1
|
Zhou Y, Hu T, Zeng H, Lin L, Xie H, Lin R, Huang M. Naringenin Inhibits Ferroptosis in Renal Tubular Epithelial Cells of Diabetic Nephropathy Through SIRT1/FOXO3a Signaling Pathway. Drug Dev Res 2025; 86:e70044. [PMID: 39799560 DOI: 10.1002/ddr.70044] [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: 10/15/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025]
Abstract
Naringenin has the potential to regulate ferroptosis and mitigate renal damage in diabetic nephropathy (DN). However, it remains unclear whether the naringenin's effects in DN are linked to its ability to regulate ferroptosis. This study investigated the potential anti-ferroptosis properties of naringenin in high glucose (HG)-induced renal tubular epithelial cell models. HK-2 cells were cultured in HG medium to establish the DN cell model. HK-2 cells were treated with different doses of naringenin to explore the effect of naringenin. The CCK-8 results show that 50 μM ~ 200 μM of naringenin do not affect the viability of HK-2 cells and the viability of HG-induced HK-2 cells increase in a dose-dependent manner with naringenin treatment. Additionally, naringenin increased the levels of IL-10 while decreasing the levels of IL-1β, TNF-α, IL-6, and ROS in HG-induced HK-2 cells. Naringenin also reduced the levels of Fe2+, oxidized lipid ROS, MDA, 4-HNE, ACSL4, and TFR1 in HG-induced HK-2 cells, while increasing the levels of non-oxidized lipid ROS, SOD, GSH-Px, SLC7A11, and GPX4. Meanwhile, naringenin restored the levels of MMP, ATP and MPTP opening, reduced OCR in HG-induced HK-2 cells. Furthermore, naringenin reversed the decreased expression of SIRT1, p-FOXO3a, Nrf2 and Nuclear Nrf2 caused by HG. SIRT1 inhibitor EX527 and Nrf2 inhibitor ML385 attenuated the effects of naringenin on ferroptosis in HG-induced HK-2 cells, with EX527 demonstrating a stronger reversal effect on ferroptosis than ML385. These results suggest that naringenin inhibits ferroptosis in HG-induced HK-2 cells mainly through SIRT1/FOXO3a signaling pathway. This finding further enhanced our understanding of the mechanism behind naringenin's protective effect on DN.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Endocrinology, Xiamen Hospital, Beijing University of Chinese Medicine/Xiamen TCM Hospital Affiliated to Fujian University of Traditional Chinese Medicine/Xiamen Hospital of Traditional Chinese Medicine, Xiamen City, People's Republic of China
| | - Tianchi Hu
- Department of Endocrinology, Xiamen Hospital, Beijing University of Chinese Medicine/Xiamen TCM Hospital Affiliated to Fujian University of Traditional Chinese Medicine/Xiamen Hospital of Traditional Chinese Medicine, Xiamen City, People's Republic of China
| | - Huarong Zeng
- Department of Endocrinology, Xiamen Hospital, Beijing University of Chinese Medicine/Xiamen TCM Hospital Affiliated to Fujian University of Traditional Chinese Medicine/Xiamen Hospital of Traditional Chinese Medicine, Xiamen City, People's Republic of China
| | - Lin Lin
- Department of Endocrinology, Xiamen Hospital, Beijing University of Chinese Medicine/Xiamen TCM Hospital Affiliated to Fujian University of Traditional Chinese Medicine/Xiamen Hospital of Traditional Chinese Medicine, Xiamen City, People's Republic of China
| | - Huan Xie
- Department of Endocrinology, Xiamen Hospital, Beijing University of Chinese Medicine/Xiamen TCM Hospital Affiliated to Fujian University of Traditional Chinese Medicine/Xiamen Hospital of Traditional Chinese Medicine, Xiamen City, People's Republic of China
| | - Rong Lin
- Department of Endocrinology, Xiamen Hospital, Beijing University of Chinese Medicine/Xiamen TCM Hospital Affiliated to Fujian University of Traditional Chinese Medicine/Xiamen Hospital of Traditional Chinese Medicine, Xiamen City, People's Republic of China
| | - Mengya Huang
- Graduate School, Fujian University of Traditional Chinese Medicine, Fuzhou City, People's Republic of China
| |
Collapse
|
2
|
Li X, Xia K, Zhong C, Chen X, Yang F, Chen L, You J. Neuroprotective effects of GPR68 against cerebral ischemia-reperfusion injury via the NF-κB/Hif-1α pathway. Brain Res Bull 2024; 216:111050. [PMID: 39147243 DOI: 10.1016/j.brainresbull.2024.111050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND G protein-coupled receptor 68 (GPR68), an orphan receptor, has emerged as a promising therapeutic target for mitigating neuronal inflammation and oxidative damage. This study explores the protective mechanisms of GPR68 in cerebral ischemia-reperfusion injury (CIRI). METHODS An in vivo middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model was established. Mice received intraperitoneal injections of Ogerin, a selective GPR68 agonist. In vitro, GPR68 was overexpressed in SH-SY5Y and HMC3 cells, and the effects of oxygen-glucose deprivation/reperfusion (OGD/R) on cell viability were assessed using real-time quantitative polymerase chain reaction (RT-qPCR), enzyme-linked immunosorbent assay (ELISA), and flow cytometry. RESULTS The expression of GPR68 was suppressed in cells subjected to OGD/R treatment, whereas its upregulation conferred protection to SH-SY5Y and HMC3 cells. In vivo, levels of GPR68 were reduced in brain tissues affected by MCAO/R, correlating with oxidative stress, inflammation, and neurological damage. Treatment with a GPR68 agonist decreased brain infarction, apoptosis, and dysregulated gene expression induced by MCAO/R. Mechanistically, GPR68 agonist treatment may inhibit the activation of the NF-κB/Hif-1α pathway, thereby reducing oxidative and inflammatory responses and enhancing protection against CIRI. CONCLUSIONS This study confirms that the GPR68/NF-κB/Hif-1α axis modulates apoptosis, inflammation, and oxidative stress in CIRI, indicating that GPR68 is a potential therapeutic target for CIRI.
Collapse
Affiliation(s)
- Xianglong Li
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China; Neurosurgical Clinical Research Center and Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, PR China; Laboratory of Neurological Diseases and Brain Functions, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Kaiguo Xia
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Chuanhong Zhong
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Xiangzhou Chen
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Fubing Yang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China; Neurosurgical Clinical Research Center and Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, PR China; Laboratory of Neurological Diseases and Brain Functions, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China.
| | - Jian You
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China; Neurosurgical Clinical Research Center and Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, PR China; Laboratory of Neurological Diseases and Brain Functions, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China.
| |
Collapse
|
3
|
Sardarabadi H, Darvishi MH, Zohrab F, Javadi H. Nanophytomedicine: A promising practical approach in phytotherapy. Phytother Res 2024; 38:3607-3644. [PMID: 38725270 DOI: 10.1002/ptr.8230] [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: 10/17/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 07/12/2024]
Abstract
The long and rich history of herbal therapeutic nutrients is fascinating. It is incredible to think about how ancient civilizations used plants and herbs to treat various ailments and diseases. One group of bioactive phytochemicals that has gained significant attention recently is dietary polyphenols. These compounds are commonly found in a variety of fruits, vegetables, spices, nuts, drinks, legumes, and grains. Despite their incredible therapeutic properties, one challenge with polyphenols is their poor water solubility, stability, and bioavailability. This means that they are not easily absorbed by the body when consumed in essential diets. Because of structural complexity, polyphenols with high molecular weight cannot be absorbed in the small intestine and after arriving in the colon, they are metabolized by gut microbiota. However, researchers are constantly working on finding solutions to enhance the bioavailability and absorption of these compounds. This study aims to address this issue by applying nanotechnology approaches to overcome the challenges of the therapeutic application of dietary polyphenols. This combination of nanotechnology and phytochemicals could cause a completely new field called nanophytomedicine or herbal nanomedicine.
Collapse
Affiliation(s)
- Hadi Sardarabadi
- Department of Physiology and Pharmacology, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Hasan Darvishi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Fatemeh Zohrab
- Department of Medical Science, Qom Branch, Islamic Azad University, Qom, Iran
| | - Hamidreza Javadi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Yuan B, Mao J, Wang J, Luo S, Luo B. Naringenin mitigates cadmium-induced cell death, oxidative stress, mitochondrial dysfunction, and inflammation in KGN cells by regulating the expression of sirtuin-1. Drug Chem Toxicol 2024; 47:445-456. [PMID: 38647073 DOI: 10.1080/01480545.2023.2288798] [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: 07/20/2023] [Accepted: 11/05/2023] [Indexed: 04/25/2024]
Abstract
The objective of this study was to examine the potential protective role of naringenin against the harmful effects induced by cadmium in KGN cell line. Cell viability was evaluated by cell counting kit-8 assay. Caspase-3/-9 activities were determined by caspase-3/-9 activity assay kits, respectively. Intracellular reactive oxygen species (ROS) level was detected by ROS-Glo™ H2O2 Assay, antioxidant capacity was determined by a total antioxidant capacity assay kit. Mitochondrial membrane potential (MMP), ATP level, and ATP synthase activity were determined by JC-1, ATP assay kit, and ATP synthase activity assay kit, respectively. The mRNA expression was determined by qRT-PCR. Cadmium reduced cell viability and increased caspase-3/-9 activities in a concentration-dependent manner. Naringenin improved cell viability and reduced caspase-3/-9 activities in cadmium-stimulated KGN cells in a concentration-dependent manner. Cadmium diminished the antioxidant capacity, increased ROS production, and induced mitochondrial dysfunction in KGN cells. These effects were ameliorated by naringenin treatment in a concentration-dependent manner. Furthermore, naringenin reduced the levels of pro-inflammatory cytokines in KGN cells exposed to cadmium. SIRT1 knockdown downregulated its expression in KGN cells and compromised the protective effects of naringenin on cell viability and caspase-3/-9 activities in cadmium-stimulated KGN cells. Naringenin prevented the reduction of MMP, ATP levels, and ATP synthase activity in cadmium-stimulated KGN cells in a concentration-dependent manner. However, these protective effects were significantly reversed by SIRT1 knockdown. In conclusion, this study suggests that naringenin protects against cadmium-induced damage by regulating oxidative stress, mitochondrial function, and inflammation in KGN cells, with SIRT1 playing a potential mediating role.
Collapse
Affiliation(s)
- Ben Yuan
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
- Huangshi Key Laboratory of Assisted Reproduction and Reproductive Medicine, Huangshi, China
| | - Junbiao Mao
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
- Huangshi Key Laboratory of Assisted Reproduction and Reproductive Medicine, Huangshi, China
| | - Junling Wang
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
- Huangshi Key Laboratory of Assisted Reproduction and Reproductive Medicine, Huangshi, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
| | - Shuhong Luo
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
- Huangshi Key Laboratory of Assisted Reproduction and Reproductive Medicine, Huangshi, China
| | - Bingbing Luo
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
- Huangshi Key Laboratory of Assisted Reproduction and Reproductive Medicine, Huangshi, China
| |
Collapse
|
5
|
Akarasereenont P, Pattanapholkornsakul S, Limsuvan S, Mamaethong D, Booranasubkajorn S, Pakaprot N, Tripatara P, Pilakasiri K. Therapeutic potential of Thai herbal formula for cognitive impairment: A metabolomics approach for Comprehensive Insights. Heliyon 2024; 10:e28027. [PMID: 38560220 PMCID: PMC10981045 DOI: 10.1016/j.heliyon.2024.e28027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Chronic cerebral ischemia hypoperfusion plays a role in the initiation and progression of vascular dementia, which causes changes in metabolites. Currently, there is no standard treatment to treat, prevent and reduce the severity of this condition. Thai herbal Yahom no.20 (YHF20) is indicated for fatigue and dizziness. The components of YHF20 have been found to have pharmacological effects related to the pathology of chronic cerebral ischemia hypoperfusion. This study aimed to investigate metabolomic changes after YHF20 administration in a rat model of permanent bilateral common carotid artery occlusion (2-VO) induced chronic cerebral ischemia hypoperfusion, and to explore its impact on spatial learning and memory. Albino Wistar rats were randomly allocated to 5 groups; sham, 2-VO, 2-VO+ 100 mg/kg YHF20, 2-VO+300 mg/kg YHF20, and 2-VO+1000 mg/kg YHF20. The rats were administered YHF20 daily by oral gavage for 56 days after 2-VO induction. Plasma was collected weekly for metabolome change analysis using LC-MS/QTof and toxicity study. The rats were evaluated for spatial learning and memory using the Morris water maze. The results showed that 78 known metabolites and 10 tentative pathways altered after chronic cerebral hypoperfusion, although it was not able to determine the effect on memory and learning behaviors of rats. Glutathione and glutathione metabolism might be metabolite-pathway that were the affect after YHF20 administration in cerebral ischemic condition. The 4 known metabolites may be the metabolites from the constituents of YHF20 could be considered and confirmed for quality control purpose. In conclusion, YHF20 administration might contribute to metabolic changes related to cerebral ischemia condition without the effect on spatial learning and memory, including hepatotoxicity and nephrotoxicity after 56 days of treatment. Alterations in the potential metabolites may provide data support for elucidating dementia pathogenesis and selecting pathways for intervention.
Collapse
Affiliation(s)
- Pravit Akarasereenont
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Saracha Pattanapholkornsakul
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Suveerawan Limsuvan
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Dollaporn Mamaethong
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Suksalin Booranasubkajorn
- Center of Applied Thai Traditional Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Narawut Pakaprot
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Pinpat Tripatara
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| | - Kajee Pilakasiri
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, 10700, Bangkok, Thailand
| |
Collapse
|