1
|
Liu Z, Cai H, Wang T, Aleem HB, Liu R, Chen H. Gallic acid protect against spinal cord ischemia-reperfusion injury in rat via activation of Nrf2/HO-1 signaling. Regen Ther 2025; 29:419-426. [PMID: 40292078 PMCID: PMC12022406 DOI: 10.1016/j.reth.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/22/2025] [Accepted: 03/29/2025] [Indexed: 04/30/2025] Open
Abstract
Object This study explores Gallic acid's (GA) neuroprotective effects against spinal cord ischemia-reperfusion injury (SCII) and its underlying mechanisms. Methods Spinal cord ischemia/reperfusion injury was induced in rats using the modified Zivin's method of clamping the abdominal aorta. The Basso-Beattie-Bresnahan scores, the inclined plane test, hematoxylin and eosin (HE) staining, and Nissl staining were used to measure locomotor activity and histological changes in the injured spinal cords. Proinflammatory factors (TNF-α and IL-1β) were examined using an ELISA kit. Moreover, In vitro oxidative stress model was induced by tBHP used to assess the cell survival rate, reactive oxygen species (ROS) levels, Malondialdehyde (MDA) levels. RT-qPCR and Western blot was used to detect the expression levels of mRNA and proteins. Results In vitro, GA inhibited tBHP-induced apoptosis in PC-12 cells, reduced ROS and MDA production, and abolished the expression of pro-apoptotic factors while enhancing the Nrf2/HO-1 signaling pathway. In vivo, GA treatment improved the behavioral and structural aspects of SCII in rats, inhibiting the production of proinflammatory factors, also reduced oxidative stress, and prevented neuronal apoptosis by enhancing the Nrf2/HO-1 signaling pathway. Conclusions GA exhibits neuroprotective effects against SCII, involving antioxidant, anti-inflammatory, and anti-apoptotic activities through Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Zhengqiang Liu
- Department of Spinal Surgery, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, Hubei, 443003, China
- Department of Orthopaedics, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, 332000, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, China
| | - Huili Cai
- Department of Hematology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, Hubei, 443003, China
| | - Tianlun Wang
- College of Medicine and Health Sciences, China Three Gorges University, Yichang 443003, China
| | - Hamza bin Aleem
- Department of Spinal Surgery, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, Hubei, 443003, China
| | - Rui Liu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, 443002, China
| | - Haidan Chen
- Department of Spinal Surgery, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, Hubei, 443003, China
| |
Collapse
|
2
|
Yang H, Zhao Y, Chen Y, Yang T, Dou X, Li J, Yang G, Feng G, Fang H, Fan H, Zhang S. Dexmedetomidine Alleviates Acute Stress-Induced Acute Kidney Injury by Attenuating Inflammation and Oxidative Stress via Inhibiting the P2X 7R/NF-κB/NLRP3 Pathway in Rats. Inflammation 2025; 48:412-425. [PMID: 38896231 DOI: 10.1007/s10753-024-02065-8] [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: 04/25/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
This study aimed to investigate the potential protective effects of Dexmedetomidine (DEX) against acute kidney injury (AKI) induced by acute stress (AS). Wistar rats were divided into five groups: Control, DEX, AS, AS + DEX, and AS + A438079. The results showed that AS led to AKI by increasing inflammatory biomarkers and oxidative stress-related indicators. The acute stress model in rats was successfully established. Renal function, histopathology, oxidative stress, and inflammation were assessed. Localization of P2X7 receptor (P2X7R) was determined by immunofluorescence. Additionally, the key inflammatory proteins of the P2X7R/NF-κB/NLRP3 signaling pathway were measured by Western blotting. DEX significantly improved kidney function, alleviated kidney injury, and reduced oxidative stress and inflammation. DEX inhibited the activation of the P2X7R, decreased the expression of NF-κB, NLRP3 inflammasome, and Caspase-1, and inhibited the expression of interleukin-1β (IL-1β) and tumor necrosis factor α (TNFα). Furthermore, DEX also alleviated AS-induced AKI by inhibiting the excessive production of reactive oxygen species (ROS) and reducing oxidative stress. In conclusion, DEX attenuates AS-induced AKI by mitigating inflammation and oxidative stress through the inhibition of the P2X7R/NF-κB/NLRP3 pathway in rats.
Collapse
Affiliation(s)
- Haotian Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Academy of Agricultural Science Branch of Animal Husbandry and Veterinary Branch, Qiqihar, China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yongping Chen
- College of Veterinary Medicine, Agricultural University, Qingdao, China
| | - Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyi Dou
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Junfeng Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Guiyan Yang
- Department of Pathology and Laboratory Medicine, Davis Health, University of California, Sacramento, CA, USA
| | - Guofeng Feng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hao Fang
- College of Optoelectronic Engineering, Chongqing University, Chongqing, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Shuai Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
3
|
Zhang S, Feng X, Yang G, Tan H, Cheng X, Tang Q, Yang H, Zhao Y, Ding X, Li S, Dou X, Li J, Kang H, Li X, Ji Y, Hou Q, An Q, Fang H, Fan H. Dexmedetomidine ameliorates acute kidney injury by regulating mitochondrial dynamics via the α2-AR/SIRT1/PGC-1α pathway activation in rats. Mol Med 2024; 30:184. [PMID: 39455916 PMCID: PMC11505563 DOI: 10.1186/s10020-024-00964-y] [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: 08/22/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Sepsis-associated acute kidney injury (AKI) is a serious complication of systemic infection with high morbidity and mortality in patients. However, no effective drugs are available for AKI treatment. Dexmedetomidine (DEX) is an alpha 2 adrenal receptor agonist with antioxidant and anti-apoptotic effects. This study aimed to investigate the therapeutic effects of DEX on sepsis-associated AKI and to elucidate the role of mitochondrial dynamics during this process. METHODS A lipopolysaccharide (LPS)-induced AKI rat model and an NRK-52E cell model were used in the study. This study investigated the effects of DEX on sepsis-associated AKI and the molecular mechanisms using histologic assessment, biochemical analyses, ultrastructural observation, western blotting, immunofluorescence, immunohistochemistry, qRT-PCR, flow cytometry, and si-mRNA transfection. RESULTS In rats, the results showed that administration of DEX protected kidney structure and function from LPS-induced septic AKI. In addition, we found that DEX upregulated the α2-AR/SIRT1/PGC-1α pathway, protected mitochondrial structure and function, and decreased oxidative stress and apoptosis compared to the LPS group. In NRK-52E cells, DEX regulated the mitochondrial dynamic balance by preventing intracellular Ca2+ overloading and activating CaMKII. CONCLUSIONS DEX ameliorated septic AKI by reducing oxidative stress and apoptosis in addition to modulating mitochondrial dynamics via upregulation of the α2-AR/SIRT1/PGC-1α pathway. This is a confirmatory study about DEX pre-treatment to ameliorate septic AKI. Our research reveals a novel mechanistic molecular pathway by which DEX provides nephroprotection.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiujing Feng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, China
| | - Guiyan Yang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haoyang Tan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xin Cheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qichao Tang
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, China
| | - Haotian Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xuanpan Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Siyao Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyi Dou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Junfeng Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Huijie Kang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xingxing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yaxin Ji
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qingdian Hou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qiuyue An
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hao Fang
- College of Optoelectronic Engineering, Chongqing University, Chongqing, China
| | - Honggang Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
4
|
Zhou Y, Bai Y, Zhang P, Weng P, Xie W. Propofol alleviates spinal cord ischemia-reperfusion injury by preserving PI3K/AKT/GIT1 axis. J Investig Med 2024; 72:705-714. [PMID: 38715211 DOI: 10.1177/10815589241254044] [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] [Indexed: 06/21/2024]
Abstract
Spinal cord ischemia-reperfusion injury (SCIRI) is a major contributor to neurological damage and mortality associated with spinal cord dysfunction. This study aims to explore the possible mechanism of Propofol and G-protein-coupled receptor-interacting protein 1 (GIT1) in regulating SCIRI in rat models. SCIRI rat models were established and injected with Propofol, over expression of GIT1 (OE-GIT1), or PI3K inhibitor (LY294002). The neurological function was assessed using Tarlov scoring system, and Hematoxylin & Eosin (H&E) staining was applied to observe morphology changes in spinal cord tissues. Cell apoptosis, blood-spinal cord barriers (BSCB) permeability, and inflammatory cytokines were determined by TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, evans blue (EB) staining, and enzyme-linked immuno sorbent assay (ELISA), respectively. Reverse transcription-quantitative polymerase chain reaction and western blot were used to detect the expression levels of GIT1, endothelial nitric oxide synthase (eNOS), PI3K/AKT signal pathway and apoptosis-related proteins. SCIRI rats had decreased expressions of GIT1 and PI3K/AKT-related proteins, whose expressions can be elevated in response to Propofol treatment. LY294002 can also decrease GIT1 expression levels in SCIRI rats. Propofol can attenuate neurological dysfunction induced by SCIRI, decrease spinal cord tissue injury and BSCB permeability in addition to suppressing cell apoptosis and inflammatory cytokines, whereas further treatment by LY294002 can partially reverse the protective effect of Propofol on SCIRI. Propofol can activate PI3K/AKT signal pathway to increase GIT1 expression level, thus attenuating SCIRI in rat models.
Collapse
Affiliation(s)
- Yilin Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, P.R. China
| | - Yuyan Bai
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, P.R. China
| | - Peisen Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, P.R. China
| | - Peiqing Weng
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, P.R. China
| | - Wenxi Xie
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, P.R. China
| |
Collapse
|
5
|
Hafezi B, Kazemi Mehrjerdi H, Moghaddam Jafari A. Effect of captopril on paraplegia caused by spinal cord ischemia-reperfusion injury in rats. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2024; 15:379-385. [PMID: 39257463 PMCID: PMC11383196 DOI: 10.30466/vrf.2024.2019729.4126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/07/2024] [Indexed: 09/12/2024]
Abstract
This study investigated the effect of captopril (Cap) on spinal cord ischemia-reperfusion injury (SCII) in rats. Twenty-four adults male Wistar rats were randomly divided into four groups of six animals each: spinal cord ischemia-reperfusion (SCI-R) with Cap (SCI-R + Cap), SCI-R, sham-operated with Cap (SHAM + Cap), and SHAM. The 24 hr and 90 min before ischemia induction, Cap was administered intragastrically (100 mg kg-1) to the SHAM + Cap and SCI-R + Cap groups. Abdominal aortic clamping was performed in the SCI-R and SCI-R + Cap groups for 40 min. Hindlimb motor function was evaluated using the Tarlov Scale at 4, 6, 12, 24, 48, and 60 hr after SCII. The malondialdehyde (MDA), the ferric-reducing ability of plasma (FRAP) and prooxidant-antioxidant balance (PAB) values were also measured. Throughout the study period, the SCI-R group had significantly lower motor function scores compared to the other groups. The MDA and PAB levels were higher and the FRAP value was lower in the SCI-R group compared to in the SHAM group. The SCI-R + Cap had higher motor function scores compared to the SCI-R group at all time points. There were no significant differences in MDA concentration, FRAP and PAB values between the SCI-R + Cap and SCI-R groups. Captopril may act as a protective agent against SCII in rats based on hind limb motor function assessment.
Collapse
Affiliation(s)
- Bahareh Hafezi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hossein Kazemi Mehrjerdi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Moghaddam Jafari
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
6
|
Wu F, Lin Y, Xiao L, Chen Q, Lin F, Li R. Administration with curcumin alleviates spinal cord ischemia-reperfusion injury by regulating anti-oxidative stress and microglia activation-mediated neuroinflammation via Nrf2/NF-κB axis. In Vitro Cell Dev Biol Anim 2024; 60:172-182. [PMID: 38228998 DOI: 10.1007/s11626-023-00846-3] [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: 08/15/2023] [Accepted: 11/08/2023] [Indexed: 01/18/2024]
Abstract
Spinal cord ischemia-reperfusion injury (SCII) ranks as the common complication after aortic surgery, usually leading to devastating post-operative paraplegia. Microglia over-activation and neuronal cell loss are key pathological features of SCII. Curcumin is involved in several I/R injuries. However, its underlying mechanism in SCII remains elusive. Here, curcumin attenuated oxygen and glucose deprivation/reoxygenation (OGD/R)-induced oxidative injury in PC12 neuronal cells by increasing cell viability, inhibiting cell apoptosis, lactate dehydrogenase, malondialdehyde levels, but elevating anti-oxidative superoxide dismutase and glutathione peroxidase levels. Furthermore, curcumin restrained OGD/R-evoked microglia M1 activation by decreasing microglia M1 polarization marker IBA-1 and iNOS transcripts. Moreover, the increased inflammatory cytokine levels of TNF-α and IL-6 in microglia under OGD/R conditions were suppressed after curcumin treatment. Importantly, neuronal cells incubated with a conditioned medium from OGD/R-treated microglia exhibited lower cell viability and higher apoptotic ratio, which were overturned when microglia were treated with curcumin. Intriguingly, curcumin could inhibit the activation of the NF-κB pathway by Nrf2 enhancement in OGD/R-treated PC12 cells and microglia. Notably, targeting Nrf2 signaling reversed the protective efficacy of curcumin against OGD/R-evoked oxidative insult in neuronal, microglia M1 activation, inflammatory response, and microglial activation-evoked neuronal death. In vivo, curcumin improved histopathologic injury and neurologic motor function in SCII rats and attenuated oxidative stress, microglia activation and neuroinflammation in spinal cord tissues, and activation of the Nrf2/NF-κB pathway. Thus, curcumin may alleviate SCII by mitigating I/R-evoked oxidative injury in neuron and microglia activation-induced neuroinflammation and neuron death through Nrf2/NF-κB signaling, supporting a promising therapeutic agent for SCII.
Collapse
Affiliation(s)
- Fengchun Wu
- Department of Orthopaedics, Third Clinical College, Fujian Medical University, Fuzhou, 350007, People's Republic of China
- Department of Orthopaedics, Fuzhou Second Hospital, Fuzhou, 350007, People's Republic of China
| | - Yu Lin
- Department of Orthopaedics, Third Clinical College, Fujian Medical University, Fuzhou, 350007, People's Republic of China
- Department of Orthopaedics, Fuzhou Second Hospital, Fuzhou, 350007, People's Republic of China
| | - Lili Xiao
- Department of Orthopaedics, Third Clinical College, Fujian Medical University, Fuzhou, 350007, People's Republic of China
- Department of Orthopaedics, Fuzhou Second Hospital, Fuzhou, 350007, People's Republic of China
| | - Qiyong Chen
- Department of Orthopaedics, Third Clinical College, Fujian Medical University, Fuzhou, 350007, People's Republic of China
| | - Fengfei Lin
- Department of Orthopaedics, Third Clinical College, Fujian Medical University, Fuzhou, 350007, People's Republic of China
- Department of Orthopaedics, Fuzhou Second Hospital, Fuzhou, 350007, People's Republic of China
| | - Renbin Li
- Department of Orthopaedics, Third Clinical College, Fujian Medical University, Fuzhou, 350007, People's Republic of China.
- Department of Orthopaedics, Fuzhou Second Hospital, Fuzhou, 350007, People's Republic of China.
| |
Collapse
|
7
|
Zhang S, Cui K, Li Y, Fan Y, Wang D, Yao X, Fang B. The m 6A methylation and expression profiles of mouse neural stem cells after hypoxia/reoxygenation. Stem Cell Res Ther 2024; 15:43. [PMID: 38360659 PMCID: PMC10870567 DOI: 10.1186/s13287-024-03658-8] [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: 12/29/2022] [Accepted: 02/07/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Ischemia-reperfusion injury to the central nervous system often causes severe complications. The activation of endogenous neural stem cells (NSCs) is considered a promising therapeutic strategy for nerve repair. However, the specific biological processes and molecular mechanisms of NSC activation remain unclear, and the role of N6-methyladenosine (m6A) methylation modification in this process has not been explored. METHODS NSCs were subjected to hypoxia/reoxygenation (H/R) to simulate ischemia-reperfusion in vivo. m6A RNA methylation quantitative kit was used to measure the total RNA m6A methylation level. Quantitative real-time PCR was used to detect methyltransferase and demethylase mRNA expression levels. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were conducted for NSCs in control and H/R groups, and the sequencing results were analyzed using bioinformatics. Finally, the migration ability of NSCs was identified by wound healing assays, and the proliferative capacity of NSCs was assessed using the cell counting kit-8, EdU assays and cell spheroidization assays. RESULTS Overall of m6A modification level and Mettl14 mRNA expression increased in NSCs after H/R treatment. The m6A methylation and expression profiles of mRNAs in NSCs after H/R are described for the first time. Through the joint analysis of MeRIP-seq and RNA-seq results, we verified the proliferation of NSCs after H/R, which was regulated by m6A methylation modification. Seven hub genes were identified to play key roles in the regulatory process. Knockdown of Mettl14 significantly inhibited the proliferation of NSCs. In addition, separate analysis of the MeRIP-seq results suggested that m6A methylation regulates cell migration and differentiation in ways other than affecting mRNA expression. Subsequent experiments confirmed the migration ability of NSCs was suppressed by knockdown of Mettl14. CONCLUSION The biological behaviors of NSCs after H/R are closely related to m6A methylation of mRNAs, and Mettl14 was confirmed to be involved in cell proliferation and migration.
Collapse
Affiliation(s)
- Shaoqiong Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Kaile Cui
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yiting Fan
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Dongxu Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Xingen Yao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
8
|
Guo L, Zhang D, Ren X, Liu D. SYVN1 attenuates ferroptosis and alleviates spinal cord ischemia-reperfusion injury in rats by regulating the HMGB1/NRF2/HO-1 axis. Int Immunopharmacol 2023; 123:110802. [PMID: 37591122 DOI: 10.1016/j.intimp.2023.110802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 07/23/2023] [Accepted: 08/11/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND The ferroptosis of neurons is an important pathological mechanism of spinal cord ischemia reperfusion injury (SCIRI). Previous studies showed that synoviolin 1 (SYVN1) is a good prognostic marker of neurodegenerative diseases, but its mechanism is still unclear. This study aims to explore the role of SYVN1 in the ferroptosis of neurons and to clarify its internal mechanism. METHODS Rat primary spinal cord neurons were treated with oxygen-glucose deprivation (OGD) for 1, 4 or 8 h, and then cell viability, ROS and MDA levels, glutathione peroxidase (GSH-Px) activity, and the expression of ferroptosis-related proteins GPX4, FTH1 and PTGS2 were detected. OGD/R-induced neurons were transfected with pcDNA-SYVN1 or si-HMGB1, and then cell functions were detected. Transmission electron microscope (TEM) was used to detect cell ferroptosis. The interplay between SYVN1 and high mobility group box 1 (HMGB1) was confirmed with Co-immunoprecipitation (Co-IP) assay. The stability of HMGB1 was measured by ubiquitination assay. Also, cells were treated with pcDNA-SYVN1 or together with ubiquitination inhibitor MG132, as well as treated with pcDNA-SYVN1 and pcDNA-HMGB1 or together with NRF2 activator dimethyl fumarate (DMF), and then Western blotting was used to detect the expression of HMGB1, nuclear NRF2 and HO-1 proteins. In addition, SD rats were occluded left common carotid artery and aortic arch to establish a SCIRI rat model. And rats were injected intrathecal with adenovirus-mediated SYVN1 overexpression vector (Ad-SYVN1, 2 μL, virus titer 5 × 1013 transduction unit [TU]/mL) to overexpress SYVN1. The motion function of rats was quantified using the Basso Rat Scale (BMS) for Locomotion. The ferroptosis and the number of neurons in the spinal cord tissue of rats were detected. RESULTS SYVN1 overexpression inhibited ferroptosis of SCIRI rats and OGD/R-treated primary spinal cord neurons, and down-regulated the expression of HMGB1. In terms of mechanism, the binding of SYVN1 and HMGB1 promoted the ubiquitination and degradation of HMGB1, and negatively regulated the expression of HMGB1. Moreover, under OGD/R conditions, MG132 treatment or HMGB1 overexpression eliminated the inhibitory effect of SYVN1 overexpression on the ferroptosis of neurons and the activation of the NRF2/HO-1 pathway, and DMF treatment abolished the inhibition of HMGB1 overexpression on the NRF2/HO-1 pathway. Finally, in vivo experiments showed that SYVN1 overexpression could alleviate the spinal cord ischemia-reperfusion injury in rats by down-regulating HMGB1 and promoting the activation of the NRF2/HO-1 pathway. CONCLUSION SYVN1 regulates ferroptosis through the HMGB1/NRF2/HO-1 axis to prevent spinal cord ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Lili Guo
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Dong Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Xiaoyan Ren
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Dingsheng Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China.
| |
Collapse
|
9
|
Yuan L, Xu J, Qin F, Yi X, Zhong Z, Gu X, Gong G. USP18 overexpression protects against spinal cord ischemia/reperfusion injury via regulating autophagy. Neurosci Lett 2023; 810:137359. [PMID: 37356565 DOI: 10.1016/j.neulet.2023.137359] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND Spinal cord ischemia-reperfusion injury (SCII) is usually caused by spinal surgery, often leading to severe neurological deficits. The ubiquitin-specific protease 18 (USP18) plays a significant role in neurological diseases. OBJECTIVE The present study was designed to assess the effects and mechanisms of USP18 on SCII. METHODS By inducing transient aortic occlusion and subsequent reperfusion, a rat model of SCII was successfully established. The Basso-Beattie-Bresnahan scores, the inclined plane test, and hematoxylin and eosin (HE) were used to measure locomotor activity and histological changes in the injured spinal cords. Moreover, the SCII cell model was established using PC12 cells under oxygen-glucose deprivation and reoxygenation (OGD/R). Proinflammatory factors (TNF-α, IL-6, and INF-α) were examined using an ELISA kit. Cell apoptosis was assessed by Annexin V-FITC/PI double-staining and TUNEL assays. Western blot was used to detect the expression levels of proteins related to apoptosis and autophagy. RESULTS USP18 expression was decreasedin vivo and in vitro SCII models. The upregulation of USP18 ameliorated hind limbs' motor function, inhibiting inflammation and apoptosis after SCII in rats. USP18 overexpression in vitro may protect PC12 cells from OGD/R-induced damage by modulating inflammatory responses and apoptosis. Moreover, Overexpression of USP18 enhanced autophagy to inhibit cell apoptosis induced by SCII in vivo and in vitro. CONCLUSIONS In summary, USP18 overexpression protects against SCII via regulating autophagy.
Collapse
Affiliation(s)
- Libang Yuan
- Department of Anesthesia, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China
| | - Jin Xu
- School of Electrical Engineering and Information, Southwest Petroleum University, Chengdu, Sichuan 610083, China
| | - Fuen Qin
- Department of Anesthesia, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China
| | - Xiaobo Yi
- Department of Anesthesia, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China
| | - Zuling Zhong
- Department of Anesthesia, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China
| | - Xiaoping Gu
- Department of Anesthesia, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China
| | - Gu Gong
- Department of Anesthesia, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| |
Collapse
|
10
|
Mercantepe F, Tumkaya L, Mercantepe T, Akyildiz K, Ciftel S, Yilmaz A. The Effects of Dexmedetomidine on Abdominal Aortic Occlusion-Induced Ovarian Injury via Oxidative Stress and Apoptosis. Cells Tissues Organs 2023; 212:554-566. [PMID: 37339613 DOI: 10.1159/000531613] [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: 03/28/2023] [Accepted: 06/14/2023] [Indexed: 06/22/2023] Open
Abstract
Ischemia/reperfusion (I/R) induced ovarian damage is caused by various diseases such as ovarian torsion, ovarian transplantation, cardiovascular surgery, sepsis, or intra-abdominal surgery. I/R-related oxidative damage can impair ovarian functions, from oocyte maturation to fertilization. This study investigated the effects of dexmedetomidine (DEX), which has been shown to exhibit antiapoptotic, anti-inflammatory, and antioxidant effects, on ovarian I/R injury. We designed four study groups: group 1 (n = 6): control group; group 2 (n = 6): only DEX group; group 3 (n = 6): I/R group; group 4 (n = 6): I/R + DEX group. Then, ovarian samples were taken and examined histologically and immunohistochemically, and tissue malondialdehyde (MDA) and glutathione (GSH) levels were measured. In the I/R group MDA levels, caspase-3, NF-κB/p65, 8-OHdG positivity, and follicular degeneration, edema, and inflammation were increased compared to the control group (p = 0.000). In addition, GSH levels were significantly decreased in the I/R group compared to the control group (p = 0.000). On the other hand, in the I/R + DEX treatment group MDA levels, caspase-3, NF-κB/p65, 8-OHdG positivity, follicular degeneration, edema, and inflammation findings were decreased than in the I/R group (p = 0.000, p = 0.005, p = 0.005, p = 0.001, p = 0.005, respectively). However, GSH levels increased significantly in the I/R + DEX treatment group compared to the I/R group (p = 0.000). DEX protects against ovarian I/R injury through antioxidation and by suppressing inflammation and apoptosis.
Collapse
Affiliation(s)
- Filiz Mercantepe
- Department of Endocrinology and Metabolism Diseases, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Kerimali Akyildiz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Serpil Ciftel
- Department of Endocrinology and Metabolism Diseases, Erzurum Regional Education and Research Hospital, Erzurum, Turkey
| | - Adnan Yilmaz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
11
|
Chen R, Ma H, Li X, Wang M, Yang Y, Wu T, Zhang Y, Kong H, Qu H, Zhao Y. A Novel Drug with Potential to Treat Hyperbilirubinemia and Prevent Liver Damage Induced by Hyperbilirubinemia: Carbon Dots Derived from Platycodon grandiflorum. Molecules 2023; 28:molecules28062720. [PMID: 36985691 PMCID: PMC10056707 DOI: 10.3390/molecules28062720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Platycodon grandiflorum (PG) is a traditional Chinese medicine with a long history, but its active compounds have not been reported. In this study, novel carbon dots (CDs), PG-based CDs (PGC-CDs), were discovered and prepared from PG via calcinations and characterized by transmission electron microscopy; high-resolution transmission electron microscopy; X-ray diffraction, fluorescence, ultraviolet-visible, and Fourier-transform infrared spectrometers; X-ray photoelectron spectroscopy; and high-performance liquid chromatography. In addition, the safety and antioxidant activity of PGC-CDs was evaluated by RAW264.7 cells and LO2 cells. The therapeutic effects of PGC-CDs on hyperbilirubinemia and liver protection were evaluated in a bilirubin-induced hyperbilirubinemia mice model. The experiment confirmed that the diameter range of PGC-CDs was from 1.2 to 3.6 nm. PGC-CDs had no toxicity to RAW264.7 cells and LO2 cells at a concentration of 3.91 to 1000 µg/mL and could reduce the oxidative damage of cells caused by H2O2. PGC-CDs could inhibit the increase levels of bilirubin and inflammation factors and increase the levels of antioxidants and survival rate, demonstrating that PGC-CDs possessed anti-inflammatory and anti-oxidation activity. PGC-CDs may reduce the content of bilirubin, so as to reduce a series of pathological lesions caused by bilirubin, which has potential in treating hyperbilirubinemia and preventing liver damage induced by hyperbilirubinemia.
Collapse
Affiliation(s)
- Rui Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Huagen Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaopeng Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Meijun Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yunbo Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tong Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yue Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Huihua Qu
- Center of Scientific Experiment, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
- Correspondence: ; Tel.: +86-010-6428-6705; Fax: +86-010-6428-6821
| |
Collapse
|
12
|
Zhang S, Tang J, Sun C, Zhang N, Ning X, Li X, Wang J. Dexmedetomidine attenuates hepatic ischemia-reperfusion injury-induced apoptosis via reducing oxidative stress and endoplasmic reticulum stress. Int Immunopharmacol 2023; 117:109959. [PMID: 36881980 DOI: 10.1016/j.intimp.2023.109959] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023]
Abstract
Dexmedetomidine (DEX) affords a hepatoprotective effect during ischemia-reperfusion (IR) injury (IRI); however, the underlying mechanism remains elusive. In this work, using a rat liver IR model and a BRL-3A cell hypoxia-reoxygenation (HR) model, we explored whether DEX protects the liver against IRI by decreasing oxidative stress (OS), endoplasmic reticulum stress (ERS), and apoptotic pathways. We found that DEX significantly increased SOD and GSH activity while decreasing ROS and MDA levels in BRL-3A cells, successfully preventing HR-induced OS damage. DEX administration reduced JNK, ERK, and P38 phosphorylation and blocked HR-induced MAPK signaling pathway activation. Additionally, DEX administration reduced the expression of GRP78, IRE1α, XBP1, TRAF2, and CHOP, which reduced HR-induced ERS. NAC prevented the MAPK pathway from being activated and inhibited the ERS pathway. Further research showed that DEX significantly reduced HR-induced apoptosis by suppressing the expression of Bax/Bcl-2 and cleaved caspase-3. Similarly, animal studies demonstrated DEX exerted a protective effect of the liver by alleviating histopathological injury and enhancing liver function, mechanically DEX reduced cell apoptosis in liver tissue by reducing oxidative stress and ERS. In conclusion, DEX mitigates OS and ERS during IR, thereby suppressing cell apoptosis, thus providing protection to the liver.
Collapse
Affiliation(s)
- Shixia Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Jilang Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China.
| | - Chen Sun
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Nuannuan Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Xiaqing Ning
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Xueqin Li
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| | - Jiaqi Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, PR China.
| |
Collapse
|
13
|
Li Y, Zhang S, Cui K, Cao L, Fan Y, Fang B. miR-872-5p/FOXO3a/Wnt signaling feed-forward loop promotes proliferation of endogenous neural stem cells after spinal cord ischemia-reperfusion injury in rats. FASEB J 2023; 37:e22760. [PMID: 36607643 DOI: 10.1096/fj.202200962rrrr] [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: 06/24/2022] [Revised: 12/17/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
The activation of endogenous neural stem cells (NSCs) is considered an important mechanism of neural repair after mechanical spinal cord injury; however, whether endogenous NSC proliferation can also occur after spinal cord ischemia-reperfusion injury (SCIRI) remains unclear. In this study, we aimed to verify the existence of endogenous NSC proliferation after SCIRI and explore the underlying molecular mechanism. NSC proliferation was observed after SCIRI in vivo and oxygen-glucose deprivation and reperfusion (OGD/R) in vitro, accompanied by a decrease in forkhead box protein O 3a (FOXO3a) expression. This downward trend was regulated by the increased expression of microRNA-872-5p (miR-872-5p). miR-872-5p affected NSC proliferation by targeting FOXO3a to increase the expression of β-catenin and T-cell factor 4 (TCF4). In addition, TCF4 in turn acted as a transcription factor to increase the expression level of miR-872-5p, and knockdown of FOXO3a enhanced the binding of TCF4 to the miR-872-5p promoter. In conclusion, SCIRI in vivo and OGD/R in vitro stimulated the miR-872-5p/FOXO3a/β-catenin-TCF4 pathway, thereby promoting NSC proliferation. At the same time, FOXO3a affected TCF4 transcription factor activity and miR-872-5p expression, forming a positive feedback loop that promotes NSC proliferation.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Shaoqiong Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Kaile Cui
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Linyan Cao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yiting Fan
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
14
|
Chen F, Wang D, Jiang Y, Ma H, Li X, Wang H. Dexmedetomidine postconditioning alleviates spinal cord ischemia-reperfusion injury in rats via inhibiting neutrophil infiltration, microglia activation, reactive gliosis and CXCL13/CXCR5 axis activation. Int J Neurosci 2023; 133:1-12. [PMID: 33499703 DOI: 10.1080/00207454.2021.1881089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE Spinal cord ischemia-reperfusion (I/R) injury is an unresolved complication and its mechanisms are still not completely understood. Here, we studied the neuroprotective effects of dexmedetomidine (DEX) postconditioning against spinal cord I/R injury in rats and explored the possible mechanisms. MATERIALS AND METHODS In the study, rats were randomly divided into five groups: sham group, I/R group, DEX0.5 group, DEX2.5 group, and DEX5 group. I/R injury was induced in experimental rats; 0.5 μg/kg, 2.5 μg/kg, 5 μg/kg DEX were intravenously injected upon reperfusion respectively. Neurological function, histological assessment, and the disruption of blood-spinal cord barrier (BSCB) were evaluated via the BBB scoring, hematoxylin and eosin staining, Evans Blue (EB) extravasation and spinal cord edema, respectively. Neutrophil infiltration was evaluated via Myeloperoxidase (MPO) activity. Microglia activation and reactive gliosis was evaluated via ionized calcium-binding adapter molecule-1(IBA-1) and glial fibrillary acidic protein (GFAP) immunofluorescence, respectively. The expression of C-X-C motif ligand 13 (CXCL13), C-X-C chemokine receptor type 5(CXCR5), caspase-3 was determined by western blotting. The expression levels of interleukin 6(IL-6), tumor necrosis factor-α(TNF-α), IL-1β were determined by ELISA assay. RESULTS DEX postconditioning preserved neurological assessment scores, improved histological assessment scores, attenuated BSCB leakage after spinal cord I/R injury. Neutrophil infiltration, microglia activation and reactive gliosis were also inhibited by DEX postconditioning. The expression of CXCL13, CXCR5, caspase-3, IL-6, TNF-α, IL-1β were reduced by DEX postconditioning. CONCLUSIONS DEX postconditioning alleviated spinal cord I/R injury, which might be mediated via inhibition of neutrophil infiltration, microglia activation, reactive gliosis and CXCL13/CXCR5 axis activation.
Collapse
Affiliation(s)
- Fengshou Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Dan Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yanhua Jiang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hong Ma
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaoqian Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - He Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
15
|
Chen Y, Dong Y, Zhang Z, Han J, Chen F, Tong X, Ma H. Fra-1 induces apoptosis and neuroinflammation by targeting S100A8 to modulate TLR4 pathways in spinal cord ischemia/reperfusion injury. Brain Pathol 2023; 33:e13113. [PMID: 36634215 PMCID: PMC9836372 DOI: 10.1111/bpa.13113] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Spinal cord ischemia/reperfusion injury (SCII) is a severe complication driven by apoptosis and neuroinflammation. An increase in the expression of c-Fos, a member of the AP-1 family, is known as a neuronal activation marker in SCII. The AP-1 family is composed of Jun, Fos, and is associated with the regulation of cytokines expression and apoptosis. Fra-1 is a member of the Fos family, however, the contribution of Fra-1 to SCII is still unclear. In our study, Fra-1 was highly upregulated especially in neurons and microglia and promoted apoptosis by changing the expression of Bax/Bcl-2 after SCII. Furthermore, we found that Fra-1 directly regulated the transcription expression of S100A8. We demonstrated that knockdown of Fra-1 alleviated S100A8 mediated neuronal apoptosis and inflammatory factor release, thus improved motor function after SCII. Interestingly, we showed that administration of TAK-242, the TLR4 inhibitor, to the ischemia/reperfusion (I/R) injury induced rats suppressed the activation of the ERK and NF-κB pathways, and further reduced Fra-1 expression. In conclusion, we found that Fra-1-targeted S100A8 was expressed the upstream of Fra-1, and the Fra-1/S100A8 interaction formed a feedback loop in the signaling pathways activated by SCII.
Collapse
Affiliation(s)
- Ying Chen
- Department of AnesthesiologyFirst Affiliated Hospital, China Medical UniversityShenyangLiaoningChina
| | - Yan Dong
- Department of AnesthesiologyFirst Affiliated Hospital, China Medical UniversityShenyangLiaoningChina
| | - Zai‐Li Zhang
- Department of AnesthesiologyFirst Affiliated Hospital, China Medical UniversityShenyangLiaoningChina
| | - Jie Han
- Department of AnesthesiologyFirst Affiliated Hospital, China Medical UniversityShenyangLiaoningChina
| | - Feng‐Shou Chen
- Department of AnesthesiologyFirst Affiliated Hospital, China Medical UniversityShenyangLiaoningChina
| | - Xiang‐Yi Tong
- Department of AnesthesiologyFirst Affiliated Hospital, China Medical UniversityShenyangLiaoningChina
| | - Hong Ma
- Department of AnesthesiologyFirst Affiliated Hospital, China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
16
|
Han GJ, Min XZ, Ma SS, Ding C, Wang XQ. Xuesaitong Combined with Dexmedetomidine Improves Cerebral Ischemia-Reperfusion Injury in Rats by Activating Keap1/Nrf2 Signaling and Mitophagy in Hippocampal Tissue. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5126042. [PMID: 36531207 PMCID: PMC9750788 DOI: 10.1155/2022/5126042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/14/2022] [Accepted: 11/25/2022] [Indexed: 08/02/2024]
Abstract
Ischemic stroke is the most common type of cerebrovascular disease with high mortality and poor prognosis, and cerebral ischemia-reperfusion (CI/R) injury is the main murderer. Here, we attempted to explore the effects and mechanism of Xuesaitong (XST) combined with dexmedetomidine (Dex) on CI/R injury in rats. First, a rat model of CI/R injury was constructed via the middle cerebral artery occlusion (MCAO) method and treated with XST and Dex alone or in combination. Then, on the 5th and 10th days of treatment, the neurological impairment was assessed using the modified neurological severity scores (mNSS), the 8-arm radial maze test (8ARMT), novel object recognition test (NORT), and fear conditioning test (FCT). H&E staining was performed to observe the pathological changes of the hippocampus. ELISA and related kits were used to assess the monoamine neurotransmitters and antioxidant enzyme activities in the hippocampus. The ATP, mitochondrial membrane potential levels, and qRT-PCR of genes related to mitochondrial function were determined to assess mitochondrial functions in the hippocampus and western blot to determine Keap1/Nrf2 signaling pathway and mitophagy-related protein expression. The results showed that XST combined with Dex significantly reduced mNSS, improved spatial memory and learning deficits, and enhanced fear memory and cognitive memory ability in CI/R rats, which was superior to single-drug treatment. Moreover, XST combined with Dex treatment improved hippocampal histopathological damage; significantly increased the levels of monoamine neurotransmitters, neurotrophic factors, ATP, and mitochondrial membrane potential; and upregulated the activities of antioxidant enzymes and the expression of mitophagy-related proteins in the hippocampus of CI/R rats. XST combined with Dex treatment also activated the Keap1/Nrf2 signaling and upregulated the protein expression of downstream antioxidant enzymes HO-1 and NQ. Altogether, this study showed that a combination of XST and Dex could activate the Keap1/Nrf2 signaling and mitophagy to protect rats from CI/R-related neurological impairment.
Collapse
Affiliation(s)
- Guo-Jie Han
- Department of Anesthesiology, Shandong Cancer Hospital and Institute Affiliated to Shandong First Medical University (Shandong Academy of Medical Science), Jinan, 250117 Shandong, China
| | - Xiang-Zhen Min
- Department of Anesthesiology, Shandong Cancer Hospital and Institute Affiliated to Shandong First Medical University (Shandong Academy of Medical Science), Jinan, 250117 Shandong, China
| | - Shuang-Shuang Ma
- Department of Anesthesiology, Shandong Cancer Hospital and Institute Affiliated to Shandong First Medical University (Shandong Academy of Medical Science), Jinan, 250117 Shandong, China
| | - Chuan Ding
- Department of Anesthesiology, Shandong Cancer Hospital and Institute Affiliated to Shandong First Medical University (Shandong Academy of Medical Science), Jinan, 250117 Shandong, China
| | - Xiu-Qin Wang
- Department of Anesthesiology, Shandong Cancer Hospital and Institute Affiliated to Shandong First Medical University (Shandong Academy of Medical Science), Jinan, 250117 Shandong, China
| |
Collapse
|
17
|
Rofaeil RR, Ahmed SM, Bahaa HA, Mahran A, Welson NN, Abdelzaher WY. Dipeptidyl peptidase-4 inhibitor: Sitagliptin down-regulated toll-like receptor 4 signaling pathway to reduce uterine injury in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1396-1401. [PMID: 36474576 PMCID: PMC9699943 DOI: 10.22038/ijbms.2022.64552.14202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 10/01/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVES Uterine ischemia is a common problem with ongoing controversy about its pathogenesis and prevention. The present study aimed to investigate the protective role of sitagliptin against uterine ischemia-reperfusion injury (IRI). MATERIALS AND METHODS Rats were allocated into 4 groups: control, sitagliptin (SIT) (5 mg/kg), IR; ischemia was induced followed by reperfusion, and IR+SIT; SIT was administered 1 hr before IRI. Uteri were removed for histopathological and biochemical observations. Malondialdehyde (MDA), total nitrites (NOx), reduced glutathione (GSH), superoxide dismutase (SOD) activity, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and toll-like receptor 4 (TLR4) were all measured. Hematoxylin and eosin (H&E) stain, Periodic acid-Schiff stain (PAS), and caspase-3 immunostaining were applied. RESULTS In the IR+SIT group; NOx, GSH, and SOD activities increased significantly. Meanwhile, the levels of MDA, TNF-α, IL-6, TLR4, and caspase-3 immunoexpression showed a significant reduction, as compared with the IR group. In the IR+SIT group, an improvement in the histopathological picture was noticed. CONCLUSION The results showed that sitagliptin confers protection against uterine IRI through anti-oxidant, anti-inflammatory, and anti-apoptotic effects with a possible role for TLR4.
Collapse
Affiliation(s)
- Remon Roshdy Rofaeil
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt, Department of Pharmacology, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Sabreen Mahmoud Ahmed
- Depatment of Human Anatomy and Embryology, Faculty of Medicine, Minia University, delegated to Deraya University-New Minia City, Egypt
| | - Haitham Ahmed Bahaa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Minia University. Minia, Egypt
| | - Ahmad Mahran
- Department of Obstetrics and Gynecology, Faculty of Medicine, Minia University. Minia, Egypt
| | - Nermeen N. Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt,Corresponding author: Nermeen N. Welson. Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt. Tel/Fax: +201157000233;
| | | |
Collapse
|
18
|
Hemsinli D, Tumkaya L, Ergene S, Karakisi SO, Mercantepe T, Yilmaz A. Dexmedetomidine attenuates pneumocyte apoptosis and inflammation induced by aortic ischemia-reperfusion injury. Clin Exp Hypertens 2022; 44:595-600. [PMID: 35787727 DOI: 10.1080/10641963.2022.2093893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Despite significant improvements in interventional vascular aneurysm repair procedures and intensive care patient management, there has been no significant decrease in mortality due to ruptured abdominal aortic aneurysm. Oxidative stress is known to play a key role in secondary organ damage due to infrarenal aortic clamping. The aim of this study was to examine the potential protective effect of the alpha-2 adrenergic receptor agonist dexmedetomidine (DMT) on aortic occlusion-induced lung injury. METHODS Thirty Sprague Dawley rats were allocated into control, ischemia-reperfusion (IR), and IR+DMT groups randomly. Vascular clamps were attached to the abdominal aorta in the IR and IR+DMT groups. Two-hour reperfusion was established 1 h after ischemia. The IR+DMT group received a single intraperitoneal 100 µg dose of DMT 30 min before infrarenal abdominal aortic clamping. RESULTS IR due to aortic occlusion led to apoptosis, widespread inflammation, alveolar septal wall thickening due to bleeding and vascular congestion were observed in both types I and II pneumocytes. Malondialdehyde levels increased while glutathione decreased. However, DMT was found to lower apoptotic pneumocytes, alveolar-septal thickness, hemorrhage, vascular congestion, and malondialdehyde levels, while glutathione levels in lung tissue increased. CONCLUSIONS This study is the first to address the effects of DMT on the lung in a ruptured abdominal aortic aneurysm model. Our findings suggest that the alpha-2 adrenergic receptor agonist DMT reduces oxidative stress and apoptosis, thus protecting against aortic occlusion-induced pulmonary injury.
Collapse
Affiliation(s)
- Dogus Hemsinli
- Faculty of Medicine, Department of Cardiovascular Surgery, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- Faculty of Medicine, Department of Histology and Embryology, Recep Tayyip Erdogan University, Rize, Turkey
| | - Saban Ergene
- Faculty of Medicine, Department of Cardiovascular Surgery, Recep Tayyip Erdogan University, Rize, Turkey
| | - S Ozan Karakisi
- Faculty of Medicine, Department of Cardiovascular Surgery, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- Faculty of Medicine, Department of Histology and Embryology, Recep Tayyip Erdogan University, Rize, Turkey
| | - Adnan Yilmaz
- Faculty of Medicine, Department of Medical Biochemistry, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
19
|
Cai Q, Liu G, Huang L, Guan Y, Wei H, Dou Z, Liu D, Hu Y, Gao M. The Role of Dexmedetomidine in Tumor-Progressive Factors in the Perioperative Period and Cancer Recurrence: A Narrative Review. Drug Des Devel Ther 2022; 16:2161-2175. [PMID: 35821701 PMCID: PMC9271281 DOI: 10.2147/dddt.s358042] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/28/2022] [Indexed: 12/20/2022] Open
Abstract
Dexmedetomidine, a specific α2 adrenergic receptor agonist, is highly frequently used in the perioperatively for its favorable pharmacology, such as mitigating postoperative cognitive dysfunction. Increasing attention has been recently focused on the effect of whether dexmedetomidine influences cancer recurrence, which urges the discussion of the role of dexmedetomidine in tumor-progressive factors. The pharmacologic characteristics of dexmedetomidine, the tumor-progressive factors in the perioperative period, and the relationships between dexmedetomidine and tumor-progressive factors were described in this review. Available evidence suggests that dexmedetomidine could reduce the degree of immune function suppression, such as keeping the number of CD3+ cells, NK cells, CD4+/CD8+ ratio, and Th1/Th2 ratio stable and decreasing the level of proinflammatory cytokine (interleukin 6 and tumor necrosis factor-alpha) during cancer operations. However, dexmedetomidine exhibits different roles in cell biological behavior depending on cancer cell types. The conclusions on whether dexmedetomidine would influence cancer recurrence could not be currently drawn for the lack of strong clinical evidence. Therefore, this is still a new area that needs further exploration.
Collapse
Affiliation(s)
- Qiang Cai
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Guoqing Liu
- Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Linsheng Huang
- Department of Hepatobiliary Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Yuting Guan
- Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Huixia Wei
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Zhiqian Dou
- Department of Obstetrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Dexi Liu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Yang Hu
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
- Yang Hu, Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, People’s Republic of China, Tel +86-13995744850, Email
| | - Meiling Gao
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
- Correspondence: Meiling Gao, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China, Tel +86-15971849819, Email
| |
Collapse
|
20
|
Hu M, Men Y, Chen L, Huang J, Duan F, Zhang Y, Dong S. Dexmedetomidine exerts its protective effect on cerebral ischemia reperfusion injury in mice by inhibiting ferroptosis. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:600-609. [PMID: 35753730 PMCID: PMC10929910 DOI: 10.11817/j.issn.1672-7347.2022.210443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Stroke is one of the major diseases that can threaten human life and health. The incidence of ischemic stroke accounts for more than 70% of stroke. The mechanism of ischemia reperfusion (IR) injury caused by ischemic stroke is extremely complex. In recent years, dexmedetomidine has been increasingly studied in anti-cerebral IR injury as a common clinical anesthetic adjunct, but its specific mechanism is not fully understood. Therefore, this study aims to explore the effects and mechanisms of dexmedetomidine on cerebral IR injury in mice. METHODS The mouse middle cerebral artery occlusion (MCAO) model was prepared by modified suture method. Male ICR mice were randomly divided into a sham group, an IR group, an IR+D1 group (IR+administered 25 µg/kg dexmedetomidine), an IR+D2 group(IR+administered 50 µg/kg dexmedetomidine), an IR+D3 group (IR+administered 100 µg/kg dexmedetomidine), and an IR+D2+ML385 group (IR+administered 50 µg/kg dexmedetomidine and 30 mg/kg ML385). The neurologic behavior of mice was evaluated by Longa's five-point method. 2,3,5-triphenyltetrazolium chloride (TTC) staining was used to detect the percentage of cerebral infarct volume in mice. The protein expressions of nuclear factor erythroid 2-related factor 2 (Nrf2), transferrin receptor 1 (TFR1), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11) in the cerebral tissues of mice were detected by Western blotting.Mitochondrial morphology was observed under the transmission electron microscope. The contents of MDA, Fe2+, and GSH in the cerebral tissues of mice were detected. RESULTS Compared with the sham group, neurobehavioral scores, cerebral infarct volume, the contents of MDA and Fe2+, as well as the protein expression of TFR1 were significantly increased; the contents of GSH and the protein expression of SLC7A11 and GPX4 were significantly reduced (all P<0.05); mitochondria in cerebral tissue were wrinkled, cristae were reduced, and membrane density was increased in the IR group. Compared with the IR group, neurobehavioral scores, cerebral infarction volume, MDA and Fe2+ contents, as well as the protein expression of TFR1 were significantly reduced; the contents of GSH and the protein expression of SLC7A11 and GPX4 were significantly increased (all P<0.05); mitochondrial damage in cerebral tissue was significantly relieved with the pre-treatment of dexmedetomidine. Compared with the IR+D2 group, neurobehavioral scores, cerebral infarction volume, MDA and Fe2+ contents, as well as the protein expression of TFR1 were significantly increased; the contents of GSH and the protein expression of SLC7A11 and GPX4 were significantly reduced (all P<0.05);mitochondria reappeared significantly damaged with the ML385 on the basis of dexmedetomidine pre-treatment. CONCLUSIONS The protective effect of dexmedetomidine on cerebral IR injury mice is related to its inhibition of ferroptosis, and the mechanism might be related to its regulation of Nrf2 expression.
Collapse
Affiliation(s)
- Miao Hu
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030.
| | - Yunzheng Men
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Lei Chen
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Fangfang Duan
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Yuxin Zhang
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030
| | - Shuying Dong
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu Anhui 233030.
- Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu Anhui 233030, China.
| |
Collapse
|
21
|
Li Y, Qu M, Xing F, Li H, Cheng D, Xing N, Zhang W. The Protective Mechanism of Dexmedetomidine in Regulating Atg14L-Beclin1-Vps34 Complex Against Myocardial Ischemia-Reperfusion Injury. J Cardiovasc Transl Res 2021; 14:1063-1074. [PMID: 33914271 DOI: 10.1007/s12265-021-10125-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/28/2021] [Indexed: 02/06/2023]
Abstract
The blood flow restoration of ischemic tissues causes myocardial injury. Dexmedetomidine (Dex) protects multi-organs against ischemia/reperfusion (I/R) injury. This study investigated the protective mechanism of Dex post-treatment in myocardial I/R injury. The rat model of myocardial I/R was established. The effects of Dex post-treatment on cardiac function and autophagy flow were observed. Dex attenuated myocardial I/R injury and reduced I/R-induced autophagy in rats. Dex weakened the interactions between Beclin1 and Vps34 and Beclin1 and Atg14L, thus downregulating Vps34 kinase activity. In vitro, the cardiomyocytes subjected to oxygen glucose deprivation/reoxygenation were treated with Dex and PI3K inhibitor LY294002. LY294002 attenuated the myocardial protective effect of DEX, indicating that Dex protected against cardiac I/R by activating the PI3K/Akt pathway. In conclusion, Dex upregulated the phosphorylation of Beclin1 at S295 site by activating the PI3K/Akt pathway and reduced the interactions of Atg14L-Beclin1-Vps34 complex, thus inhibiting autophagy and protecting against myocardial I/R injury.
Collapse
Affiliation(s)
- Yanna Li
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Mingcui Qu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Huixin Li
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Dan Cheng
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China.
| | - Wei Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China.
| |
Collapse
|
22
|
Zhang Q, Liu XM, Hu Q, Liu ZR, Liu ZY, Zhang HG, Huang YL, Chen QH, Wang WX, Zhang XK. Dexmedetomidine inhibits mitochondria damage and apoptosis of enteric glial cells in experimental intestinal ischemia/reperfusion injury via SIRT3-dependent PINK1/HDAC3/p53 pathway. J Transl Med 2021; 19:463. [PMID: 34772407 PMCID: PMC8588684 DOI: 10.1186/s12967-021-03027-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/06/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) injury commonly occurs during perioperative periods, resulting in high morbidity and mortality on a global scale. Dexmedetomidine (Dex) is a selective α2-agonist that is frequently applied during perioperative periods for its analgesia effect; however, its ability to provide protection against intestinal I/R injury and underlying molecular mechanisms remain unclear. METHODS To fill this gap, the protection of Dex against I/R injury was examined in a rat model of intestinal I/R injury and in an inflammation cell model, which was induced by tumor necrosis factor-alpha (TNF-α) plus interferon-gamma (IFN-γ) stimulation. RESULTS Our data demonstrated that Dex had protective effects against intestinal I/R injury in rats. Dex was also found to promote mitophagy and inhibit apoptosis of enteric glial cells (EGCs) in the inflammation cell model. PINK1 downregulated p53 expression by promoting the phosphorylation of HDAC3. Further studies revealed that Dex provided protection against experimentally induced intestinal I/R injury in rats, while enhancing mitophagy, and suppressing apoptosis of EGCs through SIRT3-mediated PINK1/HDAC3/p53 pathway in the inflammation cell model. CONCLUSION Hence, these findings provide evidence supporting the protective effect of Dex against intestinal I/R injury and its underlying mechanism involving the SIRT3/PINK1/HDAC3/p53 axis.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Xiao-Ming Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Qian Hu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zheng-Ren Liu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Zhi-Yi Liu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Huai-Gen Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yuan-Lu Huang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Qiu-Hong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Wen-Xiang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Xue-Kang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
23
|
Wu Y, Qiu G, Zhang H, Zhu L, Cheng G, Wang Y, Li Y, Wu W. Dexmedetomidine alleviates hepatic ischaemia-reperfusion injury via the PI3K/AKT/Nrf2-NLRP3 pathway. J Cell Mol Med 2021; 25:9983-9994. [PMID: 34664412 PMCID: PMC8572787 DOI: 10.1111/jcmm.16871] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 12/22/2022] Open
Abstract
Hepatic ischaemia-reperfusion (I/R) injury constitutes a tough difficulty in liver surgery. Dexmedetomidine (Dex) plays a protective role in I/R injury. This study investigated protective mechanism of Dex in hepatic I/R injury. The human hepatocyte line L02 received hypoxia/reoxygenation (H/R) treatment to stimulate cell model of hepatic I/R. The levels of pyroptosis proteins and inflammatory factors were detected. Functional rescue experiments were performed to confirm the effects of miR-494 and JUND on hepatic I/R injury. The levels of JUND, PI3K/p-PI3K, AKT/p-AKT, Nrf2, and NLRP3 activation were detected. The rat model of hepatic I/R injury was established to confirm the effect of Dex in vivo. Dex reduced pyroptosis and inflammation in H/R cells. Dex increased miR-494 expression, and miR-494 targeted JUND. miR-494 inhibition or JUND upregulation reversed the protective effect of Dex. Dex repressed NLRP3 inflammasome by activating the PI3K/AKT/Nrf2 pathway. In vivo experiments confirmed the protective effect of Dex on hepatic I/R injury. Overall, Dex repressed NLRP3 inflammasome and alleviated hepatic I/R injury via the miR-494/JUND/PI3K/AKT/Nrf2 axis.
Collapse
Affiliation(s)
- Yan Wu
- Department of AnesthesiologyThe First Affiliated Hospital of Anhui University of Chinese MedicineHefeiChina
| | - Gaolin Qiu
- Department of AnesthesiologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Hainie Zhang
- Department of AnesthesiologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Leilei Zhu
- Department of AnesthesiologyThe Fourth Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Gao Cheng
- Department of AnesthesiologyThe Fourth Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Yiqiao Wang
- Department of AnesthesiologyAnhui NO.2 Provincial People's HospitalHefeiChina
| | - Yuanhai Li
- Department of AnesthesiologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Weiwei Wu
- Department of AnesthesiologyThe Fourth Affiliated Hospital of Anhui Medical UniversityHefeiChina
| |
Collapse
|
24
|
Rong Z, Huang Y, Cai H, Chen M, Wang H, Liu G, Zhang Z, Wu J. Gut Microbiota Disorders Promote Inflammation and Aggravate Spinal Cord Injury Through the TLR4/MyD88 Signaling Pathway. Front Nutr 2021; 8:702659. [PMID: 34589510 PMCID: PMC8473614 DOI: 10.3389/fnut.2021.702659] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/23/2021] [Indexed: 12/02/2022] Open
Abstract
Background: In spinal cord injury (SCI), systemic inflammation and the death of nerve cells in the spinal cord are life threatening. The connection between gut microbiota and signaling pathways has been a hot research topic in recent years. The Toll-like receptor 4/Myeloid differentiation factor 88 (TLR4/MyD88) signaling pathway is closely related to the inflammatory response. This study explored whether the gut microbiota imbalance could affect the TLR4/MyD88 signaling pathway to regulate SCI to provide a new basis for SCI research and treatment. Methods: An SCI model was constructed to study the influence on the injury of gut microbiota. 16S amplicon sequencing was used to identify the diversity and abundance of gut microbes. Fecal microbiota transplantation was performed in mice with SCI. ELISA was used to detect the serum levels of pro-inflammatory and anti-inflammatory factors in mice. Hematoxylin and eosin staining was used to observe SCI in mice. Immunofluorescence was used to detect the rates of loss glial fibrillary acidic protein (GFAP), neuronal nuclear protein (NeuN), and ionized calcium-binding adapter molecule 1 (IBA1) in the spinal cord as indicators of apoptosis. The expression of the TLR4/MyD88 signaling pathway was detected by qRT-PCR and western blotting. Results: Significant differences were observed in the gut microbiota of SCI mice and normal mice. The gut microbiota of SCI mice was imbalanced. The levels of pro-inflammatory cytokines tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 in SCI mice were increased, as was the level of the toxic induced nitric oxide synthase. The levels of anti-inflammatory factors IL-4, transforming growth factor-β, and IL-10 were decreased, as was the level of arginase-1. The apoptosis rates of GFAP, NeuN, and IBA1 were increased. The TLR4/MyD88 signaling pathway was activated. In the SCI group, inflammation increased after fecal transplantation, apoptosis of GFAP, NeuN, and IBA1 increased, and SCI was more serious. Conclusion: The TLR4/MyD88 signaling pathway promotes the death of nerve cells by inducing inflammation. Gut microbiota dysregulation can lead to aggravated SCI by activating the TLR4/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Zijie Rong
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Yuliang Huang
- Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China.,Department of Orthopaedics, Huizhou Municipal Central Hospital, Huizhou, China
| | - Honghua Cai
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Min Chen
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Hao Wang
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Guihua Liu
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Zhiwen Zhang
- Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China.,Department of Orthopaedics, Huizhou Municipal Central Hospital, Huizhou, China
| | - Jiawen Wu
- Department of Spine Surgery, The People's Hospital of Longhua, Shenzhen Longhua Clinical Medical College of Guangdong Medical University, Shenzhen, China
| |
Collapse
|
25
|
Liu YB, Liu WF, Chen WC, Li W, Lin YL, Xu CJ, He HF. Dexmedetomidine alleviates traumatic spinal cord injury in rats via inhibiting apoptosis induced by endoplasmic reticulum stress. Neurol Res 2021; 44:275-284. [PMID: 34533101 DOI: 10.1080/01616412.2021.1979750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the protective effect of dexmedetomidine (Dex) on traumatic spinal cord injury (TSCI) and to evaluate the involvement of inhibition of endoplasmic reticulum (ER) stress response in the potential mechanism. METHOD Sprague-Dawley rats were randomly divided into five groups. The hind limb locomotor function of rats was evaluated at 1, 3 and 7 days after the operation. At 7 days after the operation, spinal cord specimens were obtained for hematoxylin and eosin (H&E), Nissl and TUNEL staining, as well as immunofluorescence and Western blot analyses to detect the level of apoptosis and the levels of proteins related to ER stress. RESULTS 7 days after the operation, Dex treatment promoted the recovery and also inhibited apoptosis of neurons in the spinal cord. Additionally, Dexinhibited the expression of proteins related to ER stress response after spinal cord injury. CONCLUSIONS Dex improves the neurological function of rats with TSCI and reduces apoptosis of spinal cord neurons. The potential mechanism is related to the inhibition of the ER stress response.
Collapse
Affiliation(s)
- Yi-Bin Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei-Feng Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei-Can Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei Li
- Department of ICU, Wuhan Third Hospital, Wuhan University, Wuhan, China**
| | - Yan-Ling Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chong-Jun Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - He-Fan He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
26
|
Tural K, Ozden O, Bilgi Z, Kubat E, Ermutlu CS, Merhan O, Tasoglu I. The protective effect of betanin and copper on spinal cord ischemia-reperfusion injury. J Spinal Cord Med 2021; 44:704-710. [PMID: 32223592 PMCID: PMC8477937 DOI: 10.1080/10790268.2020.1737788] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Context: Both copper and betanin have been implicated as having significant bioactivity against ischemic damage in a variety of experimental and clinical settings. The aim of this study is to investigate whether betanin and copper have any protective effect on spinal cord in an ischemia-reperfusion (I/R) model in rats.Design: Spraque-Dawley rats were used in four groups: Sham group (n = 7), control group (laparotomy and cross-clamping of aorta, n = 7), betanin treatment group (dosage of 100 mg/kg of betanin administered intraperitoneally (i.p.) 60 min before laparotomy, n = 7), copper sulfate treatment group (administered copper sulfate i.p. at a dose of 0.1 mg/kg/day for 7 days before laparotomy, n = 7). Malondialdehyde (MDA), glutathione (GSH) levels, myeloperoxidase (MPO) and superoxide dismutase (SOD) activity were measured. Terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling (TUNEL) assay was also performed to evaluate apoptosis.Setting: Kafkas University, Faculty of Medicine, Kars, Turkey.Results: I/R injury was successfully demonstrated with the surgical model. Betanin and copper treatment significantly decreased MDA levels, MPO activity and the number of apoptotic cells in the spinal cord. Betanin and copper treatment significantly increased GSH levels. Copper treatment significantly increased SOD activity, whereas betanin was not as effective. Apoptotic cells were significantly decreased in both treatment groups.Conclusion: I/R injury of the spinal cord can be successfully demonstrated by aortic clamping in this surgical model. Betanin/Copper sulphate has ameliorative effects against operative I/R injury. Low toxicity of those agents makes them ideal targets for clinical research for this purpose.
Collapse
Affiliation(s)
- Kevser Tural
- Medical Faculty, Department of Cardiovascular Surgery, Kafkas University, Kars, Turkey,Correspondence to: Kevser Tural, Medical Faculty, Department of Cardiovascular Surgery, Kafkas University, Kars, 36100, Turkey; Ph: 0474 2252105.
| | - Ozkan Ozden
- Faculty of Engineering and Architecture, Department of Bioengineering, Kafkas University, Kars, Turkey
| | - Zeynep Bilgi
- Medical Faculty, Department of Thoracic Surgery, Medeniyet University, İstanbul, Turkey
| | - Emre Kubat
- Gulhane Education and Research Hospital, Clinic of Cardiovascular Surgery, Ankara, Turkey
| | - Celal Sahin Ermutlu
- Faculty of Veterinary, Department of Surgery, Kafkas University, Kars, Turkey
| | - Oguz Merhan
- Faculty of Veterinary, Department of Biochemistry, Kafkas University, Kars, Turkey
| | - Irfan Tasoglu
- Department of Cardiovascular Surgery, Turkiye Yuksek Ihtisas Education and Research Hospital, Ankara, Turkey
| |
Collapse
|
27
|
Hong H, Huang Q, Cai Y, Lin T, Xia F, Jin Z. Dexmedetomidine preconditioning ameliorates lung injury induced by pulmonary ischemia/reperfusion by upregulating promoter histone H3K4me3 modification of KGF-2. Exp Cell Res 2021; 406:112762. [PMID: 34352276 DOI: 10.1016/j.yexcr.2021.112762] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022]
Abstract
Keratinocyte growth factor (KGF)-2 has been highlighted to play a significant role in maintaining the endothelial barrier integrity in lung injury induced by ischemia-reperfusion (I/R). However, the underlying mechanism remains largely unknown. The aims of this study were to determine whether dexmedetomidine preconditioning (DexP) modulates pulmonary I/R-induced lung injury through the alteration in KGF-2 expression. In our I/R-modeled mice, DexP significantly inhibited pathological injury, inflammatory response, and inflammatory cell infiltration, while promoted endothelial barrier integrity and KGF-2 promoter activity in lung tissues. Bioinformatics prediction and ChIP-seq revealed that I/R significantly diminished the level of H3K4me3 modification in the KGF-2 promoter, which was significantly reversed by DexP. Moreover, DexP inhibited the expression of histone demethylase JMJD3, which in turn promoted the expression of KGF-2. In addition, overexpression of JMJD3 weakened the protective effect of DexP on lung injury in mice with I/R. Collectively, the present results demonstrated that DexP ameliorates endothelial barrier dysfunction via the JMJD3/KGF-2 axis.
Collapse
Affiliation(s)
- Huisuo Hong
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, PR China
| | - Qingqing Huang
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, PR China.
| | - Yaoyao Cai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, PR China
| | - Tingting Lin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, PR China
| | - Fangfang Xia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, PR China
| | - Zhousheng Jin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, PR China.
| |
Collapse
|
28
|
Sun T, Gong Q, Wu Y, Shen Z, Zhang Y, Ge S, Duan JS. Dexmedetomidine alleviates cardiomyocyte apoptosis and cardiac dysfunction may be associated with inhibition of RhoA/ROCK pathway in mice with myocardial infarction. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:1569-1577. [PMID: 33782744 DOI: 10.1007/s00210-021-02082-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/16/2021] [Indexed: 11/25/2022]
Abstract
The global incidence of myocardial infarction has been consistently high, and it is one of the main causes of poor cardiovascular prognosis. Dexmedetomidine (DEX) is a highly selective α2 receptor agonist. Recent studies have found that DEX has a protective effect on myocardial infarction, but its specific mechanism is still unclear. In this experiment, we permanently ligated the anterior descending branch of mice to explore the protective mechanism of DEX against myocardial infarction. Our study found that intraperitoneal injection of DEX for 7 days after myocardial infarction in mice can increase the reduction of ejection fraction (EF) and fractional shortening (FS) caused by myocardial infarction and significantly reduce the release of serum markers. The results of myocardial HE and Sirius red staining suggest that the changes in the myocardial structure of mice after using DEX are reduced. Immunohistochemistry shows that DEX reduces the expression of ROCK1 protein after myocardial infarction. TUNEL staining and the protein expression levels of cleaved caspase-3 and cleaved caspase-9 were used to detect cell apoptosis and results make clear that DEX can reduce the apoptosis caused by myocardial infarction. Western blot experiments showed that DEX can reduce the expression levels of ROCK1 and ROCK2 (Rho-kinase). At the same time, it was observed that DEX improved the Bcl-2/Bax ratio. The above results indicate that DEX reduces cardiomyocyte apoptosis and improves cardiac function likely through inhibiting the RhoA/ROCK signaling pathway. This study may provide new insights into the protective effect of DEX after myocardial infarction in mice.
Collapse
Affiliation(s)
- Tao Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230032, Anhui, China
| | - Qian Gong
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230032, Anhui, China
| | - Ying Wu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230032, Anhui, China
| | - Zhiming Shen
- Yangzhou University Medical College, Yangzhou, 225001, Jiangsu, China
| | - Yan Zhang
- Department of Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230032, Anhui, China.
| | - Jing-Si Duan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230032, Anhui, China.
| |
Collapse
|
29
|
Zou HJ, Guo SW, Zhu L, Xu X, Liu JB. Methylprednisolone Induces Neuro-Protective Effects via the Inhibition of A1 Astrocyte Activation in Traumatic Spinal Cord Injury Mouse Models. Front Neurosci 2021; 15:628917. [PMID: 34135725 PMCID: PMC8200570 DOI: 10.3389/fnins.2021.628917] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/24/2021] [Indexed: 11/13/2022] Open
Abstract
Traumatic spinal cord injury (TSCI) leads to pathological changes such as inflammation, edema, and neuronal apoptosis. Methylprednisolone (MP) is a glucocorticoid that has a variety of beneficial effects, including decreasing inflammation and ischemic reaction, as well as inhibiting lipid peroxidation. However, the efficacy and mechanism of MP in TSCI therapy is yet to be deciphered. In the present study, MP significantly attenuated the apoptotic effects of H2O2 in neuronal cells. Western blot analysis demonstrated that the levels of apoptotic related proteins, Bax and cleaved caspase-3, were reduced while levels of anti-apoptotic Bcl-2 were increased. In vivo TUNEL assays further demonstrated that MP effectively protected neuronal cells from apoptosis after TSCI, and was consistent with in vitro studies. Furthermore, we demonstrated that MP could decrease expression levels of IBA1, Il-1α, TNFα, and C3 and suppress A1 neurotoxic reactive astrocyte activation in TSCI mouse models. Neurological function was evaluated using the Basso Mouse Scale (BMS) and Footprint Test. Results demonstrated that the neurological function of MP-treated injured mice was significantly increased. In conclusion, our study demonstrated that MP could attenuate astrocyte cell death, decrease microglia activation, suppress A1 astrocytes activation, and promote functional recovery after acute TSCI in mouse models.
Collapse
Affiliation(s)
- Hong-Jun Zou
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| | - Shi-Wu Guo
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| | - Lin Zhu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| | - Xu Xu
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| | - Jin-Bo Liu
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| |
Collapse
|
30
|
Oftadeh M, Ural N, LeVan P, Prabhu V, Haske M. The Evolution and Future of Spinal Drain for Thoracic Aortic Aneurysm Repair: A Review. J Cardiothorac Vasc Anesth 2021; 35:3362-3373. [PMID: 34154920 DOI: 10.1053/j.jvca.2021.04.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 11/11/2022]
Abstract
For decades, spinal drains for cerebrospinal fluid (CSF) pressure monitoring and drainage have been used as adjuncts to protect against spinal cord injury resulting from thoracic aortic aneurysm repair. There are many different approaches to placement and management of CSF drains, with no true consensus on best practice. Furthermore, the incidence of complications resulting from spinal drains largely has been stagnant. This review describes the history and rationale behind placement of CSF drains, explore various considerations, techniques, and equipment, and discuss potential considerations for developing more comprehensive protocols.
Collapse
Affiliation(s)
- Mina Oftadeh
- Department of Anesthesiology, Loyola University Medical Center, Maywood, IL.
| | - Nil Ural
- Department of Anesthesiology, Loyola University Medical Center, Maywood, IL
| | - Pierre LeVan
- Department of Anesthesiology, Franciscan Health Olympia Fields, Olympia Fields, IL
| | - Vikram Prabhu
- Department of Anesthesiology, Loyola University Medical Center, Maywood, IL
| | - Michael Haske
- Department of Anesthesiology, Loyola University Medical Center, Maywood, IL
| |
Collapse
|
31
|
Deng Y, Cai L, Wang F, Huang J, Wang H, Li L, Lv H. RETRACTED: Upregulated microRNA-381-5p strengthens the effect of dexmedetomidine preconditioning to protect against myocardial ischemia-reperfusion injury in mouse models by inhibiting CHI3L1. Int Immunopharmacol 2021; 92:107326. [PMID: 33461162 DOI: 10.1016/j.intimp.2020.107326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/31/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 1F, 2F, 3F, 4F and 5F, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0 [docs.google.com]). The journal requested the corresponding author comment on these concerns and provide the raw data. However, the authors were not responsive to the request for comment. Since original data could not be provided, the overall validity of the results could not be confirmed. Therefore, the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Yanan Deng
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054 Shaanxi, China
| | - Liang Cai
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054 Shaanxi, China
| | - Fang Wang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054 Shaanxi, China
| | - Jingyuan Huang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054 Shaanxi, China
| | - Haili Wang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054 Shaanxi, China
| | - Lu Li
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054 Shaanxi, China
| | - Haigang Lv
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054 Shaanxi, China.
| |
Collapse
|
32
|
Xiao J, Wan W, Zhang Y, Ma J, Yan L, Luo Y, Tang J. Administration of Dexmedetomidine Does Not Produce Long-Term Protective Effect on Testicular Damage Post Testicular Ischemia-Reperfusion Injury. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:315-321. [PMID: 33536744 PMCID: PMC7850429 DOI: 10.2147/dddt.s293926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022]
Abstract
Background After surgical correction of testicular torsion, up to 68% of ipsilateral testes undergo atrophy due to ischemia-reperfusion injury (IRI). Recent studies have shown that dexmedetomidine (Dex) alleviates IRI in various vital organs. However, those studies evaluated its protective effect on short-term reperfusion. Purpose We aimed to investigate whether Dex has a long-term protective effect against testicular injury after IRI. Materials and Methods A total of 24 New Zealand white rabbits were randomly divided into three groups (n = 8/group): the control group (saline-infused rabbits without IRI), the IRI group (saline-injected rabbits with IRI), and the Dex group (Dex-injected rabbits with IRI). The spermatic cord of rabbits in IRI and Dex groups was ligated for 4 h, and 1 h before reperfusion, Dex was administered intraperitoneally at a dose of 50 μg/kg body weight in group Dex, whereas saline was administered at the same dose to the IRI and control groups. Rabbits were kept alive for 4 weeks post reperfusion, then the testes were harvested, and the rabbits were euthanized. Results Four weeks post reperfusion, testicular volumes of the affected side decreased considerably in the IRI and Dex groups compared to the control group, with no significant difference between the IRI and Dex groups. Compared to the control group, the Johnson score and the mean seminiferous tubular diameters were significantly decreased in the IRI and Dex groups, but no significant differences were observed after administration of Dex. There were no significant differences in malondialdehyde and superoxide dismutase levels between the groups treated with and without Dex. Conclusion Dex administration 3 h after ischemia and 1 h before reperfusion did not demonstrate a significant protective effect against testicular injury 4 weeks after IRI in rabbits. Further research is needed to confirm the potential therapeutic effects of Dex by varying the experimental conditions.
Collapse
Affiliation(s)
- Jing Xiao
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Wenbo Wan
- Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Ying Zhang
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jun Ma
- Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lin Yan
- Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yukun Luo
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jie Tang
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
33
|
Kölükçü E, Parlaktaş BS, Kölükçü V, Firat F, Deresoy FA, Katar M, Kuyucu YE, Unsal V. Protective effects of dexmedetomidine on ischaemia-reperfusion injury in an experimental rat model of priapism. Andrologia 2021; 53:e13985. [PMID: 33474739 DOI: 10.1111/and.13985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
The study aimed to investigate the effects of dexmedetomidine against ischaemia-reperfusion injury occurring after priapism in a model of induced-priapism in rats. A total of 18 male rats were randomised into three groups. Group 1 was the control group. A priapism model was performed rats in Group 2 and then ischaemia-reperfusion injury was evaluated. Group 3 had similar procedures to the rats in Group 2. Rats in Group 3 additionally had 100 μg/kg dexmedetomidine administered intraperitoneally immediately after reperfusion. Blood and tissue samples were analysed. Biochemical analysis of blood samples revealed a decrease in the levels of the pro-inflammatory cytokines including interleukin-1 beta (IL-1 Beta), interleukin-6 (IL-6), and tumour necrosis factor-alpha (TNF-alpha) in Group 3 compared to Group 2 (p:.04, p:.009 and p:.009, respectively). Similarly, the highest malondialdehyde (MDA) level was in Group 2 (p:.002). The levels of antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities were significantly higher in Group 3 than that of Group 2 (p:.037 and p:.045, respectively). Direct microscopic examinations revealed positive changes in desquamation, oedema, inflammation and vasocongestion scores in Group 3 compared to Group 2 (p:.007, p:.008, p:.007 and p:.006, respectively). Dexmedetomidine has a protective effect against ischaemia-reperfusion injury in penile tissue.
Collapse
Affiliation(s)
- Engin Kölükçü
- Department of Urology, Gaziosmanpasa University, Tokat, Turkey
| | | | - Vildan Kölükçü
- Department of Anesthesia and Reanimation, Tokat State Hospital, Tokat, Turkey
| | - Fatih Firat
- Department of Urology, Tokat State Hospital, Tokat, Turkey
| | - Faik A Deresoy
- Department of Pathology, Gaziosmanpasa University, Tokat, Turkey
| | - Muzaffer Katar
- Department of Biochemistry, Gaziosmanpasa University, Tokat, Turkey
| | - Yunus Emre Kuyucu
- Department of Biostatistics, Gaziosmanpasa University, Tokat, Turkey
| | - Velid Unsal
- Faculty of Health Sciences and Central Research Laboratory, Mardin Artuklu University, Mardin, Turkey
| |
Collapse
|
34
|
Abstract
Ischemia/reperfusion (I/R) is a pathological process that occurs in numerous organs throughout the human body, and it is frequently associated with severe cellular damage and death. Recently it has emerged that ferroptosis, a new form of regulated cell death that is caused by iron-dependent lipid peroxidation, plays a significantly detrimental role in many I/R models. In this review, we aim to revise the pathological process of I/R and then explore the molecular pathogenesis of ferroptosis. Furthermore, we aim to evaluate the role that ferroptosis plays in I/R, providing evidence to support the targeting of ferroptosis in the I/R pathway may present as a therapeutic intervention to alleviate ischemia/reperfusion injury (IRI) associated cell damage and death.
Collapse
Affiliation(s)
- Hong-Fa Yan
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Qing-Zhang Tuo
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Qiao-Zhi Yin
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610041, China. E-mail:
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Center for Biotherapy, Chengdu, Sichuan 610041, China. E-mail:
| |
Collapse
|
35
|
Xue X, Fan J, Ma X, Liu Y, Han X, Leng Y, Yu J. Effects of local dexmedetomidine administration on the neurotoxicity of ropivacaine for sciatic nerve block in rats. Mol Med Rep 2020; 22:4360-4366. [PMID: 33000208 PMCID: PMC7533505 DOI: 10.3892/mmr.2020.11514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/14/2020] [Indexed: 11/20/2022] Open
Abstract
Dexmedetomidine, used as an adjuvant to local anesthetics (LAs), may prolong the duration of peripheral nerve block. However, the effect of dexmedetomidine on the neurotoxicity of LAs is not completely understood. The present study was designed to investigate the efficacy of two doses of dexmedetomidine as an adjuvant to ropivacaine and its protective effect against the neurotoxicity of LAs. Paw withdrawal thermal latency testing was used to detect the sensory blockade. Extensor postural thrust testing was used to detect the motor blockade. The results demonstrated that the addition of dexmedetomidine to ropivacaine prolonged the duration of sensory and motor blockade in a dose-dependent manner compared with ropivacaine alone. TUNEL staining was performed to examine apoptosis. Western blotting was used to detect the Cleaved caspase-3 expression levels. The results showed that the addition of dexmedetomidine to ropivacaine decreased the rate of apoptosis and caspase-3 expression levels in a dose-dependent manner compared with ropivacaine alone (P<0.05). In addition, the rate of apoptosis and caspase-3 expression levels were significantly lower in the high-dose dexmedetomidine group compared with the low-dose dexmedetomidine group (P<0.05). The results suggested that the addition of dexmedetomidine to ropivacaine for sciatic nerve block in rats not only prolonged the duration of sensory and motor block of the sciatic nerve, but also markedly alleviated ropivacaine-induced neurotoxicity by decreasing caspase-3-dependent sciatic nerve cell apoptosis. Furthermore, the present study indicated that dexmedetomidine was more effective at a dose of 20 µg/kg compared with 6 µg/kg.
Collapse
Affiliation(s)
- Xing Xue
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jun Fan
- Department of Anesthesiology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Xiaoli Ma
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yongqiang Liu
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xuena Han
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yufang Leng
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jinjia Yu
- School of Life Sciences, Central South University, Changsha, Hunan 410083, P.R. China
| |
Collapse
|
36
|
Dexmedetomidine: What's New for Pediatrics? A Narrative Review. J Clin Med 2020; 9:jcm9092724. [PMID: 32846947 PMCID: PMC7565844 DOI: 10.3390/jcm9092724] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past few years, despite the lack of approved pediatric labelling, dexmedetomidine’s (DEX) use has become more prevalent in pediatric clinical practice as well as in research trials. Its respiratory-sparing effects and bioavailability by various routes are only some of the valued features of DEX. In recent years the potential organ-protective effects of DEX, with the possibility for preserving neurocognitive function, has put it in the forefront of clinical and bench research. This comprehensive review focused on the pediatric literature but presents relevant, supporting adult and animal studies in order to detail the recent growing body of literature around the pharmacology, end-organ effects, organ-protective effects, alternative routes of administration, synergetic effects, and clinical applications, with considerations for the future.
Collapse
|
37
|
Effects of Pre-Cardiopulmonary Bypass Administration of Dexmedetomidine on Cardiac Injuries and the Inflammatory Response in Valve Replacement Surgery With a Sevoflurane Postconditioning Protocol: A Pilot Study. J Cardiovasc Pharmacol 2020; 74:91-97. [PMID: 31356535 PMCID: PMC6688713 DOI: 10.1097/fjc.0000000000000698] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Supplemental Digital Content is Available in the Text. Background: Preventing myocardial ischemia–reperfusion injury in on-pump cardiac surgeries remains an enormous challenge. Sevoflurane postconditioning has been effective at overcoming this challenge by modulating inflammatory mediators and ameliorating antioxidative stress. Dexmedetomidine (DEX) is a commonly used medication for cardiac patients with organ-protective properties that lead to positive outcomes. Whether DEX also has cardiac-protective properties and the associated mechanism in sevoflurane postconditioning–based valve replacement surgeries are unknown. Objective: This study was conducted to observe the effect of DEX administration before cardiopulmonary bypass (CPB) on myocardial injury, oxidative stress, and inflammatory response indicators in the peripheral blood. Methods: Twenty-eight eligible cardiac patients who underwent valve replacement surgery with standard sevoflurane postconditioning were included in the study. The patients were randomly divided into a DEX group and a non-DEX group according to whether DEX (0.5-µg/kg overload dose for 10 minutes and a 0.5-μg/kg/h maintenance dose) or saline was administered from induction to the beginning of CPB. The primary outcome was the cardiac troponin I concentration (cTnI) in the blood 24 hours after CPB. The levels of malondialdehyde (MDA), superoxide dismutase, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8) were also measured. Results: The mean cTnI at 24 hours after CPB was clearly decreased in the DEX group compared with that in the non-DEX group (4.16 ± 1.58 vs. 6.90 ± 3.73, P < 0.05). TNF-α levels were lower in the DEX group after CPB (T1–T5), with a significant difference found at 1–6 hours after CPB (1 hour, 19.03 vs. 28.09; 6 hours, 20.74 vs. 30.94, P < 0.05). The IL-6 and IL-8 concentrations in the DEX group were dramatically increased at 6 hours after CPB (P < 0.05). The MDA content and superoxide dismutase activity were comparable between the 2 groups. A lower proportion of anemia cases were noted after CPB in the DEX group than in the non-DEX group (non-DEX, 10% vs. DEX, 5%, P < 0.05). Conclusions: In valve replacement surgery with sevoflurane postconditioning, pre-CPB administration of DEX can reduce the cTnI level at 24 hours after CPB and brings synergic benefits of the inflammatory response.
Collapse
|
38
|
Zhang X, Cui J, Qian H, Wang B, Yan F, Zhao Z. CD200R Is Involved in the Anti-inflammatory Effect of Dexmedetomidine in Lipopolysaccharide-Stimulated Microglia. Inflammation 2020; 43:1707-1715. [PMID: 32394288 DOI: 10.1007/s10753-020-01244-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dexmedetomidine has been confirmed to have anti-inflammatory effect in numerous diseases. However, the detailed mechanism is still unknown. We explore the hypothesis that CD200R is related to the anti-inflammatory effect of dexmedetomidine. Primary microglia were treated with different concentrations of dexmedetomidine followed with lipopolysaccharide (LPS) for 24 h. Interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-10 (IL-10) were measured by enzyme-linked immunosorbent assay. Arginase 1 (Arg1), IL-6, and TNF-α were measured by TR-PCR. CD200R was observed by Western blot analysis. We found dexmedetomidine reduced LPS-induced IL-6 and TNF-α production and increase Arg1 in primary microglia. CD200R expression was downregulated by LPS and dexmedetomidine can attenuate the effect of LPS. Our results showed that CD200R may be related to the anti-inflammatory effect of dexmedetomidine.
Collapse
Affiliation(s)
- Xiaobao Zhang
- Department of Anesthesiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University (the First People's Hospital of Lianyungang), Lianyungang, 222000, China
| | - Jizheng Cui
- Department of Pain, Lianyungang Maternal and Child Health Hospital, Lianyungang, 222000, China
| | - Haitao Qian
- Department of Anesthesiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University (the First People's Hospital of Lianyungang), Lianyungang, 222000, China
| | - Bing Wang
- Department of Anesthesiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University (the First People's Hospital of Lianyungang), Lianyungang, 222000, China
| | - Fang Yan
- Department of Basic Medical Science, Kangda College of Nanjing Medical University, Lianyungang, 222000, China.
| | - Zhibin Zhao
- Department of Anesthesiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University (the First People's Hospital of Lianyungang), Lianyungang, 222000, China.
| |
Collapse
|
39
|
Dexmedetomidine pretreatment protects the heart against apoptosis in ischemia/reperfusion injury in diabetic rats by activating PI3K/Akt signaling in vivo and in vitro. Biomed Pharmacother 2020; 127:110188. [PMID: 32407987 DOI: 10.1016/j.biopha.2020.110188] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/26/2020] [Accepted: 04/17/2020] [Indexed: 01/26/2023] Open
Abstract
Dexmedetomidine (DEX) exerts cardioprotection against ischemia/reperfusion injury. However, the precise mechanisms underlying this cardioprotective effect in diabetic rats are still not fully understood. The aim of the present study was to investigate the cardioprotective mechanism of DEX pretreatment on myocardial ischemia/reperfusion (I/R) injury in diabetic rats. A total of 25 streptozotocin-induced diabetic rats were equally randomized into five groups: i) Sham, ii) DEX (100 μg/kg); iii) myocardial I/R; iv) myocardial I/R+DEX (10 μg/kg); and v) myocardial I/R+DEX (100 μg/kg) groups. Primary cardiomyocytes were cultured in DEX for 1 h, and then oxygen and glucose deprivation (OGD)/R for 36 h. These results showed that pretreatment with DEX significantly decreased the I/R-induced size of the myocardial infarction, structural damage, morphological changes and apoptosis in myocardial cells, as well as levels of creatinine kinase, malondialdehyde and cardiac troponin I, and increased the I/R-induced superoxide dismutase activity in vivo and in vitro. Furthermore, immunohistochemical staining and western blot analysis revealed that DEX pretreatment significantly increased the I/R-induced expression levels of B-cell lymphoma 2 (Bcl-2), phosphorylated phosphoinositide 3-kinase (pPI3K) and pAkt, and significantly decreased those of pBcl-2 associated agonist of cell death, Bcl-2-associated X protein and cleaved caspase 3 in vivo and in vitro. In addition, all of these cardioprotective effects of DEX were reversed by yohimbine and LY294002 pretreatment. These results suggested that DEX pretreatment may activate the PI3K/Akt signaling pathway in an α2 adrenoceptor-dependent manner. DEX pretreatment may exert cardioprotective effects against myocardial ischemia/reperfusion injury in diabetic rats through the I/R-induced inhibition of cell apoptosis by activating the PI3K/Akt signaling pathway.
Collapse
|
40
|
Liu XM, Chen QH, Hu Q, Liu Z, Wu Q, Liang SS, Zhang HG, Zhang Q, Zhang XK. Dexmedetomidine protects intestinal ischemia-reperfusion injury via inhibiting p38 MAPK cascades. Exp Mol Pathol 2020; 115:104444. [PMID: 32335082 DOI: 10.1016/j.yexmp.2020.104444] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/15/2022]
Abstract
Intestinal ischemia-reperfusion (I/R) is a life-threatening condition associated with high morbidity and mortality. Dexmedetomidine (DEX), an agonist of α2-adrenoceptor with sedation and analgesia effect, has recently been identified with protective function against I/R injury in multiple organs. However, the mechanism underlying the beneficial effect of DEX on intestine after I/R injury remained poorly understood. In the present study, using in both in vitro and in vivo models, we found that intestinal I/R injury was associated with the activation of p38 MAPK cascade, while DEX was capable of deactivating p38 MAPK and thus protect intestinal cells from apoptosis by inhibiting p38 MAPK-mediated mitochondrial depolarization and cytochrome c (Cyto C) release. Moreover, through inhibiting p38 MAPK activity, the downstream production of pro-inflammatory cytokines-regulated by NF-κB was also suppressed by DEX treatment, leading to the resolution of I/R-induced inflammation in intestine. In general, our study provided evidence that DEX protected intestine from I/R injury by inhibiting p38 MAPK-mediated mitochondrial apoptosis and inflammatory response.
Collapse
Affiliation(s)
- Xiao-Ming Liu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiu-Hong Chen
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qian Hu
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhen Liu
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiong Wu
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si-Si Liang
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Huai-Gen Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qin Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xue-Kang Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
41
|
Li R, Zhao K, Ruan Q, Meng C, Yin F. Bone marrow mesenchymal stem cell-derived exosomal microRNA-124-3p attenuates neurological damage in spinal cord ischemia-reperfusion injury by downregulating Ern1 and promoting M2 macrophage polarization. Arthritis Res Ther 2020; 22:75. [PMID: 32272965 PMCID: PMC7146970 DOI: 10.1186/s13075-020-2146-x] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/05/2020] [Indexed: 12/19/2022] Open
Abstract
Background Spinal cord ischemia-reperfusion injury (SCIRI) often leads to neurological damage and mortality. In this regard, understanding the pathology of SCIRI and preventing its development are of great clinic value. Methods Herein, we analyzed the role of bone marrow mesenchymal stem cell (BMMSC)-derived exosomal microRNA (miR)-124-3p in SCIRI. A SCIRI rat model was established, and the expression of Ern1 and M2 macrophage polarization markers (Arg1, Ym1, and Fizz) was determined using immunohistochemistry, immunofluorescence assay, RT-qPCR, and western blot analysis. Targeting relationship between miR-124-3p and Ern1 was predicted using bioinformatic analysis and verified by dual-luciferase reporter assay. Macrophages were co-cultured with miR-124-3p-containing BMMSC-derived exosomes. M2 macrophages were identified using flow cytometry, and the expression of Arg1, Ym1, and Fizz was determined. In addition, SCIRI rats were injected with miR-124-3p-containing exosomes, spinal cord cell apoptosis was observed using TUNEL assay, and the pathological condition was evaluated with H&E staining. Results In SCIRI, Ern1 was highly expressed and M2 polarization markers were poorly expressed. Silencing Ern1 led to elevated expression of M2 polarization markers. MiR-124-3p targeted and negatively regulated Ern1. Exosomal miR-124-3p enhanced M2 polarization. Highly expressed exosomal miR-124-3p impeded cell apoptosis and attenuated SCIRI-induced tissue impairment and nerve injury. miR-124-3p from BMMSC-derived exosomes ameliorated SCIRI and its associated nerve injury through inhibiting Ern1 and promoting M2 polarization. Conclusion In summary, exosomal miR-124-3p derived from BMMSCs attenuated nerve injury induced by SCIRI by regulating Ern1 and M2 macrophage polarization.
Collapse
Affiliation(s)
- Ran Li
- Department of Spine Surgery, China-Japan Union Hospital, Jilin University, No. 126, Xiantai Street, Changchun, 130033, Jilin Province, People's Republic of China
| | - Kunchi Zhao
- Department of Spine Surgery, China-Japan Union Hospital, Jilin University, No. 126, Xiantai Street, Changchun, 130033, Jilin Province, People's Republic of China.
| | - Qing Ruan
- Department of Spine Surgery, China-Japan Union Hospital, Jilin University, No. 126, Xiantai Street, Changchun, 130033, Jilin Province, People's Republic of China
| | - Chunyang Meng
- Department of Spine Surgery, China-Japan Union Hospital, Jilin University, No. 126, Xiantai Street, Changchun, 130033, Jilin Province, People's Republic of China
| | - Fei Yin
- Department of Spine Surgery, China-Japan Union Hospital, Jilin University, No. 126, Xiantai Street, Changchun, 130033, Jilin Province, People's Republic of China.
| |
Collapse
|
42
|
Wu SJ, Lin ZH, Lin YZ, Rao ZH, Lin JF, Wu LP, Li L. Dexmedetomidine Exerted Anti-arrhythmic Effects in Rat With Ischemic Cardiomyopathy via Upregulation of Connexin 43 and Reduction of Fibrosis and Inflammation. Front Physiol 2020; 11:33. [PMID: 32116751 PMCID: PMC7020758 DOI: 10.3389/fphys.2020.00033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/15/2020] [Indexed: 12/20/2022] Open
Abstract
Background Persistent myocardial ischemia post-myocardial infarction can lead to fatal ventricular arrhythmias such as ventricular tachycardia and fibrillation, both of which carry high mortality rates. Dexmedetomidine (Dex) is a highly selective α2-agonist used in surgery for congenital cardiac disease because of its antiarrhythmic properties. Dex has previously been reported to prevent or terminate various arrhythmias. The purpose of the present study was to determine the anti-arrhythmic properties of Dex in the context of ischemic cardiomyopathy (ICM) after myocardial infarction. Methods and Results We randomly allocated 48 rats with ICM, created by persistent ligation of the left anterior descending artery for 4 weeks, into six groups: Sham (n = 8), Sham + BML (n = 8), ICM (n = 8), ICM + BML (n = 8), ICM + Dex (n = 8), and ICM + Dex + BML (n = 8). Treatments started after ICM was confirmed (the day after echocardiographic measurement) and continued for 4 weeks (inject intraperitoneally, daily). Dex inhibited the generation of collagens, cytokines, and other inflammatory mediators in rats with ICM via the suppression of NF-κB activation and increased the distribution of connexin 43 (Cx43) via phosphorylation of adenosine 5′-monophosphate-activated protein kinase (AMPK). Dex reduced the occurrence of spontaneous ventricular arrhythmias (ventricular premature beat or ventricular tachycardia), decreased the inducibility quotient of ventricular arrhythmias induced by PES, and partly improved cardiac contraction. The AMPK antagonist BML-275 dihydrochloride (BML) partly weakened the cardioprotective effect of Dex. Conclusion Dex conferred anti-arrhythmic effects in the context of ICM via upregulation of Cx43 and suppression of inflammation and fibrosis. The anti-arrhythmic and anti-inflammatory properties of Dex may be mediated by phosphorylation of AMPK and subsequent suppression of NF-κB activation.
Collapse
Affiliation(s)
- Shu-Jie Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhong-Hao Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuan-Zheng Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-Heng Rao
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia-Feng Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lian-Pin Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
43
|
Wang YG, Liu CZ, Li YZ, Peng Y, Yan S. Cotreatments with Dex and Na2SeO3further improved antioxidant and anti-inflammatory protection of myocardial cells from I/R injury compared to their individual treatments. Free Radic Res 2020; 54:76-90. [PMID: 31909644 DOI: 10.1080/10715762.2019.1707198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yan-Gui Wang
- Department of General Practice, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Cui-Zhong Liu
- Department of General Practice, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Ying-Zhao Li
- Department of General Practice, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - You Peng
- Department of Geriatrics, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Sulan Yan
- Cardiovascular Department, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, China
| |
Collapse
|
44
|
Han H, Dai D, Hu J, Zhu J, Lu L, Tao G, Zhang R. Dexmedetomidine improves cardiac function and protects against maladaptive remodeling following myocardial infarction. Mol Med Rep 2019; 20:5183-5189. [PMID: 31661145 PMCID: PMC6854534 DOI: 10.3892/mmr.2019.10774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/09/2019] [Indexed: 01/01/2023] Open
Abstract
Dexmedetomidine (DEX), a highly specific and selective α2 adrenergic receptor agonist, has been demonstrated to possess potential cardioprotective effects. However, the mechanisms underlying this process remain to be fully illuminated. In the present study, a myocardial infarction (MI) animal model was generated by permanently ligating the left anterior descending coronary artery in mice. Cardiac function and collagen content were evaluated by transthoracic echocardiography and picrosirius red staining, respectively. Apoptosis was determined by the relative expression levels of Bax and Bcl-2 and the myocardial caspase-3 activity. Additionally, nicotinamide adenine dinucleotide phosphate oxidase (NOX)-derived oxidative stress was evaluated by the relative expression of Nox2 and Nox4, along with the myocardial contents of malondialdehyde (MDA) and superoxide dismutase (SOD) activity. It was demonstrated that intraperitoneal DEX treatment (20 µg/kg/day) improved the systolic function of the left ventricle, and decreased the fibrotic changes in post-myocardial infarction mice, which was paralleled by a decrease in the levels of apoptosis. Subsequent experiments indicated that the restoration of redox signaling was achieved by DEX administration, and the over-activation of NOXs, including Nox2 and Nox4, was markedly inhibited. In conclusion, this present study suggested that DEX was cardioprotective and limited the excess production of NOX-derived ROS in ischemic heart disease, implying that DEX is a promising novel drug, especially for patients who have suffered MI.
Collapse
Affiliation(s)
- Hui Han
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Daopeng Dai
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jinquan Hu
- Department of Orthopedics, Changzheng Hospital Affiliated with Second Military Medical University, Shanghai 200003, P.R. China
| | - Jinzhou Zhu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Lin Lu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Guorong Tao
- Department of Anesthesiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Ruiyan Zhang
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
45
|
Gao J, Sun Z, Xiao Z, Du Q, Niu X, Wang G, Chang YW, Sun Y, Sun W, Lin A, Bresnahan JC, Maze M, Beattie MS, Pan JZ. Dexmedetomidine modulates neuroinflammation and improves outcome via alpha2-adrenergic receptor signaling after rat spinal cord injury. Br J Anaesth 2019; 123:827-838. [PMID: 31623841 DOI: 10.1016/j.bja.2019.08.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/17/2019] [Accepted: 08/17/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Spinal cord injury induces inflammatory responses that include the release of cytokines and the recruitment and activation of macrophages and microglia. Neuroinflammation at the lesion site contributes to secondary tissue injury and permanent locomotor dysfunction. Dexmedetomidine (DEX), a highly selective α2-adrenergic receptor agonist, is anti-inflammatory and neuroprotective in both preclinical and clinical trials. We investigated the effect of DEX on the microglial response, and histological and neurological outcomes in a rat model of cervical spinal cord injury. METHODS Anaesthetised rats underwent unilateral (right) C5 spinal cord contusion (75 kdyne) using an impactor device. The locomotor function, injury size, and inflammatory responses were assessed. The effect of DEX was also studied in a microglial cell culture model. RESULTS DEX significantly improved the ipsilateral upper-limb motor dysfunction (grooming and paw placement; P<0.0001 and P=0.0012), decreased the injury size (P<0.05), spared white matter (P<0.05), and reduced the number of activated macrophages (P<0.05) at the injury site 4 weeks post-SCI. In DEX-treated rats after injury, tissue RNA expression indicated a significant downregulation of pro-inflammatory markers (e.g. interleukin [IL]-1β, tumour necrosis factor-α, interleukin (IL)-6, and CD11b) and an upregulation of anti-inflammatory and pro-resolving M2 responses (e.g. IL-4, arginase-1, and CD206) (P<0.05). In lipopolysaccharide-stimulated cultured microglia, DEX produced a similar inflammation-modulatory effect as was seen in spinal cord injury. The benefits of DEX on these outcomes were mostly reversed by an α2-adrenergic receptor antagonist. CONCLUSIONS DEX significantly improves neurological outcomes and decreases tissue damage after spinal cord injury, which is associated with modulation of neuroinflammation and is partially mediated via α2-adrenergic receptor signaling.
Collapse
Affiliation(s)
- Jiandong Gao
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhihua Sun
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Xiao
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Qihang Du
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Xinhuan Niu
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Gongming Wang
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yu-Wen Chang
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA
| | - Yongtao Sun
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Qianfoshan Hospital, the First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Wei Sun
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Anaesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Amity Lin
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jacqueline C Bresnahan
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mervyn Maze
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Michael S Beattie
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Jonathan Z Pan
- Department of Anaesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
46
|
Qian Y, Wang Q, Jiao J, Wang G, Gu Z, Huang D, Wang Z. Intrathecal injection of dexmedetomidine ameliorates chronic neuropathic pain via the modulation of MPK3/ERK1/2 in a mouse model of chronic neuropathic pain. Neurol Res 2019; 41:1059-1068. [PMID: 31584354 DOI: 10.1080/01616412.2019.1672391] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Objective: Despite the application of dexmedetomidine (DEX) as a perioperative adjuvant in local analgesia, the exact analgesic mechanism underpinning chronic neuropathic pain (CNP) awaits our elucidation. Methods: We investigated the molecular mechanisms of the anti-nociceptive effect of DEX on neuropathic pain in a mouse model induced by chronic constriction injury (CCI). Results: DEX administration significantly increased the paw withdrawal latency (PWL) values 0.5 to 2 h post-injection in CCI-induced CNP mice at day 5 to 21 versus dimethyl sulfoxide (DMSO)-treated mice, confirming its analgesic effect. The c-Fos expression was significantly elevated in CCI mice versus the sham-operated group, whereas the elevation was mitigated by DEX injection. Subsequently, the involvement of MKP1 and MKP3 in the pathogenesis of chronic neuropathic pain was evaluated. Western blotting analyses revealed significant decrease in both MKP1 and MKP3 in the spinal cord in CCI group versus the sham group. DEX markedly elevated the MKP3 expression and modestly reduced the MKP1 expression, with insignificant difference in the latter. Co-injection of BCI (an MKP3 inhibitor) and DEX evidently reduced the PWL values in CCI mice. Furthermore, DEX significantly downregulated the phosphorylation of extracellular-signal-regulated kinase (ERK) 1/2, down-stream effector of MKP3 in CCI mice, whereas the downregulation was reversed by BCI. Conclusion: We confirmed that DEX exerts the analgesic effect on chronic neuropathic pain via the regulation of MKP3/ERK1/2 signaling pathway, which may contribute to clarification of the molecular mechanism and novel therapy for chronic neuropathic pain.
Collapse
Affiliation(s)
- Yiling Qian
- Department of Anesthesiology, Wuxi People's Hospital of Nanjing Medical University , Wuxi , Jiangsu , China
| | - Qianlun Wang
- Department of Anesthesiology, Wuxi People's Hospital of Nanjing Medical University , Wuxi , Jiangsu , China
| | - Jiantong Jiao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University , Wuxi , Jiangsu , China
| | - Guilong Wang
- Department of Anesthesiology, Wuxi People's Hospital of Nanjing Medical University , Wuxi , Jiangsu , China
| | - Zhengfeng Gu
- Department of Anesthesiology, Wuxi People's Hospital of Nanjing Medical University , Wuxi , Jiangsu , China
| | - Dongxiao Huang
- Department of Anesthesiology, Wuxi People's Hospital of Nanjing Medical University , Wuxi , Jiangsu , China
| | - Zhiping Wang
- Department of Anesthesiology, Wuxi People's Hospital of Nanjing Medical University , Wuxi , Jiangsu , China.,Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University , Xuzhou , Jiangsu , China
| |
Collapse
|
47
|
Hao J, Wang P, Pei D, Jia B, Hu Q. Rhein lysinate improves motor function in rats with spinal cord injury via inhibiting p38 MAPK pathway. Kaohsiung J Med Sci 2019; 35:765-771. [PMID: 31483087 DOI: 10.1002/kjm2.12123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jian Hao
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| | - Ping Wang
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| | - Dai‐Ping Pei
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| | - Bin Jia
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| | - Qun‐Sheng Hu
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| |
Collapse
|
48
|
Yuan M, Meng XW, Ma J, Liu H, Song SY, Chen QC, Liu HY, Zhang J, Song N, Ji FH, Peng K. Dexmedetomidine protects H9c2 cardiomyocytes against oxygen-glucose deprivation/reoxygenation-induced intracellular calcium overload and apoptosis through regulating FKBP12.6/RyR2 signaling. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3137-3149. [PMID: 31564830 PMCID: PMC6730549 DOI: 10.2147/dddt.s219533] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/23/2019] [Indexed: 12/30/2022]
Abstract
Purpose Intracellular calcium ([Ca2+]i) overload is a major cause of cell injury during myocardial ischemia/reperfusion (I/R). Dexmedetomidine (DEX) has been shown to exert anti-inflammatory and organ protective effects. This study aimed to investigate whether pretreatment with DEX could protect H9c2 cardiomyocytes against oxygen-glucose deprivation/reoxygenation (OGD/R) injury through regulating the Ca2+ signaling. Methods H9c2 cardiomyocytes were subjected to OGD for 12 h, followed by 3 h of reoxygenation. DEX was administered 1 h prior to OGD/R. Cell viability, lactate dehydrogenase (LDH) release, level of [Ca2+]i, cell apoptosis, and the expression of 12.6-kd FK506-binding protein/ryanodine receptor 2 (FKBP12.6/RyR2) and caspase-3 were assessed. Results Cells exposed to OGD/R had decreased cell viability, increased LDH release, elevated [Ca2+]i level and apoptosis rate, down-regulated expression of FKBP12.6, and up-regulated expression of phosphorylated-Ser2814-RyR2 and cleaved caspase-3. Pretreatment with DEX significantly blocked the above-mentioned changes, alleviating the OGD/R-induced injury in H9c2 cells. Moreover, knockdown of FKBP12.6 by small interfering RNA abolished the protective effects of DEX. Conclusion This study indicates that DEX pretreatment protects the cardiomyocytes against OGD/R-induced injury by inhibiting [Ca2+]i overload and cell apoptosis via regulating the FKBP12.6/RyR2 signaling. DEX may be used for preventing cardiac I/R injury in the clinical settings.
Collapse
Affiliation(s)
- Mei Yuan
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China.,Department of Anesthesiology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, People's Republic of China
| | - Xiao-Wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Jiao Ma
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA 95817, USA
| | - Shao-Yong Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Qing-Cai Chen
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Hua-Yue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Juan Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Nan Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Fu-Hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| |
Collapse
|
49
|
Xue BB, Chen BH, Tang YN, Weng CW, Lin LN. Dexmedetomidine protects against lung injury induced by limb ischemia-reperfusion via the TLR4/MyD88/NF-κB pathway. Kaohsiung J Med Sci 2019; 35:672-678. [PMID: 31373750 DOI: 10.1002/kjm2.12115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/10/2019] [Indexed: 12/15/2022] Open
Abstract
Dexmedetomidine (DEX) can protect the lung from ischemia-reperfusion (I/R) injury, but the underlying mechanisms are not fully understood. The aims of this study were to determine whether DEX attenuates lung injury following lower extremity I/R and to investigate the related toll-like receptor 4 (TLR4) signaling pathway. Twenty-eight SD rats were divided into four groups (n = 7): Sham, I/R, I/R + DEX (25 μg/kg prior to ischemia), and I/R + DEX + Atip (250 μg/kg atipamezole before DEX treatment). Lower extremity I/R was induced by left femoral artery clamping for 3 hours and followed by 2 hours reperfusion. Quantitative alveolar damage and the wet/dry (W/D) ratio were calculated. Interleukin (IL)-1, IL-6, and tumor necrosis factor (TNF)-α in the bronchoalveolar lavage fluid (BALF) and serum and myeloperoxidase (MPO) in the lung were measured. The TLR4 and MyD88 mRNA expression levels were measured by RT-PCR, nuclear factor (NF)-κB, and phosphorylated NF-κB by western blot, respectively. Quantitative alveolar damage, W/D ratio, MPO, BALF and serum IL-1, IL-6, and TNF-α, and TLR4, MyD88, NF-κB, and p-NF-κB expression significantly increased in the I/R group relative to the Sham group. DEX preconditioning significantly reduced lung edema, and histological injury relative to the I/R group. Serum and BALF IL-1, IL-6, and TNF-α levels, MPO activity and TLR4, MyD88, NF-κB, and p-NF-κB expression were also significantly reduced in the I/R + DEX group compared with the I/R group. Atipamezole partially reversed all the aforementioned effects. DEX preconditioning protects the lungs against lower extremity I/R injury via α2-adrenoceptor-dependent and α2-adrenoceptor-independent mechanisms. It also suppresses the TLR4 pathway and reduces inflammation.
Collapse
Affiliation(s)
- Bin-Bin Xue
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bai-Hui Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ya-Ning Tang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cheng-Wei Weng
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li-Na Lin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
50
|
Ha Sen Ta Na, Nuo M, Meng QT, Xia ZY. The Pathway of Let-7a-1/2-3p and HMGB1 Mediated Dexmedetomidine Inhibiting Microglia Activation in Spinal Cord Ischemia-Reperfusion Injury Mice. J Mol Neurosci 2019; 69:106-114. [PMID: 31190218 DOI: 10.1007/s12031-019-01338-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023]
Abstract
Microglial cell activation after spinal cord ischemia-reperfusion injury (SCIRI) commonly causes the secondary nerve motion function injury. This study aims to study the mechanism by which the drug dexmedetomidine (DEX) inhibits microglial cell activation and improves motion function of SCIRI mice. Mice SCIRI model was established, and microglia from spinal cord were isolated and cultured for subsequent molecule analysis of let-7a-1-3p, let-7a-2-3p, HMGB1, TNF-α, and IL-6. DEX was given by intraperitoneal injection. Mice motion function was evaluated by Basso mouse score. In vitro microglial cells were subjected to oxygen and glucose deprivation/reoxygenation (OGD/R) to imitate ischemia-reperfusion injury stimulation. DEX injection improves the mouse motion function in SCIRI model and upregulates let-7a-1/2-3p expression in the isolated activated microglia from SCIRI mice. In OGD/R-stimulated microglia, DEX treatment also caused the inactivation of cells, the upregulation of let-7a-1/2-3p expression, and the downregulation of HMGB1 expression. While the co-silencing of let-7a-1/2-3p in microglia in addition to DEX treatment restored the activation of microglia. HMGB1 is a targeted gene for let-7a-1/2-3p and negatively regulated by them. HMGB1 knockdown abrogates the pro-activation impact on microglial cell by let-7a-1/2-3p silencing. DEX inhibits the activation of microglial cell in the spinal cord of SCIRI mice, mediated by the let-7a-1/2-3p/HMGB1 pathway.
Collapse
Affiliation(s)
- Ha Sen Ta Na
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Ming Nuo
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|