1
|
Ge L, Ma J, Xu J, Wang B, Adil A, Xu H. The mechanism of lncRNA PVT1 targeting the miR-30a/Beclin-1 axis to mediate ventricular remodeling in spontaneously hypertensive rats. Cell Signal 2025; 130:111650. [PMID: 39923929 DOI: 10.1016/j.cellsig.2025.111650] [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: 05/14/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVES Hypertension poses a great health threat globally. We probed the mechanisms of long non-coding RNA plasmacytoma variant translocation 1 (lncRNA PVT1) mediating ventricular remodeling (VR) in spontaneously hypertensive rats (SHR). METHODS PVT1 was down-regulated or miR-30a was inhibited in SHR in vivo. Hypertensive injury model was established in vitro. VR, fibrosis and autophagy-related indicators were detected by echocardiography, HE/WGA/Masson staining, ELISA, and immunohistochemistry. Cell viability, fibrosis markers, autophagy-related markers, and lncRNA PVT1 and miR-30a levels were assessed. Interactions between PVT1, Beclin-1 and miR-30a were verified. RESULTS PVT1 was up-regulated in myocardial tissues of SHR. PVT1 knockdown alleviated VR and myocardial fibrosis (MF) in SHR, as evidenced by decreased systolic blood pressure, left ventricular end-systolic diameter, left ventricular end-systolic diameter, and heart weight index, boosted left ventricular fractional shortening and left ventricular ejection fraction, abated inflammatory infiltration of myocardial tissues, decreased myocardial hypertrophy and interstitial fibrosis, reduced serum angiotensin II (Ang II) and atrial natriuretic peptide, and downregulated collagen I, collagen II, α-smooth muscle actin, and fibronectin protein. PVT1 knockdown down-regulated Beclin 1 and LC3B-II/LC3B-I and up-regulated p62 protein. In vitro, PVT1 knockdown improved fibrosis by inhibiting Ang II-induced cardiomyocyte autophagy. PVT1 acted as a competitive endogenous RNA to competitively bind to miR-30a to target Beclin-1 expression. PVT1 targeted the miR-30a/Beclin-1 axis to mediate autophagy to affect VR and MF in SHR. CONCLUSIONS LncRNA PVT1 promotes cellular autophagy by targeting the miR-30a/Beclin-1 axis, thereby promoting VR and MF in SHR. Knockdown of lncRNA PVT1 attenuates VR and MF in SHR.
Collapse
Affiliation(s)
- Li Ge
- Department of Hypertension, The Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Jianjun Ma
- Department of Hypertension, The Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Jingxuan Xu
- School of Optometry and Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Bo Wang
- Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830000, China
| | - Abdusalam Adil
- Department of Hypertension, The Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Hongfeng Xu
- Department of Hypertension, The Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
2
|
Wang Z, Zhang LN, Wu T, Pan X, Li L, Yang X, Zhang M, Liu Y, Liu Y. Actions of dexmedetomidine in regulating NLRP3 in postoperative cognitive dysfunction in aged mice via the autophagy-lysosome pathway. Br J Pharmacol 2025; 182:1683-1703. [PMID: 39815423 DOI: 10.1111/bph.17378] [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: 01/12/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND AND PURPOSE Autophagy-lysosomal pathway dysfunction leads to postoperative cognitive dysfunction (POCD). Dexmedetomidine (Dex) improves POCD, and we probed the effects of Dex on autophagy-lysosomal pathway dysfunction in a POCD model. EXPERIMENTAL APPROACH A POCD mouse model was established and intraperitoneally injected with Dex. Cognitive function was evaluated by Morris water maze/open field test/novel object recognition assay. Levels of neurotransmitters/inflammatory cytokines in hippocampus, and NLRP3/ASC/Cleaved Caspase-1 proteins were determined by ELISA/Western blot. NLRP3 inflammasome-mediated microglial activation/astrocyte A1 differentiation in the hippocampal CA1 region were assessed by immunofluorescence assay. BV-2 cells were treated with lipopolysaccharide (LPS) and Dex and/or the NLRP3 inflammasome activator Nigericin, and transfected with si-TFEB for co-culture with primary reactive astrocytes (RAs) to verify the function of Dex in vitro. KEY RESULTS Dex alleviated cognitive dysfunction in POCD mice and repressed NLRP3 inflammasome-mediated microglial activation and astrocyte A1 differentiation. NLRP3 inflammasome activation partially reversed the protective effect of Dex on the POCD condition. In vitro experiments verified the inhibitory properties of Dex on microglial activation and astrocyte A1 differentiation. Dex induces TFEB nuclear translocation, microglial autophagy and lysosomal biogenesis. By activating the autophagy-lysosome pathway, Dex regulated NLRP3 inflammasome-mediated microglial activation, inhibited astrocyte A1 differentiation and alleviated POCD in vivo. CONCLUSION AND IMPLICATIONS Dex regulates NLRP3 inflammasome-mediated hippocampal microglial activation by promoting TFEB nuclear translocation and activating the autophagy-lysosome pathway and inhibits astrocyte A1 differentiation, thereby alleviating POCD.
Collapse
Affiliation(s)
- Zhi Wang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Li-Na Zhang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Ting Wu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Xu Pan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Le Li
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Xin Yang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Miao Zhang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Ying Liu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
3
|
Liu Q, Li F, Hu S, Ding N, Ma F, Hao Y, Li G, Xiong J, Zhang H, Jiang Y. Pyruvate dehydrogenase alleviates macrophage autophagy in Hcy-induced ApoE -/- mice. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 39980347 DOI: 10.3724/abbs.2025021] [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: 02/22/2025] Open
Abstract
Macrophages play a protective role in atherosclerosis, whereas homocysteine (Hcy) is recognized as an independent risk factor for atherosclerosis. Defects in macrophage autophagy contribute to the formation of atherosclerotic plaques, and dysregulated energy metabolism is closely linked to the process of autophagy. However, the regulation of macrophage autophagy by pyruvate dehydrogenase (PDH), a key component of the PDH complex involved in energy and metabolic homeostasis, remains poorly understood in the context of atherosclerosis induced by Hcy. In our study, proteomic profiling identifies 748 upregulated proteins and 760 downregulated proteins in Hcy-treated macrophages. KEGG pathway analysis reveals significant enrichment of differentially expressed proteins in metabolism-related pathways, including those related to the biosynthesis of amino acids, carbon metabolism, and glycolysis/gluconeogenesis. Additionally, we explore the role of PDH in mediating Hcy-induced atherosclerosis in ApoE -/- mice. The results show a marked reduction in PDH expression and activity in Hcy-treated macrophages, leading to impaired autophagy. Notably, PDH activation enhances the assembly of the autophagy initiator ULK1-FIP200-Atg13 complex through the modulation of the AMPK/mTOR signaling pathway, suggesting a potential therapeutic target for Hcy-induced atherosclerosis.
Collapse
Affiliation(s)
- Qiujun Liu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Feng Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
- Center of Laboratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Shutong Hu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Ning Ding
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Fang Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Yinju Hao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China
| | - Guizhong Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Jiantuan Xiong
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Huiping Zhang
- Department of Medical Genetics, Maternal and Child Health of Hunan Province, Changsha 410008, China
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Yideng Jiang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
4
|
Shen L, Zhu Y, Chen Z, Shen F, Yu W, Zhang L. Isoliquiritigenin attenuates myocardial ischemia reperfusion through autophagy activation mediated by AMPK/mTOR/ULK1 signaling. BMC Cardiovasc Disord 2024; 24:415. [PMID: 39123142 PMCID: PMC11311884 DOI: 10.1186/s12872-024-04054-z] [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: 02/29/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Ischemia reperfusion (IR) causes impaired myocardial function, and autophagy activation ameliorates myocardial IR injury. Isoliquiritigenin (ISO) has been found to protect myocardial tissues via AMPK, with exerting anti-tumor property through autophagy activation. This study aims to investigate ISO capacity to attenuate myocardial IR through autophagy activation mediated by AMPK/mTOR/ULK1 signaling. METHODS ISO effects were explored by SD rats and H9c2 cells. IR rats and IR-induced H9c2 cell models were established by ligating left anterior descending (LAD) coronary artery and hypoxia/re-oxygenation, respectively, followed by low, medium and high dosages of ISO intervention (Rats: 10, 20, and 40 mg/kg; H9c2 cells: 1, 10, and 100 μmol/L). Myocardial tissue injury in rats was assessed by myocardial function-related index, HE staining, Masson trichrome staining, TTC staining, and ELISA. Autophagy of H9c2 cells was detected by transmission electron microscopy (TEM) and immunofluorescence. Autophagy-related and AMPK/mTOR/ULK1 pathway-related protein expressions were detected with western blot. RESULTS ISO treatment caused myocardial function improvement, and inhibition of myocardial inflammatory infiltration, fibrosis, infarct area, oxidative stress, CK-MB, cTnI, and cTnT expression in IR rats. In IR-modeled H9c2 cells, ISO treatment lowered apoptosis rate and activated autophagy and LC3 fluorescence expression. In vivo and in vitro, ISO intervention exhibited enhanced Beclin1, LC3II/LC3I, and p-AMPK/AMPK levels, whereas inhibited P62, p-mTOR/mTOR and p-ULK1(S757)/ULK1 protein expression, activating autophagy and protecting myocardial tissues from IR injury. CONCLUSION ISO treatment may induce autophagy by regulating AMPK/mTOR/ULK1 signaling, thereby improving myocardial IR injury, as a potential candidate for treatment of myocardial IR injury.
Collapse
Affiliation(s)
- Liying Shen
- Department of Cardiology, Huzhou Central Hospital, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China
| | - Yingwei Zhu
- Department of Cardiology, Huzhou Central Hospital, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China
| | - Zhenfeng Chen
- Department of Cardiology, Huzhou Central Hospital, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China
| | - Feng Shen
- Department of Cardiology, Huzhou Central Hospital, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China
| | - Weiwei Yu
- Department of Cardiology, Huzhou Central Hospital, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China
| | - Li Zhang
- Department of Cardiology, Huzhou Central Hospital, No. 1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang, China.
| |
Collapse
|
5
|
Wu X, Liu Y, Li Y, Tang Z, Li A, Zhang H. Molecular mechanism of thiram-induced abnormal chondrocyte proliferation via lncRNA MSTRG.74.1-BNIP3 axis. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 201:105847. [PMID: 38685209 DOI: 10.1016/j.pestbp.2024.105847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 05/02/2024]
Abstract
Thiram, a widely used organic pesticide in agriculture, exhibits both bactericidal and insecticidal effects. However, prolonged exposure to thiram has been linked to bone deformities and cartilage damage, contributing to the development of tibial dyschondroplasia (TD) in broilers and posing a significant threat to global agricultural production. TD, a prevalent nutritional metabolic disease, manifests as clinical symptoms like unstable standing, claudication, and sluggish movement in affected broilers. In recent years, there has been growing recognition of the regulatory role of long non-coding RNA (lncRNA) in tibial cartilage formation among broilers through diverse signaling pathways. This study employs in vitro experimental models, growth performance analysis, and clinical observation to assess broilers' susceptibility to thiram pollution. Transcriptome sequencing analysis revealed a significant elevation in the expression of lncRNA MSTRG.74.1 in both the con group and the thiram-induced in vitro group. The results showed that lncRNA MSTRG.74.1 plays a pivotal role in influencing the proliferation and abnormal differentiation of chondrocytes. This regulation occurs through the negative modulation of apoptotic genes, including Bax, Cytc, Bcl2, Apaf1, and Caspase3, along with genes Atg5, Beclin1, LC3b, and protein p62. Moreover, the overexpression of lncRNA MSTRG.74.1 was found to regulate broiler chondrocyte development by upregulating BNIP3. In summary, this research sheds light on thiram-induced abnormal chondrocyte proliferation in TD broilers, emphasizing the significant regulatory role of the lncRNA MSTRG.74.1-BNIP3 axis, which will contribute to our understanding of the molecular mechanisms underlying TD development in broilers exposed to thiram.
Collapse
Affiliation(s)
- Xiaomei Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yingwei Liu
- Guangzhou National Laboratory, Guangzhou 510000, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Aoyun Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, PR China.
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
6
|
Marcondes-de-Castro IA, Reis-Barbosa PH, Marinho TS, Aguila MB, Mandarim-de-Lacerda CA. AMPK/mTOR pathway significance in healthy liver and non-alcoholic fatty liver disease and its progression. J Gastroenterol Hepatol 2023; 38:1868-1876. [PMID: 37438882 DOI: 10.1111/jgh.16272] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Obesity is related to several organs, but the liver is particularly affected. Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor and regulator of liver lipid dysfunction and glucose metabolism. The mechanistic target of rapamycin (mTOR) is a protein kinase regulating cell growth, survival, metabolism, and immunity. Together, these pathways are involved in obesity, insulin resistance, non-alcoholic fatty liver disease (NAFLD) and its progression, and autophagy. During energy demand, liver kinase B (LKB) phosphorylation helps activate the AMPK/mTOR pathways. Likewise, the protein forkhead box O family (FOXO) negatively regulates adipogenesis by binding to the promoter sites of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha, initiating adipogenesis. In addition, acetyl-CoA carboxylase, which regulates de novo lipogenesis, is linked to LKB and FOXO in developing NAFLD. The kinase complex, consisting of Unc-51-like autophagy-activating kinase 1 or 2 (ULK1, ULK2) by stimulating autophagy, and eliminating fat droplets in NAFLD, is regulated by mTORC1 and negatively regulated by AMPK that suppresses liver lipogenesis and increases fatty acid oxidation. Also, ULK1 is essential for initiating phagophore formation, establishing macrophagy, and generating autophagosomes. The selective breakdown of lipid droplets through macroautophagy, or macrolipophagy, occurs on a cellular energy level using free fatty acids. In addition, mTORC1 promotes lipogenesis by activating sterol regulatory element-binding protein. Finding new components and novel regulatory modes in signaling is significant for a better understanding of the AMPK/mTOR pathways, potentially facilitating the development of future diagnostic and therapeutic strategies for NAFLD and its progression to non-alcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ilitch Aquino Marcondes-de-Castro
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Pedro Henrique Reis-Barbosa
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Centre, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Ma K, Liu W, Liu Q, Hu P, Bai L, Yu M, Yang Y. Naringenin facilitates M2 macrophage polarization after myocardial ischemia-reperfusion by promoting nuclear translocation of transcription factor EB and inhibiting the NLRP3 inflammasome pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:1405-1419. [PMID: 36988289 DOI: 10.1002/tox.23774] [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: 09/20/2022] [Revised: 02/22/2023] [Accepted: 02/25/2023] [Indexed: 05/18/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) remains an unsolved puzzle in medical circles. Naringenin (NAR) is a flavonoid with cardioprotective potential. The purpose of this article was to discuss the protective mechanism of NAR in MIRI by regulating macrophage polarization. The MIRI mouse model was established and perfused with NAR before surgery. In the in vitro experiment, macrophages RAW264.7 were treated with lipopolysaccharide to induce M1 polarization after pretreatment with NAR. Rescue experiments were carried out to validate the functions of transcription factor EB (TFEB), the NLR pyrin domain containing 3 (NLRP3) inflammasome, and autophagy in macrophage polarization. NAR reduced histopathological injury and infarction of myocardial tissues in MIRI mice, inhibited M1 polarization and promoted M2 polarization of macrophages, diminished levels of pro-inflammatory factors, and augmented levels of anti-inflammatory factors. NAR facilitated TFEB nuclear translocation and inhibited the NLRP3 inflammasome pathway. Silencing TFEB or Nigericin partly nullified the effect of NAR on macrophage polarization. NAR increased autophagosome formation, autophagy flux, and autophagy level. Autophagy inhibitor 3-methyladenine partly invalidated the inhibition of NAR on the NLRP3 inflammasome pathway. In animal experiments, NAR protected MIRI mice through the TFEB-autophagy-NLRP3 inflammasome pathway. Collectively, NAR inhibited NLRP3 inflammasome activation and facilitated M2 macrophage polarization by stimulating TFEB nuclear translocation, thus protecting against MIRI.
Collapse
Affiliation(s)
- Kuiying Ma
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Wenqing Liu
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Qi Liu
- Emergency Department, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Pengfei Hu
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Lingyu Bai
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Miao Yu
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Yan Yang
- Department of General Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| |
Collapse
|
8
|
Yang H, Huang Z, Luo Y, Lei D, Yan P, Shen A, Liu W, Li D, Wu Z. TRIM37 exacerbates hepatic ischemia/reperfusion injury by facilitating IKKγ translocation. Mol Med 2023; 29:62. [PMID: 37158850 PMCID: PMC10165779 DOI: 10.1186/s10020-023-00653-2] [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: 01/26/2023] [Accepted: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Hepatic ischemia/reperfusion (I/R) injury is one of the major pathological processes associated with various liver surgeries. However, there is still a lack of strategies to protect against hepatic I/R injury because of the unknown underlying mechanism. The present study aimed to identify a potential strategy and provide a fundamental experimental basis for treating hepatic I/R injury. METHOD A classic 70% ischemia/reperfusion injury was established. Immunoprecipitation was used to identify direct interactions between proteins. The expression of proteins from different subcellular localizations was detected by Western blotting. Cell translocation was directly observed by immunofluorescence. HE, TUNEL and ELISA were performed for function tests. RESULT We report that tripartite motif containing 37 (TRIM37) aggravates hepatic I/R injury through the reinforcement of IKK-induced inflammation following dual patterns. Mechanistically, TRIM37 directly interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6), inducing K63 ubiquitination and eventually leading to the phosphorylation of IKKβ. TRIM37 enhances the translocation of IKKγ, a regulatory subunit of the IKK complex, from the nucleus to the cytoplasm, thereby stabilizing the cytoplasmic IKK complex and prolonging the duration of inflammation. Inhibition of IKK rescued the function of TRIM37 in vivo and in vitro. CONCLUSION Collectively, the present study discloses some potential function of TRIM37 in hepatic I/R injury. Targeting TRIM37 might be potential for treatment against hepatic I/R injury.Targeting TRIM37 might be a potential treatment strategy against hepatic I/R injury.
Collapse
Affiliation(s)
- Hang Yang
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Zuotian Huang
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yunhai Luo
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Dengliang Lei
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Ping Yan
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ai Shen
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Wenbin Liu
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Dewei Li
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China.
| | - Zhongjun Wu
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
9
|
Mao B, Yuan W, Wu F, Yan Y, Wang B. Autophagy in hepatic ischemia-reperfusion injury. Cell Death Discov 2023; 9:115. [PMID: 37019879 PMCID: PMC10076300 DOI: 10.1038/s41420-023-01387-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 04/07/2023] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is a major complication of liver resection or liver transplantation that can seriously affect patient's prognosis. There is currently no definitive and effective treatment strategy for HIRI. Autophagy is an intracellular self-digestion pathway initiated to remove damaged organelles and proteins, which maintains cell survival, differentiation, and homeostasis. Recent studies have shown that autophagy is involved in the regulation of HIRI. Numerous drugs and treatments can change the outcome of HIRI by controlling the pathways of autophagy. This review mainly discusses the occurrence and development of autophagy, the selection of experimental models for HIRI, and the specific regulatory pathways of autophagy in HIRI. Autophagy has considerable potential in the treatment of HIRI.
Collapse
Affiliation(s)
- Benliang Mao
- College of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Wei Yuan
- Department of General Surgery, Guangzhou Red Cross Hospital affiliated to Jinan University, Guangzhou, China
| | - Fan Wu
- Department of General Surgery, Guangzhou Red Cross Hospital affiliated to Jinan University, Guangzhou, China
| | - Yong Yan
- Department of General Surgery, Guangzhou Red Cross Hospital affiliated to Jinan University, Guangzhou, China
| | - Bailin Wang
- College of Clinical Medicine, Guizhou Medical University, Guiyang, China.
- Department of General Surgery, Guangzhou Red Cross Hospital affiliated to Jinan University, Guangzhou, China.
| |
Collapse
|
10
|
Tang B, Luo Z, Zhang R, Zhang D, Nie G, Li M, Dai Y. An update on the molecular mechanism and pharmacological interventions for Ischemia-reperfusion injury by regulating AMPK/mTOR signaling pathway in autophagy. Cell Signal 2023; 107:110665. [PMID: 37004834 DOI: 10.1016/j.cellsig.2023.110665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
AMP-activated protein kinase (5'-adenosine monophosphate-activated protein kinase, AMPK)/mammalian target of rapamycin (mTOR) is an important signaling pathway maintaining normal cell function and homeostasis in vivo. The AMPK/mTOR pathway regulates cellular proliferation, autophagy, and apoptosis. Ischemia-reperfusion injury (IRI) is secondary damage that frequently occurs clinically in various disease processes and treatments, and the exacerbated injury during tissue reperfusion increases disease-associated morbidity and mortality. IRI arises from multiple complex pathological mechanisms, among which cell autophagy is a focus of recent research and a new therapeutic target. The activation of AMPK/mTOR signaling in IRI can modulate cellular metabolism and regulate cell proliferation and immune cell differentiation by adjusting gene transcription and protein synthesis. Thus, the AMPK/mTOR signaling pathway has been intensively investigated in studies focused on IRI prevention and treatment. In recent years, AMPK/mTOR pathway-mediated autophagy has been found to play a crucial role in IRI treatment. This article aims to elaborate the action mechanisms of AMPK/mTOR signaling pathway activation in IRI and summarize the progress of AMPK/mTOR-mediated autophagy research in the field of IRI therapy.
Collapse
Affiliation(s)
- Bin Tang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Zhijian Luo
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Rong Zhang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Dongmei Zhang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Guojun Nie
- The First Outpatient Department of People's Liberation Army Western Theater General Hospital, Cheng Du, Sichuan Province 61000, China
| | - Mingxing Li
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| | - Yan Dai
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
11
|
Chen L, Kang X, Meng X, Huang L, Du Y, Zeng Y, Liao C. MALAT1-mediated EZH2 Recruitment to the GFER Promoter Region Curbs Normal Hepatocyte Proliferation in Acute Liver Injury. J Clin Transl Hepatol 2023; 11:97-109. [PMID: 36406327 PMCID: PMC9647095 DOI: 10.14218/jcth.2021.00391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/12/2021] [Accepted: 03/04/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS The goal of this study was to investigate the mechanism by which the long noncoding RNA MALAT1 inhibited hepatocyte proliferation in acute liver injury (ALI). METHODS Lipopolysaccharide (LPS) was used to induce an ALI cellular model in HL7702 cells, in which lentivirus vectors containing MALAT1/EZH2/GFER overexpression or knockdown were introduced. A series of experiments were performed to determine their roles in liver injury, oxidative stress injury, and cell biological processes. The interaction of MALAT1 with EZH2 and enrichment of EZH2 and H3K27me3 in the GFER promoter region were identified. Rats were treated with MALAT1 knockdown or GFER overexpression before LPS induction to verify the results derived from the in vitro assay. RESULTS MALAT1 levels were elevated and GFER levels were reduced in ALI patients and the LPS-induced cell model. MALAT1 knockdown or GFER overexpression suppressed cell apoptosis and oxidative stress injury induced cell proliferation, and reduced ALI. Functionally, MALAT1 interacted directly with EZH2 and increased the enrichment of EZH2 and H3K27me3 in the GFER promoter region to reduce GFER expression. Moreover, MALAT1/EZH2/GFER was activated the AMPK/mTOR signaling pathway. CONCLUSION Our study highlighted the inhibitory role of reduced MALAT1 in ALI through the modulation of EZH2-mediated GFER.
Collapse
Affiliation(s)
- Li Chen
- Department of Infectious Diseases, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Correspondence to: Li Chen, Department of Infectious Diseases, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China. ORCID: https://orcid.org/0000-0003-2385-2858. Tel: +86-13755192409, E-mail:
| | - Xintong Kang
- Department of Hepatology, Public Health Clinical Center of Chengdu, Chengdu, Sichuan, China
| | - Xiujuan Meng
- Hospital-Acquired Infection Control Center, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Liang Huang
- Department of Hepatology, Public Health Clinical Center of Chengdu, Chengdu, Sichuan, China
| | - Yiting Du
- Department of Emergency, Chengdu Women’s and Children’s Central Hospital, Chengdu, Sichuan, China
| | - Yilan Zeng
- Department of Hepatology, Public Health Clinical Center of Chengdu, Chengdu, Sichuan, China
| | - Chunfeng Liao
- Department of Cardiovascular Medicine, The First Hospital of Changsha, Changsha, Hunan, China
| |
Collapse
|
12
|
Cai J, Chen X, Liu X, Li Z, Shi A, Tang X, Xia P, Zhang J, Yu P. AMPK: The key to ischemia-reperfusion injury. J Cell Physiol 2022; 237:4079-4096. [PMID: 36134582 DOI: 10.1002/jcp.30875] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/08/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022]
Abstract
Ischemia-reperfusion injury (IRI) refers to a syndrome in which tissue damage is further aggravated and organ function further deteriorates when blood flow is restored after a period of tissue ischemia. Acute myocardial infarction, stress ulcer, pancreatitis, intestinal ischemia, intermittent claudication, acute tubular necrosis, postshock liver failure, and multisystem organ failure are all related to reperfusion injury. AMP-activated protein kinase (AMPK) has been identified in multiple catabolic and anabolic signaling pathways. The functions of AMPK during health and diseases are intriguing but still need further research. Except for its conventional roles as an intracellular energy switch, emerging evidence reveals the critical role of AMPK in IRI as an energy-sensing signal molecule by regulating metabolism, autophagy, oxidative stress, inflammation, and other progressions. At the same time, drugs based on AMPK for the treatment of IRI are constantly being researched and applied in clinics. In this review, we summarize the mechanisms underlying the effects of AMPK in IRI and describe the AMPK-targeting drugs in treatment, hoping to increase the understanding of AMPK in IRI and provide new insights into future clinical treatment.
Collapse
Affiliation(s)
- Jie Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xinyue Chen
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xingyu Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ao Shi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Biochemistry and Molecular Biology, Mayo Graduate School of Biomedical Science, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiaoyi Tang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Panpan Xia
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| |
Collapse
|
13
|
Cao Y, Jiao Y, Zhan S, Liang X, Li Z, Chen J, Xiong X, Gu Z, Du X, Zheng Z. Polyamine Putrescine Regulates Oxidative Stress and Autophagy of Hemocytes Induced by Lipopolysaccharides in Pearl Oyster Pinctada fucata martensii. Front Physiol 2021; 12:781324. [PMID: 34955892 PMCID: PMC8703005 DOI: 10.3389/fphys.2021.781324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/11/2021] [Indexed: 12/28/2022] Open
Abstract
The polyamine putrescine (Put) is a ubiquitous small cationic amine. It plays an essential role in controlling the innate immune response. However, little is known about its function in mollusks. In this study, the Put content was observed to increase in the serum of pearl oyster Pinctada fucata martensii after 6 and 24 h of lipopolysaccharide (LPS) stimulation. Activities of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) increased, and nitric oxide synthase was downregulated in the Put group (i.e., combined treatment with Put and LPS) compared with that in the LPS group (i.e., combined treatment with phosphate-buffered saline and LPS). Furthermore, activities of alkaline phosphatase and acid phosphatase were inhibited after 6 h of LPS stimulation. The expression levels of the nuclear factor kappa B, IκB kinase, Janus kinase, and signal transducer and activator of transcription proteins genes were all significantly suppressed at 12 and 24 h in the Put group. Pseudomonas aeruginosa and Bacillus subtilis grew better after being incubated with the serum from the Put group than that from the LPS group. Additionally, the Put treatment remarkably inhibited the autophagy of hemocytes mediated by the AMP-activated protein kinase-mammalian target of rapamycin-Beclin-1 pathway. This study demonstrated that Put can effectively inhibit the inflammatory response induced by LPS in pearl oysters. These results provide useful information for further exploration of the immunoregulatory functions of polyamines in bivalves and contribute to the development of immunosuppressive agents.
Collapse
Affiliation(s)
- Yanfei Cao
- Fishery College, Guangdong Ocean University, Zhanjiang, China
| | - Yu Jiao
- Fishery College, Guangdong Ocean University, Zhanjiang, China
- Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, China
- Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, China
- Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Zhanjiang, China
| | - Shuzhi Zhan
- Fishery College, Guangdong Ocean University, Zhanjiang, China
| | - Xueru Liang
- Fishery College, Guangdong Ocean University, Zhanjiang, China
| | - Zhixin Li
- Fishery College, Guangdong Ocean University, Zhanjiang, China
| | - Jiayi Chen
- Fishery College, Guangdong Ocean University, Zhanjiang, China
| | - Xinwei Xiong
- Fishery College, Guangdong Ocean University, Zhanjiang, China
| | - Zefeng Gu
- Fishery College, Guangdong Ocean University, Zhanjiang, China
| | - Xiaodong Du
- Fishery College, Guangdong Ocean University, Zhanjiang, China
- Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, China
- Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, China
- Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Zhanjiang, China
| | - Zhe Zheng
- Fishery College, Guangdong Ocean University, Zhanjiang, China
- Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, China
- Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, China
- Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Zhanjiang, China
| |
Collapse
|
14
|
Hong T, Zhou Y, Peng L, Wu X, Li Y, Li Y, Zhao Y. Knocking Down Peroxiredoxin 6 Aggravates Cerebral Ischemia-Reperfusion Injury by Enhancing Mitophagy. Neuroscience 2021; 482:30-42. [PMID: 34863856 DOI: 10.1016/j.neuroscience.2021.11.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 01/18/2023]
Abstract
Cerebral ischemia-reperfusion injury (IRI) is caused by reperfusion following ischemia. Mitophagy is closely related to cerebral IRI. Mitophagy disorder or excess may be harmful and lead to neuronal apoptosis. Peroxiredoxin 6 (PRDX6) is an antioxidant protein and plays an important role in ischemic stroke. However, the relationship between PRDX6 and mitophagy is not clear at present. In order to explore and solve this problem. We have established a middle cerebral artery occlusion (MCAO) model of cerebral ischemia-reperfusion in SD rats and knockdown PRDX6 and PINK1 with lentivirus. Knocking down PRDX6 led to further aggravation of cerebral IRI. Our research found that knockdown PRDX6 increased the expression of mitophagy-related and apoptosis-related proteins. Knocking down PINK1 relieved mitophagy and apoptosis caused by knocking down PRDX6. In conclusion, knockdown of PRDX6 could aggravate cerebral IRI by enhancing PINK1/PARKIN pathway mediated mitophagy, and this effect could increase neuronal apoptosis.
Collapse
Affiliation(s)
- Toushen Hong
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Yang Zhou
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Molecular Medical Laboratory, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.
| | - Li Peng
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Molecular Medical Laboratory, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Xiaoying Wu
- Department of Gastroenterology, Qijiang District People's Hospital, 401420 Chongqing, People's Republic of China
| | - Yixin Li
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Molecular Medical Laboratory, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Yumei Li
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Yong Zhao
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Molecular Medical Laboratory, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.
| |
Collapse
|
15
|
Wang ZY, Lin JY, Feng YR, Liu DS, Zhao XZ, Li T, Li SY, Sun JC, Li SF, Jia WY, Jing HR. Recombinant angiopoietin-like protein 4 attenuates intestinal barrier structure and function injury after ischemia/reperfusion. World J Gastroenterol 2021; 27:5404-5423. [PMID: 34539141 PMCID: PMC8409166 DOI: 10.3748/wjg.v27.i32.5404] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/17/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Intestinal barrier breakdown, a frequent complication of intestinal ischemia-reperfusion (I/R) including dysfunction and the structure changes of the intestine, is characterized by a loss of tight junction and enhanced permeability of the intestinal barrier and increased mortality. To develop effective and novel therapeutics is important for the improvement of outcome of patients with intestinal barrier deterioration. Recombinant human angiopoietin-like protein 4 (rhANGPTL4) is reported to protect the blood-brain barrier when administered exogenously, and endogenous ANGPTL4 deficiency deteriorates radiation-induced intestinal injury. AIM To identify whether rhANGPTL4 may protect intestinal barrier breakdown induced by I/R. METHODS Intestinal I/R injury was elicited through clamping the superior mesenteric artery for 60 min followed by 240 min reperfusion. Intestinal epithelial (Caco-2) cells and human umbilical vein endothelial cells were challenged by hypoxia/ reoxygenation to mimic I/R in vitro. RESULTS Indicators including fluorescein isothiocyanate-conjugated dextran (4 kilodaltons; FD-4) clearance, ratio of phosphorylated myosin light chain/total myosin light chain, myosin light chain kinase and loss of zonula occludens-1, claudin-2 and VE-cadherin were significantly increased after intestinal I/R or cell hypoxia/reoxygenation. rhANGPTL4 treatment significantly reversed these indicators, which were associated with inhibiting the inflammatory and oxidative cascade, excessive activation of cellular autophagy and apoptosis and improvement of survival rate. Similar results were observed in vitro when cells were challenged by hypoxia/reoxygenation, whereas rhANGPTL4 reversed the indicators close to normal level in Caco-2 cells and human umbilical vein endothelial cells significantly. CONCLUSION rhANGPTL4 can function as a protective agent against intestinal injury induced by intestinal I/R and improve survival via maintenance of intestinal barrier structure and functions.
Collapse
Affiliation(s)
- Zi-Yi Wang
- Emergent Intensive Care Unit, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| | - Jian-Yu Lin
- Department of Gastrointestinal Surgery, Weihai Central Hospital, Weihai 264200, Shandong Province, China
| | - Yang-Rong Feng
- Graduate College, Shandong First Medical University, Jinan 271000, Shandong Province, China
| | - De-Shun Liu
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, Liaoning Province, China
| | - Xu-Zi Zhao
- Department of Pharmacology, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Tong Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100000, China
| | - Si-Yuan Li
- Department of General Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Jing-Chao Sun
- Department of General Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Shu-Feng Li
- Department of General Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Wen-Yan Jia
- Physiological Examination Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Hui-Rong Jing
- Department of General Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| |
Collapse
|