1
|
Lv R, Zhao Y, Wang X, He Y, Dong N, Min X, Liu X, Yu Q, Yuan K, Yue H, Yin Q. GLP-1 analogue liraglutide attenuates CIH-induced cognitive deficits by inhibiting oxidative stress, neuroinflammation, and apoptosis via the Nrf2/HO-1 and MAPK/NF-κB signaling pathways. Int Immunopharmacol 2024; 142:113222. [PMID: 39321702 DOI: 10.1016/j.intimp.2024.113222] [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: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Obstructive sleep apnea (OSA) is a common clinical condition linked to cognitive impairment, mainly characterized by chronic intermittent hypoxia (CIH). GLP-1 receptor agonist, known for promoting insulin secretion and reducing glucose levels, has demonstrated neuroprotective effects in various experimental models such as stroke, Alzheimer's disease, and Parkinson's disease. This study aims to investigate the potential role and mechanisms of the GLP-1 receptor agonist liraglutide in ameliorating OSA-induced cognitive deficits. CIH exposure, a well-established and mature OSA pathological model, was used both in vitro and in vivo. In vitro, CIH significantly activated oxidative stress, inflammation, and apoptosis in SH-SY5Y cells. Liraglutide enhanced the nuclear translocation of Nrf2, activating its downstream pathways, thereby mitigating CIH-induced injury in SH-SY5Y cells. Additionally, liraglutide modulated the MAPK/NF-κB signaling pathway, reducing the expression of inflammatory factors and proteins. In vivo, we subjected mice to an intermittent hypoxia incubator to mimic the pathogenesis of human OSA. The Morris water maze test revealed that CIH exposure substantially impaired spatial memory. Subsequent western blot analyses and histopathological examinations indicated that liraglutide could activate the Nrf2/HO-1 axis and inhibit the MAPK/NF-κB signaling pathway, thereby alleviating OSA-associated cognitive dysfunction in mice. These findings suggest that GLP-1 receptor agonists may offer a promising preventive strategy for OSA-associated cognitive impairment. By refining these findings, we provide new insights into GLP-1's protective mechanisms in combating cognitive deficits associated with CIH, underscoring its potential as a therapeutic agent for conditions linked to OSA.
Collapse
Affiliation(s)
- Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xiangzhen Min
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xueying Liu
- Jinan Third People's Hospital, Jinan, Shandong 250132, China
| | - Qin Yu
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing, China
| | - Hongmei Yue
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China; Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| |
Collapse
|
2
|
Ma CY, Yu AC, Sheng XH, Wang XG, Xing K, Xiao LF, Lv XZ, Guo Y, Long C, Qi XL. Supplementing ageing male laying breeders with lycopene alleviates oxidative stress in testis and improves testosterone secretion. Theriogenology 2024; 230:220-232. [PMID: 39341034 DOI: 10.1016/j.theriogenology.2024.09.026] [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: 10/15/2023] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Reproductive performance is a crucial aspect of poultry production and is carefully controlled by endocrine, paracrine, and autocrine factors. This study aimed to investigate the effect of lycopene on testosterone synthesis in Leydig cells of laying breeder roosters, clarify the mechanism of lycopene improving Leydig cells function and promoting testosterone production, and explore the role of related signal transduction pathways in testosterone synthesis. RESULTS A total of 96 healthy 55-week-old breeding roosters were randomly assigned to one of five dietary treatments. They were provided with a corn-soybean meal-based diet containing different levels of lycopene: 0 mg/kg (control), 50 mg/kg, 100 mg/kg, or 200 mg/kg. The experiment lasted for 6 weeks. With the increase in lycopene levels, the testosterone content in the plasma was significantly higher than in the control group. Testicular Leydig cells were isolated and cultured from fresh testicular tissue of 45-wk-old to 60-wk-old breeding roosters. Various doses of lycopene were administered to Leydig cells, and subsequently, cells were collected for the detection of cell viability and testosterone content. The optimal concentration of lycopene to be added was determined, and changes in mRNA expression and protein levels of key proteins involved in testosterone synthesis were investigated. The results showed that lycopene treatment significantly increased testosterone secretion, mRNA expression, and protein levels of steroid-producing enzymes. Cells were collected to measure the activity of antioxidant enzymes, the mRNA transcription level of apoptotic factors, and the protein expression of apoptotic factors after treatment with lycopene. The results showed that lycopene significantly increased the activities of antioxidant enzymes, and the ability to inhibit oxygen radicals, and decreased the content of malondialdehyde. Apoptosis was inhibited by regulating the expression of apoptosis-inducing and anti-apoptosis factors. After that, the MAPK signaling pathway and downstream SF-1, Nrf2 gene, and protein expression levels were detected. The results showed that lycopene treatment significantly increased the gene and protein expression of JNK, SF-1, and Nrf2, and significantly decreased the gene and protein expression of p38. CONCLUSIONS Lycopene treatment could promote testosterone synthesis of testicular Leydig cells by activating MAPK-SF-1 (increasing steroid-producing enzyme level) and MAPK-Nrf2 pathways (resisting oxidative damage).
Collapse
Affiliation(s)
- Chun-Yu Ma
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Ao-Chuan Yu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Xi-Hui Sheng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Xiang-Guo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Kai Xing
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Long-Fei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Xue-Ze Lv
- Department of Livestock and Poultry Products Testing, Beijing General Station of Animal Husbandry, Beijing, 100107, China
| | - Yong Guo
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Cheng Long
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China.
| | - Xiao-Long Qi
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China.
| |
Collapse
|
3
|
Pan X, Zhu R, Peng J, Liu H, Pan W, Jin Y, Pei J, Zhang L. Molecular mechanisms and potential targets of lycopene for alleviating renal ischemia-reperfusion injury revealed by network pharmacology and animal experiments. Int Immunopharmacol 2024; 143:113421. [PMID: 39442187 DOI: 10.1016/j.intimp.2024.113421] [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: 09/11/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Renal IRI is one of the leading causes of AKI. How to effectively mitigate renal IRI is important for the recovery of renal function. The regulatory mechanism of lycopene, a natural antioxidant, in renal IRI is currently unknown. Therefore, we utilized network pharmacology and animal experiments to explore the possible mechanisms and potential targets of lycopene for alleviating renal IRI. METHODS We obtained lycopene-regulated genes and renal IRI-related genes from the CTD database and GeneCards database, respectively. Subsequently, the two were intersected and the intersecting genes we defined as lycopene-regulated genes in renal IRI. Next, we explored their potential biological functions and mechanisms through enrichment analysis. Meanwhile, we constructed a rat renal IRI model and validated the protective effects of lycopene and related mechanisms. To further explore the Hub genes regulated by lycopene, we constructed a PPI protein interactions network and characterized the Hub genes using Cytoscape software. We also verified the expression of Hub genes using animal experiments and molecular docking techniques. Finally, we constructed TF-Hub gene and miRNA-Hub gene regulatory networks. RESULTS We obtained a total of 255 lycopene-regulated genes and 327 renal IRI-related genes. The enrichment analysis revealed that they were closely related to the regulation of oxidative stress as well as the regulation of inflammatory factors. At the same time, the MAPK signaling pathway was significantly enriched. Next, we found in animal experiments that lycopene significantly alleviated the level of oxidative stress and inflammation during renal IRI, and had a protective effect on kidney damage. Also, we found that this protective effect may be achieved by inhibiting the MAPK signaling pathway. Next, we identified a total of five Hub genes using Cytoscape software: TNF, AKT1, MAPK3, IL6 and CASP3. Both animal experiments and molecular docking techniques demonstrated that lycopene can effectively regulate the expression of Hub genes. Finally, our constructed TF-Hub gene and miRNA-Hub gene regulatory network provide a theoretical basis for further regulation of Hub genes in follow-up. CONCLUSIONS This study suggests that lycopene is a promising option in mitigating renal IRI. Lycopene may exert protective effects by inhibiting the MAPK signaling pathway.
Collapse
Affiliation(s)
- Xingyu Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Rong Zhu
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Jinpu Peng
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Hongli Liu
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Wenqing Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Yuhan Jin
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Jun Pei
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| | - Li Zhang
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China.
| |
Collapse
|
4
|
Wang J, Shen Y, Li L, Li L, Zhang J, Li M, Qiu F. Lycopene attenuates D-galactose-induced memory and behavioral deficits by mediating microbiota-SCFAs-gut-brain axis balance in female CD-1 mice. J Nutr Biochem 2024; 135:109777. [PMID: 39370012 DOI: 10.1016/j.jnutbio.2024.109777] [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: 05/06/2024] [Revised: 08/16/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Aging impairs cognitive function, whereas nutritional intervention can delay aging and age-related diseases. Lycopene (LYC), a naturally occurring carotenoid, posses multiple health-promoting properties, including neuroprotective function. Here, the effects of LYC on memory and behavioral deficits induced by D-galactose (D-gal) treatment and the relative contribution of LYC-derived gut microbiota in these process were investigated. Results demonstrated that LYC showed effective protection on D-gal induced cognitive deficit and neuronal damage. Moreover, LYC treatment has beneficial effects on gut barrier damage, microbiota dysbiosis and levels of SCFAs in D-gal-induced subacute aging mice. Next, fecal microbiota transplantation (FMT) experiment was performed and increased SCFAs were observed in mice received stools from D-gal+LYC group when compared with D-gal-FMT group. Thus, we added SCFAs treatment served as a control group in order to evaluated whether the alterations of gut-brain axis could be attributed to LYC-reshaped gut microbiota and SCFAs. Results showed that recipient mice received SCFAs and stools from D-gal+LYC group have similar beneficial effects in improving gut and brain function, demonstrated as: improved intestinal health via elevating antioxidant enzymes contents, increasing the expressions of tight junctions proteins and protecting gut barrier, enhanced mice working memory capacity via alleviating hippocampal neurons impairment, improving synaptic function and enhancing mitochondrial function in the intestinal pseudo-aseptic mice. In conclusion, our results demonstrated that LYC-derived microbiome played a pivotal role in the regulation of cognitive functions during aging and enhanced SCFAs formation might be an important signaling molecule connecting gut microbiome and brain.
Collapse
Affiliation(s)
- Jia Wang
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan, China.
| | - Yuqi Shen
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Lu Li
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Li Li
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Juan Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengling Li
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Fubin Qiu
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
5
|
Zhang A, Wang X, Fan M, Guan Y, Jiang Y, Jin S, Liu L, Huangfu H, Song C. Lycopene alleviates zearalenone-induced oxidative stress, apoptosis, and NLRP3 inflammasome activation in mice kidneys. Toxicon 2024; 249:108078. [PMID: 39181415 DOI: 10.1016/j.toxicon.2024.108078] [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: 07/09/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
The aim of this study was to investigate the protective effects of lycopene on renal damage caused by zearalenone (ZEN). Male Kunming mice were treated daily for 4 weeks by intragastric administration with 40 mg/kg ZEN in the presence or absence of lycopene (2.5 or 5 mg/kg). The results showed that lycopene markedly alleviated the damage of renal structure and function in mice induced by ZEN, as indicated by the reduced degree of pathological damage and the decreased levels of urea nitrogen and creatinine. Meanwhile, results of dihydroethidine (DHE) staining and biochemical markers revealed that ZEN exposure notably increased the levels of reactive oxygen species (ROS) and malondialdehyde (MDA), decreased the level of GSH, and reduced the activities of catalase (CAT) and superoxide dismutase (SOD). Administration of lycopene alleviated the increased oxidative stress induced by ZEN. Moreover, ZEN ingestion notably resulted in apoptosis, increased the protein levels of BCL2 associated X protein (Bax) and cleaved caspase-3, and decreased the protein levels of apoptosis regulator Bcl-2 (Bcl-2), which were reversed by lycopene intervention. Results of immunofluorescence demonstrated that lycopene reversed ZEN-induced the upregulation of NOD-like receptor pyrin domain-containing protein 3 (NLRP3), Caspase-1, and interleukin-1 beta (IL-1β) in mice kidneys. Lycopene supplementation could alleviate ZEN-induced renal toxicity by inhibiting oxidative stress, apoptosis, and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Aiguo Zhang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Xuefei Wang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Mingming Fan
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Yibo Guan
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Yuchen Jiang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Shuangxing Jin
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Lingling Liu
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Heping Huangfu
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Chao Song
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China.
| |
Collapse
|
6
|
Liu Y, Lv X, Yuan H, Wang X, Huang J, Wang L. Selenomethionine and Allicin Synergistically Mitigate Intestinal Oxidative Injury by Activating the Nrf2 Pathway. TOXICS 2024; 12:719. [PMID: 39453138 PMCID: PMC11510923 DOI: 10.3390/toxics12100719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Oxidative stress frequently contributes to intestinal barrier injury in animals and humans. It was reported that both Selenomethionine (SeMet) and allicin exhibit protective effects against a range of diseases caused by oxidative stress. This study aimed to investigate the synergistic antioxidant effects and underlying mechanisms of SeMet and allicin on a H2O2-induced intestinal barrier injury model using IPEC-J2 cells and mice. The results showed that H2O2 induced severe oxidative stress, including a decrease in cell viability, antioxidant level, migration capacity, and cell integrity. SeMet and allicin exhibited significant synergistic anti-oxidative effects on intestinal epithelial cells. The combined use of SeMet and allicin increased SOD activity, GSH content, and GSH/GSSG ratio while decreasing MDA, NO, and ROS content levels. Furthermore, we found that SeMet and allicin synergistically activated the nuclear factor erythroid-related factor 2 (Nrf2)-NAD(P)H dehydrogenase [quinone] 1 (NQO1) signaling pathway and down-regulated endoplasmic reticulum stress (ER stress)-related proteins. However, the synergistic antioxidative and intestinal barrier protective effects of SeMet and allicin were abolished by Nrf2 inhibitor ML385 in vitro and in vivo. In conclusion, SeMet and allicin synergistically attenuate intestinal barrier injury induced by excessively oxidative stress through the activation of the Nrf2 signaling pathway and inhibition ER stress. These findings support that the combined use of SeMet and allicin could enhance antioxidative properties and alleviate intestinal injury in further clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | - Liping Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (X.L.); (H.Y.); (X.W.); (J.H.)
| |
Collapse
|
7
|
Li D, Li F, Zhou Y, Tang Y, Hu Z, Wu Q, Xie T, Lin Q, Wang H, Luo F. Role and Mechanism of Sialic Acid in Alleviating Acute Lung Injury through In Vivo and In Vitro Models. Foods 2024; 13:2984. [PMID: 39335912 PMCID: PMC11431537 DOI: 10.3390/foods13182984] [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: 08/10/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Excessive inflammatory reactions are the most important pathological injury factor in acute lung injury (ALI). Our recent study found that sialic acid had an anti-colitis effect. In this study, the effect of sialic acid (SA) on acute lung inflammation was investigated. A lipopolysaccharide (LPS)-induced ALI animal model and LPS-stimulated HUVEC cell model were used to evaluate the anti-inflammatory effect of SA and study its molecular mechanisms. Compared with the LPS group, the lung index of the SA group decreased from 0.79 ± 0.05% to 0.58 ± 0.06% (LPS + 50 SA) and 0.62 ± 0.02% (LPS + 100 SA), with p < 0.01, suggesting that SA could improve the pulmonary edema of mice and alleviate LPS-induced lung injury. Transcriptome research identified 26 upregulated genes and 25 downregulated genes involved in the protection of SA against ALI. These genes are mainly related to the MAPK and NF-κB signaling pathways. Our study also proved that SA markedly downregulated the expression of inflammatory factors and blocked the JNK/p38/PPAR-γ/NF-κB pathway. Meanwhile, SA treatment also upregulated the expression of HO-1 and NQO1 in ALI mice. In vitro, SA obviously repressed the expressions of inflammatory cytokines and the JNK/p38-NF-κB/AP-1 pathway. SA also regulated the expression of oxidative stress-related genes through the Nrf2 pathway. Taken together, SA exhibits a protective role by modulating the anti-inflammatory and anti-oxidation pathways in ALI, and it may be a promising candidate for functional foods to prevent ALI.
Collapse
Affiliation(s)
- Dan Li
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
- Hunan Engineering Research Center of Full Life-Cycle Energy-Efficient Buildings and Environmental Health, School of Civil Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Fangyan Li
- Hunan Engineering Research Center of Full Life-Cycle Energy-Efficient Buildings and Environmental Health, School of Civil Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Yiping Tang
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Qi Wu
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Tiantian Xie
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Qinlu Lin
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Hanqing Wang
- Hunan Engineering Research Center of Full Life-Cycle Energy-Efficient Buildings and Environmental Health, School of Civil Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Feijun Luo
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| |
Collapse
|
8
|
Xu X, Zhu W, Miao M, Bai M, Fan J, Niu Y, Li Y, Zhang A, Jia Z, Wu M. Activation of LONP1 by 84-B10 alleviates aristolochic acid nephropathy via re-establishing mitochondrial and peroxisomal homeostasis. Chin J Nat Med 2024; 22:808-821. [PMID: 39326975 DOI: 10.1016/s1875-5364(24)60608-4] [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: 02/09/2024] [Indexed: 09/28/2024]
Abstract
Pharmaceutical formulations derived from Aristolochiaceae herbs, which contain aristolochic acids (AAs), are widely used for medicinal purposes. However, exposure to these plants and isolated AAs is linked to renal toxicity, known as AA nephropathy (AAN). Currently, the mechanisms underlying AAN are not fully understood, leading to unsatisfactory treatment strategies. In this study, we explored the protective role of 84-B10 (5-[[2-(4-methoxyphenoxy)-5-(trifluoromethyl) phenyl] amino]-5-oxo-3-phenylpentanoic acid) against AAN. RNA-seq analysis revealed that the mitochondrion and peroxisome were the most affected cellular components following 84-B10 treatment in AAN mice. Consistently, 84-B10 treatment preserved mitochondrial ultrastructure, restored mitochondrial respiration, enhanced the expression of key transporters (carnitine palmitoyltransferase 2) and enzymes (acyl-Coenzyme A dehydrogenase, medium chain) involved in mitochondrial fatty acid β-oxidation, and reduced mitochondrial ROS generation in both aristolochic acid I (AAI)-challenged mice kidneys and cultured proximal tubular epithelial cells. Additionally, 84-B10 treatment increased the expression of key transporters (ATP binding cassette subfamily D) and rate-limiting enzymes (acyl-CoA oxidase 1) involved in peroxisomal fatty acid β-oxidation and restored peroxisomal redox balance. Knocking down LONP1 expression diminished the protective effects of 84-B10 against AAN, suggesting LONP1-dependent protection. In conclusion, our study provides evidence that AAN is associated with significant disturbances in both mitochondrial and peroxisomal functions. The LONP1 activator 84-B10 demonstrates therapeutic potential against AAN, likely by maintaining homeostasis in both mitochondria and peroxisomes.
Collapse
Affiliation(s)
- Xinyue Xu
- School of Medicine, Southeast University, Nanjing 210009, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China
| | - Wenping Zhu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China
| | - Mengqiu Miao
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China
| | - Mi Bai
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China
| | - Jiaojiao Fan
- School of Medicine, Southeast University, Nanjing 210009, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China
| | - Yujia Niu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China
| | - Yuting Li
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China
| | - Aihua Zhang
- School of Medicine, Southeast University, Nanjing 210009, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China.
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China.
| | - Mengqiu Wu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China; Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in Children, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
9
|
Liu C, Li L, Li L, Li Q, Liu J, Zhang C, Cao Z, Ma L, Zeng X, Fu P. Ultrasmall magnolol/ebselen nanomicelles for preventing renal ischemia/reperfusion injury. Biomater Sci 2024; 12:4407-4426. [PMID: 39034872 DOI: 10.1039/d4bm00614c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Renal ischemia/reperfusion injury (RIRI) is an inevitable complication following kidney transplantation surgery, accompanied by the generation of a large amount of free radicals. A cascade of events including oxidative stress, extreme inflammation, cellular apoptosis, and thrombosis disrupts the microenvironment of renal cells and the hematological system, ultimately leading to the development of acute kidney injury (AKI). The current research primarily focuses on reducing inflammation and mitigating damage to renal cells through antioxidative approaches. However, studies on simultaneously modulating the renal hematologic system remain unreported. Herein, potent and novel drug-loaded nanomicelles can be efficiently self-assembled with magnolol (MG) and ebselen (EBS) by π-π conjugation, hydrophobic action and the surfactant properties of Tween-80. The ultrasmall MG/EBS nanomicelles (average particle size: 10-25 nm) not only fully preserve the activity of both drugs, but also greatly enhance drug utilization (encapsulation rates: MG: 90.1%; EBS: 49.3%) and reduce drug toxicity. Furthermore, EBS, as a glutathione peroxidase mimic and NO catalyst, combines with the multifunctional MG to scavenge free radicals and hydroperoxides, significantly inhibiting inflammation and thrombosis while effectively preventing apoptosis of vascular endothelial cells and renal tubular epithelial cells. This study provides a new strategy and theoretical foundation for the simultaneous regulation of kidney cells and blood microenvironment stability.
Collapse
Affiliation(s)
- Chang Liu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Linhua Li
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Li Li
- Institute of Clinical Pathology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Qingyin Li
- Department of Nephrology, Institute of Kidney Diseases, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jing Liu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Chunle Zhang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Zhengjiang Cao
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Liang Ma
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Xiaoxi Zeng
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Ping Fu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Liu ZH, Xu QY, Wang Y, Gao HX, Min YH, Jiang XW, Yu WH. Catalpol from Rehmannia glutinosa Targets Nrf2/NF-κB Signaling Pathway to Improve Renal Anemia and Fibrosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1451-1485. [PMID: 39075978 DOI: 10.1142/s0192415x24500575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Rehmannia glutinosa is widely recognized as a prominent medicinal herb employed by practitioners across various generations for the purpose of fortifying kidney yin. Within Rehmannia glutinosa, the compound known as catalpol (CAT) holds significant importance as a bioactive constituent. However, the protective effects of CAT on kidneys, including ameliorative effects on chronic kidney disease - most prominently renal anemia and renal fibrosis - have not been clearly defined. In this study, the kidney injury model of NRK-52E cells and C57BL/6N male mice was prepared by exposure to aristolochic acid I (AA-I), and it was discovered that CAT could ameliorate oxidative stress injury, inflammatory injury, apoptosis, renal anemia, renal fibrosis, and other renal injuries both in vivo and in vitro. Further treatment of NRK-52E cells with Nrf2 inhibitors (ML385) and activators (ML334), as well as NF-κB inhibitors (PDTC), validated CAT's ability to target Nrf2 activation. Furthermore, the expression of phosphorylated NF-κB p65, IL-6, and Cleaved-Caspase3 protein was inhibited. CAT also inhibited NF-κB, and then inhibited the expression of IL-6, p-STAS3, TGF-β1 protein. Therefore, CAT can regulate Nrf2/NF-κB signaling pathway, significantly correct renal anemia and renal fibrosis, and is conducive to the preservation of renal structure and function, thus achieving a protective effect on the kidneys.
Collapse
Affiliation(s)
- Zhi-Hui Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang Province, 150030, P. R. China
| | - Qing-Yang Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang Province, 150030, P. R. China
| | - Yu Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang Province, 150030, P. R. China
| | - Hong-Xin Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang Province, 150030, P. R. China
| | - Ya-Hong Min
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang Province, 150030, P. R. China
| | - Xiao-Wen Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang Province, 150030, P. R. China
| | - Wen-Hui Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang Province, 150030, P. R. China
- Chinese Veterinary Research Institute, Northeast Agricultural University, Harbin, Heilongjiang Province, 150030, P. R. China
- Heilongjiang Key Laboratory for the Prevention and Control of Common Animal Diseases, Harbin, Heilongjiang Province, 150030, P. R. China
| |
Collapse
|
11
|
Yao Y, Liu X, Niu X, Li Y, Han L. Lycopene Regulates Macrophage Immune Response through the Autophagy Pathway Mediated by RIPK1. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14747-14759. [PMID: 38889306 DOI: 10.1021/acs.jafc.4c02531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
The effects of lycopene (LP) on macrophage immune responses were evaluated in this study. Compared with the control treatment, LP treatment significantly increased cell vitality, phagocytic activity, and chemokine production in RAW264.7 cells. Additionally, compared with the control treatment, 4 μM LP treatment significantly activated autophagy, enhanced mitochondrial membrane potential, and upregulated receptor-interacting protein kinase 1 (RIPK1), while necrostatin-1 significantly reversed these effects of LP. Furthermore, compared with that in the control group, RIPK1 was significantly upregulated in the 4 μM LP and 4 μM LP + spautin-1 groups, whereas p-mTOR levels were reduced. More importantly, compared with that in the control group, p62 was significantly downregulated, and Beclin1, LC3-II, and Atg7 were upregulated in the 4 μM LP group, while spautin-1 significantly reversed these effects of LP. These results confirm that LP activates the mTOR/Beclin1/LC3/p62 autophagy signaling pathway through RIPK1, thereby enhancing the immune response of macrophages.
Collapse
Affiliation(s)
- Yupei Yao
- Hebei Key Laboratory of Public Health Safety, Ministry of Education & College of Public Health, Hebei University, Baoding 071002, China
| | - Xiaoran Liu
- Hebei Key Laboratory of Public Health Safety, Ministry of Education & College of Public Health, Hebei University, Baoding 071002, China
| | - Xiaoyan Niu
- Hebei Key Laboratory of Public Health Safety, Ministry of Education & College of Public Health, Hebei University, Baoding 071002, China
| | - Yaping Li
- Hebei Key Laboratory of Public Health Safety, Ministry of Education & College of Public Health, Hebei University, Baoding 071002, China
| | - Lirong Han
- Hebei Key Laboratory of Public Health Safety, Ministry of Education & College of Public Health, Hebei University, Baoding 071002, China
| |
Collapse
|
12
|
Yi BJ, Wang CC, Li XW, Xu YR, Ma XY, Jian PA, Talukder M, Li XN, Li JL. Lycopene Protects against Atrazine-Induced Kidney STING-Dependent PANoptosis through Stabilizing mtDNA via Interaction with Sam50/PHB1. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14956-14966. [PMID: 38820047 DOI: 10.1021/acs.jafc.4c02820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Atrazine (ATR) is a widely used herbicide worldwide that can cause kidney damage in humans and animals by accumulation in water and soil. Lycopene (LYC), a carotenoid with numerous biological activities, plays an important role in kidney protection due to its potent antioxidant and anti-inflammatory effects. The current study sought to investigate the role of interactions between mtDNA and the cGAS-STING signaling pathway in LYC mitigating PANoptosis and inflammation in kidneys induced by ATR exposure. In our research, 350 mice were orally administered LYC (5 mg/kg BW/day) and ATR (50 or 200 mg/kg BW/day) for 21 days. Our results reveal that ATR exposure induces a decrease in mtDNA stability, resulting in the release of mtDNA into the cytoplasm through the mPTP pore and the BAX pore and the mobilization of the cGAS-STING pathway, thereby inducing renal PANoptosis and inflammation. LYC can inhibit the above changes caused by ATR. In conclusion, LYC inhibited ATR exposure-induced histopathological changes, renal PANoptosis, and inflammation by inhibiting the cGAS-STING pathway. Our results demonstrate the positive role of LYC in ATR-induced renal injury and provide a new therapeutic target for treating renal diseases in the clinic.
Collapse
Affiliation(s)
- Bao-Jin Yi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Chi-Chiu Wang
- Department of Obstetrics & Gynaecology; Li Ka Shing Institute of Health Sciences; School of Biomedical Sciences; and The Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao-Wei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Ya-Ru Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xiang-Yu Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Ping-An Jian
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
- Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Xue-Nan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
- Department of Obstetrics & Gynaecology; Li Ka Shing Institute of Health Sciences; School of Biomedical Sciences; and The Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P. R. China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P. R. China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| |
Collapse
|
13
|
Xing F, Liu N, Wang C, Wang XD. Caffeic acid phenethyl ester promotes oxaliplatin sensitization in colon cancer by inhibiting autophagy. Sci Rep 2024; 14:14624. [PMID: 38918541 PMCID: PMC11199620 DOI: 10.1038/s41598-024-65409-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Colon cancer ranks as the third most prevalent form of cancer globally, with chemotherapy remaining the primary treatment modality. To mitigate drug resistance and minimize adverse effects associated with chemotherapy, selection of appropriate adjuvants assumes paramount importance. Caffeic acid phenethyl ester (CAPE), a naturally occurring compound derived from propolis, exhibits a diverse array of biological activities. We observed that the addition of CAPE significantly augmented the drug sensitivity of colon cancer cells to oxaliplatin. In SW480 and HCT116 cells, oxaliplatin combined with 10 µM CAPE reduced the IC50 of oxaliplatin from 14.24 ± 1.03 and 84.16 ± 3.02 µM to 2.11 ± 0.15 and 3.92 ± 0.17 µM, respectively. We then used proteomics to detect differentially expressed proteins in CAPE-treated SW480 cells and found that the main proteins showing changes in expression after CAPE treatment were p62 (SQSTM1) and LC3B (MAP1LC3B). Gene ontology analysis revealed that CAPE exerted antitumor and chemotherapy-sensitization effects through the autophagy pathway. We subsequently verified the differentially expressed proteins using immunoblotting. Simultaneously, the autophagy inhibitor bafilomycin A1 and the mCherry-EGFP-LC3 reporter gene were used as controls to detect the effect of CAPE on autophagy levels. Collectively, the results indicate that CAPE may exert antitumor and chemotherapy-sensitizing effects by inhibiting autophagy, offering novel insights for the development of potential chemosensitizing agents.
Collapse
Affiliation(s)
- Fei Xing
- Department of Gastrointestinal Nutrition Surgery, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Ning Liu
- Academic Center, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Can Wang
- Department of Gastrointestinal Nutrition Surgery, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Xu-Dong Wang
- Department of Gastrointestinal Nutrition Surgery, The Second Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
14
|
Abdolmaleki A, Karimian A, Khoshnazar SM, Asadi A, Samarein ZA, Smail SW, Bhattacharya D. The role of Nrf2 signaling pathways in nerve damage repair. Toxicol Res (Camb) 2024; 13:tfae080. [PMID: 38799411 PMCID: PMC11116835 DOI: 10.1093/toxres/tfae080] [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: 10/25/2023] [Revised: 03/05/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
The protein, Nuclear factor-E2-related factor 2 (Nrf2), is a transitory protein that acts as a transcription factor and is involved in the regulation of many cytoprotective genes linked to xenobiotic metabolism and antioxidant responses. Based on the existing clinical and experimental data, it can be inferred that neurodegenerative diseases are characterized by an excessive presence of markers of oxidative stress (OS) and a reduced presence of antioxidant defense systems in both the brain and peripheral tissues. The presence of imbalances in the homeostasis between oxidants and antioxidants has been recognized as a substantial factor in the pathogenesis of neurodegenerative disorders. The dysregulations include several cellular processes such as mitochondrial failure, protein misfolding, and neuroinflammation. These dysregulations all contribute to the disruption of proteostasis in neuronal cells, leading to their eventual mortality. A noteworthy component of Nrf2, as shown by recent research undertaken over the last decade, is to its role in the development of resistance to OS. Nrf2 plays a pivotal role in regulating systems that defend against OS. Extant research offers substantiation for the protective and defensive roles of Nrf2 in the context of neurodegenerative diseases. The purpose of this study is to provide a comprehensive analysis of the influence of Nrf2 on OS and its function in regulating antioxidant defense systems within the realm of neurodegenerative diseases. Furthermore, we evaluate the most recent academic inquiries and empirical evidence about the beneficial and potential role of certain Nrf2 activator compounds within the realm of therapeutic interventions.
Collapse
Affiliation(s)
- Arash Abdolmaleki
- Department of Biophysics, Faculty of Advanced Technologies, University of Mohaghegh Ardabili, PO Box: 179, Ardabil, 11367-56199, Iran
| | - Aida Karimian
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, PO Box: 179, Ardabil, 11367-56199, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Imam Khomeini Highway, Mustafa Khomeini Boulevard, Ibn Sina, Kerman, 9986598, Iran
| | - Asadollah Asadi
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, PO Box: 179, Ardabil, 11367-56199, Iran
| | - Zahra Akhavi Samarein
- Department of Counseling, Faculty of Education and Psychology, University of Mohaghegh Ardabili, PO Box: 179, Ardabil, 11367-56199, Iran
| | - Shukur Wasman Smail
- Department of Medical Microbiology, College of Science, Cihan University-Erbil, Kurdistan Region, 1235897, Iraq
| | - Deepak Bhattacharya
- Ph.D., Policy, Nursing, At Fight-Cancer at Home, Medicinal Toxicology & QC, Sri Radha Krishna Raas Mandir, KedarGouri Road, Bhubaneswar, Odisa 751002, India
| |
Collapse
|
15
|
Figueiredo ID, Lima TFO, Carlstrom PF, Assis RP, Brunetti IL, Baviera AM. Lycopene in Combination with Insulin Triggers Antioxidant Defenses and Increases the Expression of Components That Detoxify Advanced Glycation Products in Kidneys of Diabetic Rats. Nutrients 2024; 16:1580. [PMID: 38892513 PMCID: PMC11173891 DOI: 10.3390/nu16111580] [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: 04/21/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Biochemical events provoked by oxidative stress and advanced glycation may be inhibited by combining natural bioactives with classic therapeutic agents, which arise as strategies to mitigate diabetic complications. The aim of this study was to investigate whether lycopene combined with a reduced insulin dose is able to control glycemia and to oppose glycoxidative stress in kidneys of diabetic rats. METHODS Streptozotocin-induced diabetic rats were treated with 45 mg/kg lycopene + 1 U/day insulin for 30 days. The study assessed glycemia, insulin sensitivity, lipid profile and paraoxonase 1 (PON-1) activity in plasma. Superoxide dismutase (SOD) and catalase (CAT) activities and the protein levels of advanced glycation end-product receptor 1 (AGE-R1) and glyoxalase-1 (GLO-1) in the kidneys were also investigated. RESULTS An effective glycemic control was achieved with lycopene plus insulin, which may be attributed to improvements in insulin sensitivity. The combined therapy decreased the dyslipidemia and increased the PON-1 activity. In the kidneys, lycopene plus insulin increased the activities of SOD and CAT and the levels of AGE-R1 and GLO-1, which may be contributing to the antialbuminuric effect. CONCLUSIONS These findings demonstrate that lycopene may aggregate favorable effects to insulin against diabetic complications resulting from glycoxidative stress.
Collapse
Affiliation(s)
- Ingrid Delbone Figueiredo
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara 14800-903, SP, Brazil; (I.D.F.); (T.F.O.L.); (P.F.C.); (R.P.A.); (I.L.B.)
| | - Tayra Ferreira Oliveira Lima
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara 14800-903, SP, Brazil; (I.D.F.); (T.F.O.L.); (P.F.C.); (R.P.A.); (I.L.B.)
| | - Paulo Fernando Carlstrom
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara 14800-903, SP, Brazil; (I.D.F.); (T.F.O.L.); (P.F.C.); (R.P.A.); (I.L.B.)
| | - Renata Pires Assis
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara 14800-903, SP, Brazil; (I.D.F.); (T.F.O.L.); (P.F.C.); (R.P.A.); (I.L.B.)
- Institute of Health Sciences, Paulista University (Unip), Araraquara 14804-300, SP, Brazil
| | - Iguatemy Lourenço Brunetti
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara 14800-903, SP, Brazil; (I.D.F.); (T.F.O.L.); (P.F.C.); (R.P.A.); (I.L.B.)
| | - Amanda Martins Baviera
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara 14800-903, SP, Brazil; (I.D.F.); (T.F.O.L.); (P.F.C.); (R.P.A.); (I.L.B.)
| |
Collapse
|
16
|
Xu C, Wang Q, Du C, Chen L, Zhou Z, Zhang Z, Cai N, Li J, Huang C, Ma T. Histone deacetylase-mediated silencing of PSTPIP2 expression contributes to aristolochic acid nephropathy-induced PANoptosis. Br J Pharmacol 2024; 181:1452-1473. [PMID: 38073114 DOI: 10.1111/bph.16299] [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: 02/16/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND AND PURPOSE Aristolochic acid nephropathy (AAN) is a progressive kidney disease caused by using herbal medicines. Currently, no therapies are available to treat or prevent aristolochic acid nephropathy. Histone deacetylase (HDAC) plays a crucial role in the development and progression of renal disease. We tested whether HDAC inhibitors could prevent aristolochic acid nephropathy and determined the underlying mechanism. EXPERIMENTAL APPROACH HDACs expression in the aristolochic acid nephropathy model was examined. The activation of PANoptosis of mouse kidney and renal tubular epithelial cell were assessed after exposure to HDAC1 and HDAC2 blockade. Kidney-specific knock-in of proline-serine-threonine-phosphatase-interacting protein 2 (PSTPIP2) mice were used to investigate whether PSTPIP2 affected the production of PANoptosome. KEY RESULTS Aristolochic acid upregulated the expression of HDAC1 and HDAC2 in the kidneys. Notably, the HDAC1 and HDAC2 specific inhibitor, romidepsin (FK228, depsipeptide), suppressed aristolochic acid-induced kidney injury, epithelial cell pyroptosis, apoptosis and necroptosis (PANoptosis). Moreover, romidepsin upregulated PSTPIP2 in renal tubular epithelial cells, which was enhanced by aristolochic acid treatment. Conditional knock-in of PSTPIP2 in the kidney protected against aristolochic acid nephropathy. In contrast, the knockdown of PSTPIP2 expression in PSTPIP2-knock-in mice restored kidney damage and PANoptosis. PSTPIP2 function was determined in vitro using PSTPIP2 knockdown or overexpression in mouse renal tubular epithelial cells (mTECs). Additionally, PSTPIP2 was found to regulate caspase 8 in aristolochic acid nephropathy. CONCLUSION AND IMPLICATIONS HDAC-mediated silencing of PSTPIP2 may contribute to aristolochic acid nephropathy. Hence, HDAC1 and HDAC2 specific inhibitors or PSTPIP2 could be valuable therapeutic agents for preventing aristolochic acid nephropathy.
Collapse
Affiliation(s)
- Chuanting Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, Hefei, China
| | - Qi Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Changlin Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Lu Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Zhongnan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Zhenming Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Na Cai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Taotao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, Hefei, China
- Anhui Provincial Institute of Translational Medicine, Hefei, China
| |
Collapse
|
17
|
Wang Y, Ping Z, Gao H, Liu Z, Xv Q, Jiang X, Yu W. LYC inhibits the AKT signaling pathway to activate autophagy and ameliorate TGFB-induced renal fibrosis. Autophagy 2024; 20:1114-1133. [PMID: 38037248 PMCID: PMC11135866 DOI: 10.1080/15548627.2023.2287930] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Renal fibrosis is a typical pathological change in chronic kidney disease (CKD). Epithelial-mesenchymal transition (EMT) is the predominant stage. Activation of macroautophagy/autophagy plays a crucial role in the process of EMT. Lycopene (LYC) is a highly antioxidant carotenoid with pharmacological effects such as anti-inflammation, anti-apoptosis and mediation of autophagy. In this study, we demonstrated the specific mechanism of LYC in activating mitophagy and improving renal fibrosis. The enrichment analysis results of GO and KEGG showed that LYC had high enrichment values with autophagy. In this study, we showed that LYC alleviated aristolochic acid I (AAI)-induced intracellular expression of PINK1, TGFB/TGF-β, p-SMAD2, p-SMAD3, and PRKN/Parkin, recruited expression of MAP1LC3/LC3-II and SQSTM1/p62, decreased mitochondrial membrane potential (MMP), and ameliorated renal fibrosis in mice. When we simultaneously intervened NRK52E cells using bafilomycin A1 (Baf-A1), AAI, and LYC, intracellular MAP1LC3-II and SQSTM1 expression was significantly increased. A similar result was seen in renal tissue and cells when treated in vitro and in vivo with CQ, AAI, and LYC, and the inhibitory effect of LYC on the AAI-activated SMAD2-SMAD3 signaling pathway was attenuated. Molecular docking simulation experiments showed that LYC stably bound to the AKT active site. After intervention of cells with AAI and GSK-690693, the expression of PINK1, PRKN, MAP1LC3-II, BECN1, p-SMAD2 and p-SMAD3 was increased, and the expression of SQSTM1 was decreased. However, SC79 inhibited autophagy and reversed the inhibitory effect of LYC on EMT. The results showed that LYC could inhibit the AKT signaling pathway to activate mitophagy and reduce renal fibrosis.Abbreviation: AA: aristolochic acid; ACTA2/α-SMA: actin alpha 2, smooth muscle, aorta; ACTB: actin beta; AKT/protein kinase B: thymoma viral proto-oncogene; BAF-A1: bafilomycin A1; BECN1: beclin 1, autophagy related; CCN2/CTGF: cellular communication network factor 2; CDH1/E-Cadherin: cadherin 1; CKD: chronic kidney disease; COL1: collagen, type I; COL3: collagen, type III; CQ: chloroquine; ECM: extracellular matrix; EMT: epithelial-mesenchymal transition; FN1: fibronectin 1; LYC: lycopene; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MMP: mitochondrial membrane potential; MTOR: mechanistic target of rapamycin kinase ; PI3K: phosphoinositide 3-kinase; PINK1: PTEN induced putative kinase 1; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; PPI: protein-protein interaction; SMAD2: SMAD family member 2; SMAD3: SMAD family member 3; SQSTM1/p62: sequestosome 1; TGFB/TGFβ: transforming growth factor, beta; VIM: vimentin.
Collapse
Affiliation(s)
- Yu Wang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhenlei Ping
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hongxin Gao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhihui Liu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qingyang Xv
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of Animal Pathogenesis and Comparative Medicine in Heilongjiang Province, Northeast Agricultural University, Harbin, China
| |
Collapse
|
18
|
Nayak SPRR, Boopathi S, Chandrasekar M, Yamini B, Chitra V, Almutairi BO, Arokiyaraj S, Guru A, Arockiaraj J. Indole-3 acetic acid induced cardiac hypertrophy in Wistar albino rats. Toxicol Appl Pharmacol 2024; 486:116917. [PMID: 38555004 DOI: 10.1016/j.taap.2024.116917] [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: 01/05/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Indole-3-acetic acid (IAA) is the most widely utilized plant growth regulator. Despite its extensive usage, IAA is often overlooked as an environmental pollutant. Due to its protein-binding nature, it also functions as a uremic toxin, contributing to its association with chronic kidney disease (CKD). While in vitro and epidemiological research have demonstrated this association, the precise impact of IAA on cardiovascular disease in animal models is unknown. The main objective of this study is to conduct a mechanistic analysis of the cardiotoxic effects caused by IAA using male Wistar albino rats as the experimental model. Three different concentrations of IAA (125, 250, 500 mg/kg) were administered for 28 days. The circulating IAA concentration mimicked previously observed levels in CKD patients. The administration of IAA led to a notable augmentation in heart size and heart-to-body weight ratio, indicating cardiac hypertrophy. Echocardiographic assessments supported these observations, revealing myocardial thickening. Biochemical and gene expression analyses further corroborated the cardiotoxic effects of IAA. Dyslipidemia, increased serum c-Troponin-I levels, decreased SOD and CAT levels, and elevated lipid peroxidation in cardiac tissue were identified. Moreover, increased expression of cardiac inflammatory biomarkers, including ANP, BNP, β-MHC, Col-III, TNF-α, and NF-κB, was also found in the IAA-treated animals. Histopathological analysis confirmed the cardiotoxic nature of IAA, providing additional evidence of its adverse effects on cardiovascular health. These results offer insights into the potential negative impact of IAA on cardiovascular function, and elucidating the underlying mechanisms of its cardiotoxicity.
Collapse
Affiliation(s)
- S P Ramya Ranjan Nayak
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Seenivasan Boopathi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Munisamy Chandrasekar
- Resident Veterinary Services Section, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai 600007, Tamil Nadu, India
| | - B Yamini
- International Center for Cardio Thoracic and Vascular Diseases, Dr K M Cherian Heart Foundation, Anna Nagar, Chennai 600040, Tamil Nadu, India
| | - Vellapandian Chitra
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Bader O Almutairi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Selvaraj Arokiyaraj
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
19
|
Gao X, Lin B, Chen C, Fang Z, Yang J, Wu S, Chen Q, Zheng K, Yu Z, Li Y, Gao X, Lin G, Chen L. Lycopene from tomatoes and tomato products exerts renoprotective effects by ameliorating oxidative stress, apoptosis, pyroptosis, fibrosis, and inflammatory injury in calcium oxalate nephrolithiasis: the underlying mechanisms. Food Funct 2024; 15:4021-4036. [PMID: 38584465 DOI: 10.1039/d4fo00042k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Several mechanisms underlying nephrolithiasis, one of the most common urological diseases, involve calcium oxalate formation, including oxidative stress, inflammatory reactions, fibrosis, pyroptosis, and apoptosis. Although lycopene has strong antioxidant activity, its protective effects against CaOx-induced injury have not yet been reported. This study aimed to systematically investigate the protective effects of lycopene and explore its mechanisms and molecular targets. Crystal deposition, renal function, oxidative stress, inflammatory response, fibrosis, pyroptosis, and apoptosis were assessed to evaluate the renoprotective effects of lycopene against crystal formation in a CaOx rat model and oxalate-stimulated NRK-52E and HK-2 cells. Lycopene markedly ameliorated crystal deposition, restored renal function, and suppressed kidney injury by reducing oxidative stress, apoptosis, inflammation, fibrosis, and pyroptosis in the rats. In cell models, lycopene pretreatment reversed reactive oxygen species increase, apoptotic damage, intracellular lactate dehydrogenase release, cytotoxicity, pyroptosis, and extracellular matrix deposition. Network pharmacology and proteomic analyses were performed to identify lycopene target proteins under CaOx-exposed conditions, and the results showed that Trappc4 might be a pivotal target gene for lycopene, as identified by cellular thermal shift assay and surface plasmon resonance analyses. Based on molecular docking, molecular dynamics simulations, alanine scanning mutagenesis, and saturation mutagenesis, we observed that lycopene directly interacts with Trappc4 via hydrophobic bonds, which may be attributed to the PHE4 and PHE142 residues, preventing ERK1/2 or elevating AMPK signaling pathway phosphorylation events. In conclusion, lycopene might ameliorate oxalate-induced renal tubular epithelial cell injury via the Trappc4/ERK1/2/AMPK pathway, indicating its potential for the treatment of nephrolithiasis.
Collapse
Affiliation(s)
- Xiaomin Gao
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Southern Baixiang, OuHai District, Wenzhou, Zhejiang, 325006, P.R. China.
| | - Binwei Lin
- Department of Urology, Rui'an People's Hospital, The Third Affiliated Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang province, 325200, P.R. China
| | - Chen Chen
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, P.R. China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, P.R. China
| | - Ziyu Fang
- Department of Urology, Changhai Hospital, Navy Medical University, Changhai Road, YangPu District, Shanghai, 200433, P.R. China.
| | - Jinzhao Yang
- The Department of Pharmacy, The Third Clinical Institute Affiliated to Wenzhou Medical University (Wenzhou People's Hospital), Wenzhou, Zhejiang, 325006, P.R. China
| | - Shuzhi Wu
- The Department of Neurology, The Third Clinical Institute Affiliated to Wenzhou Medical University (Wenzhou People's Hospital), Wenzhou, Zhejiang, 325006, P.R. China
| | - Qing Chen
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, P.R. China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, P.R. China
| | - Kewen Zheng
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Southern Baixiang, OuHai District, Wenzhou, Zhejiang, 325006, P.R. China.
| | - Zhixian Yu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Southern Baixiang, OuHai District, Wenzhou, Zhejiang, 325006, P.R. China.
| | - Yeping Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Southern Baixiang, OuHai District, Wenzhou, Zhejiang, 325006, P.R. China.
| | - Xiaofeng Gao
- Department of Urology, Changhai Hospital, Navy Medical University, Changhai Road, YangPu District, Shanghai, 200433, P.R. China.
| | - Guanyang Lin
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, P.R. China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, P.R. China
| | - Lianguo Chen
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, P.R. China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, P.R. China
| |
Collapse
|
20
|
Li M, Tang S, Peng X, Sharma G, Yin S, Hao Z, Li J, Shen J, Dai C. Lycopene as a Therapeutic Agent against Aflatoxin B1-Related Toxicity: Mechanistic Insights and Future Directions. Antioxidants (Basel) 2024; 13:452. [PMID: 38671900 PMCID: PMC11047733 DOI: 10.3390/antiox13040452] [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: 03/13/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Aflatoxin (AFT) contamination poses a significant global public health and safety concern, prompting widespread apprehension. Of the various AFTs, aflatoxin B1 (AFB1) stands out for its pronounced toxicity and its association with a spectrum of chronic ailments, including cardiovascular disease, neurodegenerative disorders, and cancer. Lycopene, a lipid-soluble natural carotenoid, has emerged as a potential mitigator of the deleterious effects induced by AFB1 exposure, spanning cardiac injury, hepatotoxicity, nephrotoxicity, intestinal damage, and reproductive impairment. This protective mechanism operates by reducing oxidative stress, inflammation, and lipid peroxidation, and activating the mitochondrial apoptotic pathway, facilitating the activation of mitochondrial biogenesis, the endogenous antioxidant system, and the nuclear factor erythroid 2-related factor 2 (Nrf2)/kelch-like ECH-associated protein 1 (KEAP1) and peroxisome proliferator-activated receptor-γ coactivator-1 (PGC-1) pathways, as well as regulating the activities of cytochrome P450 (CYP450) enzymes. This review provides an overview of the protective effects of lycopene against AFB1 exposure-induced toxicity and the underlying molecular mechanisms. Furthermore, it explores the safety profile and potential clinical applications of lycopene. The present review underscores lycopene's potential as a promising detoxification agent against AFB1 exposure, with the intent to stimulate further research and practical utilization in this domain.
Collapse
Affiliation(s)
- Meng Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (M.L.); (S.T.); (S.Y.); (Z.H.)
| | - Shusheng Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (M.L.); (S.T.); (S.Y.); (Z.H.)
| | - Xinyan Peng
- College of Life Sciences, Yantai University, Yantai 264000, China;
| | - Gaurav Sharma
- Cardiovascular and Thoracic Surgery, Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Shutao Yin
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (M.L.); (S.T.); (S.Y.); (Z.H.)
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (M.L.); (S.T.); (S.Y.); (Z.H.)
| | - Jichang Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, China;
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (M.L.); (S.T.); (S.Y.); (Z.H.)
| | - Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (M.L.); (S.T.); (S.Y.); (Z.H.)
| |
Collapse
|
21
|
Long Y, Paengkoum S, Lu S, Niu X, Thongpea S, Taethaisong N, Han Y, Paengkoum P. Physicochemical properties, mechanism of action of lycopene and its application in poultry and ruminant production. Front Vet Sci 2024; 11:1364589. [PMID: 38562916 PMCID: PMC10983797 DOI: 10.3389/fvets.2024.1364589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Lycopene is a kind of natural carotenoid that could achieve antioxidant, anti-cancer, lipid-lowering and immune-improving effects by up-regulating or down-regulating genes related to antioxidant, anti-cancer, lipid-lowering and immunity. Furthermore, lycopene is natural, pollution-free, and has no toxic side effects. The application of lycopene in animal production has shown that it could improve livestock production performance, slaughter performance, immunity, antioxidant capacity, intestinal health, and meat quality. Therefore, lycopene as a new type of feed additive, has broader application prospects in many antibiotic-forbidden environments. This article serves as a reference for the use of lycopene as a health feed additive in animal production by going over its physical and chemical characteristics, antioxidant, lipid-lowering, anti-cancer, and application in animal production.
Collapse
Affiliation(s)
- Yong Long
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Siwaporn Paengkoum
- Program in Agriculture, Faculty of Science and Technology, Nakhon Ratchasima Rajabhat University, Nakhon Ratchasima, Thailand
| | - Shengyong Lu
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Xinran Niu
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Sorasak Thongpea
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Nittaya Taethaisong
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Yong Han
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Pramote Paengkoum
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| |
Collapse
|
22
|
Wang Y, Xiong Z, Qiao Y, Zhang Q, Zhou G, Zhou C, Ma X, Jiang X, Yu W. Acetyl-11-keto-beta-boswellic acid modulates macrophage polarization and Schwann cell migration to accelerate spinal cord injury repair in rats. CNS Neurosci Ther 2024; 30:e14642. [PMID: 38430464 PMCID: PMC10908365 DOI: 10.1111/cns.14642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Inhibiting secondary inflammatory damage caused by glial cells and creating a stable microenvironment is one of the main strategies to investigate drugs for the treatment of spinal cord injury. Acetyl-11-keto-beta-boswellic acid (AKBA) is the active component of the natural drug boswellia, which has anti-inflammatory and antioxidant effects and offers a possible therapeutic option for spinal cord injury. METHODS In this study, a spinal cord injury model was established by crushing spinal cord, respectively, to detect the M1 macrophage inflammatory markers: iNOS, TNF-α, IL-1β, and the M2 macrophage markers CD206, ARG-1, IL-10, and the detection of antioxidant enzymes and MDA. In vitro, macrophages were cultured to verify the main mechanism of the macrophage switch from Nrf2/HO-1 to M2 type by flow cytometry, immunofluorescence, and other techniques. Macrophage and Schwann cell co-culture validated the migration mechanism of Schwann cells promoted by AKBA. RESULTS AKBA significantly enhanced the antioxidant enzyme activities of CAT, GSH-Px, T-AOC, and SOD, reduced MDA content, and reduced oxidative damage caused by spinal cord injury via the Nrf2/HO-1 signaling pathway; AKBA mediates Nrf2/HO-1/IL-10, converts macrophages from M1 to M2 type, reduces inflammation, and promotes Schwann cell migration, thereby accelerating the repair of spinal cord injury in rats. CONCLUSIONS Our work demonstrates that AKBA can attenuate oxidative stress as well as the secondary inflammatory injury caused by macrophages after SCI, promote Schwann cell migration to the injury site, and thus accelerate the repair of the injured spinal cord.
Collapse
Affiliation(s)
- Yao Wang
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Zongliang Xiong
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Yuncong Qiao
- School of Life SciencesNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Qiyuan Zhang
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Guanghu Zhou
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Chong Zhou
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Xianglin Ma
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Xiaowen Jiang
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| | - Wenhui Yu
- Department of Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and TreatmentNortheast Agricultural UniversityHarbinHeilongjiangChina
- Institute of Chinese Veterinary MedicineNortheast Agricultural UniversityHarbinHeilongjiangChina
| |
Collapse
|
23
|
Zhang Q, Tian L, Hu Y, Jiang W, Wang X, Chen L, Cheng S, Ying J, Jiang B, Zhang L. Aristolochic acid I aggravates oxidative stress-mediated apoptosis by inhibiting APE1/Nrf2/HO-1 signaling. Toxicol Mech Methods 2024; 34:20-31. [PMID: 37621060 DOI: 10.1080/15376516.2023.2250429] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Nephrotoxicity induced by aristolochic acid I (AAI) is related to redox stress and apoptosis. Apurinic/apyrimidine endonuclease 1 (APE1) has antioxidant and anti-apoptotic effects. This study investigated the potential role of APE1 in AAI-induced nephrotoxicity. Renal injury was successfully induced in C57BL/6J mice by intraperitoneal injection of AAI every other day for 28 days. Expressions of APE1, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase 1 (HO-1) in renal tissues of the model mice was inhibited, accompanied by oxidative damage and apoptosis. Similar results were obtained in vitro in human proximal tubular (HK-2) cells damaged by AAI. In the presence of a low concentration of the APE1 inhibitor E3330, expression of Nrf2 and HO-1 proteins in HK-2 cells was decreased and AAI-induced apoptosis was aggravated. Overexpression of APE1 in HK-2 cells promoted the expression of Nrf2 and HO-1, and alleviated apoptosis and renal injury induced by AAI. The collective findings demonstrate that AAI can inhibit the induction of oxidative stress and apoptosis by the APE1/Nrf2/HO-1 axis, leading to AAI renal injury. Targeting APE1 may be an effective therapeutic strategy to treat AA nephrotoxicity.
Collapse
Affiliation(s)
- Qi Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Tian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yongkang Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenjuan Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xian Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Langqun Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyu Cheng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiahui Ying
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Baoping Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liang Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
24
|
Wu YF, Tang ZY, Deng YX, Liu K, Gu XR, Zhou GL, Huang YJ, Lin XQ, Zhou LY, Zuo XC. Identification and analysis of differently expressed transcription factors in aristolochic acid nephropathy. Environ Health Prev Med 2024; 29:30. [PMID: 38777778 PMCID: PMC11157247 DOI: 10.1265/ehpm.23-00245] [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: 09/05/2023] [Accepted: 03/30/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Aristolochic acid nephropathy (AAN) is a rapidly progressive interstitial nephropathy caused by Aristolochic acid (AA). AAN is associated with the development of nephropathy and urothelial carcinoma. It is estimated that more than 100 million people worldwide are at risk of developing AAN. However, the underlying mechanisms driving renal deterioration in AAN remain poorly understood, and the treatment options are limited. METHODS We obtained GSE27168 and GSE136276 series matrix data from the Gene Expression Omnibus (GEO) related to AAN. Using the R Studio environment, we applied the limma package and WGCNA package to identify co-differently expressed genes (co-DEGs). By GO/KEGG/GSVA analysis, we revealed common biological pathways. Subsequently, co-DEGs were subjected to the String database to construct a protein-protein interaction (PPI) network. The MCC algorithms implemented in the Cytohubba plugin were employed to identify hub genes. The hub genes were cross-referenced with the transcription factor (TF) database to identify hub TFs. Immune infiltration analysis was performed to identify key immune cell groups by utilizing CIBERSORT. The expressions of AAN-associated hub TFs were verified in vivo and in vitro. Finally, siRNA intervention was performed on the two TFs to verify their regulatory effect in AAN. RESULTS Our analysis identified 88 co-DEGs through the "limma" and "WGCNA" R packages. A PPI network comprising 53 nodes and 34 edges was constructed with a confidence level >0.4. ATF3 and c-JUN were identified as hub TFs potentially linked to AAN. Additionally, expressions of ATF3 and c-JUN positively correlated with monocytes, basophils, and vessels, and negatively correlated with eosinophils and endothelial cells. We observed a significant increase in protein and mRNA levels of these two hub TFs. Furthermore, it was found that siRNA intervention targeting ATF3, but not c-JUN, alleviated cell damage induced by AA. The knockdown of ATF3 protects against oxidative stress and inflammation in the AAN cell model. CONCLUSION This study provides novel insights into the role of ATF3 in AAN. The comprehensive analysis sheds light on the molecular mechanisms and identifies potential biomarkers and drug targets for AAN treatment.
Collapse
Affiliation(s)
- Yi-Feng Wu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Zhi-Yao Tang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Yi-Xuan Deng
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Kun Liu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Xu-Rui Gu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Guang-Liang Zhou
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Yu-Jie Huang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Xiao-Qing Lin
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Lin-Yun Zhou
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Xiao-Cong Zuo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410000, China
| |
Collapse
|
25
|
Fang G, Li X, Yang F, Huang T, Qiu C, Peng K, Yang Y, Lan C. Galangin attenuates doxorubicin-induced cardiotoxicity via activating nuclear factor erythroid 2-related factor 2/heme oxygenase 1 signaling pathway to suppress oxidative stress and inflammation. Phytother Res 2023; 37:5854-5870. [PMID: 37655750 DOI: 10.1002/ptr.7991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023]
Abstract
Doxorubicin (DOX) has aroused contradiction between its potent anti-tumor capacity and severe cardiotoxicity. Galangin (Gal) possesses antioxidant, anti-inflammatory, and antiapoptotic activities. We aimed to explore the role and underlying mechanisms of Gal on DOX-induced cardiotoxicity. Mice were intraperitoneally injected with DOX (3 mg/kg, every 2 days for 2 weeks) to generate cardiotoxicity model and Gal (15 mg/kg, 2 weeks) was co-administered via gavage daily. Nuclear factor erythroid 2-related factor 2 (Nrf2) specific inhibitor, ML385, was employed to explore the underlying mechanisms. Compared to DOX-insulted mice, Gal effectively improved cardiac dysfunction and ameliorated myocardial damage. DOX-induced increase of reactive oxygen species, malondialdehyde, and NADPH oxidase activity and downregulation of superoxide dismutase (SOD) activity were blunted by Gal. Gal also markedly blocked increase of IL-1β, IL-6, and TNF-α in DOX-insulted heart. Mechanistically, Gal reversed DOX-induced downregulation of Nrf2, HO-1, and promoted nuclear translocation of Nrf2. ML385 markedly blunted the cardioprotective effects of Gal, as well as inhibitive effects on oxidative stress and inflammation. Gal ameliorates DOX-induced cardiotoxicity by suppressing oxidative stress and inflammation via activating Nrf2/HO-1 signaling pathway. Gal may serve as a promising cardioprotective agent for DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Guangyao Fang
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Xiuchuan Li
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| | - Fengyuan Yang
- Department of Nephrology, General Hospital of Western Theater Command, Chengdu, China
| | - Ting Huang
- Department of Medical Oncology, People's Hospital of Luotian County, Huanggang, China
| | - Chenming Qiu
- Department of Burn and Plastic Surgery, General Hospital of Western Theater Command, Chengdu, China
| | - Ke Peng
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| | - Yongjian Yang
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Cong Lan
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
26
|
Xie D, Song C, Qin T, Zhai Z, Cai J, Dai J, Sun T, Xu Y. Moschus ameliorates glutamate-induced cellular damage by regulating autophagy and apoptosis pathway. Sci Rep 2023; 13:18586. [PMID: 37903904 PMCID: PMC10616123 DOI: 10.1038/s41598-023-45878-7] [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: 11/16/2022] [Accepted: 10/25/2023] [Indexed: 11/01/2023] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, causes short-term memory and cognition declines. It is estimated that one in three elderly people die from AD or other dementias. Chinese herbal medicine as a potential drug for treating AD has gained growing interest from many researchers. Moschus, a rare and valuable traditional Chinese animal medicine, was originally documented in Shennong Ben Cao Jing and recognized for its properties of reviving consciousness/resuscitation. Additionally, Moschus has the efficacy of "regulation of menstruation with blood activation, relief of swelling and pain" and is used for treating unconsciousness, stroke, coma, and cerebrovascular diseases. However, it is uncertain whether Moschus has any protective effect on AD patients. We explored whether Moschus could protect glutamate (Glu)-induced PC12 cells from cellular injury and preliminarily explored their related action mechanisms. The chemical compounds of Moschus were analyzed and identified by GC-MS. The Glu-induced differentiated PC12 cell model was thought to be the common AD cellular model. The study aims to preliminarily investigate the intervention effect of Moschus on Glu-induced PC12 cell damage as well as their related action mechanisms. Cell viability, lactate dehydrogenase (LDH), mitochondrial reactive oxygen species, mitochondrial membrane potential (MMP), cell apoptosis, autophagic vacuoles, autolysosomes or autophagosomes, proteins related to apoptosis, and the proteins related to autophagy were examined and analyzed. Seventeen active compounds of the Moschus sample were identified based on GC-MS analysis. In comparison to the control group, Glu stimulation increased cell viability loss, LDH release, mitochondrial damage, loss of MMP, apoptosis rate, and the number of cells containing autophagic vacuoles, and autolysosomes or autophagosomes, while these results were decreased after the pretreatment with Moschus and 3-methyladenine (3-MA). Furthermore, Glu stimulation significantly increased cleaved caspase-3, Beclin1, and LC3II protein expression, and reduced B-cell lymphoma 2/BAX ratio and p62 protein expression, but these results were reversed after pretreatment of Moschus and 3-MA. Moschus has protective activity in Glu-induced PC12 cell injury, and the potential mechanism might involve the regulation of autophagy and apoptosis. Our study may promote research on Moschus in the field of neurodegenerative diseases, and Moschus may be considered as a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Caiyou Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jie Cai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jingyi Dai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
27
|
Radic Savic Z, Coric V, Vidovic S, Vidovic V, Becarevic J, Milovac I, Reljic Z, Mirjanic-Azaric B, Skrbic R, Gajanin R, Matic M, Simic T. GPX3 rs8177412 Polymorphism Modifies Risk of Upper Urothelial Tumors in Patients with Balkan Endemic Nephropathy. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1421. [PMID: 37629712 PMCID: PMC10456338 DOI: 10.3390/medicina59081421] [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: 07/10/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023]
Abstract
Current data suggest that aristolochic acid (AA) exposure is a putative cause of Balkan endemic nephropathy (BEN), a chronic kidney disease strongly associated with upper tract urothelial carcinoma. The cellular metabolism of AA is associated with the production of reactive oxygen species, resulting in oxidative distress. Purpose: Therefore, the aim of this study was to analyze individual, combined and cumulative effect of antioxidant gene polymorphisms (Nrf2 rs6721961, KEAP1 rs1048290, GSTP1AB rs1695, GSTP1CD rs1138272, GPX3 rs8177412 and MDR1 rs1045642), as well as GSTP1ABCD haplotypes with the risk for BEN development and associated urothelial cell carcinoma in 209 BEN patients and 140 controls from endemic areas. Experimental method: Genotyping was performed using polymerase chain reaction (PCR) and PCR with confronting two-pair primers (PCR-CTTP) methods. Results: We found that female patients carrying both variant GPX3 rs8177412 and MDR1 rs1045642 genotypes in combination exhibited significant risk towards BEN (OR 1 = 3.34, 95% CI = 1.16-9.60, p = 0.025; OR 2 = 3.79, 95% CI = 1.27-11.24, p = 0.016). Moreover, significant association was determined between GPX3rs8174412 polymorphism and risk for urothelial carcinoma. Carriers of variant GPX3*TC + CC genotype were at eight-fold increased risk of BEN-associated urothelial tumors development. There was no individual or combined impact on BEN development and BEN-associated tumors among all examined polymorphisms. The haplotype consisting of variant alleles for both polymorphisms G and T was associated with 1.6-fold increased risk although statistically insignificant (OR = 1.64; 95% CI = 0.75-3.58; p = 0.21). Conclusions: Regarding GPX3 rs8177412 polymorphism, the gene variant that confers lower expression is associated with significant increase in upper urothelial carcinoma risk. Therefore, BEN patients carrying variant GPX3 genotype should be more frequently monitored for possible upper tract urothelial carcinoma development.
Collapse
Affiliation(s)
- Zana Radic Savic
- Department of Medical Biochemistry, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.R.S.); (B.M.-A.)
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (S.V.); (V.V.); (J.B.); (I.M.)
| | - Vesna Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Center of Excellence for Redox Medicine, Biotech Place, 2W-017, 575 North Patterson Avenue, Winston-Salem, NC 27157, USA
| | - Stojko Vidovic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (S.V.); (V.V.); (J.B.); (I.M.)
- Department of Human Genetics, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Vanja Vidovic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (S.V.); (V.V.); (J.B.); (I.M.)
- Department of Human Genetics, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Jelena Becarevic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (S.V.); (V.V.); (J.B.); (I.M.)
| | - Irina Milovac
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (S.V.); (V.V.); (J.B.); (I.M.)
- Department of Human Genetics, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Zorica Reljic
- Medical Laboratory “PAN LAB”, 36000 Kraljevo, Serbia;
| | - Bosa Mirjanic-Azaric
- Department of Medical Biochemistry, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.R.S.); (B.M.-A.)
| | - Ranko Skrbic
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina;
- Academy of Sciences and Arts of the Republic of Srpska, 78000 Banja Luka, The Republic of Srpska, Bosnia and Herzegovina
| | - Radoslav Gajanin
- Department of Pathological Anatomy, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina;
| | - Marija Matic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Center of Excellence for Redox Medicine, Biotech Place, 2W-017, 575 North Patterson Avenue, Winston-Salem, NC 27157, USA
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Center of Excellence for Redox Medicine, Biotech Place, 2W-017, 575 North Patterson Avenue, Winston-Salem, NC 27157, USA
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| |
Collapse
|
28
|
Li S, Xie J, Zhang D, Zhao G, Bai Y, Li K, Li X, Li Q, Tang X, Ge X. Lycopene abolishes typical polyhalogenated carbazoles (PHCZs)-induced hepatic injury in yellow catfish (Pelteobagrus fulvidraco): Involvement of ROS/PI3K-AKT/NF-κB signaling. FISH & SHELLFISH IMMUNOLOGY 2023:108897. [PMID: 37301309 DOI: 10.1016/j.fsi.2023.108897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/07/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
Aquatic ecosystems are being more contaminated with polyhalogenated carbazoles (PHCZs), which raising concerns about their impact on aquatic organisms. Lycopene (LYC) exhibits several beneficial properties for fish via enhance antioxidant defenses and improve immunity. In this study, we attempted to investigate the hepatotoxic effects of typical PHCZs 3, 6-dichlorocarbazole (3,6-DCCZ) and the protective mechanisms of LYC. In this study, we found that yellow catfish (Pelteobagrus fulvidraco) exposure to 3,6-DCCZ (1.2 mg/L) resulted in hepatic inflammatory infiltration and disordered hepatocyte arrangement. Besides, we observed that 3,6-DCCZ exposure resulted in hepatic reactive oxygen species (ROS) overproduction and excessive autophagosome accumulation, accompanied with inhibition of phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) pathway. Subsequently, we confirmed that 3,6-DCCZ exposure triggered hepatic uncontrolled inflammatory response via activation of nuclear factor-κB (NF-κB) pathway, along with decreased plasma complement C3 (C3) and complement C4 (C4) levels. Meanwhile, yellow catfish exposed to 3,6-DCCZ exhibit an increased hepatic apoptosis phenomenon, as evidenced by the elevated number of positive TUNEL cells and upregulated expression of caspase3 and cytochrome C (CytC). In contrast, LYC treatment could alleviate the 3,6-DCCZ-induced pathological changes, hepatic ROS accumulation, autophagy, inflammatory response and apoptosis. To sum up, this study provided the demonstration that LYC exerts hepatoprotective effects to alleviate 3,6-DCCZ-induced liver damage by inihibiting ROS/PI3K-AKT/NF-κB signaling in yellow catfish.
Collapse
Affiliation(s)
- Siwen Li
- Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan Province, PR China
| | - Jiaqi Xie
- Hunan Food and Drug Vocational College, Changsha, 410078, Hunan Province, PR China
| | - Dongfang Zhang
- Department of Pathology, Jilin Medical University, Jilin, 130013, Jilin Province, PR China
| | - Guifang Zhao
- Department of Pathology, Jilin Medical University, Jilin, 130013, Jilin Province, PR China
| | - Yiang Bai
- Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan Province, PR China
| | - Keman Li
- Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan Province, PR China
| | - Xinlian Li
- Department of Physiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Qiuyue Li
- Department of Physiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Xiaoqing Tang
- Department of Physiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, PR China
| | - Xiaofeng Ge
- Jilin People's Hospital, Jilin, 130013, Jilin Province, PR China.
| |
Collapse
|
29
|
Azırak S, Özgöçmen M. Linalool prevents kidney damage by inhibiting rifampicin-induced oxidative stress and apoptosis. Tissue Cell 2023; 82:102097. [PMID: 37104973 DOI: 10.1016/j.tice.2023.102097] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/30/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023]
Abstract
Today, kidney diseases are increasing day by day and life quality is decreasing. In hospitalized patients of all ages, acute kidney injury (AKI) is commonly observed and associated with high rates of morbidity and mortality. Rifampicin (RF) or rifampin is an antibiotic drug from the rifamycin group with a bactericidal effect. RF causes acute kidney injury, often anemia, thrombocytopenia, liver damage and side effect such as cell death. RF causes tissue damage by means of oxidative stress and apoptosis. Thus, in this study, it was examined whether linalool (LN) which had antinociceptive, antimicrobial, antioxidant and anti-inflammatory effects, was beneficial for kidney damage in order to eliminate the side effects of RF. NGAL mRNA, creatinine (Cr), blood urea nitrogen (BUN), Caspase 9 (CAS-9) and nuclear factor-κB (NF-κB) levels increased in the group treated with RF compared to the control group, while the levels of albumin, uric acid and total protein were decreased in the RF-treated group. NGAL mRNA, BUN, Cr, CAS-9 and NF-κB levels decreased significantly in RF+LN administered rats, while it was observed that there was an increase in the levels of albumin, uric acid and total protein. From the results obtained, it was observed that LN was determined to be very effective in preventing tissue damage in kidneys caused by oxidative stress by RF.
Collapse
Affiliation(s)
- Sebile Azırak
- Vocational School of Health Services, University of Adıyaman, Adıyaman, Turkey.
| | - Meltem Özgöçmen
- Suleyman Demirel University, Faculty of Medicine, Department of Histology and Embryology, Isparta, Turkey
| |
Collapse
|
30
|
Combined Effects of Lycopene and Metformin on Decreasing Oxidative Stress by Triggering Endogenous Antioxidant Defenses in Diet-Induced Obese Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238503. [PMID: 36500596 PMCID: PMC9737677 DOI: 10.3390/molecules27238503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022]
Abstract
Since lycopene has antioxidant activity, its combination with metformin may be useful to contrast diabetic complications related to oxidative stress. This study aimed to investigate the effects of metformin combined with lycopene on high-fat diet (HFD)-induced obese mice. Seventy-two C57BL-6J mice were divided into six groups: C (control diet-fed mice), H (HFD-fed mice for 17 weeks), H-V (HFD-fed mice treated with vehicle), H-M (HFD-fed mice treated with 50 mg/kg metformin), H-L (HFD-fed mice treated with 45 mg/kg lycopene), and H-ML (HFD-fed mice treated with 50 mg/kg metformin + 45 mg/kg lycopene). Treatments were administered for 8 weeks. Glucose tolerance, insulin sensitivity, fluorescent AGEs (advanced glycation end products), TBARS (thiobarbituric acid-reactive substances), and activities of antioxidant enzymes paraoxonase-1 (PON-1; plasma), superoxide dismutase, catalase and glutathione peroxidase (liver and kidneys) were determined. Metformin plus lycopene reduced body weight; improved insulin sensitivity and glucose tolerance; and decreased AGEs and TBARS in plasma, liver and kidneys. Combined therapy significantly increased the activities of antioxidant enzymes, mainly PON-1. Lycopene combined with metformin improved insulin resistance and glucose tolerance, and caused further increases in endogenous antioxidant defenses, arising as a promising therapeutic strategy for combating diabetic complications resulting from glycoxidative stress.
Collapse
|
31
|
Albrahim T. Lycopene Modulates Oxidative Stress and Inflammation in Hypercholesterolemic Rats. Pharmaceuticals (Basel) 2022; 15:1420. [PMID: 36422550 PMCID: PMC9693203 DOI: 10.3390/ph15111420] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
The complicated disorder of hypercholesterolemia has several underlying factors, including genetic and lifestyle factors. Low LDL cholesterol and elevated serum total cholesterol are its defining features. A carotenoid with antioxidant quality is lycopene. Examining lycopene activity in an animal model of hypercholesterolemia induced using food was the aim of this investigation. Triglycerides, LDL cholesterol, HDL cholesterol, and plasma total cholesterol were all measured. Biomarkers of renal and cardiac function were also examined. Apoptotic indicators, pro-inflammatory markers, and oxidative stress were also assessed. Additionally, the mRNA expression of paraoxonase 1 (PON-1), peroxisome proliferator-activated receptor gamma (PPAR-γ), and PPAR-γ coactivator 1 alpha (PGC-1α) in cardiac and renal tissues was examined. Rats showed elevated serum lipid levels, renal and cardiac dysfunction, significant oxidative stress, and pro-inflammatory and apoptotic markers at the end of the study. Treatment with lycopene significantly corrected and restored these changes. Additionally, lycopene markedly increased the mRNA expression of PGC-1α and PON-1, and decreased PPAR-γ expression. It was determined that lycopene has the capacity to modulate the PPAR-γ and PON-1 signaling pathway in order to preserve the cellular energy metabolism of the heart and kidney, which in turn reduces tissue inflammatory response and apoptosis. According to these findings, lycopene may be utilized as a medication to treat hypercholesterolemia. However, further studies should be conducted first to determine the appropriate dose and any adverse effects that may appear after lycopene usage in humans.
Collapse
Affiliation(s)
- Tarfa Albrahim
- Department of Health Sciences, Clinical Nutrition, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| |
Collapse
|