functional index (p < 0.05) in walking track analysis, tail flick latency (p ≤ 0.05) in the cold tail immersion test, and paw withdrawal threshold (p ≤ 0.05) in the Von Frey test compared to CCI control. In addition, a nonsignificant difference in all these above-mentioned variables between the rats with CCI plus AILE and the CCI plus AILE plus glibenclamide group indicated that the KATP channel was not involved in the beneficial analgesic effects of AILE. CONCLUSIONS The outcome of the present study indicates that AILE prevented worsening of neuropathic pain after chronic constriction injury in the sciatic nerve of Wistar rats in which the KATP channel was not involved.
Collapse
Affiliation(s)
- Parijat Biswas
- Department of Physiology, Ashiyan Medical College Hospital, Dhaka, Bangladesh
| | - Monira Shahnaz
- Department of Physiology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | | | - Arifa Parvin Ripa
- Department of Pharmacology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Taskina Ali
- Department of Physiology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Kazi Rafiq
- Department of Pharmacology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| |
Collapse
|
10
|
Huangqi Guizhi Wuwu Decoction can prevent and treat oxaliplatin-induced neuropathic pain by TNFα/IL-1β/IL-6/MAPK/NF-kB pathway. Aging (Albany NY) 2022; 14:5013-5022. [PMID: 35759577 PMCID: PMC9271291 DOI: 10.18632/aging.203794] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 08/17/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study explored the effects and mechanisms of Huangqi Guizhi Wuwu Decoction on chemotherapy-induced neuropathic pain (CINP). METHODS Bodyweight and related behavioral testing of the rat model were utilized to investigate the effects of Huangqi Guizhi Wuwu Decoction on CINP. ELISA was used to measure the levels of TNF-α, IL-1β, and IL-6, in the serum of chronic CINP rats. Immunohistochemistry and Western blot analysis were performed to detect the expression of MAPK pathway related-proteins namely ERK1/2, p38, and JNK, and the expression of downstream essential proteins such as c-Fos, CREB, and NF-κB. RESULTS Body weight and related behavioral testing of the rat model suggests that Huangqi Guizhi Wuwu Decoction can improve the slow weight gain of oxaliplatin-induced chronic CINP model rats and effectively prevent and treat oxaliplatin-induced regular CIPN rat model of hyperalgesia. It can also oppress the mechanical pain threshold, cold pain threshold, and heat pain threshold decreased. Furthermore, by ELISA, immunohistochemistry, and western blot analysis, we found that Huangqi Guizhi Wuwu Decoction can down-regulate the levels of TNF-α, IL-1β, and IL-6 in the serum of chronic CINP rats induced by oxaliplatin. It also suppresses the expression of MAPK pathway related-proteins ERK1/2, p38, and JNK. This results in a decrease in the expression of downstream essential proteins, c-Fos, CREB, and Nf-κB. CONCLUSIONS In conclusion, we found that Huangqi Guizhi Wuwu Decoction can combat nerve cell injury, reduce pain sensitization, and prevent and repair the damage of nerve cells in the oxaliplatin CINP model rats via TNFα/IL-1β/IL-6/MAPK/NF-kB pathway.
Collapse
|
11
|
Abdallah NM, Bakeer AH. A Multimodal Analgesic Protocol with Gabapentin-dexmedetomidine for Post-operative Pain Management after Modified Radical Mastectomy Surgery: A Randomized Placebo-Controlled Study. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background and Aim: Modified radical mastectomy (MRM) is accompanied by severe acute postoperative pain. This study evaluated the safety and efficacy of oral gabapentin plus dexmedetomidine infusion as an analgesic multimodal protocol in patients undergoing MRM.
Methods: This prospective randomized, double-blind placebo-controlled study included 30 females scheduled for MRM from June 2021 to December 2021. They were randomly divided into two groups. GD Group (n=15) received oral gabapentin 400 mg and IV infusion of Dexmedetomidine 0.4 µg/kg/h over 10 min after a bolus of 0.5 µg/kg before induction of general anesthesia. Placebo Group (n=15) received a placebo capsule and saline infusion identical to the GD Group. The primary outcome measure was total morphine consumption, and secondary outcomes were pain and sedation scores and intraoperative fentanyl consumption.
Results: Pain score was significantly lower in the GD Group than the Placebo group, starting immediately postoperative up to 24 hours except after 18 hours. The total intraoperative fentanyl consumption and postoperative morphine consumption were significantly lower in the GD Group. The sedation score was significantly higher in the GD Group compared to the Placebo group immediately postoperative and after 2 hours. The heart rate and mean arterial pressure were within the clinically accepted ranges intra- and postoperatively in the two groups.
Conclusion: Preemptive oral gabapentin plus dexmedetomidine IV infusion is a safe and effective analgesic alternative for patients undergoing MRM.
Collapse
|
12
|
Effect of intratracheal dexmedetomidine combined with ropivacaine on postoperative sore throat: a prospective randomised double-blinded controlled trial. BMC Anesthesiol 2022; 22:144. [PMID: 35568822 PMCID: PMC9107121 DOI: 10.1186/s12871-022-01694-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 05/11/2022] [Indexed: 11/10/2022] Open
Abstract
Background The present study aimed to investigate whether intratracheal dexmedetomidine combined with ropivacaine reduces the severity and incidence of postoperative sore throat after tracheal intubation under general anaesthesia. Methods Two hundred patients with American Society of Anaesthesiologists physical status I-II who were subjected to general anaesthesia were randomly divided into four groups, namely, Group D, Group R, Group DR and Group S; these groups received intratracheal dexmedetomidine (1 µg/kg), 0.8% ropivacaine (40 mg), dexmedetomidine (1 µg/kg) combined with 0.8% ropivacaine (40 mg) and normal saline before endotracheal intubation, respectively. The primary outcomes were the incidence and grade of sore throat and hoarseness at 2 h and 24 h after surgery. Moreover, the modified Observer's Assessment of Alertness/Sedation Scale results were recorded at each time point. The secondary outcomes were intraoperative haemodynamic fluctuations, intraoperative anaesthetic drug requirements, and adverse reactions during and after surgery. The patients’ vital signs before induction, before superficial anaesthesia, after superficial anaesthesia, before intubation, after intubation, and 1 min after intubation were recorded. The use of anaesthetic drugs and occurrence of adverse effects were also recorded. Results The incidence and severity of sore throat were significantly lower in Group DR than in the other three groups 2 h after the operation, but they were only significantly lower in Group DR than in the control group 24 h after the operation. Moreover, compared with Group S and Group D, Group DR exhibited more stable haemodynamics during intubation. The doses of remifentanil and propofol were significantly lower in Group DR than in the other groups. Conclusion The combined use of dexmedetomidine and ropivacaine for surface anaesthesia before intubation significantly reduced the incidence and severity of postoperative sore throat. This treatment also decreased anaesthetic drug requirements and intraoperative haemodynamic fluctuations and caused no adverse effects. Trial registration This clinical research was registered at the Chinese Clinical Trial Registry (ChiCTR1900022907, Registration date 30/04/2019).
Collapse
|
13
|
Liu Y, Kuai S, Ding M, Wang Z, Zhao L, Zhao P. Dexmedetomidine and Ketamine Attenuated Neuropathic Pain Related Behaviors via STING Pathway to Induce ER-Phagy. Front Synaptic Neurosci 2022; 14:891803. [PMID: 35645765 PMCID: PMC9136071 DOI: 10.3389/fnsyn.2022.891803] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Our previous work indicated that ER-phagy level had altered in spinal nerve ligation (SNL) rats. In this study, we investigated whether dexmedetomidine or ketamine exhibits anti-anxiety or anti-nociceptive effects via modulation of the spinal STING/TBK pathway to alter ER-phagy in SNL rats. We evaluated the analgesic and anti-anxiety effects of ketamine and dexmedetomidine in SNL rats. 2’3’-cGAMP (a STING pathway agonist) was administrated to investigate whether enhanced spinal STING pathway activation could inhibit dexmedetomidine or ketamine treatment effects in SNL rats. Analgesic effects were assessed with the mechanical withdrawal threshold (MWT) and anti-anxiety effects were measured via an open field test (OFT). Protein expression levels were evaluated by immunoblotting. Distribution and cellular localization of Grp78 (ER stress marker) were evaluated by confocal immunofluorescence. SNL induced mechanical hypersensitivity and anxiety in rats; dexmedetomidine and ketamine both provided analgesia and anti-anxiety effects in SNL rats. Furthermore, the STING pathway was involved in the modulation of ER stress and ER-phagy in SNL rats and dexmedetomidine and ketamine alleviated ER stress by inhibiting STING pathway to enhance ER-phagy. Thus, both ketamine and dexmedetomidine provided anti-anxiety and anti-nociceptive effects by alleviating ER stress through the inhibition of the STING/TBK pathway to modulate spinal ER-phagy in SNL rats.
Collapse
Affiliation(s)
- Yongda Liu
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shihui Kuai
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mengmeng Ding
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhibin Wang
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Limei Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Limei Zhao Ping Zhao
| | - Ping Zhao
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Limei Zhao Ping Zhao
| |
Collapse
|
14
|
Qiu Y, Tang Z. Dexmedetomidine Attenuates LPS-Induced Acute Lung Injury in Rats by Activating the Nrf2/ARE Pathway. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:4185195. [PMID: 35449859 PMCID: PMC9017427 DOI: 10.1155/2022/4185195] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022]
Abstract
Background To investigate the effect of dexmedetomidine (Dex) on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in rats and its mechanism. Methods Eighteen SD rats were randomly divided into 3 groups (6 rats in each group): control group (intratracheal instillation of saline), ALI group (intratracheal instillation of 5 mg/kg LPS), and ALI-Dex group (tail vein injection of 50 μg/kg/h Dex + intratracheal instillation of LPS). Subsequently, the water content of lung tissues was assessed using the wet-dry (W/D) ratio and the histopathological changes of lung tissues using H&E staining. Further activities of ROS, SOD, and GSH-Px in lung tissues of rats were measured by an automatic biochemistry analyzer. ELISA was performed to detect TNF-α, IL-1β, and IL-6 expression in alveolar lavage fluid (BALF) and Western blot to detect the expression of Nrf2/ARE pathway-related proteins. Results After Dex treatment, a reduction in water content in lung tissue and an improvement of lung injury were found in the ALI rats. Compared with the ALI group, rats in the ALI-Dex group had decreased ROS activity and increased activities of SOD and GSH-Px in lung tissues. Dex-treated rats were also associated with a decrease in TNF-α, IL-1β, and IL-6 expression in alveolar lavage fluid (BALF). Additionally, increased expression levels of HO-1 and NQO1 in lung tissues and elevated expression of Nrf2 in the nucleus were shown in the ALI-Dex group compared with the ALI group. Conclusion Dex alleviates LPS-induced ALI by activating the Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Yuandong Qiu
- Department of Anesthesiology, Koiqeung Memorial Hospital, Guangzhou, Guangdong, China
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510900, China
| | - Zhiwei Tang
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510900, China
| |
Collapse
|
15
|
Esu KD, Bakare AO, Owoyele BV. Effects of co-administration of vitamin E and lithium chloride on chronic constriction injury-induced neuropathy in male Wistar rats: Focus on antioxidant and anti-inflammatory mechanisms. Pain Pract 2022; 22:148-158. [PMID: 34351685 DOI: 10.1111/papr.13064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/21/2021] [Accepted: 07/29/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES This study investigated the antinociceptive effects of co-administration of lithium chloride (LiCl) and vitamin E (Vit E) on chronic constriction injury (CCI)-induced peripheral neuropathy in male Wistar rats. It further explored the anti-inflammatory and neuroprotective properties of LiCl and Vit E, which may be complementary to the antinociceptive effects of the two substances. METHODS Thirty-six male Wistar rats, 190.00 ± 10.00 g of body weight were randomly assigned to 6 experimental groups and administered with normal saline, Vit E, LiCl, or their combination, once daily for 21 days. CCI was used to induce neuropathic pain (NP) and mechanical allodynia was assessed using von Frey filaments and pinprick test. Open field maze (OFM) was used to assess the exploratory behavior. Biochemical parameters were assessed in the dorsal root ganglion after 21 days of treatment. RESULTS Mechanical allodynia was developed in rats following CCI. Co-administration of LiCl and Vit E synergistically reduced mechanical hyperalgesia in rats which were significantly different compared with the single administration of either Vit E or LiCl. Combined doses of Vit E and LiCl significantly increases the explorative behavior in the OFM. CCI increased malondialdehyde (MDA), tumor necrotic factor-alpha (TNF-α), calcitonin gene-related polypeptide, calcium ion (Ca2+ ), and reduced superoxide dismutase (SOD) activities. Co-administration of LiCl and Vit E significantly reduced MDA, TNF-α, but increased SOD compared with ligated control. DISCUSSION The findings revealed that the synergistic effects of the co-administration of Vit E and LiCl in ameliorating NP are mediated by their anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
- Kingsley Dominic Esu
- Neuroscience and Inflammation Unit, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| | - Ahmed Olalekan Bakare
- Neuroscience and Inflammation Unit, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| | - Bamidele Victor Owoyele
- Neuroscience and Inflammation Unit, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
16
|
Naveed M, Ullah R, Khan A, Shal B, Khan AU, Khan SZ, Rehman ZU, Khan S. Anti-neuropathic pain activity of a cationic palladium (II) dithiocarbamate by suppressing the inflammatory mediators in paclitaxel-induced neuropathic pain model. Mol Biol Rep 2021; 48:7647-7656. [PMID: 34734371 DOI: 10.1007/s11033-021-06754-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neuropathic pain is a chronic pain state that negatively impacts the quality of life. Currently, available therapies for the treatment of neuropathic pain often lack efficacy and tolerability. Therefore, the search for novel drugs is crucial to obtain treatments that effectively suppress neuropathic pain. OBJECTIVES The present study was undertaken to investigate the antinociceptive properties of (1,4-bis-(diphenylphosphino) butane) palladium (II) chloride monohydrate (Compound 1) in a paclitaxel (PTX)-induced neuropathic pain model. METHODS Initially, behavioral tests such as mechanical and cold allodynia as well as thermal and tail immersion hyperalgesia were performed to investigate the antinociceptive potential of Compound 1 (5 and 10 mg/kg, b.w). RT-PCR was performed to determine the effect of Compound 1 on the mRNA expression level of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and proinflammatory cytokines such as tumor necrosis factor-alpha (TNF)-α, interleukin (IL)-1β, and IL-6. In addition, antioxidant protein, nitric oxide (NO), and malondialdehyde (MDA) levels were also determined. RESULTS The results demonstrated that once-daily dosing of Compound 1 significantly suppressed the PTX-induced behavioral pain responses dose-dependently. The mRNA gene expressions of iNOS, COX-2, and inflammatory cytokines were markedly reduced by Compound 1. Furthermore, it enhanced the level of antioxidant enzymes and lowered the level of MDA and NO production. CONCLUSION These findings suggest that the antinociceptive potential of Compound 1 in the PTX-induced neuropathic pain model is via suppression of oxidative stress and inflammation. Thus, Compound 1 might be a potential candidate for the therapeutic management of PTX induced neuropathic pain.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Rahim Ullah
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Adnan Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Bushra Shal
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Ashraf Ullah Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Shahan Zeb Khan
- Department of Chemistry, Quaid-I-Azam University, Islamabad, 45320, Pakistan
- Department of Chemistry, University of Science and Technology, KPK, Bannu, 28100, Pakistan
| | - Zia Ur Rehman
- Department of Chemistry, Quaid-I-Azam University, Islamabad, 45320, Pakistan.
| | - Salman Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan.
| |
Collapse
|
17
|
Liao J, Zhang F, Qing W, Yu R, Hu Z. Mechanism of Incisional Pain: Novel Finding on Long Noncoding RNA XIST/miR-340-5p/RAB1A Axis. ASN Neuro 2021; 13:17590914211049056. [PMID: 34806436 PMCID: PMC8613904 DOI: 10.1177/17590914211049056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The objective of this study is to investigate the effect of long noncoding RNA (lncRNA) XIST on postoperative pain and inflammation of plantar incision pain (PIP) in rats and its underlying mechanisms. PIP rat models were established by plantar incision. Rats in the sham group were subjected to povidone-iodine scrubbing, and no incision was made. To explore the role of XIST/miR-340-5p/RAB1A in postoperative pain and inflammation, PIP rats were separately or simultaneously injected with lentivirus containing sh-NC, sh-XIST, mimic NC, miR-340-5p mimic, inhibitor NC, miR-340-5p inhibitor, pcDNA3.1, or pcDNA3.1-RAB1A through an intrathecal catheter. The paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) values of rats in each group were assessed to evaluate the pain behavior. RT-qPCR and Western blot were utilized to determine the levels of XIST, miR-340-5p, RAB1A, and NF-κB pathway-related proteins (p-IκBα, IκBα, p-p65, and p65). The concentrations of inflammatory cytokines (TNF-α, IL-1β, and IL-6) in rat spinal dorsal horn tissues were inspected by ELISA. H and E staining was applied to observe the pathological changes of neurons in the spinal dorsal horn, TUNEL staining to detect neuronal apoptosis, and immunohistochemistry to measure RAB1A level. Plantar incision surgery caused decreased PWT and PWL values, enhanced levels of XIST, RAB1A, and inflammatory cytokines, along with an increased proportion of apoptotic neurons. The pain sensitivity and inflammation of rats were motivated after plantar incision surgery. Intrathecal injection of sh-XIST or miR-340-5p mimic ameliorated the pain and inflammation of PIP rats, while silencing of miR-340-5p or overexpression of RAB1A partly reversed the effect of sh-XIST on PIP rats. XIST targeted miR-340-5p and miR-340-5p negatively regulated RAB1A. The XIST/miR-340-5p/RAB1A axis activated the NF-κB signaling pathway. LncRNA XIST aggravates inflammatory response and postoperative pain of PIP rats by activating the NF-κB pathway via the miR-340-5p/RAB1A axis.
Collapse
Affiliation(s)
- Juan Liao
- Department of Anesthesiology, the Third Xiangya Hospital, 504354Central South University, Changsha, Hunan 410013, P.R. China
| | - Fan Zhang
- Department of Anesthesiology, the Third Xiangya Hospital, 504354Central South University, Changsha, Hunan 410013, P.R. China
| | - Wenxiang Qing
- Department of Anesthesiology, the Third Xiangya Hospital, 504354Central South University, Changsha, Hunan 410013, P.R. China
| | - Rili Yu
- Department of Anesthesiology, the Third Xiangya Hospital, 504354Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhonghua Hu
- Department of Anesthesiology, the Third Xiangya Hospital, 504354Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
18
|
Shan W, Liao X, Tang Y, Liu J. Dexmedetomidine alleviates inflammation in neuropathic pain by suppressing NLRP3 via Nrf2 activation. Exp Ther Med 2021; 22:1046. [PMID: 34434260 PMCID: PMC8353619 DOI: 10.3892/etm.2021.10479] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
The aim of the present study was to investigate the mechanism by which dexmedetomidine (DEX) alleviates neuropathic pain in a chronic constriction injury (CCI) model in rats. A CCI rat model was established through sciatic nerve ligation. CCI rats were treated with DEX, the nuclear factor erythroid 2-related factor 2 (Nrf2) inhibitor ML385, the NLR family pyrin domain containing 3 (NLRP3) antagonist MCC950 and/or the NLRP3 activator nigericin. The mechanical withdrawal threshold (MWT) was measured to assess the pain sensitivity of CCI rats. Hematoxylin and eosin staining and TUNEL staining were used to examine spinal injury and apoptosis, respectively. ELISA was used to quantify the levels of inflammatory factors. The expression levels of Nrf2 and NLRP3 were also examined. The results indicated that a decrease in MWT and increases in spinal cord injury, apoptosis and inflammatory factors were detected in CCI rats compared with control rats. Spinal inflammation was abrogated in DEX-treated CCI rats. Compared with the model group, an increase in MWT and decreases in spinal cord injury, apoptosis and inflammatory factors were detected in rats treated with MCC950, while the opposite effects were observed in rats treated with nigericin. The opposite effects on these indicators were observed in the DEX + ML385 and MCC950 + ML385 groups compared with the DEX and MCC950 groups, respectively. MWT was increased, while spinal cord injury, apoptosis and inflammation decreased in the nigericin + DEX group compared with the nigericin group. In summary, the results of the present study indicated that DEX reduced neuropathic pain in CCI rats by suppressing NLRP3 through Nrf2 activation.
Collapse
Affiliation(s)
- Wenyan Shan
- Department of Anesthesiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, P.R. China.,Clinical Research Center for Anesthesiology of ERAS in Hunan Province, Changsha, Hunan 410005, P.R. China
| | - Xiaoyun Liao
- Department of Anesthesiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, P.R. China.,Clinical Research Center for Anesthesiology of ERAS in Hunan Province, Changsha, Hunan 410005, P.R. China
| | - Yixun Tang
- Department of Anesthesiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, P.R. China.,Clinical Research Center for Anesthesiology of ERAS in Hunan Province, Changsha, Hunan 410005, P.R. China
| | - Jitong Liu
- Department of Anesthesiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, P.R. China.,Clinical Research Center for Anesthesiology of ERAS in Hunan Province, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
19
|
Wang X, Liu Q. Dexmedetomidine relieved neuropathic pain and inflammation response induced by CCI through HMGB1/TLR4/NF-κB signal pathway. Biol Pharm Bull 2021:b21-00329. [PMID: 34421084 DOI: 10.1248/bpb.b21-00329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuropathic pain is one of the most intractable diseases. The lack of effective therapy measures remains a critical problem due to the poor understanding of the cause of neuropathic pain. The aim of this study was to investigate the effect of dexmedetomidine (Dex) in trigeminal neuropathic pain and the underlying molecular mechanism in order to identify possible therapeutic targets. We used a chronic constriction injury (CCI) model of mice to investigate whether Dex prevents neuropathic pain and the inflammation response. The α 2-adrenoceptors (α2AR) inhibitor BRL44408 and adenovirus for knocking down High mobility group box 1 (HMGB1) was administrated to confirm whether Dex exert its effect through targeting α2AR and HMGB1. The results indicated that Dex significantly inhibited CCI induced neuropathic pain through targeting α2AR and HMGB1. Dex inhibited the inflammatory response through decreasing the release and the mRNA expression of IL-1β, IL-6, and TNF-ɑ while increasing that of IL-10. Moreover, Dex participates in the regulation of HMGB1, Toll-like receptor 4 (TLR4), NFκb (p-65) expression and the phosphorylation of IκB-ɑ. In conclusion, Dex could relieve neuropathic pain through α2AR and HMGB1 and attenuate inflammation response.
Collapse
Affiliation(s)
- Xin Wang
- Department of Anesthesiology, Affiliated Hospital of traditional Chinese medicine, Southwest Medical University
| | - Qing Liu
- Department of Anesthesiology, Affiliated Hospital of traditional Chinese medicine, Southwest Medical University
| |
Collapse
|
20
|
Bakare AO, Owoyele BV. Bromelain reduced pro-inflammatory mediators as a common pathway that mediate antinociceptive and anti-anxiety effects in sciatic nerve ligated Wistar rats. Sci Rep 2021; 11:289. [PMID: 33432004 PMCID: PMC7801445 DOI: 10.1038/s41598-020-79421-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/08/2020] [Indexed: 02/02/2023] Open
Abstract
The involvement of pro-inflammatory mediators complicates the complex mechanism in neuropathic pain (NP). This study investigated the roles of bromelain against pro-inflammatory mediators as a mechanism that underpins its antinociceptive and anti-anxiety effects in the peripheral model of NP. Sixty-four male Wistar rats randomly divided into eight groups, were used for the study. A chronic constriction injury model of peripheral neuropathy was used to induce NP. Tail-immersion and von Frey filaments tests were used to assess hyperalgesia while open field and elevated plus mazes were used to assess anxiety-like behaviour. NF-кB, iNOS, nitrate, and pro-inflammatory cytokines were investigated in the plasma, sciatic nerve, and brain tissues using ELISA, spectrophotometer, and immunohistochemistry techniques after twenty-one days of treatment. Bromelain significantly (p < 0.05) improved the cardinal signs of NP and inhibited anxiety-like behaviours in ligated Wistar rats. It mitigated the increases in cerebral cortex interleukin (IL) -1β, IL-6, and PGE2 levels. Bromelain reduced NF-кB, IL-1β, IL-6, TNF-α, PGE2, and nitrate concentrations as well as the expression of iNOS in the sciatic nerve. Hence, the antinociceptive and anxiolytic effects of bromelain in the sciatic nerve ligation model of NP is in part due to its ability to reduce nitrosative and inflammatory activities.
Collapse
Affiliation(s)
- Ahmed O Bakare
- Department of Physiology, Neuroscience and Inflammation Unit, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Bamidele V Owoyele
- Department of Physiology, Neuroscience and Inflammation Unit, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria.
| |
Collapse
|
21
|
Motaghi E, Ghasemi Pirbalooti M, Bozorgi H, Eslami M, Rashidi M. Safety and Efficacy of Dexmedetomidine in Breast Surgeries: A Systematic Review and Meta-Analysis. J Perianesth Nurs 2020; 36:179-186. [PMID: 33303343 DOI: 10.1016/j.jopan.2020.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/22/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE Pain control during and after breast surgery is still a challenging task. Dexmedetomidine (DEX) is considered as a sedative agent that is widely used perineurally or intravenously as an adjuvant in general anesthesia and critical care medicine practice. The aim of this study is to evaluate the efficacy of perineural DEX and intravenous (IV) DEX and their effects on postoperative complications in breast surgeries. DESIGN Systematic review and meta-analysis. METHODS The present study systematically reviewed all identified randomized controlled trials for efficacy and safety of IV and perineural use of DEX in breast surgeries. Databases were searched for articles published before October 2019. FINDINGS Twelve trials were identified including 803 patients undergoing breast surgery. Although administration of IV DEX and its use with pectoral nerve (Pecs) block significantly postponed time for first analgesic request and decreased pain score at 1 and 12 hours after surgery, paravertebral use of DEX had no statistically significant effect. Pooled data about perineural DEX showed no significant effect on postoperative nausea and vomiting (PONV), whereas IV DEX significantly reduced PONV. Pooled analysis also showed that DEX administration did not significantly affect postoperative complications, such as postoperative itching, bradycardia, and pneumothorax in patients undergoing breast surgery. CONCLUSIONS The results showed that unlike paravertebral DEX, both DEX use with Pecs blocks and IV DEX were effective in control of postoperative pain in patients undergoing breast surgeries. Unlike perineural DEX, IV DEX significantly reduced PONV.
Collapse
Affiliation(s)
- Ehsan Motaghi
- Department of Physiology and Pharmacology, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - Hooman Bozorgi
- Department of Pharmacology, Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Mohsen Rashidi
- Faculty of Medicine, Department of Pharmacology, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
22
|
Urits I, Virgen CG, Alattar H, Jung JW, Berger AA, Kassem H, Shehata IM, Elhassan A, Kaye AD, Viswanath O. A Comprehensive Review and Update of the Use of Dexmedetomidine for Regional Blocks. PSYCHOPHARMACOLOGY BULLETIN 2020; 50:121-141. [PMID: 33633422 PMCID: PMC7901136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
PURPOSE OF REVIEW This evidence-based systematic review will focus on the use of dexmedetomidine and its role as adjuvant anesthetics in regional blocks to help better guide physicians in their practice. This review will cover background and mechanism of dexmedetomidine as well as the use in various regional blocks. RECENT FINDINGS Local anesthetics are preferred for nerve blocks over opioids; however, both due come with its own side effects. Local anesthetics may be toxic as they disrupt cell membrane and proteins, but by using adjuvants such as dexmedetomidine, that can prolong sensory and motor blocks can reduce total amount of local anesthetics needed. Dexmedetomidine is an alpha-2-adrenergic agonist used as additive for regional nerve block. It has a relatively low side effect profile and have been researched in various regional blocks (intrathecal, paravertebral, axillary, infraclavicular brachial plexus, interscalene). Dexmedetomidine shows promising results as adjuvant anesthetics in most regional blocks. SUMMARY Many studies have been done and many show promising results for the use of dexmedetomidine in regional blocks. It may significantly increase in duration of sensory and motor blocks that correlates with lower pain scores and less need of morphine in various regional blocks.
Collapse
Affiliation(s)
- Ivan Urits
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Celina Guadalupe Virgen
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Hamed Alattar
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Jai Won Jung
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Amnon A Berger
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Hisham Kassem
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Islam Mohammad Shehata
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Amir Elhassan
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Alan D Kaye
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| | - Omar Viswanath
- Urits, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA. Berger, MD, PhD, Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA. Kaye, MD, PhD, Departments of Anesthesiology and Pharmacology, Toxicology and Neurosciences, Louisiana State University School of Medicine, Shreveport, LA. Virgen, BS, Alattar, BS, University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ. Jung, BS, Georgetown University School of Medicine, MedStar Georgetown University Hospital, Washington, DC. Kassem, MD, Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL. Shehata, MD, Ain Shams University, Department of Anesthesiology, Cairo, Egypt. Elhassan, MD, Desert Regional Medical Center, Department of Anesthesiology, Palm Springs, CA. Viswanath, MD, Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE Department of Anesthesiology, Louisiana State University School of Medicine, Shreveport, LA; Valley Pain Consultants - Envision Physician Services, Phoenix, AZ; University of Arizona College of Medicine-Phoenix, Department of Anesthesiology, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE
| |
Collapse
|
23
|
1,3,4-Oxadiazole Derivative Attenuates Chronic Constriction Injury Induced Neuropathic Pain: A Computational, Behavioral, and Molecular Approach. Brain Sci 2020; 10:brainsci10100731. [PMID: 33066162 PMCID: PMC7601954 DOI: 10.3390/brainsci10100731] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/21/2022] Open
Abstract
The production and up-regulation of inflammatory mediators are contributing factors for the development and maintenance of neuropathic pain. In the present study, the post-treatment of synthetic 1,3,4 oxadiazole derivative (B3) for its neuroprotective potential in chronic constriction injury-induced neuropathic pain was applied. In-silico studies were carried out through Auto Dock, PyRx, and DSV to obtain the possible binding and interactions of the ligands (B3) with COX-2, IL-6, and iNOS. The sciatic nerve of the anesthetized rat was constricted with sutures 3/0. Treatment with 1,3,4-oxadiazole derivative was started a day after surgery and continued until the 14th day. All behavioral studies were executed on day 0, 3rd, 7th, 10th, and 14th. The sciatic nerve and spinal cord were collected for further molecular analysis. The interactions in the form of hydrogen bonding stabilizes the ligand target complex. B3 showed three hydrogen bonds with IL-6. B3, in addition to correcting paw posture/deformation induced by CCI, attenuates hyperalgesia (p < 0.001) and allodynia (p < 0.001). B3 significantly raised the level of GST and GSH in both the sciatic nerve and spinal cord and reduced the LPO and iNOS (p < 0.001). B3 attenuates the pathological changes induced by nerve injury, which was confirmed by H&E staining and IHC examination. B3 down-regulates the over-expression of the inflammatory mediator IL-6 and hence provides neuroprotective effects in CCI-induced pain. The results demonstrate that B3 possess anti-nociceptive and anti-hyperalgesic effects and thus minimizes pain perception and inflammation. The possible underlying mechanism for the neuroprotective effect of B3 probably may be mediated through IL-6.
Collapse
|
24
|
Effect of PKC/NF- κB on the Regulation of P2X 3 Receptor in Dorsal Root Ganglion in Rats with Sciatic Nerve Injury. Pain Res Manag 2020; 2020:7104392. [PMID: 33014214 PMCID: PMC7519985 DOI: 10.1155/2020/7104392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/01/2020] [Indexed: 11/17/2022]
Abstract
Background Protein kinase C (PKC), nuclear factor-kappa B p65 (NF-κB p65), and P2X3 receptor (P2X3R) play significant roles in the sensitization and transduction of nociceptive signals, which are considered as potential targets for the treatment of neuropathic pain. However, the mechanisms and relationships among them have not been clearly clarified. Methods 80 rats were randomized and divided into 10 groups (n = 8). Sciatic chronic constriction injury (CCI) rats were intrathecally administered with bisindolylmaleimide I (GF109203X), a PKC-selective antagonist once a day, or pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor twice a day. Sham-operated rats were intrathecally administered with saline. Thermal withdrawal latency (TWL) and mechanical withdrawal threshold (MWT) were evaluated in all the groups before CCI operation (baseline) and on the 1st, 3rd, 7th, 10th, and 14th day after CCI operation. Protein levels of p-PKCα, p-NF-κB p65, and P2X3R were analyzed in the CCI ipsilateral L4-6 dorsal root ganglions (DRGs). Results Intrathecal injection of GF109203X or PDTC alleviated the TWL and MWT in the following 2 weeks after CCI surgery. The protein levels of p-PKCα, p-NF-κB p65, and P2X3R in the ipsilateral DRGs significantly increased after CCI operation, which could be partly reversed by intrathecal administration of GF109203X or PDTC. Conclusion The upregulation of p-PKCα, p-NF-κB p65, and P2X3R expression in the DRGs of CCI rats was involved in the occurrence and development of neuropathic pain. Phosphorylated PKCα and phosphorylated NF-κB p65 regulated with each other. Phosphorylated NF-κB p65 and PKCα have a mutual regulation relationship with P2X3R, respectively, while the specific regulatory mechanism needs further research.
Collapse
|
25
|
Mirasheh MH, Zohrehvand MR, Kazemi R, Bahari Z, Bahrami F, Jangravi Z, Graily M. The Analgesic and Anxiolytic Activity of Resveratrol Mediated by Different Sub-Types of α-Adrenoceptors of Anterior Cingulate Cortex Following Neuropathic Pain in Male Rats. ACTA ACUST UNITED AC 2020. [DOI: 10.30699/jambs.28.129.183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
26
|
Zhao E, Bai L, Li S, Li L, Dou Z, Huang Y, Li Y, Lv Y. Dexmedetomidine Alleviates CCI-Induced Neuropathic Pain via Inhibiting HMGB1-Mediated Astrocyte Activation and the TLR4/NF-κB Signaling Pathway in Rats. Neurotox Res 2020; 38:723-732. [PMID: 32607919 DOI: 10.1007/s12640-020-00245-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/21/2020] [Accepted: 06/17/2020] [Indexed: 01/16/2023]
Abstract
To investigate the effects of dexmedetomidine on chronic constriction injury (CCI)-induced neuropathic pain and to further explore its mechanism. A CCI rat model was established and treatment with dexmedetomidine. The paw withdrawal mechanical threshold (PWMT) and paw withdrawal thermal latency (PWTL) were monitored at different time points, and the effects of hematoxylin-eosin staining on the sciatic nerve morphology of rats were observed. Immunohistochemical and immunofluorescence analyses were used to detect the expression of high mobility group box-1 (HMGB1) protein and glial fibrillary acidic protein (GFAP), and protein fluorescence intensity of GFAP in spinal cord tissue, respectively. Moreover, the expression of HMGB1 and Toll-like receptor-4/nuclear factor kappa-B (TLR4/NF-κB) pathway-related proteins were detected by western blot assay. To verify whether dexmedetomidine alleviates CCI-induced neuropathic pain by inhibiting HMGB1-mediated astrocyte activation and the TLR4/NF-κB signaling pathway, the rats were further treated with an HMGB1 activator or antagonist. Dexmedetomidine was found to improve the pathological changes of the sciatic nerve and alleviate pain in the CCI rats. The expression of HMGB1, GFAP, TLR4, TRAF6, MyD88, and p-P65 were greatly downregulated in the spinal cord tissues of the CCI rats. In addition, a further study showed that an HMGB1 activator can reverse the inhibition of neuropathic pain behaviors of dexmedetomidine. Overexpression of HMGB1 downregulated the PWMT and PWTL and enhanced the astrocyte activity and the TLR4/NF-κB signaling pathway in CCI rats. These results indicated that dexmedetomidine can alleviate neuropathic pain in CCI rats by inhibiting HMGB1-mediated astrocyte activation and the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Erxian Zhao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Liying Bai
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Shurong Li
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100000, China
| | - Li Li
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Zhongci Dou
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Yunli Huang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Yan Li
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Yunqi Lv
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan Province, China.
| |
Collapse
|
27
|
Zhao Y, He J, Yu N, Jia C, Wang S. Mechanisms of Dexmedetomidine in Neuropathic Pain. Front Neurosci 2020; 14:330. [PMID: 32431587 PMCID: PMC7214625 DOI: 10.3389/fnins.2020.00330] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/20/2020] [Indexed: 12/23/2022] Open
Abstract
Dexmedetomidin is a new-generation, highly selective α2 adrenergic receptor agonist with a large number of advantages, including its sedative and analgesic properties, its ability to inhibit sympathetic nerves, its reduced anesthetic dosage, its hemodynamic stability, its mild respiratory depression abilities, and its ability to improve postoperative recognition. Its safety and effectiveness, as well as its ability to provide a certain degree of comfort to patients, make it a useful anesthetic adjuvant for a wide range of clinical applications. For example, dexmedetomidine is commonly used in patients undergoing general anesthesia, and it also exerts sedative effects during tracheal intubation or mechanical ventilation in intensive care unit patients. In recent years, with the deepening of clinical research on dexmedetomidine, the drug is still applied in the treatment of spastic pain, myofascial pain, neuropathic pain, complex pain syndrome, and chronic headache, as well as for multimodal analgesia. However, we must note that the appropriateness of patient and dose selection should be given attention when using this drug; furthermore, patients should be observed for adverse reactions such as hypotension and bradycardia. Therefore, the safety and effectiveness of this drug for long-term use remain to be studied. In addition, basic experimental studies have also found that dexmedetomidine can protect important organs, such as the brain, heart, kidney, liver, and lung, through various mechanisms, such as antisympathetic effects, the inhibition of apoptosis and oxidative stress, and a reduction in the inflammatory response. Moreover, the neuroprotective properties of dexmedetomidine have received the most attention from scholars. Hence, in this review, we mainly focus on the characteristics and clinical applications of dexmedetomidine, especially the role of dexmedetomidine in the nervous system and the use of dexmedetomidine in the relief of neuropathic pain.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianshuai He
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ning Yu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changxin Jia
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shilei Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
28
|
Liu J, Huang X, Hu S, He H, Meng Z. Dexmedetomidine attenuates lipopolysaccharide induced acute lung injury in rats by inhibition of caveolin-1 downstream signaling. Biomed Pharmacother 2019; 118:109314. [DOI: 10.1016/j.biopha.2019.109314] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/27/2019] [Accepted: 07/31/2019] [Indexed: 10/26/2022] Open
|
29
|
Tumor necrosis factor receptor 1 inhibition is therapeutic for neuropathic pain in males but not in females. Pain 2019; 160:922-931. [PMID: 30586024 DOI: 10.1097/j.pain.0000000000001470] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor necrosis factor (TNF) is a proinflammatory cytokine, which is involved in physiological and pathological processes and has been found to be crucial for pain development. In the current study, we were interested in the effects of blocking Tumor necrosis factor receptor 1 (TNFR1) signaling on neuropathic pain after peripheral nerve injury with the use of transgenic mice and pharmacological inhibition. We have previously shown that TNFR1 mice failed to develop neuropathic pain and depressive symptoms after chronic constriction injury (CCI). To investigate the therapeutic effects of inhibiting TNFR1 signaling after injury, we delivered a drug that inactivates soluble TNF (XPro1595). Inhibition of solTNF signaling resulted in an accelerated recovery from neuropathic pain in males, but not in females. To begin exploring a mechanism, we investigated changes in N-methyl-D-aspartate (NMDA) receptors because neuropathic pain has been shown to invoke an increase in glutamatergic signaling. In male mice, XPro1595 treatment reduces elevated NMDA receptor levels in the brain after injury, whereas in female mice, NMDA receptor levels decrease after CCI. We further show that estrogen inhibits the therapeutic response of XPro1595 in females. Our results suggest that TNFR1 signaling plays an essential role in pain induction after CCI in males but not in females.
Collapse
|
30
|
Dexmedetomidine relieves neuropathic pain by inhibiting hyperpolarization-activated cyclic nucleotide-gated currents in dorsal root ganglia neurons. Neuroreport 2019; 29:1001-1006. [PMID: 29912028 DOI: 10.1097/wnr.0000000000001068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study was designed to examine the effect and mechanism of dexmedetomidine (Dex) on neuropathic pain (NP). The NP model was established by performing chronic sciatic nerve constriction injury (CCI). Seven days after CCI surgery, the rats were injected intraperitoneally with Dex, ZD7288 (an HCN channel inhibitor), and saline, respectively. The paw withdrawal threshold to mechanical stimulation and the thermal withdrawal latency tests were performed. After administration, the L4, L5 dorsal root ganglia (DRG) neurons of rats were isolated. In addition, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels subtype plasmids were transfected into human embryonic kidney (HEK)293 cells. Whole-cell clamp recordings were used to examine the properties of HCN currents (Ih) expressed in HEK293 cells and DRG neurons. After surgery, the paw withdrawal threshold to mechanical stimulation and thermal withdrawal latency were reduced, the HCN currents (Ih) amplitude of DRG neurons was increased, and the semiactivated voltage (V1/2) value was decreased in CCI rats (P<0.05). CCI rats treated with Dex or ZD7288 had reduced mechanical and thermal hyperalgesia. The Ih amplitude was lower and the V1/2 value was increased in DRG neurons in CCI rats treated with Dex or ZD7288 (P<0.05). In addition, Dex inhibited HCN1 and HCN2 currents in HEK293 cells; caused a decrease in maximal currents, an increase in the inhibition rate of Ih, and a negative shift in V1/2 (P<0.05). Taken together, our finding suggested that Dex alleviates NP and the effect is likely because of the inhibition of HCN currents.
Collapse
|
31
|
Dexmedetomidine protects against lipopolysaccharide-induced early acute kidney injury by inhibiting the iNOS/NO signaling pathway in rats. Nitric Oxide 2019; 85:1-9. [PMID: 30659917 DOI: 10.1016/j.niox.2019.01.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 12/14/2022]
Abstract
Increasing evidence has demonstrated that dexmedetomidine (DEX) possesses multiple pharmacological actions. Herein, we explored the protective effect and potential molecular mechanism of DEX on lipopolysaccharide (LPS)-induced early acute kidney injury (AKI) from the perspective of antioxidant stress. We found that DEX (30 μg/kg, i.p.) ameliorated the renal dysfunction and histopathological damage (tubular necrosis, vacuolar degeneration, infiltration of inflammatory cells and cast formation) induced by LPS (10 mg/kg). DEX also attenuated renal oxidative stress remarkably in LPS-induced early AKI, as evidenced by reduction in production of reactive nitrogen species, decreasing malondialdehyde levels, as well as increasing superoxide dismutase activity and glutathione content. DEX prevented activator protein-1 translocation, inhibited phosphorylation of I-kappa B (IκB) and activation of nuclear factor kappa B (NF-κB) in LPS-induced early AKI, as assessed by real-time quantitative polymerase chain reaction and protein levels of c-Jun, c-Fos, IκB and NF-κB. Notably, DEX pretreatment had the same effect as intraperitoneal injection of an inhibitor of inducible nitric oxide synthase inhibitor (1400W; 15 mg/kg), and inhibited the activity of renal inducible nitric oxide synthase (iNOS) and decreased the expression of iNOS mRNA and NO production. However, the protective effect of DEX on LPS-induced early AKI was reversed by the alpha 2 adrenal receptor (α2-AR) inhibitor atipamezole, whereas the imidazoline receptor inhibitor idazoxan did not. Taken together, DEX protects against LPS-induced early AKI in rats by inhibiting the iNOS/NO signaling pathway, mainly by acting on α2-ARs instead of IRs.
Collapse
|
32
|
Shi J, Jiang K, Li Z. MiR-145 ameliorates neuropathic pain via inhibiting inflammatory responses and mTOR signaling pathway by targeting Akt3 in a rat model. Neurosci Res 2017; 134:10-17. [PMID: 29162479 DOI: 10.1016/j.neures.2017.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 12/19/2022]
Abstract
Neuropathic pain perplexes a large population of patients with various diseases. Inflammation plays a key role in the physiopathology of neuropathic pain. Anti-inflammatory can be a promising strategy to treat neuropathic pain. We generated a chronic constriction injury rat model to mimic neuropathic pain by ligating the left ischiadic nerves of rats. Then we performed intrathecal injection of miR-145 mimics to treat these rats for seven consecutive days. Pain behavior tests including mechanical allodynia and thermal hyperalgesia, pro-inflammatory cytokines including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 were analyzed. Quantitative polymerase chain reaction and immunoblotting were performed to detect the changes of signaling pathway after miR-145 mimic treatment. Targeting of Akt3 by miR-145 was studied by dual-luciferase reporter gene assays. MiR-145 mimics injection significantly mollified both mechanical allodynia and thermal hyperalgesia in rats, and down-regulated secretion of TNF-α, IL-1β and IL-6. We confirmed that miR-145 directly targeted Akt3, inhibiting NF-κB and mTOR downstream genes in rat dorsal root ganglia. MiR-145 can mollify neuropathic pain in a chronic constriction injury rat model by reducing inflammation and ion channel overexpression through Akt3/mTOR and Akt3/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Jinshan Shi
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| | - Ke Jiang
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Zhaoduan Li
- Department of Anesthesiology, Tianjin Nankai Hospital, Tianjin 300100, China
| |
Collapse
|
33
|
Tang C, Xia Z. Dexmedetomidine in perioperative acute pain management: a non-opioid adjuvant analgesic. J Pain Res 2017; 10:1899-1904. [PMID: 28860845 PMCID: PMC5565238 DOI: 10.2147/jpr.s139387] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Many nociceptive, inflammatory, and neuropathic pathways contribute to perioperative pain. Although opioids have long been a mainstay for perioperative analgesia, other non-opioid therapies, and dexmedetomidine, in particular, have been increasingly used as part of a multimodal analgesic regimen to provide improved pain control while minimizing opioid-related side effects. This article reviews the evidence supporting the preoperative, intraoperative, and postoperative efficacy of dexmedetomidine as an adjuvant, and the efficacy of intravenous, spinal canal, and nerve block analgesia with dexmedetomidine for perioperative acute pain treatment. While there have not been any large-scale clinical trials conducted, the current body of evidence suggests that dexmedetomidine is suitable for use as an adjuvant analgesic at all perioperative stages. However, there are potential adverse effects, such as hypotension and bradycardia, which must be taken into consideration by clinicians.
Collapse
Affiliation(s)
- Chaoliang Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|