1
|
Zhang Y, Wang D, Wu X, Zhao T, He M, He Y, Meng C. Targeting the lncRNA GAS5/TLR4/NLRP3 signaling cascade inhibits endometrial stromal cell pyroptosis and prevents the progression of intrauterine adhesions. J Reprod Immunol 2025; 168:104450. [PMID: 39951898 DOI: 10.1016/j.jri.2025.104450] [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: 07/22/2024] [Revised: 12/04/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
Intrauterine adhesion (IUA) poses a serious threat to women's health, and its specific pathogenesis has not yet been elucidated. Our study found through high-throughput sequencing that differentially expressed genes of the endometrial tissues from healthy individuals or IUA patients were enriched in the toll-like receptor (TLR), nuclear factor-kappa B (NF-kB), and nucleotide-binding oligomerization domain-like receptor (NLR) signaling pathways. Meanwhile, we observed that compared to the controls, long non-coding RNA (lncRNA) growth arrest-specific transcripts 5 (GAS5) was significantly upregulated in the endometrial tissue of IUA patients and scratching/lipopolysaccharide (LPS)-induced IUA model mice. Subsequently, results from the functional verification assay, including hematoxylin-eosin staining, enzyme-linked immunosorbent assay, and western blot, showed that knockdown of GAS5 improved endometrial injury and uterine adhesions, decreased the levels of TIMP1, α-SMA, Vimentin, and COL1A1, but elevated MMP9 level to reduce excessive accumulation of extracellular matrix (ECM), and inhibited the expression of NLRP3, cleaved caspase-1, GSDMD, and nuclear p65 to ameliorate pyroptosis in IUA model mice. As confirmed by bioinformatics analysis and dual luciferase reporter gene system, GAS5 sponged microRNA (miR)-205-5p to upregulate TLR4, further activating the NF-kB and NLRP3 signaling in endometrial stromal cells (ESCs). The in vitro functional recovery experiments suggested that GAS5 knockdown alleviated LPS-induced activation of the NF-kB and NLRP3 signaling, pyroptotic cell death, and ECM deposition in ESCs, which was counteracted by overexpressing TLR4 and NLRP3. In a word, our study proved that targeting the GAS5/TLR4/NLRP3 signaling cascade inhibits ESCs pyroptosis and prevents the progression of IUA, providing promising therapeutic strategies for IUA disease.
Collapse
Affiliation(s)
- Yifeng Zhang
- Gynecology Department, Yunnan First People's Hospital, Kunming, Yunan 650032, China; The Affiliated Hospital of Kunming University of Science and technology, Kunming, Yunan 650032, China.
| | - Dongjie Wang
- Gynecology Department, Yunnan First People's Hospital, Kunming, Yunan 650032, China; The Affiliated Hospital of Kunming University of Science and technology, Kunming, Yunan 650032, China.
| | - Xiaomei Wu
- Gynecology Department, Yunnan First People's Hospital, Kunming, Yunan 650032, China; The Affiliated Hospital of Kunming University of Science and technology, Kunming, Yunan 650032, China.
| | - Ting Zhao
- Gynecology Department, Yunnan First People's Hospital, Kunming, Yunan 650032, China; The Affiliated Hospital of Kunming University of Science and technology, Kunming, Yunan 650032, China.
| | - Ming He
- Kunming Medical University, Kunming, Yunan 650500, China.
| | - Yunyu He
- Gynecology Department, Yunnan First People's Hospital, Kunming, Yunan 650032, China; The Affiliated Hospital of Kunming University of Science and technology, Kunming, Yunan 650032, China.
| | - Chunmei Meng
- Gynecology Department, Yunnan First People's Hospital, Kunming, Yunan 650032, China; The Affiliated Hospital of Kunming University of Science and technology, Kunming, Yunan 650032, China.
| |
Collapse
|
2
|
Wang K, Wang Y, Zhang T, Chang B, Fu D, Chen X. The Role of Intravenous Anesthetics for Neuro: Protection or Toxicity? Neurosci Bull 2025; 41:107-130. [PMID: 39153174 PMCID: PMC11748649 DOI: 10.1007/s12264-024-01265-4] [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: 12/15/2023] [Accepted: 03/15/2024] [Indexed: 08/19/2024] Open
Abstract
The primary intravenous anesthetics employed in clinical practice encompass dexmedetomidine (Dex), propofol, ketamine, etomidate, midazolam, and remimazolam. Apart from their established sedative, analgesic, and anxiolytic properties, an increasing body of research has uncovered neuroprotective effects of intravenous anesthetics in various animal and cellular models, as well as in clinical studies. However, there also exists conflicting evidence pointing to the potential neurotoxic effects of these intravenous anesthetics. The role of intravenous anesthetics for neuro on both sides of protection or toxicity has been rarely summarized. Considering the mentioned above, this work aims to offer a comprehensive understanding of the underlying mechanisms involved both in the central nerve system (CNS) and the peripheral nerve system (PNS) and provide valuable insights into the potential safety and risk associated with the clinical use of intravenous anesthetics.
Collapse
Affiliation(s)
- Kaixin Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Bingcheng Chang
- The Second Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550003, China
| | - Daan Fu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
3
|
He Y, Wang J, Ying C, Xu KL, Luo J, Wang B, Gao J, Yin Z, Zhang Y. The interplay between ferroptosis and inflammation: therapeutic implications for cerebral ischemia-reperfusion. Front Immunol 2024; 15:1482386. [PMID: 39582857 PMCID: PMC11583640 DOI: 10.3389/fimmu.2024.1482386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/14/2024] [Indexed: 11/26/2024] Open
Abstract
Stroke ranks as the second most significant contributor to mortality worldwide and is a major factor in disability. Ischemic strokes account for 71% of all stroke incidences globally. The foremost approach to treating ischemic stroke prioritizes quick reperfusion, involving methods such as intravenous thrombolysis and endovascular thrombectomy. These techniques can reduce disability but necessitate immediate intervention. After cerebral ischemia, inflammation rapidly arises in the vascular system, producing pro-inflammatory signals that activate immune cells, which in turn worsen neuronal injury. Following reperfusion, an overload of intracellular iron triggers the Fenton reaction, resulting in an excess of free radicals that cause lipid peroxidation and damage to cellular membranes, ultimately leading to ferroptosis. The relationship between inflammation and ferroptosis is increasingly recognized as vital in the process of cerebral ischemia-reperfusion (I/R). Inflammatory processes disturb iron balance and encourage lipid peroxidation (LPO) through neuroglial cells, while also reducing the activity of antioxidant systems, contributing to ferroptosis. Furthermore, the lipid peroxidation products generated during ferroptosis, along with damage-associated molecular patterns (DAMPs) released from ruptured cell membranes, can incite inflammation. Given the complex relationship between ferroptosis and inflammation, investigating their interaction in brain I/R is crucial for understanding disease development and creating innovative therapeutic options. Consequently, this article will provide a comprehensive introduction of the mechanisms linking ferroptosis and neuroinflammation, as well as evaluate potential treatment modalities, with the goal of presenting various insights for alleviating brain I/R injury and exploring new therapeutic avenues.
Collapse
Affiliation(s)
- Yuxuan He
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jingyi Wang
- Faculty of Chinese Medicine of Macau University of Science and
Technology, Macao, Macao SAR, China
| | - Chunmiao Ying
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Kang Li Xu
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jingwen Luo
- Faculty of Chinese Medicine of Macau University of Science and
Technology, Macao, Macao SAR, China
| | - Baiqiao Wang
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jing Gao
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zaitian Yin
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yunke Zhang
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Tao Z, Li P, Zhao X. Progress on the Mechanisms and Neuroprotective Benefits of Dexmedetomidine in Brain Diseases. Brain Behav 2024; 14:e70116. [PMID: 39482839 PMCID: PMC11527817 DOI: 10.1002/brb3.70116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/27/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024] Open
Abstract
INTRODUCTION Dexmedetomidine, a highly specific α2 agonist, has been extensively utilized in clinical sedation and surgical anesthesia since its introduction in 2000 due to its excellent sympatholytic, sedative, and analgesic effects. This review aimed to identify new approaches for the treatment of patients with brain disorders by thoroughly describing the mechanism of action of dexmedetomidine and examining its neuroprotective effects from the standpoints of basic and clinical research. METHODS The PubMed and Web of Science databases were searched using the keywords dexmedetomidine and related brain diseases, although relevant articles from the last decade were included for detailed summarization and analysis. RESULTS Dexmedetomidine has shown strong neuroprotective effects, such as protection of the blood-brain barrier, decreased neuronal death, maintained hemodynamic stability, and reduced postoperative agitation and cognitive dysfunction. Furthermore, dexmedetomidine has been shown to exert various neuroprotective effects, including anti-inflammatory and antioxidative stress effects, modulation of autophagy, and reduction of apoptosis in cerebral diseases. CONCLUSIONS Dexmedetomidine acts as a neuroprotective agent against brain diseases during all phases of treatment. However, clinical trials with larger sample sizes are required to optimize dosage and dosing strategies.
Collapse
Affiliation(s)
- Zhenxing Tao
- Wuxi Medical SchoolJiangnan UniversityWuxiChina
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
| | - Pengpeng Li
- Wuxi Medical SchoolJiangnan UniversityWuxiChina
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
| | - Xudong Zhao
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
- Wuxi Neurosurgical InstituteWuxiChina
| |
Collapse
|
5
|
Chen B, Di B. Endogenous Ligands of TLR4 in Microglia: Potential Targets for Related Neurological Diseases. Curr Drug Targets 2024; 25:953-970. [PMID: 39234911 DOI: 10.2174/0113894501316051240821060249] [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: 04/08/2024] [Revised: 07/09/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Chronic inflammation mediated by microglia is a cause of some neuroinflammatory diseases. TLR4, a natural immune receptor on microglia, plays an important role in the occurrence of inflammation and the process of diseases. TLR4 can be activated by a variety of ligands to trigger inflammatory responses, including endogenous ligands HMGB1, S100A8/9, Heme, and Fetuin-A. As ligands derived from the body itself, they have the ability to bind directly to TLR4 and can be used as inducers of aseptic inflammation. In the past 20 years, targeting ligands rather than receptors has become an emerging therapeutic strategy for the treatment of diseases, so understanding the relationship between microglia, TLR4, TLR4 ligands, and corresponding diseases may have new implications for the treatment of diseases. In the article, we will discuss the TLR4 and the endogenous substances that can activate the TLR4 signaling pathway and present literature support for their role in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Bo Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P.R. China
- Office of China National Narcotics Control Commission, China Pharmaceutical University, Joint Laboratory on Key Technologies of Narcotics Control, Nanjing, 210009, P.R. China
| | - Bin Di
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P.R. China
- Office of China National Narcotics Control Commission, China Pharmaceutical University, Joint Laboratory on Key Technologies of Narcotics Control, Nanjing, 210009, P.R. China
| |
Collapse
|
6
|
Seong H, Jeong D, Kim EH, Yoon KS, Na D, Yoon SZ, Cho JE. MicroRNA-323-5p Involved in Dexmedetomidine Preconditioning Impart Neuroprotection. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1518. [PMID: 37763638 PMCID: PMC10532972 DOI: 10.3390/medicina59091518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
Background and Objectives: Cerebral ischemia is one of the major preoperative complications. Dexmedetomidine is a well-known sedative-hypnotic agent that has potential organ-protective effects. We examine the miRNAs associated with preconditioning effects of dexmedetomidine in cerebral ischemia. Materials and Methods: Transient infarcts were induced in mice via reperfusion after temporary occlusion of one side of the middle cerebral artery. A subset of these mice was exposed to dexmedetomidine prior to cerebral infarction and miRNA profiling of the whole brain was performed. We administered dexmedetomidine and miRNA-323-5p mimic/inhibitor to oxygen-glucose deprivation/reoxygenation astrocytes. Additionally, we administered miR-323-5p mimic and inhibitor to mice via intracerebroventricular injection 2 h prior to induction of middle cerebral artery occlusion. Results: The infarct volume was significantly lower in the dexmedetomidine-preconditioned mice. Analysis of brain samples revealed an increased expression of five miRNAs and decreased expression of three miRNAs in the dexmedetomidine-pretreated group. The viability of cells significantly increased and expression of miR-323-5p was attenuated in the dexmedetomidine-treated oxygen-glucose deprivation/reoxygenation groups. Transfection with anti-miR-323-5p contributed to increased astrocyte viability. When miRNA-323-5p was injected intraventricularly, infarct volume was significantly reduced when preconditioned with the miR-323-5p inhibitor compared with mimic and negative control. Conclusions: Dexmedetomidine has a protective effect against transient neuronal ischemia-reperfusion injury and eight specific miRNAs were profiled. Also, miRNA-323-5p downregulation has a cell protective effect under ischemic conditions both in vivo and in vitro. Our findings suggest the potential of the miR-323-5p inhibitor as a therapeutic agent against cerebral infarction.
Collapse
Affiliation(s)
- Hyunyoung Seong
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Daun Jeong
- Institute for Healthcare Service Innovation, Korea University, Seoul 02841, Republic of Korea
| | - Eung Hwi Kim
- Institute for Healthcare Service Innovation, Korea University, Seoul 02841, Republic of Korea
| | - Kyung Seob Yoon
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Donghyun Na
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seung Zhoo Yoon
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jang Eun Cho
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
7
|
Zheng J, Chen X, Zhan JB, Li CW, Wei X, Jiang HY. CircARF3 Mitigates Allergic Rhinitis through Targeting microRNA-205-5p/Sirtuin 5 Axis. Int Arch Allergy Immunol 2023; 184:1056-1070. [PMID: 37343537 DOI: 10.1159/000530351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/13/2023] [Indexed: 06/23/2023] Open
Abstract
INTRODUCTION Circular RNAs (circRNAs) are essential in the progression of allergic rhinitis (AR). The purpose of this research was to examine the role of circRNA ADP-ribosylation factor 3 (circARF3) in the pathogenesis of AR. METHODS To generate an animal model of AR, mice were treated with house dust mite (HDM), and mice nasal epithelial cells (NEpCs) were treated with IL-4/IL-13 to imitate the inflammatory damage of AR in vitro. Sanger sequencing, qRT-PCR, and RNAse R digestion assays all validated the circularization structure of circARF3. The levels of circARF3, miR-205-5p, and sirtuin 5 (SIRT5) were determined by qRT-PCR or Western blotting. Luciferase reporter, RNA immunoprecipitation, and pull-down experiments were used to investigate the regulatory network. Flow cytometry was used to investigate the rate of cell apoptosis, and Western blotting was used to determine the levels of apoptotic-related proteins (cleaved caspase 3, cleaved polyadenosine-diphosphate-ribose polymerase) and HMGB1, TLR4, and MyD88. Enzyme-linked immunosorbent assay was used to assess the inflammatory response. Hematoxylin-eosin staining and TUNEL were used to detect the histology of injury and apoptosis of nasal mucosa tissues. RESULTS CircARF3 and SIRT5 levels were reduced in HDM-treated animals and IL-4/IL-13-treated NEpCs, while miR-205-5p expression was increased. CircARF3 was generated by back-splicing exons 3-5 with a stable circular shape. CircARF3 overexpression mitigated IL-4/IL-13-induced apoptosis in NEpCs by inhibiting miR-205-5p. SIRT5 upregulation attenuated IL-4/IL-13-induced inflammatory injury in NEpCs, and SIRT5 knockdown induced opposite effects. miR-205-5p silencing reversed the effects of SIRT5 knockdown on IL-4/IL-13-induced inflammatory injury. Furthermore, circARF3 overexpression alleviated histological abnormalities, apoptosis, inflammatory response, and HMGB1/TLR4 signaling activation in HDM-treated animals. CONCLUSION CircARF3 inhibited cell apoptosis and inflammation via the miR-205-5p/SIRT5 axis in IL-4/IL-13-treated NEpCs and HDM-treated mice.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Xi Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Jia-Bin Zhan
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Chang-Wu Li
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Xin Wei
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Hong-Yan Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| |
Collapse
|
8
|
Zhao R, Tang X, Lin H, Xing C, Xu N, Dai B, Wang P, Shao W, Liu M, Shen J, Deng S, Ren C. Knocking Down Gm16685 Decreases Liver Granuloma in Murine Schistosomiasis Japonica. Microorganisms 2023; 11:microorganisms11030796. [PMID: 36985369 PMCID: PMC10058064 DOI: 10.3390/microorganisms11030796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) can regulate key genes and pathways in liver disease development. Moreover, macrophages are speculated to play an important role in regulating granulomatous inflammation during schistosomiasis. However, the role of lncRNAs in the formation of liver granulomas by influencing the polarization of macrophages in Schistosoma japonicum infection is unclear. Our study aimed to determine whether lncRNAs can play a role in S. japonicum-induced hepatic egg granulomas and elucidate their effect on macrophages. We established S. japonicum infection models and screened the target lncRNA Gm16685 highly expressed in schistosomiasis mice using high-throughput sequencing. Hematoxylin and eosin staining revealed that the knockdown of Gm16685 reduced the area of egg granulomas. Moreover, M1 macrophage factor genes were significantly downregulated in Gm16685 knockdown livers. Meanwhile, M2 macrophage factor genes were significantly upregulated, which was consistent with the protein detection results. Hepatocytes, hepatic stellate cells, and macrophages were isolated from mouse models infected with S. japonicum, with Gm16685 being significantly upregulated in macrophages. Moreover, the knockdown of Gm16685 in RAW264.7 cells revealed similar results to in liver tissue. RNA fluorescence in situ hybridization (FISH) and nucleocytoplasmic separation experiments revealed that Gm16685 was predominantly localized in the cytoplasm of cells. We found that miR-205-5p was upregulated after Gm16685 was knocked down. After overexpression of miR-205-5p, the expression of Gm16685 and inflammatory factors was significantly downregulated. These results indicate that Gm16685 can participate in the pathogenesis of hepatic disease in schistosomiasis and promote M1 macrophage polarization by regulating miR-205-5p. Thus, our study may provide a new target for schistosomiasis japonica treatment.
Collapse
Affiliation(s)
- Ruyu Zhao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xiaoxue Tang
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Huiyao Lin
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Chen Xing
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Na Xu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Bingxin Dai
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Pingping Wang
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Wei Shao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Miao Liu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jijia Shen
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Shengqun Deng
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Cuiping Ren
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, Anhui Key Laboratory of Zoonosis of High Institution, Laboratory of Tropical and Parasitic Diseases Control, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
9
|
Yıldızhan K, Nazıroğlu M. NMDA Receptor Activation Stimulates Hypoxia-Induced TRPM2 Channel Activation, Mitochondrial Oxidative Stress, and Apoptosis in Neuronal Cell Line: Modular Role of Memantine. Brain Res 2023; 1803:148232. [PMID: 36610553 DOI: 10.1016/j.brainres.2023.148232] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023]
Abstract
TRPM2 channel is activated by the increase of hypoxia (HYP)-mediated excessive mitochondrial (mROS) and cytosolic (cROS) free reactive oxygen species generation and intracellular free Ca2+ ([Ca2+]i) influx. The stimulations of the N-methyl-d-aspartate(NMDA) receptor and TRPM2 channel induce mROS and apoptosis in the neurons, although their inhibitions via the treatments of memantine (MEM) and MK-801 decrease mROS and apoptosis. However, the molecular mechanisms underlying MEM treatment and NMDA inhibition' neuroprotection via TRPM2 inhibition in the HYP remain elusive. We investigated the modulator role of MEM and NMDA via the modulation of TRPM2 on oxidative neurodegeneration and apoptosis in SH-SY5Y neuronal cells. Six groups were induced in the SH-SY5Y and HEK293 cells as follows: Control, MEM, NMDA blocker (MK-801), HYP (CoCl2), HYP + MEM, and HYP + MK-801. The HYP caused to the increases of TRPM2 and PARP-1 expressions, and TRPM2 agonist (H2O2 and ADP-ribose)-induced TRPM2 current density and [Ca2+]i concentration via the upregulation of mitochondrial membrane potential, cROS, and mROS generations. The alterations were not observed in the absence of TRPM2 in the HEK293 cells. The increase of cROS, mROS, lipid peroxidation, cell death (propidium iodide/Hoechst) rate, apoptosis, caspase -3, caspase -8, and caspase -9 were restored via upregulation of glutathione and glutathione peroxidase by the treatments of TRPM2 antagonists (ACA or 2-APB), MEM, and MK-801. In conclusion, the inhibition of NMDA receptor via MEM treatment modulated HYP-mediated mROS, apoptosis, and TRPM2-induced excessive [Ca2+]i and may provide an avenue for protecting HYP-mediated neurodegenerative diseases associated with the increase of mROS, [Ca2+]i, and apoptosis.
Collapse
Affiliation(s)
- Kenan Yıldızhan
- Department of Biophysics, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey; Drug Discovery Unit, BSN Health, Analyses, Innov., Consult., Org., Agricul., Trade Ltd, Isparta, Turkey; Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.
| |
Collapse
|
10
|
Shafik MS, Bishr A, El-Tanbouly DM, Attia AS. Modulation of miR-205/ EGLN2 by rosuvastatin mitigates colistin-induced nephrotoxicity in rats: Involvement of ATF4/ CHOP and Nrf2 pathways. Biomed Pharmacother 2023; 157:114042. [PMID: 36436490 DOI: 10.1016/j.biopha.2022.114042] [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] [Received: 10/31/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022] Open
Abstract
Although the beneficial role of microRNA has been investigated thoroughly, the reno-protective role of microRNA-205 (miR-205) against colistin-induced nephrotoxicity has not yet been tackled. Hence, our study sought to study the possible modulatory effect of rosuvastatin on miR-205 and its downstream target, Egl-9 family hypoxia-inducible factor 2 (EGLN2) to combat oxidative and endoplasmic reticulum (ER) stresses as pivotal contributors to colistin-associated renal injury. Rats were randomly divided into four groups; normal, colistin (300 000 IU/Kg/day; i.p), colistin pretreated with rosuvastatin (10 mg/kg; p.o) and colistin pretreated with rosuvastatin (20 mg/kg; p.o) for 6 successive days. Pretreatment with rosuvastatin attenuated renal injury induced by colistin and enhanced kidney function with a marked reduction in renal injury markers, neutrophil gelatinase-associated lipocalin, and kidney injury molecule-1. Besides, rosuvastatin upregulated renal miR-205 expression and suppressed gene expression of EGLN2. In addition, it downregulated ER stress-related genes (activation transcription factor 4 (ATF4) and C/EBP homologous protein (CHOP)) along with caspases 12 and 3. It also induced the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) as detected by immunohistochemical examination besides increased renal antioxidants, reduced glutathione, and superoxide dismutase. In conclusion, rosuvastatin triggered a series of protective mechanisms against colistin-induced nephrotoxicity through modulating miR-205 and EGLN2 expression. Rosuvastatin suppressed ATF4/ CHOP trajectory and activated the Nrf2 pathway to substantiate its antioxidant and anti-apoptotic capacities.
Collapse
Affiliation(s)
- Marihan S Shafik
- Pharmacology and Toxicology department, Faculty of Pharmacy, Ahram Canadian University, Egypt
| | - Abeer Bishr
- Pharmacology and Toxicology department, Faculty of Pharmacy, Ahram Canadian University, Egypt
| | - Dalia M El-Tanbouly
- Pharmacology and Toxicology department, Faculty of Pharmacy, Cairo University, Egypt.
| | - Amina S Attia
- Pharmacology and Toxicology department, Faculty of Pharmacy, Cairo University, Egypt
| |
Collapse
|
11
|
Xiao Z, Xie Y, Huang F, Yang J, Liu X, Lin X, Zhu P, Zheng S. MicroRNA-205-5p plays a suppressive role in the high-fat diet-induced atrial fibrosis through regulation of the EHMT2/IGFBP3 axis. GENES & NUTRITION 2022; 17:11. [PMID: 35858845 PMCID: PMC9297569 DOI: 10.1186/s12263-022-00712-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
Objective MicroRNAs (miRNAs) targeting has been revealed to be an appealing strategy for the treatment and management of atrial fibrillation (AF). In this research, we aimed to explore the mechanisms of miR-205-5p in reducing the high-fat diet (HFD)-induced atrial fibrosis through the EHMT2/IGFBP3 axis. Methods Expression levels of miR-205-5p, IGFBP3 and EHMT2 were determined in AF patients, cell fibrosis models and mouse atrial fibrosis models. Luciferase activity and RIP assays were performed to detect the binding between miR-205-5p and EHMT2, and ChIP assays were implemented to detect the enrichment of H3K9me2 and H3K4me3 in the promoter region of IGFBP3 in cells. The related experiments focusing on the inflammatory response, atrial fibrosis, mitochondrial damage, and metabolic abnormalities were performed to figure out the roles of miR-205-5p, IGFBP3, and EHMT2 in cell and mouse atrial fibrosis models. Results Low expression levels of miR-205-5p and IGFBP3 and a high expression of EHMT2 were found in AF patients, cell fibrosis models and mouse atrial fibrosis models. Upregulation of miR-205-5p reduced the expression of TGF-β1, α-SMA, Col III and other fibrosis-related proteins. miR-205-5p overexpression targeted EHMT2 to regulate the methylation of H3 histones to promote IGFBP3 expression, which in turn affected the fibrosis of atrial muscle cells. In HFD-induced atrial fibrosis mice, upregulated miR-205-5p or elevated IGFBP3 alleviated atrial fibrosis, mitochondrial damage, and metabolic abnormalities. Conclusion This study suggests that miR-205-5p attenuates HFD-induced atrial fibrosis via modulating the EHMT2/IGFBP3 axis. Graphical Abstract miR-205-5p alleviates high-fat diet-induced atrial fibrosis in mice via EHMT2/IGFBP3. ![]()
Collapse
|
12
|
The Crosstalk between the Blood–Brain Barrier Dysfunction and Neuroinflammation after General Anaesthesia. Curr Issues Mol Biol 2022; 44:5700-5717. [PMID: 36421670 PMCID: PMC9689502 DOI: 10.3390/cimb44110386] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
As we know, with continuous medical progress, the treatment of many diseases can be conducted via surgery, which often relies on general anaesthesia for its satisfactory performance. With the widespread use of general anaesthetics, people are beginning to question the safety of general anaesthesia and there is a growing interest in central nervous system (CNS) complications associated with anaesthetics. Recently, abundant evidence has suggested that both blood–brain barrier (BBB) dysfunction and neuroinflammation play roles in the development of CNS complications after anaesthesia. Whether there is a crosstalk between BBB dysfunction and neuroinflammation after general anaesthesia, and whether this possible crosstalk could be a therapeutic target for CNS complications after general anaesthesia needs to be clarified by further studies.
Collapse
|
13
|
Dexmedetomidine prevents hemorrhagic brain injury by reducing damage induced by ferroptosis in mice. Neurosci Lett 2022; 788:136842. [PMID: 35995304 DOI: 10.1016/j.neulet.2022.136842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/20/2022] [Accepted: 08/14/2022] [Indexed: 11/20/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating condition with significant morbidity and mortality for which few effective treatments are clinically available. After ICH, iron overload within the perihaematomal region can induce lethal reactive oxygen species (ROS) production and lipid peroxidation, which contribute to secondary brain injury. An iron-dependent form of non-apoptotic cell death known as ferroptosis was recently identified. Ferroptosis plays an important role in ICH pathology. It is characterized by an accumulation of iron-induced lipid ROS, which leads to intracellular oxidative stress. Dexmedetomidine (DEX), an α2-adrenergic agonist, is widely used for anesthesia, pain control, and intensive care unit sedation. DEX has numerous beneficial activities, including anti-inflammatory, anti-oxidative, and anti-cell death activities. Here, we established a mouse model of ICH using collagenase VII and evaluated the effect of DEX in preventing ICH-induced brain injury. Our study showed that administering DEX reduced the damage induced by ferroptosis after ICH by regulating iron metabolism, amino acid metabolism and lipid peroxidation processes.
Collapse
|
14
|
Burlacu CC, Neag MA, Mitre AO, Sirbu AC, Badulescu AV, Buzoianu AD. The Role of miRNAs in Dexmedetomidine's Neuroprotective Effects against Brain Disorders. Int J Mol Sci 2022; 23:5452. [PMID: 35628263 PMCID: PMC9141783 DOI: 10.3390/ijms23105452] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 02/04/2023] Open
Abstract
There are limited neuroprotective strategies for various central nervous system conditions in which fast and sustained management is essential. Neuroprotection-based therapeutics have become an intensively researched topic in the neuroscience field, with multiple novel promising agents, from natural products to mesenchymal stem cells, homing peptides, and nanoparticles-mediated agents, all aiming to significantly provide neuroprotection in experimental and clinical studies. Dexmedetomidine (DEX), an α2 agonist commonly used as an anesthetic adjuvant for sedation and as an opioid-sparing medication, stands out in this context due to its well-established neuroprotective effects. Emerging evidence from preclinical and clinical studies suggested that DEX could be used to protect against cerebral ischemia, traumatic brain injury (TBI), spinal cord injury, neurodegenerative diseases, and postoperative cognitive disorders. MicroRNAs (miRNAs) regulate gene expression at a post-transcriptional level, inhibiting the translation of mRNA into functional proteins. In vivo and in vitro studies deciphered brain-related miRNAs and dysregulated miRNA profiles after several brain disorders, including TBI, ischemic stroke, Alzheimer's disease, and multiple sclerosis, providing emerging new perspectives in neuroprotective therapy by modulating these miRNAs. Experimental studies revealed that some of the neuroprotective effects of DEX are mediated by various miRNAs, counteracting multiple mechanisms in several disease models, such as lipopolysaccharides induced neuroinflammation, β-amyloid induced dysfunction, brain ischemic-reperfusion injury, and anesthesia-induced neurotoxicity models. This review aims to outline the neuroprotective mechanisms of DEX in brain disorders by modulating miRNAs. We address the neuroprotective effects of DEX by targeting miRNAs in modulating ischemic brain injury, ameliorating the neurotoxicity of anesthetics, reducing postoperative cognitive dysfunction, and improving the effects of neurodegenerative diseases.
Collapse
Affiliation(s)
- Codrin-Constantin Burlacu
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Andrei-Otto Mitre
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandru-Constantin Sirbu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Andrei-Vlad Badulescu
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
15
|
Deng W, Huang D, Xie H, Wang L, Shen Q, Zeng R, Huang Y, Li J, Yang B. Danhong injection represses diabetic retinopathy and nephropathy advancement in diabetic mice by upregulating microRNA-30d-5p and targeting JAK1. Bioengineered 2022; 13:8187-8200. [PMID: 35297304 PMCID: PMC9162027 DOI: 10.1080/21655979.2021.2006964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Danhong injection (DHI) restrains diabetic retinopathy and nephropathy (DR and DN) advancement in diabetic mice. However, the downstream mechanism of its modulation is not fully studied. Diabetic model mice (db/db mice) were intravenously injected with DHI and corresponding virus particles. MiR-30d-5p and JAK1 were detected. The body weight and fasting blood glucose mice were measured every 4 weeks. The renal tissues and serum of mice were collected, and the contents of creatinine and blood urea nitrogen were biochemically analyzed. IL-6, IFN-γ and TNF-α were detected by ELISA, with the pathological conditions of renal tissues in mice by He staining, and the adjustment conditions by TUNEL. Human retinal pigment epithelium (ARPE-19) cells were selected to induce DR model in vitro by high glucose, and exposed to DHI for treatment. The corresponding plasmids were transfected, and miR-30d-5p and JAK1 were detected, with the proliferation ability by plate cloning, apoptosis by flow cytometry, and cell migration ability by Transwell. The angiogenesis ability of cells was assessed by tube formation assay. The targeting relationship between miR-30d-5p and JAK1 was detected. The results manifested that miR-30d-5p was declined in DR and DN, while JAK1 expression was elevated. DHI was able to improve DR and renal injury. DHI could regulate the miR-30d-5p-JAK1 axis in vivo, and miR-30d-5p targeted and regulated JAK1. Upregulation of miR-30d-5p or inhibition of JAK1 could improve DR and renal injury. The results implies that DHI can repress the development of DR and DN by elevating miR-30d-5p and targeting JAK1.
Collapse
Affiliation(s)
- Wei Deng
- Department of Nephrology, The Fourth People's Hospital of Chenzhou City, Hunan Province, 423001, China
| | - Dan Huang
- Department of Ophthalmology, The Fourth People's Hospital of Chenzhou City, Hunan Province, 423001, China
| | - HongWu Xie
- Department of Endocrinology, The Fourth People's Hospital of Chenzhou City, Hunan Province, 423001, China
| | - LiMin Wang
- Department of Nephrology, The Fourth People's Hospital of Chenzhou City, Hunan Province, 423001, China
| | - Qun Shen
- Department of Endocrinology, The Fourth People's Hospital of Chenzhou City, Hunan Province, 423001, China
| | - RongRong Zeng
- Department of Endocrinology, The Fourth People's Hospital of Chenzhou City, Hunan Province, 423001, China
| | - YuanLian Huang
- Department of Nephrology, The Fourth People's Hospital of Chenzhou City, Hunan Province, 423001, China
| | - JianHua Li
- Department of Nephrology, The Fourth People's Hospital of Chenzhou City, Hunan Province, 423001, China
| | - Bo Yang
- Department of Nephrology, The First Affiliated Hospital, University of South China, Hengyang City, Hunan Province, 421001,China
| |
Collapse
|
16
|
Su G, Qu Y, Li G, Deng M. Sevoflurane protects against cerebral ischemia/reperfusion injury via microrna-30c-5p modulating homeodomain-interacting protein kinase 1. Bioengineered 2021; 12:11858-11871. [PMID: 34709114 PMCID: PMC8810137 DOI: 10.1080/21655979.2021.1999551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 12/21/2022] Open
Abstract
Sevoflurane (SEV) has been reported to be an effective neuroprotective agent for cerebral ischemia/reperfusion injury (CIRI). However, the precise molecular mechanisms of Sev preconditioning in CIRI remain largely unknown. Therefore, CIRI model was established via middle cerebral artery occlusion method. SEV was applied before modeling. after successful modeling, lentivirus was injected into the lateral ventricle of the brain. Neurological impairment score was performed in each group, and histopathologic condition, infarct volume, apoptosis, inflammation, oxidative stress, microRNA (miR)-30 c-5p and homeodomain-interacting protein kinase 1 (HIPK1) were detected. Mouse hippocampal neuronal cell line HT22 cells were pretreated with SEV, and the in vitro model was stimulated via oxygen-glucose deprivation and reoxygenation. The corresponding plasmids were transfected, and the cell growth was detected, including inflammation and oxidative stress, etc. The targeting of miR-30 c-5p with HIPK1 was examined. The results clarified that reduced miR-30 c-5p and elevated HIPK1 were manifested in CIRI. SEV could improve CIRI and modulate the miR-30 c-5p-HIPK1 axis in vitro and in vivo, and miR-30 c-5p could target HIPK1. Depressed miR-30 c-5p could eliminate the protection of SEV in vitro and in vivo. Repression of HIPK1 reversed the effect of reduced miR-30 c-5p on CIRI. Therefore, it is concluded that SEV is available to depress CIRI via targeting HIPK1 through upregulated miR-30 c-5p.
Collapse
Affiliation(s)
- Guoning Su
- Department of Anesthesia, Affiliated Hospital of Yunnan University, KunMing City, China
| | - Yan Qu
- Department of Anesthesia, Affiliated Hospital of Yunnan University, KunMing City, China
| | - Gang Li
- Department of Anesthesia, Affiliated Hospital of Yunnan University, KunMing City, China
| | - Min Deng
- Department of Anesthesia, Affiliated Hospital of Yunnan University, KunMing City, China
| |
Collapse
|
17
|
Jakaria M, Belaidi AA, Bush AI, Ayton S. Ferroptosis as a mechanism of neurodegeneration in Alzheimer's disease. J Neurochem 2021; 159:804-825. [PMID: 34553778 DOI: 10.1111/jnc.15519] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 01/19/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, with complex pathophysiology that is not fully understood. While β-amyloid plaque and neurofibrillary tangles define the pathology of the disease, the mechanism of neurodegeneration is uncertain. Ferroptosis is an iron-mediated programmed cell death mechanism characterised by phospholipid peroxidation that has been observed in clinical AD samples. This review will outline the growing molecular and clinical evidence implicating ferroptosis in the pathogenesis of AD, with implications for disease-modifying therapies.
Collapse
Affiliation(s)
- Md Jakaria
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdel Ali Belaidi
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|