1
|
Li Y, Li XM, Wei LS, Ye JF. Advancements in mitochondrial-targeted nanotherapeutics: overcoming biological obstacles and optimizing drug delivery. Front Immunol 2024; 15:1451989. [PMID: 39483479 PMCID: PMC11524880 DOI: 10.3389/fimmu.2024.1451989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
In recent decades, nanotechnology has significantly advanced drug delivery systems, particularly in targeting subcellular organelles, thus opening new avenues for disease treatment. Mitochondria, critical for cellular energy and health, when dysfunctional, contribute to cancer, neurodegenerative diseases, and metabolic disorders. This has propelled the development of nanomedicines aimed at precise mitochondrial targeting to modulate their function, marking a research hotspot. This review delves into the recent advancements in mitochondrial-targeted nanotherapeutics, with a comprehensive focus on targeting strategies, nanocarrier designs, and their therapeutic applications. It emphasizes nanotechnology's role in enhancing drug delivery by overcoming biological barriers and optimizing drug design for specific mitochondrial targeting. Strategies exploiting mitochondrial membrane potential differences and specific targeting ligands improve the delivery and mitochondrial accumulation of nanomedicines. The use of diverse nanocarriers, including liposomes, polymer nanoparticles, and inorganic nanoparticles, tailored for effective mitochondrial targeting, shows promise in anti-tumor and neurodegenerative treatments. The review addresses the challenges and future directions in mitochondrial targeting nanotherapy, highlighting the need for precision, reduced toxicity, and clinical validation. Mitochondrial targeting nanotherapy stands at the forefront of therapeutic strategies, offering innovative treatment perspectives. Ongoing innovation and research are crucial for developing more precise and effective treatment modalities.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xiao-meng Li
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Li-si Wei
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Yu Q, Zhang J, Li J, Song Y, Pan J, Mei C, Cui M, He Q, Wang H, Li H, Cheng B, Zhang Y, Guo W, Zhu C, Chen S. Sirtuin 5-Mediated Desuccinylation of ALDH2 Alleviates Mitochondrial Oxidative Stress Following Acetaminophen-Induced Acute Liver Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402710. [PMID: 39159058 PMCID: PMC11497042 DOI: 10.1002/advs.202402710] [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: 03/14/2024] [Revised: 08/02/2024] [Indexed: 08/21/2024]
Abstract
Acetaminophen (APAP) overdose is a major cause of drug-induced liver injury. Sirtuins 5 (SIRT5) has been implicated in the development of various liver diseases. However, its involvement in APAP-induced acute liver injury (AILI) remains unclear. The present study aimed to explore the role of SIRT5 in AILI. SIRT5 expression is dramatically downregulated by APAP administration in mouse livers and AML12 hepatocytes. SIRT5 deficiency not only exacerbates liver injury and the inflammatory response, but also worsens mitochondrial oxidative stress. Conversely, the opposite pathological and biochemical changes are observed in mice with SIRT5 overexpression. Mechanistically, quantitative succinylome analysis and site mutation experiments revealed that SIRT5 desuccinylated aldehyde dehydrogenase 2 (ALDH2) at lysine 385 and maintained the enzymatic activity of ALDH2, resulting in the suppression of inflammation and mitochondrial oxidative stress. Furthermore, succinylation of ALDH2 at lysine 385 abolished its protective effect against AILI, and the protective effect of SIRT5 against AILI is dependent on the desuccinylation of ALDH2 at K385. Finally, virtual screening of natural compounds revealed that Puerarin promoted SIRT5 desuccinylase activity and further attenuated AILI. Collectively, the present study showed that the SIRT5-ALDH2 axis plays a critical role in AILI progression and might be a strategy for therapeutic intervention.
Collapse
Affiliation(s)
- Qiwen Yu
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Jiakai Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Jiye Li
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Yaodong Song
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Jie Pan
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Chaopeng Mei
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Mengwei Cui
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Qianqian He
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Haifeng Wang
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Huihui Li
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Bo Cheng
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Yan Zhang
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Key Laboratory for Digestive Organ TransplantationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Changju Zhu
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| | - Sanyang Chen
- Department of Emergency MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Henan Medical Key Laboratory of Emergency and Trauma ResearchZhengzhouHenan450052China
- Henan Emergency and Trauma Medicine Engineering Research CenterZhengzhouHenan450052China
| |
Collapse
|
3
|
Bejenaru LE, Biţă A, Mogoşanu GD, Segneanu AE, Radu A, Ciocîlteu MV, Bejenaru C. Polyphenols Investigation and Antioxidant and Anticholinesterase Activities of Rosmarinus officinalis L. Species from Southwest Romania Flora. Molecules 2024; 29:4438. [PMID: 39339433 PMCID: PMC11434282 DOI: 10.3390/molecules29184438] [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: 07/31/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Rosemary is one of the most important medicinal plants for natural therapy due to its multiple pharmacological properties, such as antioxidant, anti-inflammatory, neuroprotective, antiproliferative, antitumor, hepato- and nephroprotective, hypolipidemic, hypocholesterolemic, antihypertensive, anti-ischemic, hypoglycemic, radioprotective, antimicrobial, antiviral, antiallergic, and wound healing properties. Our study reports for the first time, over a 12-month period, the identification and quantification of polyphenols and the investigation of the antioxidant and acetylcholinesterase (AChE) inhibitory activities of the Rosmarinus officinalis L. species harvested at flowering from the flora of southwestern Romania (Oltenia Region). Identification and quantification of polyphenolic acids was made by ultra-high-performance liquid chromatography/mass spectrometry (UHPLC/MS). Total phenolic content was determined using the spectrophotometric method. In situ antioxidant and anticholinesterase activities were evaluated using 2,2-diphenyl-1-picrylhydrazyl (DPPH) and AChE inhibitory assay, respectively, on high-performance thin-layer chromatography (HPTLC) plates. DPPH radical scavenging activity was also assessed spectrophotometrically. The results revealed significant correlations between specific polyphenolic compounds and the measured biological activities, understanding the role of seasonal variations and providing insights into the optimal harvesting times and medicinal benefits of rosemary. Our research brings new information on the phytochemical profile of R. officinalis as a natural source of polyphenols with antioxidant and AChE inhibitory properties.
Collapse
Affiliation(s)
- Ludovic Everard Bejenaru
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Andrei Biţă
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - George Dan Mogoşanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Adina-Elena Segneanu
- Institute for Advanced Environmental Research, West University of Timişoara (ICAM-WUT), 4 Oituz Street, 300086 Timişoara, Romania
| | - Antonia Radu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Maria Viorica Ciocîlteu
- Department of Analytical Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Cornelia Bejenaru
- Department of Pharmaceutical Botany, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| |
Collapse
|
4
|
Luo G, Li H, Lu Q, Cao J, Lv H, Jiang Y. Effects of protoscoleces excretory-secretory products of Echinococcus granulosus on hepatocyte growth, function, and glucose metabolism. Acta Trop 2024; 249:107066. [PMID: 37944837 DOI: 10.1016/j.actatropica.2023.107066] [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: 02/23/2023] [Revised: 05/21/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
Cystic echinococcosis (CE) is one of the most widespread and harmful zoonotic parasitic diseases, which most commonly affects the liver. In this study, we characterized multiple changes in mouse hepatocytes following treatment with excretory-secretory products (ESPs) of Echinococcus granulosus protoscoleces (Eg-PSCs) by a factorial experiment. The cell counting kit-8 assay (CCK-8), the 5-ethynyl-2'-deoxyuridine (EdU) assay, and flow cytometry were used to detect the growth of hepatocytes. Inverted microscopy, scanning electron microscopy (SEM), and transmission electron microscopy (TEM) were used to observe the morphology and ultrastructure of hepatocytes. An automatic biochemical analyzer and an ELISA detection kit were used to determine six conventional hepatocyte enzymatic indices, the levels of five hepatocyte-synthesized substances, and the contents of glucose and lactate. Western blot analysis was conducted to analyze the protein expression of three apoptosis-related proteins, Bax, Bcl-2, cleaved caspase-3, and six glucose metabolism pathways rate-limiting enzymes in hepatocytes. The results showed that ESPs inhibited hepatocyte proliferation and promoted hepatocyte apoptosis. The cell membrane and microvilli of hepatocytes changed, and the nucleus, mitochondria and rough endoplasmic reticulum were damaged to varying degrees. The contents of iron, albumin (ALB), uric acid (UA) and urea were increased, and the activities of six enzymes in hepatocytes were increased except for the decrease of transferrin (TRF). The expression levels of all six key enzymes in the glucose metabolism pathway in hepatocytes were reduced. Our characterization provides a basis for further research on the pathogenesis, prevention and treatment of CE.
Collapse
Affiliation(s)
- Guangyi Luo
- Section for Hepatopancreatobiliary Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of Hepatopancreatobiliary Surgery, Anyue County People's Hospital, Ziyang, 642350, Sichuan, China
| | - Haiwen Li
- School of Basic Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Qiong Lu
- Department of Infectious Diseases, Anyue County People's Hospital, Ziyang, 642350, Sichuan, China
| | - Jiangtao Cao
- School of Basic Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Hailong Lv
- Section for Hepatopancreatobiliary Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| | - Yufeng Jiang
- School of Basic Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan, China.
| |
Collapse
|
5
|
Wang M, He Z, Xiong Z, Liu H, Zhou X, He J. Supplementation with grape seed extract, onion peel extract, or rosemary extract in the diet alleviates growth inhibition, liver damage, and oxidative stress induced by diquat in Lohmann chicks. Anim Biotechnol 2023; 34:5067-5074. [PMID: 37878368 DOI: 10.1080/10495398.2023.2271532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
The present study aimed to assess the impact of grape seed extract (GSE), onion peel extract (OPE), and rosemary extract (ROE) on Diquat-induced growth restriction and oxidative stress in Lohmann chicks. A total of 200 chicks were randomly assigned to 5 diets: the positive control (PC) group, the negative control (NC) group, GSE group, OPE group, and ROE group. During the first 7 d of trial, compared with NC and PC groups, the GSE group enhanced average daily feed intake (ADFI). From day 8-21, diquat injection resulted in reduced growth performance, increased platelet volume distribution width (PWD), malondialdehyde (MDA) concentration, and activities of alanine aminotransferase (ALT) in chick serum; it also decreased total protein (TP), albumin (ALB), globulin (GLB) concentration, activities of superoxide dismutase (SOD) and glutathione S-transferase (GST) in chick serum; furthermore, it increased MDA concentration while decreasing GST activities in liver. The NC group exhibited lower average daily gain (ADG) than other groups. Compared with NC group, GSE group reduced ALT activities, MDA levels, and red cell distribution width (RDW), and PDW concentration; it also increased SOD, GST activities. The ROE group lowered ALT activities and MDA concentration. The OPE group decreased ALT activities, and MDA levels, RDW, and PDW concentration, and increased SOD activities of chicks. These results suggest that supplementing antioxidants in diets alleviated oxidative stress in chicks challenged by improving antioxidant capacity and liver function.
Collapse
Affiliation(s)
- Man Wang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, P.R. China
| | - Zongze He
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, P.R. China
| | - Zhaolong Xiong
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, P.R. China
| | - Hongwei Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, P.R. China
| | - Xiang Zhou
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, P.R. China
| | - Jian He
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, P.R. China
| |
Collapse
|
6
|
Francolino R, Martino M, Caputo L, Amato G, Chianese G, Gargiulo E, Formisano C, Romano B, Ercolano G, Ianaro A, De Martino L, Feo VD. Phytochemical Constituents and Biological Activity of Wild and Cultivated Rosmarinus officinalis Hydroalcoholic Extracts. Antioxidants (Basel) 2023; 12:1633. [PMID: 37627628 PMCID: PMC10451299 DOI: 10.3390/antiox12081633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Rosmarinus officinalis L. is an aromatic evergreen plant from the Lamiaceae family. The purpose of this study was to compare the chemical profile and bioactivities of hydroalcoholic extracts derived from wild and cultivated R. officinalis. The chemical composition of the extracts was evaluated via LC-MS analysis, which revealed the presence of a wide range of phenolic compounds, including flavonoids, phenolic and terpenes. Both extracts showed a similar interesting antioxidant activity, probably related to their content of phenol and flavonoids. The analysis of anti-acetylcholinesterase (AChE), anti-butyrylcholinesterase (BChE), and anti-α-amylase activities showed analogous inhibition, except for AChE, in which the wild type was more active than the cultivated one. Finally, in vitro studies were performed using the J774A.1 murine macrophage cell line, to characterize the anti-inflammatory and the antioxidant effects of the extracts. As expected, pretreatment with the extracts significantly reduced the production proinflammatory cytokines and ROS through modulation of the nitric oxide pathway and the mitochondrial activity. Importantly, it is observed that the anti-inflammatory effect of the extracts was explicated through the inhibition of NF-kB and its downstream mediator COX-2. Collectively, these results demonstrated that these extracts could represent a starting point for developing novel therapeutic strategies for the treatment of inflammation-based diseases. Moreover, since no significant changes were observed in terms of composition and activity, both wild and cultivated R. officinalis extracts can be recommended for food and pharmaceutical purposes.
Collapse
Affiliation(s)
- Rosaria Francolino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (R.F.); (M.M.); (L.C.); (G.A.); (V.D.F.)
| | - Mara Martino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (R.F.); (M.M.); (L.C.); (G.A.); (V.D.F.)
| | - Lucia Caputo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (R.F.); (M.M.); (L.C.); (G.A.); (V.D.F.)
| | - Giuseppe Amato
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (R.F.); (M.M.); (L.C.); (G.A.); (V.D.F.)
| | - Giuseppina Chianese
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via D. Montesano, 49, 80131 Napoli, Italy; (G.C.); (E.G.); (B.R.); (G.E.); (A.I.)
| | - Ernesto Gargiulo
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via D. Montesano, 49, 80131 Napoli, Italy; (G.C.); (E.G.); (B.R.); (G.E.); (A.I.)
| | - Carmen Formisano
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via D. Montesano, 49, 80131 Napoli, Italy; (G.C.); (E.G.); (B.R.); (G.E.); (A.I.)
| | - Benedetta Romano
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via D. Montesano, 49, 80131 Napoli, Italy; (G.C.); (E.G.); (B.R.); (G.E.); (A.I.)
| | - Giuseppe Ercolano
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via D. Montesano, 49, 80131 Napoli, Italy; (G.C.); (E.G.); (B.R.); (G.E.); (A.I.)
| | - Angela Ianaro
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via D. Montesano, 49, 80131 Napoli, Italy; (G.C.); (E.G.); (B.R.); (G.E.); (A.I.)
| | - Laura De Martino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (R.F.); (M.M.); (L.C.); (G.A.); (V.D.F.)
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (R.F.); (M.M.); (L.C.); (G.A.); (V.D.F.)
- Institute of Food Science, National Research Council (C.N.R.), Via Roma, n. 60, 83100 Avellino, Italy
| |
Collapse
|
7
|
Xu J, Zhao L, Zhang X, Ying K, Zhou R, Cai W, Wu X, Jiang H, Xu Q, Miao D, Zeng Y, Yu F. Salidroside ameliorates acetaminophen-induced acute liver injury through the inhibition of endoplasmic reticulum stress-mediated ferroptosis by activating the AMPK/SIRT1 pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115331. [PMID: 37556956 DOI: 10.1016/j.ecoenv.2023.115331] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/14/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
Acetaminophen (APAP) overdose has long been considered a major cause of drug-induced liver injury. Ferroptosis is a type of programmed cell death mediated by iron-dependent lipid peroxidation. Endoplasmic reticulum (ER) stress is a systemic response triggered by the accumulation of unfolded or misfolded proteins in the ER. Ferroptosis and ER stress have been proven to contribute to the progression of APAP-induced acute liver injury (ALI). It was reported that salidroside protects against APAP-induced ALI, but the potential mechanism remain unknown. In this study, male C57BL/6 J mice were intraperitoneally (i.p.) injected APAP (500 mg/kg) to induce an ALI model. Salidroside was i.p. injected at a dose of 100 mg/kg 2 h prior to APAP administration. Mice were sacrificed 12 h after APAP injection and the liver and serum of the mice were obtained for histological and biochemistry analysis. AML12 cells were used in in vitro assays. The results indicated that salidroside mitigated glutathione degradation via inhibiting cation transport regulator homolog 1 (CHAC1) to attenuate ferroptosis, and simultaneously suppressing PERK-eIF2α-ATF4 axis-mediated ER stress, thus alleviating APAP-induced ALI. However, PERK activator CCT020312 and overexpression of ATF4 inhibited the protective function of salidroside on CHAC1-mediated ferroptosis. Besides this, activation of the AMPK/SIRT1 signaling pathway by salidroside was demonstrated to have a protective effect against APAP-induced ALI. Interestingly, selective inhibition of SIRT1 ameliorated the protective effects of salidroside on ER stress and ferroptosis. Overall, salidroside plays a significant part in the mitigation of APAP-induced ALI by activating the AMPK/SIRT1 signaling to inhibit ER stress-mediated ferroptosis in the ATF4-CHAC1 axis.
Collapse
Affiliation(s)
- Jun Xu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Luying Zhao
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangting Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kanglei Ying
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ruoru Zhou
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weimin Cai
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao Wu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haoran Jiang
- Department of Urology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qian Xu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dan Miao
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuan Zeng
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Fujun Yu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|