1
|
Yao H, Wu R, Du D, Ai F, Yang F, Li Y, Qi S. Flavonoids from Polypodium hastatum as neuroprotective agents attenuate cerebral ischemia/reperfusion injury in vitro and in vivo via activating Nrf2. Redox Rep 2025; 30:2440204. [PMID: 39702961 DOI: 10.1080/13510002.2024.2440204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVES Cerebral ischemic stroke is a leading cause of death worldwide. Though timely reperfusion reduces the infarction size, it exacerbates neuronal apoptosis due to oxidative stress. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor regulating the expression of antioxidant enzymes. Activating Nrf2 gives a therapeutic approach to ischemic stroke. METHODS Herein we explored flavonoids identified from Polypodium hastatum as Nrf2 activators and their protective effects on PC12 cells injured by oxygen and glucose deprivation/restoration (OGD/R) as well as middle cerebral artery occlusion (MCAO) mice. RESULTS The results showed among these flavonoids, AAKR significantly improved the survival of PC12 cells induced by OGD/R and activated Nrf2 in a Keap1-dependent manner. Further investigations have disclosed AAKR attenuated oxidative stress, mitochondrial dysfunction and following apoptosis resulting from OGD/R. Meanwhile, activation of Nrf2 by AAKR was involved in the protective effects. Finally, it was found that AAKR could protect MCAO mice brains against ischemia/reperfusion injury via activating Nrf2. DISCUSSION This investigation could provide lead compounds for the discovery of novel Nrf2 activators targeting ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Huankai Yao
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Ruiqing Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Dan Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Fengwei Ai
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Feng Yang
- School of Stomatology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yan Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Suhua Qi
- School of Medical Technology & Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|
2
|
Ursu GM, Krawic C, Zhitkovich A. Nuclear SUMOylation and Proteotoxic Stress Responses to Metals with Different Ligand Preferences. Chem Res Toxicol 2025. [PMID: 40243484 DOI: 10.1021/acs.chemrestox.5c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Proteins are vulnerable to damage by a broad range of electrophiles, and cells contain several proteotoxic stress-monitoring systems. Main transcriptional responses to protein damage are driven by cytosolic HSF1 and NRF2 using soft nucleophile Cys-SH as sensors of electrophiles. It is unclear what stress responses are activated by poorly SH-reactive hard electrophiles. We examined protein damage responses in normal human lung cells with equitoxic doses of three carcinogenic metals with different electrophilic softness: soft, cadmium(II), intermediate, cobalt(II), and hard, chromium(III) delivered into cells using chromium(VI)/chromate. Cd(II) strongly activated cytosolic NRF2 and HSF1, produced soluble and insoluble polyubiquitinated proteins in the cytosol, and moderately elevated ER and mitochondrial unfolded protein responses and nuclear polySUMOylation. Cr(III) primarily induced nuclear protein damage and polySUMOylation and was negative for the activation of all cytoplasmic stress responses. Co(II) triggered HSF1, NRF2, and other responses seen with both Cr(III) and Cd(II) except for cytosolic polyubiquitin aggregates. Physiological levels of the antioxidant ascorbate inhibited but did not eliminate NRF2 activation by Co(II) and enhanced polySUMOylation by Cr(VI/III). For all three metals, SUMOylated proteins accumulated in nuclear PML bodies, and their formation was suppressed by PML knockdown. Inhibition of SUMOylation decreased transcription and, even more severely, protein expression of NRF2 and HSF1 targets by Cd(II) and Co(II), revealing the importance of this nuclear response in the functionality of cytosolic stress-activated pathways. Our findings demonstrate that soft and hard metal electrophiles elicit distinct proteotoxic stress responses, with the notable inability of the hard electrophile Cr(III) to trigger cytosolic damage-monitoring systems.
Collapse
Affiliation(s)
- Giorgiana Madalina Ursu
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02903, United States
| | - Casey Krawic
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02903, United States
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02903, United States
| |
Collapse
|
3
|
Hu Z, Chen X, Hu Q, Zou M, Liu Z. Role of Chinese Medicine Monomers in Dry Eye Disease: Breaking the Vicious Cycle of Inflammation. Pharmacol Res Perspect 2025; 13:e70077. [PMID: 39979080 PMCID: PMC11842162 DOI: 10.1002/prp2.70077] [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: 04/23/2024] [Revised: 12/05/2024] [Accepted: 01/24/2025] [Indexed: 02/22/2025] Open
Abstract
Dry eye disease (DED) is a chronically inflammatory ocular surface disorder of unknown pathogenesis. Anti-inflammatory medications, artificial tears, autologous serum, and LipiFlow have been shown to be highly beneficial in alleviating symptoms. Nevertheless, these interventions often provide only short-term results and do not address the underlying problems of the disease. There is growing evidence that the risk of DED is associated with a vicious cycle of inflammation. This vicious cycle of inflammation is produced by the interaction of several factors, including tear film hyperosmolarity, tear film instability, inflammation, and apoptosis. Chinese medicine monomers, distinguished by their multicomponent and multitarget advantages, have been shown to help treat DED by modulating tear film status, and inhibiting inflammatory responses, and apoptosis, providing a new way of thinking of the management of DED in Chinese medicine.
Collapse
Affiliation(s)
- Zhuoyu Hu
- The First Hospital of Hunan University of Chinese MedicineChangshaHunanChina
| | - Xiangdong Chen
- The First Hospital of Hunan University of Chinese MedicineChangshaHunanChina
| | - Qi Hu
- Graduate School of Hunan University of Chinese MedicineChangshaHunanChina
| | - Menglong Zou
- Graduate School of Hunan University of Chinese MedicineChangshaHunanChina
| | - Zhimin Liu
- The First Hospital of Hunan University of Chinese MedicineChangshaHunanChina
| |
Collapse
|
4
|
Augello FR, Lombardi F, Ciafarone A, Altamura S, Marazzato M, Artone S, Cinque B, Palumbo P. Streptococcus thermophilus CNCM I-5570 lysate counteracts the aging process in human dermal fibroblast cells by neutralizing harmful free radicals and impacting antioxidant and anti-inflammatory pathways, thus restoring their physiological functions. Biomed Pharmacother 2025; 185:117975. [PMID: 40081000 DOI: 10.1016/j.biopha.2025.117975] [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: 11/27/2024] [Revised: 02/25/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025] Open
Abstract
Previous studies have highlighted the in vitro and in vivo anti-aging potential of Streptococcus thermophilus prompting us to investigate the biomolecular mechanisms underlying its effects. We evaluated the reparative ability of S. thermophilus lysate in a hydrogen peroxide (H2O2)-induced senescence model of human dermal fibroblasts (HDFs). Cell proliferation, cell number, and senescence level were evaluated by IncuCyte® Live Cell Imager system, trypan blue dye exclusion test and β-galactosidase activity, respectively. We analyzed p21, prolyl 4-hydroxylase A1, intracellular collagen I, nuclear factor E2-related factor 2 (Nrf2), nuclear factor kappa B (NF-κB) and heme oxygenase-1 expression through western blot. Extracellular levels of collagen I, interleukin-1β, and IL-6 were assessed by ELISA. The oxidative stress markers were assayed using standard methods. The direct antioxidant activity of probiotic was quantified using multiple techniques. The presence of antioxidant genes in probiotic was detected via PCR assay. Probiotic lysate exposure increased the proliferation rate, counteracted the aging by reducing β-galactosidase activity and p21 levels, promoted collagen I synthesis and neutralized oxidative stress by activating Nrf2. The probiotic lysate inhibited the NF-κB pathway with pro-inflammatory marker downregulation. Notably, we revealed that probiotic exhibited strong free radical scavenging ability, iron-chelating properties, and significant ferric reducing power in a concentration-dependent manner. We identified seven genes with antioxidant function in its genome. Our results show that S. thermophilus lysate is efficacious in suppressing the biomolecular events associated with H2O2-induced cellular aging, thus supporting the reparative action of S. thermophilus, helpful in treating skin aging.
Collapse
Affiliation(s)
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessia Ciafarone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy; PhD School in Health & Environmental Sciences, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila 67100, Italy
| | - Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy; PhD School in Medicine and Public Health, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila 67100, Italy
| | - Massimiliano Marazzato
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome 00185, Italy
| | - Serena Artone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy; PhD School in Medicine and Public Health, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila 67100, Italy
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
5
|
Selvaraj NR, Nandan D, Nair BG, Nair VA, Venugopal P, Aradhya R. Oxidative Stress and Redox Imbalance: Common Mechanisms in Cancer Stem Cells and Neurodegenerative Diseases. Cells 2025; 14:511. [PMID: 40214466 PMCID: PMC11988017 DOI: 10.3390/cells14070511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/14/2025] Open
Abstract
Oxidative stress (OS) is an established hallmark of cancer and neurodegenerative disorders (NDDs), which contributes to genomic instability and neuronal loss. This review explores the contrasting role of OS in cancer stem cells (CSCs) and NDDs. Elevated levels of reactive oxygen species (ROS) contribute to genomic instability and promote tumor initiation and progression in CSCs, while in NDDs such as Alzheimer's and Parkinson's disease, OS accelerates neuronal death and impairs cellular repair mechanisms. Both scenarios involve disruption of the delicate balance between pro-oxidant and antioxidant systems, which leads to chronic oxidative stress. Notably, CSCs and neurons display alterations in redox-sensitive signaling pathways, including Nrf2 and NF-κB, which influence cell survival, proliferation, and differentiation. Mitochondrial dynamics further illustrate these differences: enhanced function in CSCs supports adaptability and survival, whereas impairments in neurons heighten vulnerability. Understanding these common mechanisms of OS-induced redox imbalance may provide insights for developing interventions, addressing aging hallmarks, and potentially mitigating or preventing both cancer and NDDs.
Collapse
Affiliation(s)
| | | | | | | | - Parvathy Venugopal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| |
Collapse
|
6
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. The Janus Face of Oxidative Stress and Hydrogen Sulfide: Insights into Neurodegenerative Disease Pathogenesis. Antioxidants (Basel) 2025; 14:360. [PMID: 40227410 PMCID: PMC11939184 DOI: 10.3390/antiox14030360] [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: 02/27/2025] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Oxidative stress plays an essential role in neurodegenerative pathophysiology, acting as both a critical signaling mediator and a driver of neuronal damage. Hydrogen sulfide (H2S), a versatile gasotransmitter, exhibits a similarly "Janus-faced" nature, acting as a potent antioxidant and cytoprotective molecule at physiological concentrations, but becoming detrimental when dysregulated. This review explores the dual roles of oxidative stress and H2S in normal cellular physiology and pathophysiology, focusing on neurodegenerative disease progression. We highlight potential therapeutic opportunities for targeting redox and sulfur-based signaling systems in neurodegenerative diseases by elucidating the intricate balance between these opposing forces.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
7
|
Cheng Y, Zheng G, Huang H, Ni J, Zhao Y, Sun Y, Chang Y, Liu S, He F, Li D, Guo Y, Miao Y, Xu M, Wang D, Zhang Y, Hua Y, Yang S, Fan G, Ma C. GLSP mitigates vascular aging by promoting Sirt7-mediated Keap1 deacetylation and Keap1-Nrf2 dissociation. Theranostics 2025; 15:4345-4367. [PMID: 40225574 PMCID: PMC11984382 DOI: 10.7150/thno.110324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/07/2025] [Indexed: 04/15/2025] Open
Abstract
Background and Purpose: Vascular aging is a prior marker of human aging and a significant contributor to atherosclerosis and vascular calcification. However, there are limited pharmacological options available to mitigate vascular aging. Thus, understanding the mechanisms underlying vascular aging and age-related atherosclerosis and vascular calcification is crucial. This study investigates the targets of vascular aging and elucidates the role and mechanisms of Ganoderma lucidum spore powder (GLSP) in mitigating vascular aging and aging-associated atherosclerosis as well as vascular calcification. Methods: The anti-vascular aging effects of GLSP was determined in aged C57BL/6J mice and the targets of GLSP was identified through transcriptome sequencing. Additionally, the protective effects of GLSP on the aged vasculature were assessed by examining atherosclerosis in apoE-/- mice and vascular calcification in VD3 and nicotine-induced mice. In vitro, the protective effects of GLSP triterpenes against vascular aging and calcification was determined in vascular smooth muscle cells (VSMCs). Results: GLSP exerted anti-vascular aging effects by regulating the cell cycle and senescence-associated secretory phenotype (SASP), mitigating DNA damage, reducing oxidative stress, improving mitochondrial function and modulating metabolic levels. Furthermore, GLSP improved vascular aging-associated atherosclerosis and vascular calcification in vivo. Mechanistically, RNA sequencing revealed an upregulation of Sirt7 expression after GLSP treatment. Sirt7 inhibitor exacerbated VSMCs senescence and calcification in senescent VSMCs and abolished the anti-senescence and the inhibitory effect of GLSP triterpenes on VSMCs senescence and calcification. Innovatively, we found that Sirt7 interacted with Keap1 and facilitated Keap1 deacetylation, which promoted Keap1-Nrf2 dissociation and consequently enhanced Nrf2 nuclear translocation and activation. Conclusion: GLSP alleviates vascular aging by exerting antioxidant effects through the activation of the Sirt7-Nrf2 axis, providing a promising new strategy for delaying vascular aging, atherosclerosis and vascular calcification.
Collapse
Affiliation(s)
- Yanfei Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Guobin Zheng
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin 300134, China
| | - Heming Huang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuting Sun
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yingxin Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shangjing Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng He
- Culture and Industry Research Center of Li Shizhen Traditional Chinese Medicine, Li Shizhen College of Traditional Chinese Medicine, Huanggang Normal University, Huanggang, 438000, China
| | - Dan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuying Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengxin Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Dongyue Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunsha Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shu Yang
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
8
|
Kang L, Bao S, Li P, Zhang G, Zhu X, Ji M, Guan H. METTL14-mediated depression of NEIL1 aggravates oxidative damage and mitochondrial dysfunction of lens epithelial cells through regulating KEAP1/NRF2 pathways. Cell Signal 2025; 127:111623. [PMID: 39855533 DOI: 10.1016/j.cellsig.2025.111623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Abnormal base excision repair (BER) pathway and N6-methyladenosine (m6A) of RNA have been proved to be significantly related to age-related cataract (ARC) pathogenesis. However, the relationship between the Nei Endonuclease VIII-Like1 (NEIL1) gene (a representative DNA glycosylase of BER pathway) and its m6A modification remains unclear. Here, we showed that the expression of NEIL1 was decreased in the ARC anterior lens capsules and H2O2-stimulated SRA01/04 cells. Our findings demonstrated that ectopic expression of NEIL1 alleviated DNA oxidative damage, apoptosis and mitochondrial dysfunction through disturbing KEAP1/NRF2 interaction. Furthermore, silencing NEIL1 aggravated H2O2-induced lens opacity, whereas ML334 could mitigate lens cloudy ex vitro in rat lenses. Besides, intravitreal injection of AAV2-NEIL1 alleviated lens opacity in Emory mice in vivo. Mechanistically, the N(6)-Methyladenosine (m6A) methyltransferase-like 14 (METTL14) was identified as a factor in promoting m6A modification of NEIL1, which resulted in the recruitment of YTHDF2 to recognize and impair NEIL1 RNA stability. Collectively, these findings highlight the critical role of the m6A modification in NEIL1 on regulating oxidative stress and mitochondrial homeostasis through KEAP1/NRF2 pathways, providing a new way to explore the pathogenesis of ARC.
Collapse
Affiliation(s)
- Lihua Kang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Sijie Bao
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Pengfei Li
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Guowei Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xi Zhu
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Min Ji
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
9
|
Lei XY, Wang X, Cao X, Li YH. Silymarin mediates the gut-liver axis pathway to alleviate Carassius auratus hepatic lipid metabolism disorders caused by carbonate exposure. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101457. [PMID: 40024209 DOI: 10.1016/j.cbd.2025.101457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
An 8-week feeding trial was conducted to investigate the mechanism of silymarin alleviating the abnormal lipid metabolism of Hefang Crucian Carp (HCC) (13.43 ± 0.059 g) liver caused by carbonate exposure. The fish were randomly divided into three groups: Control group (group B, 0 g/L carbonate, 0 mg/kg silymarin), carbonate stress group (group CA, 3 g/L carbonate, 0 mg/kg silymarin) and silymarin group (group SI, 3 g/L carbonate, 60 mg/kg silymarin). The results showed that the growth performance of group CA was significantly increased compared with group B. Compared with CA group, brush villi in SI group recovered significantly, and the width of submucosa decreased. Compared with group B, the intestinal barrier was damaged and permeability increased in group CA, while the damage was alleviated in group SI. Intestinal microbiota analysis showed that the bacterial community function genes related to lipopolysaccharide biosynthesis protein and lipopolysaccharide biosynthesis in CA group were higher than those in B and SI groups, and it was found that the change of LPS content in fish was echoed by the results of intestinal microflora. Compared with group B, the liver of group CA was damaged and the lipid metabolism process was abnormal, resulting in lipid metabolism disorder. SI group alleviated the liver damage caused by carbonate exposure, promoted the process of liver lipid synthesis, and balanced the body's lipid metabolism. More than 50 % of the metabolites are closely related to lipids and lipid molecules. The most metabolites in metabolism are oxidative phosphorylation and pyruvate metabolism. In summary, this study demonstrated that silymarin alleviating carbonate exposure altered intestinal microbiota homeostasis in HCC, leading to intestinal inflammation and increased mucosal barrier permeability, inhibiting LPS synthesis and absorption, preventing it from entering the liver through the intestinal liver, and increasing oxidative stress in the liver and abnormal lipid metabolism in the liver, thereby leading to liver injury. To provide theoretical basis for the development and utilization of silymarin functional feed additives and the mitigation strategy of carbonate exposure to liver damage in fish.
Collapse
Affiliation(s)
- Xin-Yu Lei
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Xin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Xue Cao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Yue-Hong Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
10
|
Li J, Li J, Liu Y, Hu C, Xu H, Cao D, Zhang R, Zhang K. Nrf2 Ameliorates Sevoflurane-Induced Cognitive Deficits in Aged Mice by Inhibiting Neuroinflammation in the Hippocampus. Mol Neurobiol 2025:10.1007/s12035-025-04777-w. [PMID: 39969679 DOI: 10.1007/s12035-025-04777-w] [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: 04/30/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Perioperative neurocognitive disorders (PND), common complications that occur after anesthetized surgery in elderly patients, are major challenges to our rapidly growing aging population. The transcription factor known as nuclear factor erythroid-2-related factor 2 (Nrf2) is an essential component of the cellular antioxidant response, purportedly contributing to the preservation of cognitive functions such as learning and memory. Nevertheless, the function and intracellular processes involving Nrf2 in PND remain largely unknown. Therefore, we evaluate the influence and fundamental mechanism of Nrf2 on PND in aged mice. To establish the postoperative neurocognitive dysfunction (PND) model, aged mice were subjected to anesthesia via inhalation of 3% sevoflurane for a duration of 2 h. The role of Nrf2 in PND was investigated by administering microinjections of either the adeno-associated virus (AAV)-Nrf2 vector or a null virus vector into the hippocampal CA1 region of aged mice 28 days before exposure to sevoflurane. Various assays including enzyme-linked immunosorbent assay (ELISA), immunofluorescence staining, and western blotting were employed to assess levels of pro-inflammatory cytokines, microglial activation, and the oxidative stress response. Furthermore, synaptic plasticity was evaluated through long-term potentiation (LTP) recording and Golgi staining techniques. Elevated expression of Nrf2 within the hippocampal CA1 region ameliorated sevoflurane-induced cognitive deficits, synaptic plasticity anomalies, and the oxidative stress reaction in aged mice. Furthermore, the activation of microglia and the release of pro-inflammatory cytokines (including IL-6, TNF-α, and IL-1β) within the hippocampus post-sevoflurane exposure were modulated in an Nrf2-dependent fashion. Based on the findings from present research, we conclude that Nrf2 ameliorates sevoflurane-induced cognitive dysfunction by inhibiting hippocampal neuroinflammation, thereby proposing a potential therapeutic target for PND.
Collapse
Affiliation(s)
- Junhua Li
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jinfeng Li
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (the Second Clinical Medical College of Guangzhou University of Chinese Medicine), Guangzhou, China
| | - Yafang Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Chuwen Hu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Hui Xu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Dong Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Rong Zhang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Kun Zhang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
11
|
Cocksedge SP, Mantecón L, Castaño E, Infante C, Bailey SJ. The Potential of Superoxide Dismutase-Rich Tetraselmis chuii as a Promoter of Cellular Health. Int J Mol Sci 2025; 26:1693. [PMID: 40004157 PMCID: PMC11855123 DOI: 10.3390/ijms26041693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Tetraselmis chuii (T. chuii) is a green, marine, eukaryotic, microalgae that was authorized in the European Union (EU) as a novel food for human consumption in 2014, and as a food supplement in 2017. This narrative review will provide an overview of preclinical and clinical trials assessing the efficacy of a T. chuii-derived ingredient, characterized by a high superoxide dismutase (SOD) activity (SOD-rich T. chuii), to improve various aspects of cellular health. Collectively, results from in vitro, and more importantly in vivo research, support SOD-rich T. chuii as a potential promoter of cellular health. Principally, the ingredient appears to function as an indirect antioxidant by boosting intracellular antioxidant systems. Moreover, it can positively modulate inflammatory status by up-regulating anti-inflammatory and down-regulating pro-inflammatory cytokines and factors. In addition, SOD-rich T. chuii appears to promote cellular health though protecting from DNA damage, boosting immune function, strengthening cell structure and integrity, and positively modulating cell signaling pathways. There is also some evidence to suggest that SOD-rich T. chuii may improve aspects of mitochondrial function through the up-regulation of genes linked to mitochondrial biogenesis and ATP synthesis. From the trials conducted to date, transcriptional activation of nuclear factor erythroid 2-related factor 2 (NRF2) and sirtuin 1 (SIRT1) appear to be important in mediating the effects of SOD-rich T. chuii on cellular health. These exciting preliminary observations suggest that SOD-rich T. chuii may represent a natural blue food supplement with the potential to enhance various aspects of cellular health.
Collapse
Affiliation(s)
- Stuart P. Cocksedge
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
- Centre for Physical Activity, Sport and Exercise Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Lalia Mantecón
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Enrique Castaño
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Carlos Infante
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Stephen J. Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
| |
Collapse
|
12
|
Torres-Isidro O, González-Montoya M, Vargas-Vargas MA, Florian-Rodriguez U, García-Berumen CI, Montoya-Pérez R, Saavedra-Molina A, Calderón-Cortés E, Rodríguez-Orozco AR, Cortés-Rojo C. Anti-Aging Potential of Avocado Oil via Its Antioxidant Effects. Pharmaceuticals (Basel) 2025; 18:246. [PMID: 40006059 PMCID: PMC11858862 DOI: 10.3390/ph18020246] [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: 01/23/2025] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Aging is a process characterized by tissue degeneration, increased susceptibility to chronic degenerative diseases, infections, and the appearance of neoplasms, which leads to disability and a reduction in the length and quality of life. This phenomenon is the result of the convergence of multiple processes, including mitochondrial dysfunction, fibrosis, inflammation, dysregulation of cell death processes, and immunosenescence. These processes have as their point of convergence an increase in the production of ROS. Avocado oil (Persea americana Mill.) contains a diverse array of bioactive compounds, including oleic acid, phytosterols, chlorophylls, xanthones, xanthines, and carotenoids. These bioactive compounds have the capacity to modulate the excessive production of ROS, thereby reducing the progression of age-related diseases and extending lifespan in experimental models of aging. In addition, several studies have demonstrated the efficacy of avocado oil in mitigating age-related diseases, including hypertension; insulin resistance; diabetes; non-alcoholic liver disease; and degenerative processes such as hearing loss, cognitive decline, neurodegeneration, and impaired wound healing. In light of these findings, it is hypothesized that avocado oil is a promising agent capable of promoting healthspan in later stages of life owing to its direct antioxidant actions and the activation of pathways that enhance endogenous antioxidant levels.
Collapse
Affiliation(s)
- Olin Torres-Isidro
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (O.T.-I.); (M.G.-M.); (M.A.V.-V.); (C.I.G.-B.); (R.M.-P.); (A.S.-M.)
| | - Marcela González-Montoya
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (O.T.-I.); (M.G.-M.); (M.A.V.-V.); (C.I.G.-B.); (R.M.-P.); (A.S.-M.)
| | - Manuel Alejandro Vargas-Vargas
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (O.T.-I.); (M.G.-M.); (M.A.V.-V.); (C.I.G.-B.); (R.M.-P.); (A.S.-M.)
| | - Ulises Florian-Rodriguez
- Facultad de Químico Farmacobiología, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58240, Michoacán, Mexico;
| | - Claudia Isabel García-Berumen
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (O.T.-I.); (M.G.-M.); (M.A.V.-V.); (C.I.G.-B.); (R.M.-P.); (A.S.-M.)
| | - Rocío Montoya-Pérez
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (O.T.-I.); (M.G.-M.); (M.A.V.-V.); (C.I.G.-B.); (R.M.-P.); (A.S.-M.)
| | - Alfredo Saavedra-Molina
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (O.T.-I.); (M.G.-M.); (M.A.V.-V.); (C.I.G.-B.); (R.M.-P.); (A.S.-M.)
| | - Elizabeth Calderón-Cortés
- Facultad de Enfermería, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58260, Michoacán, Mexico;
| | - Alain Raimundo Rodríguez-Orozco
- Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58020, Michoacán, Mexico;
| | - Christian Cortés-Rojo
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (O.T.-I.); (M.G.-M.); (M.A.V.-V.); (C.I.G.-B.); (R.M.-P.); (A.S.-M.)
| |
Collapse
|
13
|
Du K, Su Y, Song Q, Chen S, Wu R, Teng X, Huang R, Wang L, Zou C. 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione protects against MPP +-induced neurotoxicity by ameliorating oxidative stress, apoptosis and autophagy in SH-SY5Y cells. Metab Brain Dis 2025; 40:113. [PMID: 39878879 DOI: 10.1007/s11011-025-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione (DMDD) is a cyclohexanedione compound extracted from the roots of Averrhoa carambola L. Several studies have documented its beneficial effects on diabetes, Alzheimer's disease, and cancer. However, its potential neuroprotective effects on Parkinson's disease (PD) have not yet been explored. The present study aimed to investigate the protective effects and underlying mechanisms of DMDD in a cellular model of PD. In this study, SH-SY5Y cells were incubated with or without DMDD following intoxication with the parkinsonian neurotoxin 1-methyl-4-phenylpyridine (MPP+). Cell viability and apoptosis were evaluated using 3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2 H-tetrazolium (MTS) assay and Hoechst 33,342 staining, respectively. The mitochondrial membrane potential (Δψm) was assessed through the JC-10 assay. The activities of superoxide dismutase (SOD) and the levels of reactive oxygen species (ROS) were measured using WST-8 and DCFH-DA assays. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to explore significant biological processes and pathways influenced by DMDD. Molecular docking was employed to predict the domains of potential protein targets interacting with DMDD. Western blotting was subsequently conducted to determine the protein expression levels of TH, Nrf2, Bax, Bcl-2, Caspase-3, Beclin-1, PARP, LC3-II, LC3-I, p-PI3K, PI3K, p-mTOR and mTOR. Our study showed that DMDD treatment significantly increased cell viability and reduced apoptosis in MPP+-treated SH-SY5Y cells. In addition, DMDD treatment reversed the loss of TH expression and Δψm in MPP+-exposed SH-SY5Y cells. Moreover, DMDD treatment reduced MPP+-induced ROS production by promoting SOD activity. Additionally, compared with those in the MPP+ group, the protein expression levels of Beclin-1, Caspase-3, and PARP and the LC3II/I ratio were significantly decreased, whereas the protein expression levels of Nrf2 and the Bcl-2/Bax, p-PI3K/PI3K, and p-mTOR/mTOR ratios were significantly increased in the DMDD-treated group. In conclusion, DMDD protects against MPP+-induced cytotoxicity by mitigating oxidative stress, apoptosis, and autophagy. PI3K/mTOR signaling at least partly mediates the cytoprotective effect of DMDD.
Collapse
Affiliation(s)
- Kechen Du
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying Su
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Qiong Song
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shuai Chen
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ribao Wu
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiahong Teng
- School of International Education, Guangxi Medical University, Nanning, Guangxi, China
- School of International Education, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Renbin Huang
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Lihui Wang
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China.
- Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Chunlin Zou
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China.
- Department of Human Anatomy, Institute of Neuroscience and Guangxi Key Laboratory of Brain Science, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China.
- Center for Translational Medicine, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
14
|
Sun D, Wang W, Fan Q, Wang X, Xu X, Jiang W, Zhao L, Li H, Fu Z, Zhao L, Jiao H. Protective Effects of Black Rice Anthocyanins on D-Galactose-Induced Renal Injury in Mice: The Role of Nrf2 and NF-κB Signaling and Gut Microbiota Modulation. Nutrients 2025; 17:502. [PMID: 39940360 PMCID: PMC11820437 DOI: 10.3390/nu17030502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to evaluate the renal protective effects of black rice anthocyanins (BRAs) against renal injury in mice induced by D-galactose (D-gal). METHODS The renal aging mouse model was established by thirteen consecutive weeks of subcutaneous injections of D-gal. The serum levels of urea nitrogen (BUN), creatinine (CRE), uric acid (UA), antioxidant enzymes (e.g., GSH-Px and SOD), and total antioxidant capacity (T-AOC), as well as the contents of inflammatory factors (IL-1β, IL-6, and TNF-α) in kidney tissues were evaluated. Additionally, the relative expression of the NQO1, HO-1, IKKβ, NF-kBp65, and TLR4 proteins was examined. RESULTS BRA treatment significantly reduced serum levels of BUN, and CRE increased the concentrations of antioxidant enzymes and total antioxidant capacity in renal tissues, and reduced the levels of inflammatory factors. Furthermore, BRAs restored the relative expression of the NQO1, HO-1, IKKβ, NF-kBp65, and TLR4 proteins to normal levels and promoted the recovery of the renal tissue architecture. CONCLUSIONS It was demonstrated that BRAs could potentially prevent and protect against kidney injury by modulating the Nrf2 and NF-κB signaling pathways, attenuating oxidative stress and inflammatory responses, and modulating the gut microflora. These findings provide a scientific basis for the application of BRAs as a natural bioactive substance in the field of nephroprotection, especially against the renal degeneration that accompanies the aging process.
Collapse
Affiliation(s)
- Dan Sun
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, China; (D.S.); (H.L.); (Z.F.)
| | - Wei Wang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (W.W.); (X.W.)
| | - Qian Fan
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (Q.F.); (X.X.); (W.J.); (L.Z.)
| | - Xinyi Wang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (W.W.); (X.W.)
| | - Xinyan Xu
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (Q.F.); (X.X.); (W.J.); (L.Z.)
| | - Weiye Jiang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (Q.F.); (X.X.); (W.J.); (L.Z.)
| | - Liang Zhao
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (Q.F.); (X.X.); (W.J.); (L.Z.)
| | - Hong Li
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, China; (D.S.); (H.L.); (Z.F.)
| | - Zhifang Fu
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, China; (D.S.); (H.L.); (Z.F.)
| | - Lei Zhao
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (Q.F.); (X.X.); (W.J.); (L.Z.)
| | - Hongmei Jiao
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, China; (D.S.); (H.L.); (Z.F.)
| |
Collapse
|
15
|
Wang D, Li Z, Jiang Z, Li Y, Chen Q, Zhou Z. Polymethoxylated flavone variations and in vitro biological activities of locally cultivated Citrus varieties in China. Food Chem 2025; 463:141047. [PMID: 39236394 DOI: 10.1016/j.foodchem.2024.141047] [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: 05/27/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Citrus peels are rich in polymethoxylated flavones (PMFs), which have beneficial health and pharmacological properties. In this study, the profiles, variations, and biological activities of PMFs in the peel extracts of 27 Citrus varieties (eight species) native to China were investigated. UPLC-QTOF-MS/MS analysis revealed that mandarin accumulated more diversity and higher detectable PMF contents. Wangcangzhoupigan (ZPG) possessed the highest antioxidant capacity. Gailiangcheng (GLC) and Bingtangcheng (BTC), sweet oranges showed excellent inhibitory effects against pancreatic lipase and α-glucosidase, respectively. Most citrus extracts effectively inhibited the production of ROS and pro-inflammatory cytokines, while increasing the accumulation of anti-inflammatory cytokines. In addition, Limeng (LM), Cupig-oushigan (GSG), and Yanxiwanlu (YXWL) showed anti-proliferative effects against DU145 and PC3 cancer cells. This study provides a comprehensive PMF profile and biological activities of various citrus species and will benefit future functional citrus breeding practices aimed at designing plants rich in total or specific PMFs for health benefits.
Collapse
Affiliation(s)
- Dan Wang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621010, China; Tianfu Institute of Research and Innovation, Southwest University of Science and Technology, Chengdu 610299, China
| | - Zhenqing Li
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Chongqing 400715, China
| | - Zixiao Jiang
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Chongqing 400715, China
| | - Yi Li
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Chongqing 400715, China
| | - Qiyang Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621010, China.
| | - Zhiqin Zhou
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Chongqing 400715, China; The Southwest Institute of Fruits Nutrition, Banan District, Chongqing 400054, China.
| |
Collapse
|
16
|
Sethi P, Mehan S, Khan Z, Maurya PK, Kumar N, Kumar A, Tiwari A, Sharma T, Das Gupta G, Narula AS, Kalfin R. The SIRT-1/Nrf2/HO-1 axis: Guardians of neuronal health in neurological disorders. Behav Brain Res 2025; 476:115280. [PMID: 39368713 DOI: 10.1016/j.bbr.2024.115280] [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: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
SIRT1 (Sirtuin 1) is a NAD+-dependent deacetylase that functions through nucleoplasmic transfer and is present in nearly all mammalian tissues. SIRT1 is believed to deacetylate its protein substrates, resulting in neuroprotective actions, including reduced oxidative stress and inflammation, increased autophagy, increased nerve growth factors, and preserved neuronal integrity in aging or neurological disease. Nrf2 is a transcription factor that regulates the genes responsible for oxidative stress response and substance detoxification. The activation of Nrf2 guards cells against oxidative damage, inflammation, and carcinogenic stimuli. Several neurological abnormalities and inflammatory disorders have been associated with variations in Nrf2 activation caused by either pharmacological or genetic factors. Recent evidence indicates that Nrf2 is at the center of a complex cellular regulatory network, establishing it as a transcription factor with genuine pleiotropy. HO-1 is most likely a component of a defense mechanism in cells under stress, as it provides negative feedback for cell activation and mediator synthesis. This mediator is upregulated by Nrf2, nitric oxide (NO), and other factors in various inflammatory states. HO-1 or its metabolites, such as CO, may mitigate inflammation by modulating signal transduction pathways. Neurological diseases may be effectively treated by modulating the activity of HO-1. Multiple studies have demonstrated that SIRT1 and Nrf2 share an important connection. SIRT1 enhances Nrf2, activates HO-1, protects against oxidative injury, and decreases neuronal death. This has been associated with numerous neurodegenerative and neuropsychiatric disorders. Therefore, activating the SIRT1/Nrf2/HO-1 pathway may help treat various neurological disorders. This review focuses on the current understanding of the SIRT1 and Nrf2/HO-1 neuroprotective processes and the potential therapeutic applications of their target activators in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Pranshul Sethi
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Pankaj Kumar Maurya
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Aakash Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, Sofia 1113, Bulgaria; Department of Healthcare, South-West University "NeofitRilski", Ivan Mihailov St. 66, Blagoevgrad 2700, Bulgaria
| |
Collapse
|
17
|
Helgueta S, Heurtaux T, Sciortino A, Gui Y, Ohnmacht J, Mencke P, Boussaad I, Halder R, Garcia P, Krüger R, Mittelbronn M, Buttini M, Sauter T, Sinkkonen L. Park7 deletion leads to sex-specific transcriptome changes involving NRF2-CYP1B1 axis in mouse midbrain astrocytes. NPJ Parkinsons Dis 2025; 11:8. [PMID: 39755720 DOI: 10.1038/s41531-024-00851-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/09/2024] [Indexed: 01/06/2025] Open
Abstract
Loss-of-function mutations in PARK7, encoding for DJ-1, can lead to early onset Parkinson's disease (PD). In mice, Park7 deletion leads to dopaminergic deficits during aging, and increased sensitivity to oxidative stress. However, the severity of the reported phenotypes varies. To understand the early molecular changes upon loss of DJ-1, we performed transcriptomic profiling of midbrain sections from young mice. While at 3 months the transcriptomes of both male and female mice were unchanged compared to their wildtype littermates, an extensive deregulation was observed in 8 month-old males. The affected genes are involved in processes like focal adhesion, extracellular matrix interaction, and epithelial-to-mesenchymal transition (EMT), and enriched for primary target genes of NRF2. Consistently, the antioxidant response was altered specifically in the midbrain of male DJ-1 deficient mice. Many of the misregulated genes are known target genes of estrogen and retinoic acid signaling and show sex-specific expression in wildtype mice. Depletion of DJ-1 or NRF2 in male primary astrocytes recapitulated many of the in vivo changes, including downregulation of CYP1B1, an enzyme involved in estrogen and retinoic acid metabolism. Interestingly, knock-down of CYP1B1 led to gene expression changes in focal adhesion and EMT in primary male astrocytes. Finally, male iPSC-derived astrocytes with loss of function mutation in the PARK7 gene also showed changes in the EMT pathway and NRF2 target genes. Taken together, our data indicate that loss of Park7 leads to sex-specific gene expression changes through astrocytic alterations in the NRF2-CYP1B1 axis, suggesting higher sensitivity of males to loss of DJ-1.
Collapse
Affiliation(s)
- Sergio Helgueta
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg, Luxembourg
| | - Tony Heurtaux
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg, Luxembourg
| | - Alessia Sciortino
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Yujuan Gui
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
| | - Jochen Ohnmacht
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
| | - Pauline Mencke
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Ibrahim Boussaad
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Michel Mittelbronn
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
| | - Manuel Buttini
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Thomas Sauter
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg
| | - Lasse Sinkkonen
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
18
|
Monadi T, Mohajer Z, Soltani A, Khazeei Tabari MA, Manayi A, Azadbakht M. The influence of apigenin on cellular responses to radiation: From protection to sensitization. Biofactors 2025; 51:e2113. [PMID: 39134426 DOI: 10.1002/biof.2113] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 07/24/2024] [Indexed: 12/29/2024]
Abstract
Apigenin, a dietary flavonoid, has gained increasing attention for its potential therapeutic applications in radiation protection and radiosensitization. Ionizing radiation (IR) can harm healthy cells, but as radiotherapy remains crucial in cancer treatment. Owing to the remarkable application of radiotherapy in the treatment of cancers, it is vital to protect healthy cells from radiation hazards while increasing the sensitivity of cancer cells to radiation. This article reviews the current understanding of apigenin's radioprotective and radiosensitive properties with a focuses on the involved signaling pathways and key molecular targets. When exposed to irradiation, apigenin reduces inflammation via cyclooxygenase-2 inhibition and modulates proapoptotic and antiapoptotic biomarkers. Apigenin's radical scavenging abilities and antioxidant enhancement mitigate oxidative DNA damage. It inhibits radiation-induced mammalian target of rapamycin activation, vascular endothelial growth factor (VEGF), matrix metalloproteinase-2 (MMP), and STAT3 expression, while promoting AMPK, autophagy, and apoptosis, suggesting potential in cancer prevention. As a radiosensitizer, apigenin inhibits tumor growth by inducing apoptosis, suppressing VEGF-C, tumor necrosis factor alpha, and STAT3, reducing MMP-2/9 activity, and inhibiting cancer cell glucose uptake. Cellular and animal studies support apigenin's radioprotective and anticancer potential, making it a potential candidate for further research. Investigation into apigenin's therapeutic efficacy in diverse cancer types and radiation damage is essential.
Collapse
Affiliation(s)
- Taha Monadi
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Mohajer
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Soltani
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
- USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Azadeh Manayi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Azadbakht
- Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Medicinal Plants Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
19
|
Rockhold JD, Marszalkowski H, Sannella M, Gibney K, Murphy L, Zukowski E, Kalantar GH, SantaCruz-Calvo S, Hart SN, Kuhn MK, Yu J, Stefanik O, Chase G, Proctor EA, Hasturk H, Nikolajczyk BS, Bharath LP. Everolimus alleviates CD4 + T cell inflammation by regulating autophagy and cellular redox homeostasis. GeroScience 2024; 46:5681-5699. [PMID: 38761287 PMCID: PMC11493941 DOI: 10.1007/s11357-024-01187-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/30/2024] [Indexed: 05/20/2024] Open
Abstract
Aging is associated with the onset and progression of multiple diseases, which limit health span. Chronic low-grade inflammation in the absence of overt infection is considered the simmering source that triggers age-associated diseases. Failure of many cellular processes during aging is mechanistically linked to inflammation; however, the overall decline in the cellular homeostasis mechanism of autophagy has emerged as one of the top and significant inducers of inflammation during aging, frequently known as inflammaging. Thus, physiological or pharmacological interventions aimed at improving autophagy are considered geroprotective. Rapamycin analogs (rapalogs) are known for their ability to inhibit mTOR and thus regulate autophagy. This study assessed the efficacy of everolimus, a rapalog, in regulating inflammatory cytokine production in T cells from older adults. CD4+ T cells from older adults were treated with a physiological dose of everolimus (0.01 µM), and indices of autophagy and inflammation were assessed to gain a mechanistic understanding of the effect of everolimus on inflammation. Everolimus (Ever) upregulated autophagy and broadly alleviated inflammatory cytokines produced by multiple T cell subsets. Everolimus's ability to alleviate the cytokines produced by Th17 subsets of T cells, such as IL-17A and IL-17F, was dependent on autophagy and antioxidant signaling pathways. Repurposing the antineoplastic drug everolimus for curbing inflammaging is promising, given the drug's ability to restore multiple cellular homeostasis mechanisms.
Collapse
Affiliation(s)
- Jack Donato Rockhold
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | | | - Marco Sannella
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Kaleigh Gibney
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Lyanne Murphy
- Department of Biology, Merrimack College, North Andover, MA, USA
| | - Emelia Zukowski
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Gabriella H Kalantar
- Dept of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Sara SantaCruz-Calvo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Samantha N Hart
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Madison K Kuhn
- Department of Neurosurgery, Pharmacology, and Biomedical Engineering and Center for Neural Engineering, Pennsylvania State University, Hershey, PA, USA
| | - Jingting Yu
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Olivia Stefanik
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Gabrielle Chase
- Department of Chemistry and Biochemistry, Merrimack College, North Andover, MA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Pharmacology, and Biomedical Engineering and Center for Neural Engineering, Pennsylvania State University, Hershey, PA, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA
| | | | - Barbara S Nikolajczyk
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Leena P Bharath
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA.
| |
Collapse
|
20
|
Chikhaoui A, Zayoud K, Kraoua I, Bouchoucha S, Tebourbi A, Turki I, Yacoub-Youssef H. Supplementation with nicotinamide limits accelerated aging in affected individuals with cockayne syndrome and restores antioxidant defenses. Aging (Albany NY) 2024; 16:13271-13287. [PMID: 39611850 PMCID: PMC11719109 DOI: 10.18632/aging.206160] [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: 03/11/2024] [Accepted: 10/21/2024] [Indexed: 11/30/2024]
Abstract
Cockayne syndrome (CS) is a segmental progeroid syndrome characterized by defects in the DNA excision repair pathway, predisposing to neurodegenerative manifestations. It is a rare genetic disorder and an interesting model for studying premature aging. Oxidative stress and autophagy play an important role in the aging process. The study of these two processes in a model of accelerated aging and the means to counteract them would lead to the identification of relevant biomarkers with therapeutic value for healthy aging. Here we investigated the gene expression profiles of several oxidative stress-related transcripts derived from CS-affected individuals and healthy elderly donors. We also explored the effect of nicotinamide supplementation on several genes related to inflammation and autophagy. Gene expression analysis revealed alterations in two main pathways. This involves the activation of arachidonic acid metabolism and the repression of the NRF2 pathway in affected individuals with CS. The supplementation with nicotinamide adjusted these abnormalities by enhancing autophagy and decreasing inflammation. Furthermore, CSA/CSB-dependent depletion of the mitochondrial DNA polymerase-γ catalytic subunit (POLG1) was restored following nicotinamide supplementation in CS-affected individuals' fibroblasts. This study reveals the link between oxidative stress and accelerated aging in affected individuals with CS and highlights new biomarkers of cellular senescence. However, further analyses are needed to confirm these results, which could not be carried out, mainly due to the unavailability of crucial samples of this rare disease.
Collapse
Affiliation(s)
- Asma Chikhaoui
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| | - Kouloud Zayoud
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| | - Ichraf Kraoua
- Department of Neuropediatrics, National Institute of Neurology Mongi Ben Hamida, Tunis 2092, Tunisia
| | - Sami Bouchoucha
- Orthopedics Department, Béchir Hamza Children’s Hospital, Tunis 2092, Tunisia
| | - Anis Tebourbi
- Orthopedic and Trauma Surgery Department, Mongi Slim Hospital, La Marsa 2046, Tunisia
| | - Ilhem Turki
- Department of Neuropediatrics, National Institute of Neurology Mongi Ben Hamida, Tunis 2092, Tunisia
| | - Houda Yacoub-Youssef
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| |
Collapse
|
21
|
Franco FN, Peixoto BE, de Araújo GR, Chaves MM. Silencing of the Nrf2 pathway in aging promotes a decrease in the anti-inflammatory effect of resveratrol. Arch Gerontol Geriatr 2024; 129:105694. [PMID: 39541750 DOI: 10.1016/j.archger.2024.105694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/21/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
During aging, in addition to increased oxidative stress, inflammation also occurs. A chronic and low-grade inflammation - called "inflammaging" - develops, which contributes to the etiology of diseases related to aging. Resveratrol (Resv.) is a polyphenol well known for its biologically active properties, such as antioxidant and anti-inflammatory properties. This balance can be regulated by Nrf2 - a transcription factor that regulates cellular defense against oxidative agents through the expression or inhibition of certain genes. The objective was to evaluate the effect of Nrf2 on the production of cytokines in leukocytes of different ages treated with resveratrol (5µm). The subjects were divided into three groups: 20-39, 40-59 and 60-80 years old. After separation of the leukocytes, a 24-hour treatment was carried out with and without ML385 inhibitor with the treatments: Control, Resv, Peroxide and Peroxide+Resv. 150 µM peroxide was set to develop an oxidative environment. Cytokines were measured by ELISA (*p < 0.05). In general, there was an increase in TNF and IL-6 in cells stimulated with peroxide compared to controls. A decrease in these two cytokines was also observed in cells treated with resveratrol, both at basal levels and in an oxidizing environment (with peroxide). The polyphenol was able to increase IL-10 only in the youngest age groups. The same profile was observed comparing the same groups when the Nrf2 pathway was inhibited with ML385. It is concluded that resveratrol may have a better effect on preventing oxidation and inflammation present in aging, especially through the antioxidant and anti-inflammatory Nrf2 pathway.
Collapse
Affiliation(s)
- Filipe Nogueira Franco
- Biochemistry Laboratory of Aging and Correlated Diseases, Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Av. Antônio Carlos 6627, 30161-970, Belo Horizonte MG Brazil
| | - Brenda Evangelista Peixoto
- Biochemistry Laboratory of Aging and Correlated Diseases, Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Av. Antônio Carlos 6627, 30161-970, Belo Horizonte MG Brazil
| | - Glaucy Rodrigues de Araújo
- Biochemistry Laboratory of Aging and Correlated Diseases, Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Av. Antônio Carlos 6627, 30161-970, Belo Horizonte MG Brazil
| | - Miriam Martins Chaves
- Biochemistry Laboratory of Aging and Correlated Diseases, Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Av. Antônio Carlos 6627, 30161-970, Belo Horizonte MG Brazil.
| |
Collapse
|
22
|
Zhang X, Li H, Chen L, Wu Y, Li Y. NRF2 in age-related musculoskeletal diseases: Role and treatment prospects. Genes Dis 2024; 11:101180. [PMID: 39281838 PMCID: PMC11400624 DOI: 10.1016/j.gendis.2023.101180] [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: 05/10/2023] [Revised: 10/06/2023] [Accepted: 10/31/2023] [Indexed: 09/18/2024] Open
Abstract
The NRF2 pathway is a metabolic- and redox-sensitive signaling axis in which the transcription factor controls the expression of a multitude of genes that enable cells to survive environmental stressors, such as oxidative stress, mainly by inducing the expression of cytoprotective genes. Basal NRF2 levels are maintained under normal physiological conditions, but when exposed to oxidative stress, cells activate the NRF2 pathway, which is crucial for supporting cell survival. Recently, the NRF2 pathway has been found to have novel functions in metabolic regulation and interplay with other signaling pathways, offering novel insights into the treatment of various diseases. Numerous studies have shown that targeting its pathway can effectively investigate the development and progression of age-related musculoskeletal diseases, such as sarcopenia, osteoporosis, osteoarthritis, and intervertebral disc degeneration. Appropriate regulation of the NRF2 pathway flux holds promise as a means to improve musculoskeletal function, thereby providing a new avenue for drug treatment of age-related musculoskeletal diseases in clinical settings. The review summarized an overview of the relationship between NRF2 and cellular processes such as oxidative stress, apoptosis, inflammation, mitochondrial dysfunction, ferroptosis, and autophagy, and explores the potential of targeted NRF2 regulation in the treatment of age-related musculoskeletal diseases.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei 430056, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
23
|
Zhang Y, Li Y, Ren T, Duan JA, Xiao P. Promising tools into oxidative stress: A review of non-rodent model organisms. Redox Biol 2024; 77:103402. [PMID: 39437623 PMCID: PMC11532775 DOI: 10.1016/j.redox.2024.103402] [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: 07/28/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
Oxidative stress is a crucial concept in redox biology, and significant progress has been made in recent years. Excessive levels of reactive oxygen species (ROS) can lead to oxidative damage, heightening vulnerability to various diseases. By contrast, ROS maintained within a moderate range plays a role in regulating normal physiological metabolism. Choosing suitable animal models in a complex research context is critical for enhancing research efficacy. While rodents are frequently utilized in medical experiments, they pose challenges such as high costs and ethical considerations. Alternatively, non-rodent model organisms like zebrafish, Drosophila, and C. elegans offer promising avenues into oxidative stress research. These organisms boast advantages such as their small size, high reproduction rate, availability for live imaging, and ease of gene manipulation. This review highlights advancements in the detection of oxidative stress using non-rodent models. The oxidative homeostasis regulatory pathway, Kelch-like ECH-associated protein 1-Nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2), is systematically reviewed alongside multiple regulation of Nrf2-centered pathways in different organisms. Ultimately, this review conducts a comprehensive comparative analysis of different model organisms and further explores the combination of novel techniques with non-rodents. This review aims to summarize state-of-the-art findings in oxidative stress research using non-rodents and to delineate future directions.
Collapse
Affiliation(s)
- Yuhao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yun Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tianyi Ren
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ping Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
24
|
Guo H, Li C, Zhao J, Guo T, Chen S, Qin X, Zhu K, Zhang W. Mechanism of Gastrodin against neurotoxicity based on network pharmacology, molecular docking and experimental verification. Biomed Pharmacother 2024; 180:117611. [PMID: 39461014 DOI: 10.1016/j.biopha.2024.117611] [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: 06/20/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Disorders of glutamate metabolism and excessive release participat in multiple neuronal pathologies including ischemic stroke (IS), Alzheimer's disease (AD), or Parkinson's disease (PD). Recently, herbal medicines have been widely used and have shown satisfactory results in the treatment of neurological disorders. Gastrodin is a traditional Chinese medicine (TCM) used for the treatment of nerve injuries, spinal cord injuries, and some central nervous system diseases as well. This research examines the neuroprotective effects of Gastrodin against glutamate-induced neurotoxicity in neuronal cells. METHODS The HERB database was used to explore the active ingredients and target genes of Gastrodia Elata. The STRING database and Cytoscape software were used to screen and construct the Protein-Protein Interaction (PPI). Furthermore, we used molecular docking to predict the potential targets of Gastrodin. The effects of Gastrodin were revealed by western blot, calcium imaging, membrane clamp, CCK8 and flow cytometry. Neuronal oxidative stress and damage were assessed by measuring malondialdehyde (MDA) levels and superoxide dismutase (SOD) activity. Neuronal morphology was examined using Golgi-Cox staining. Finally, animal behavior was examined using novel object recognition and fear conditioning tests. RESULTS We have obtained 22 components such as TM10, TM17, TM25 (Gastrodin), and 281 targets such as AKT, EGFR, and CDK1 through network pharmacology. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed these genes were significantly enriched in protein phosphorylation, protein serine/threonine/tyrosine kinase activity, apoptosis and HIF-1 signaling pathways, etc. A higher affinity between Gastrodin and AKT was revealed by PPI analysis and molecular docking. Further, Gastrodin significantly inhibited Ca2+ influxes and excitatory synaptic transmission in cortical neurons. In addition, Gastrodin effectively alleviated neuron apoptosis, oxidative stress and damage. CONCLUSION Gastrodin has neuroprotective effects against glutamate-induced neurotoxicity.
Collapse
Affiliation(s)
- Han Guo
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Shijiazhuang, Hebei 050017, China; Department of Oral Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Chenyang Li
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Shijiazhuang, Hebei 050017, China
| | - Jiaojiao Zhao
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Shijiazhuang, Hebei 050017, China
| | - Tianyuan Guo
- Department of plastic surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Siruan Chen
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Shijiazhuang, Hebei 050017, China
| | - Xia Qin
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Shijiazhuang, Hebei 050017, China
| | - Kangsheng Zhu
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Shijiazhuang, Hebei 050017, China
| | - Wei Zhang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Shijiazhuang, Hebei 050017, China.
| |
Collapse
|
25
|
Murphy BÓ, Latimer C, Dobani S, Pourshahidi LK, Fontana M, Ternan NG, McDougall G, Rowland I, Pereira-Caro G, Tuohy KM, Del Rio D, Almutairi TM, Crozier A, Naumovski N, Gill CIR. Microbially mediated phenolic catabolites exert differential genoprotective activities in normal and adenocarcinoma cell lines. Int J Food Sci Nutr 2024; 75:673-686. [PMID: 39261459 DOI: 10.1080/09637486.2024.2397055] [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: 03/18/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Age-associated decline of nuclear factor erythroid 2-related factor 2 (Nrf2) activity and DNA repair efficiency leads to the accumulation of DNA damage and increased risk of cancer. Understanding the mechanisms behind increased levels of damaged DNA is crucial for developing interventions to mitigate age-related cancer risk. Associated with various health benefits, (poly)phenols and their microbially mediated phenolic catabolites represent a potential means to reduce DNA damage. Four colonic-microbiota-derived phenolic catabolites were investigated for their ability to reduce H2O2-induced oxidative DNA damage and modulate the Nrf2-Antixoidant Response Element (ARE) pathway, in normal (CCD 841 CoN) and adenocarcinoma (HT29) colonocyte cell lines. Each catabolite demonstrated significant (p < .001) genoprotective activity and modulation of key genes in the Nrf2-ARE pathway. Overall, the colon-derived phenolic metabolites, when assessed at physiologically relevant concentrations, reduced DNA damage in both normal and adenocarcinoma colonic cells in response to oxidative challenge, mediated in part via upregulation of the Nrf2-ARE pathway.
Collapse
Affiliation(s)
- Brian Óg Murphy
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK
| | - Cheryl Latimer
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK
| | - Sara Dobani
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK
| | - L Kirsty Pourshahidi
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK
| | - Massimilano Fontana
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK
| | - Nigel G Ternan
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK
| | - Gordon McDougall
- Environmental and Biochemical Sciences Department, The James Hutton Institute, Dundee, UK
| | - Ian Rowland
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | - Gema Pereira-Caro
- Department of Food Science and Health, IFAPA-Alameda Del Obispo, Córdoba, Spain
| | - Kieran M Tuohy
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
| | | | - Alan Crozier
- Department of Chemistry, King Saud University Riyadh, Saudi Arabia
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Nenad Naumovski
- School of Rehabilitation and Exercise Sciences, Faculty of Health, University of Canberra, Canberra, Australia
| | - Chris I R Gill
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK
| |
Collapse
|
26
|
Yang Y, Lu X, Liu N, Ma S, Zhang H, Zhang Z, Yang K, Jiang M, Zheng Z, Qiao Y, Hu Q, Huang Y, Zhang Y, Xiong M, Liu L, Jiang X, Reddy P, Dong X, Xu F, Wang Q, Zhao Q, Lei J, Sun S, Jing Y, Li J, Cai Y, Fan Y, Yan K, Jing Y, Haghani A, Xing M, Zhang X, Zhu G, Song W, Horvath S, Rodriguez Esteban C, Song M, Wang S, Zhao G, Li W, Izpisua Belmonte JC, Qu J, Zhang W, Liu GH. Metformin decelerates aging clock in male monkeys. Cell 2024; 187:6358-6378.e29. [PMID: 39270656 DOI: 10.1016/j.cell.2024.08.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/10/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024]
Abstract
In a rigorous 40-month study, we evaluated the geroprotective effects of metformin on adult male cynomolgus monkeys, addressing a gap in primate aging research. The study encompassed a comprehensive suite of physiological, imaging, histological, and molecular evaluations, substantiating metformin's influence on delaying age-related phenotypes at the organismal level. Specifically, we leveraged pan-tissue transcriptomics, DNA methylomics, plasma proteomics, and metabolomics to develop innovative monkey aging clocks and applied these to gauge metformin's effects on aging. The results highlighted a significant slowing of aging indicators, notably a roughly 6-year regression in brain aging. Metformin exerts a substantial neuroprotective effect, preserving brain structure and enhancing cognitive ability. The geroprotective effects on primate neurons were partially mediated by the activation of Nrf2, a transcription factor with anti-oxidative capabilities. Our research pioneers the systemic reduction of multi-dimensional biological age in primates through metformin, paving the way for advancing pharmaceutical strategies against human aging.
Collapse
Affiliation(s)
- Yuanhan Yang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyong Lu
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ning Liu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Ma
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Zhiyi Zhang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Kuan Yang
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengmeng Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zikai Zheng
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yicheng Qiao
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qinchao Hu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou 510060, China
| | - Ying Huang
- Chongqing Fifth People's Hospital, Chongqing 400060, China
| | - Yiyuan Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Muzhao Xiong
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lixiao Liu
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pradeep Reddy
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | - Xueda Dong
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fanshu Xu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiaoran Wang
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhao
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Jinghui Lei
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Shuhui Sun
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ying Jing
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Jingyi Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Yusheng Cai
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanling Fan
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Kaowen Yan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yaobin Jing
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; International Center for Aging and Cancer, Hainan Medical University, Haikou 571199, China
| | - Amin Haghani
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | - Mengen Xing
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guodong Zhu
- Institute of Gerontology, Guangzhou Geriatric Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weihong Song
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Steve Horvath
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | | | - Moshi Song
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Si Wang
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing 100053, China; National Medical Center for Neurological Diseases, Beijing 100053, China; Beijing Municipal Geriatric Medical Research Center, Beijing 100053, China
| | - Wei Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Jing Qu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| | - Weiqi Zhang
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| | - Guang-Hui Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| |
Collapse
|
27
|
Sawicka D, Maciak S, Sadowska A, Sokołowska E, Gohal S, Guzińska-Ustymowicz K, Niemirowicz-Laskowska K, Car H. Metabolic Rate and Oxidative Stress as a Risk Factors in the Development of Colorectal Cancer. Int J Mol Sci 2024; 25:10713. [PMID: 39409042 PMCID: PMC11476475 DOI: 10.3390/ijms251910713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
There is growing evidence that the body's energy expenditures constitute a significant risk factor for the development of most deadly diseases, including cancer. Our aim was to investigate the impact of basal metabolic rate (BMR) on the growth and progression of colorectal cancer (CRC). To do so, we used a unique model consisting of three lines of laboratory mice (Mus musculus) artificially selected for high (HBMR) and low (LBMR) basal metabolic rate and randomly bred individuals (non-selected, NSBMR). The experimental individuals were implanted with human colorectal cancer cells DLD-1. The variation in BMR between the lines allowed for testing the impact of whole-body metabolism on oxidative and antioxidant parameters in the liver throughout the cancerogenesis process. We investigated the dependence between metabolic values, reactive oxygen species (ROS) levels, and Kelch-like ECH-associated protein 1-based E3 ligase complexes (Keap1) gene activity in these animals. We found that the HBMR strain had a higher concentration of oxidative enzymes compared to the LBMR and NSBMR. Furthermore, the growth rate of CRC tumors was associated with alterations in the levels of oxidative stress enzymes and Keap1 expression in animals with a high metabolic rate. Our results indicate that a faster growth and development of CRC line DLD-1 is associated with enzymatic redox imbalance in animals with a high BMR.
Collapse
Affiliation(s)
- Diana Sawicka
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna Street 37, 15-295 Bialystok, Poland; (A.S.); (S.G.); (K.N.-L.); (H.C.)
| | - Sebastian Maciak
- Department of Evolutionary and Physiological Ecology, Faculty of Biology, University of Bialystok, Ciolkowskiego Street 1J, 15-245 Bialystok, Poland;
| | - Anna Sadowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna Street 37, 15-295 Bialystok, Poland; (A.S.); (S.G.); (K.N.-L.); (H.C.)
| | - Emilia Sokołowska
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona Street 15A, 15-274 Bialystok, Poland;
| | - Sylwia Gohal
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna Street 37, 15-295 Bialystok, Poland; (A.S.); (S.G.); (K.N.-L.); (H.C.)
| | - Katarzyna Guzińska-Ustymowicz
- Department of General Pathomorphology, Medical University of Bialystok, Waszyngtona Street 13, 15-269 Bialystok, Poland;
| | - Katarzyna Niemirowicz-Laskowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna Street 37, 15-295 Bialystok, Poland; (A.S.); (S.G.); (K.N.-L.); (H.C.)
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna Street 37, 15-295 Bialystok, Poland; (A.S.); (S.G.); (K.N.-L.); (H.C.)
| |
Collapse
|
28
|
Medoro A, Saso L, Scapagnini G, Davinelli S. NRF2 signaling pathway and telomere length in aging and age-related diseases. Mol Cell Biochem 2024; 479:2597-2613. [PMID: 37917279 PMCID: PMC11455797 DOI: 10.1007/s11010-023-04878-x] [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: 08/19/2023] [Accepted: 10/07/2023] [Indexed: 11/04/2023]
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is well recognized as a critical regulator of redox, metabolic, and protein homeostasis, as well as the regulation of inflammation. An age-associated decline in NRF2 activity may allow oxidative stress to remain unmitigated and affect key features associated with the aging phenotype, including telomere shortening. Telomeres, the protective caps of eukaryotic chromosomes, are highly susceptible to oxidative DNA damage, which can accelerate telomere shortening and, consequently, lead to premature senescence and genomic instability. In this review, we explore how the dysregulation of NRF2, coupled with an increase in oxidative stress, might be a major determinant of telomere shortening and age-related diseases. We discuss the relevance of the connection between NRF2 deficiency in aging and telomere attrition, emphasizing the importance of studying this functional link to enhance our understanding of aging pathologies. Finally, we present a number of compounds that possess the ability to restore NRF2 function, maintain a proper redox balance, and preserve telomere length during aging.
Collapse
Affiliation(s)
- Alessandro Medoro
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy
| | - Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy.
| |
Collapse
|
29
|
Liu W, Liu R, Qin Q, Wang H, Zhang X, Meng G. Molecular docking and molecular dynamics simulation of wheat gluten-derived antioxidant peptides acting through the Keap1-Nrf2 pathway. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:8150-8161. [PMID: 38837798 DOI: 10.1002/jsfa.13647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/03/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND In our previous study, we successfully identified five peptides from wheat gluten: Ala-Pro-Ser-Tyr (APSY), Leu-Tyr (LY), Pro-Tyr (PY), Arg-Gly-Gly-Tyr (RGGY) and Tyr-Gln (YQ). Molecular docking and molecular dynamics simulation methods were employed to investigate the interaction between these antioxidant peptides and the Kelch-like ECH-associated protein 1 (Keap1 protein), revealing the molecular mechanism of their non-competitive binding. In addition, the total antioxidant capacity of the five peptides was determined using the 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt (ABTS) method. RESULTS The affinities of APSY, LY, PY, RGGY and YQ were -8.9, -8.3, -8.5, -9.1 and - 7.9 kcal mol-1, respectively. The five peptides effectively bound to Keap1 protein through hydrogen, π-σ, π-alkyl and alkyl interactions. Significant roles were observed for the P1 pocket residue ARG-415 and the P3 pocket residue ALA-556 in the interactions of the Keap1-peptide complexes. Molecular dynamics simulations further elucidated the dynamic process of peptide binding to the Keap1 protein. All five peptides formed stable complexes with Keap1 protein, with van der Waals forces playing crucial roles in these complex systems, indicative of the peptides' strong binding ability to Keap1 protein. The van der Waals forces were -178.74, -123.11, -134.36, -132.59, and -121.44 kJ mol-1 for the Keap1-APSY, Keap1-LY, Keap1-PY, Keap1-RGGY and Keap1-YQ complexes, respectively. These peptides exhibited excellent antioxidant effects. Among them, the YQ peptide exhibited the highest total antioxidant capacity, with an activity value of 1.18 ± 0.06 mmol Trolox equivalent (TE) L-1 at a concentration of 0.10 mg mL-1. The RGGY, PY, LY and APSY peptides followed in descending order, with activity values of 0.91 ± 0.05, 0.72 ± 0.06, 0.62 ± 0.04 and 0.60 ± 0.05 mmol TE L-1, respectively. CONCLUSION These results unveiled the molecular mechanism by which the five antioxidant peptides act on active pockets through the Keap1-Nrf2 signaling pathway, providing a theoretical basis for the development of antioxidants. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wenying Liu
- Engineering Laboratory for Agro Biomass Recycling and Valorizing, College of Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Rui Liu
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries Co., Ltd, Beijing, People's Republic of China
| | - Qingyu Qin
- Engineering Laboratory for Agro Biomass Recycling and Valorizing, College of Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Hualei Wang
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries Co., Ltd, Beijing, People's Republic of China
| | - Xinxue Zhang
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries Co., Ltd, Beijing, People's Republic of China
| | - Ganlu Meng
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries Co., Ltd, Beijing, People's Republic of China
| |
Collapse
|
30
|
Burtscher J, Samaja M. Healthy Aging at Moderate Altitudes: Hypoxia and Hormesis. Gerontology 2024; 70:1152-1160. [PMID: 39348814 DOI: 10.1159/000541216] [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: 06/11/2024] [Accepted: 08/27/2024] [Indexed: 10/02/2024] Open
Abstract
BACKGROUND Aging is associated with cellular and tissue responses that collectively lead to functional and structural deterioration of tissues. Poor tissue oxygenation, or hypoxia, is involved in such responses and contributes to aging. Consequently, it could be speculated that living at higher altitude, and therefore in hypoxic conditions, accelerates aging. This assumption is indeed supported by evidence from populations residing at very high altitudes (>3,500 m). In contrast, accumulating evidence suggests that living at moderate altitudes (1,500-2,500 m) is protective rather than injurious, at least for some body systems. SUMMARY In this review, we critically evaluate the hypothesis that the physiological responses to mild hypoxic stress associated to life at moderate altitudes provide protection from many hypoxia-related diseases through hormesis. Hormesis means that a low dose of a stressor (here hypoxia) elicits beneficial outcomes, while a higher dose can be toxic and might explain at least in part the dose-dependent contrasting effects of hypoxia on the aging processes. The lack of well-designed longitudinal studies focusing on the role of the altitude of residence, and difficulties in accounting for potentially confounding factors such as migration, ethnicity/genetics, and socioeconomic and geoclimatic conditions, currently hampers translation of related research into uncontroversial paradigms. KEY MESSAGES Deeper investigations are required to understand the impact of altitude-related hypoxia on age-related diseases and to develop molecular markers of ageing/senescence in humans that are linked to hypoxia. However, the presented emerging evidence supports the view that hypoxia conditioning has the potential to improve life quality and expectancy.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Michele Samaja
- Department of Health Science, University of Milan, Milan, Italy
| |
Collapse
|
31
|
Li H, Bao L, Pan Y, Zhu X, Cheng J, Zhang J, Chu W. The role of miR-216a-mediated Nrf2 pathway in muscle oxidative stress of Siniperca chuatsi induced by cadmium. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116863. [PMID: 39128454 DOI: 10.1016/j.ecoenv.2024.116863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Cadmium (Cd) is a toxic heavy metal pollutant in the environment. Excessive Cd in water has toxic effects on fish, endangering their healthy growth and ultimately affecting the quality and safety of aquatic products. To evaluate the toxicity of excessive Cd to fish through potential oxidative damage, Siniperca chuatsi was exposed to Cd in water for 15 days. It was found that Cd exposure significantly decreased the survival rate of S. chuatsi and Cd was detected in their muscle. Meanwhile, Cd disrupts the redox balance by reducing antioxidant enzyme activities, increasing reactive oxygen species (ROS) and malondialdehyde (MDA) levels in muscle, and promoting oxidative damage. Histomorphology showed that enlargement of muscle fiber gaps, cell swelling and vacuolar degeneration after Cd exposure. In addition, Cd toxicity induced up-regulating the expression of miR-216a, while down-regulation of Nrf2 protein and its downstream antioxidant enzyme genes expression. Further analysis revealed that miR-216a was significantly negatively correlated with the expression of Nrf2, and injection of miR-216a antagomir significantly enhanced the expression of Nrf2 and antioxidant enzyme genes, as well as the activity of antioxidant enzymes, thereby reducing the damage of Cd to fish. These results suggested that miR-216a-mediated Nrf2 signaling pathway plays an important role in Cd-induced oxidative stress of S. chuatsi muscle.
Collapse
Affiliation(s)
- Honghui Li
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China.
| | - Lingsheng Bao
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China.
| | - Yaxiong Pan
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China
| | - Xin Zhu
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China
| | - Jia Cheng
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China
| | - Jianshe Zhang
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China
| | - Wuying Chu
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China.
| |
Collapse
|
32
|
Lu R, Zhou X, Zhang L, Hao M, Yang X. Nrf2 Deficiency Exacerbates Parkinson's Disease by Aggravating NLRP3 Inflammasome Activation in MPTP-Induced Mouse Models and LPS-Induced BV2 Cells. J Inflamm Res 2024; 17:6277-6295. [PMID: 39281779 PMCID: PMC11401530 DOI: 10.2147/jir.s478683] [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: 05/17/2024] [Accepted: 09/04/2024] [Indexed: 09/18/2024] Open
Abstract
Background Parkinson's disease (PD) is a movement disorder characterized by the progressive loss of dopamine neurons. Microglia-mediated neuroinflammation drives disease progression and becomes a critical factor in neuronal degeneration. Recent studies have found that nuclear factor-erythroid 2-related-2 (Nrf2) expression levels are reduced during aging and neurodegenerative diseases, but its regulatory mechanism on microglia-induced neuroinflammation has not been fully elucidated. Methods In vivo, we used the intraperitoneal injection of the neurotoxic drug neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to establish an animal model of PD and, at the same time, administered Nrf2 inhibitors ML385 and dimethyl fumarate to regulate Nrf2 protein levels. In vitro, we used si-RNA to knock out the Nrf2 gene to intervene in BV2 cells and used lipopolysaccharide (LPS) to stimulate and induce the cell model. Results The study found that inhibition of Nrf2 expression aggravated the motor defects of PD mice, accompanied by a significant loss of dopaminergic neurons in the substantia nigra and striatum of the brain. In addition, after inhibition of Nrf2, the malondialdehyde (MDA) level in the substantia nigra of the midbrain of mice increased, and the levels of superoxide dismutase (SOD) and heme oxygenase-1 (HO-1) decreased, accompanied by the proliferation of microglia and astrocytes. In addition, the activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, the assembly of apoptosis-associated speck-like protein containing a CARD (ASC) protein in microglia, and the release of downstream inflammatory factors caspase-1 and interleukin (IL)-1β, were aggravated. At the cellular level, it was found that knocking out the expression of Nrf2 would aggravate the activation of NLRP3 inflammasomes and the assembly of ASC in LPS-induced BV2 cells. Conclusion Inhibited Nrf2 activity can reduce the downstream antioxidant enzyme HO-1 and antioxidant levels, induce NLRP3 inflammasome activation and ASC protein assembly in microglia, and ultimately aggravate PD inflammatory response and dopamine neuron degeneration.
Collapse
Affiliation(s)
- Ranran Lu
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, People's Republic of China
- Xinjiang Key Laboratory of Neurological Disease Research, Ürümqi, Xinjiang, People's Republic of China
| | - Xu Zhou
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, People's Republic of China
- Xinjiang Key Laboratory of Neurological Disease Research, Ürümqi, Xinjiang, People's Republic of China
| | - Lijie Zhang
- Xinjiang Production and Construction Corps Hospital, Ürümqi, Xinjiang, People's Republic of China
| | - Mengdie Hao
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, People's Republic of China
- Xinjiang Key Laboratory of Neurological Disease Research, Ürümqi, Xinjiang, People's Republic of China
| | - Xinling Yang
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, People's Republic of China
- Xingjiang Medical University, Ürümqi, Xinjiang, People's Republic of China
| |
Collapse
|
33
|
Santos DF, Simão S, Nóbrega C, Bragança J, Castelo-Branco P, Araújo IM. Oxidative stress and aging: synergies for age related diseases. FEBS Lett 2024; 598:2074-2091. [PMID: 39112436 DOI: 10.1002/1873-3468.14995] [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: 03/04/2024] [Revised: 06/03/2024] [Accepted: 06/24/2024] [Indexed: 10/04/2024]
Abstract
Aging is characterized by a progressive decline in physiological function and underlies several disabilities, including the increased sensitivity of cells and tissues to undergo pathological oxidative stress. In recent years, efforts have been made to better understand the relationship between age and oxidative stress and further develop therapeutic strategies to minimize the impact of both events on age-related diseases. In this work, we review the impact of the oxidant and antioxidant systems during aging and disease development and discuss the crosstalk of oxidative stress and other aging processes, with a focus on studies conducted in elderly populations.
Collapse
Affiliation(s)
- Daniela F Santos
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, Portugal
| | - Sónia Simão
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
| | - Clévio Nóbrega
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- ABC Collaborative Laboratory, Association for Integrated Aging and Rejuvenation Solutions (ABC CoLAB), Loulé, Portugal
| | - José Bragança
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- ABC Collaborative Laboratory, Association for Integrated Aging and Rejuvenation Solutions (ABC CoLAB), Loulé, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Pedro Castelo-Branco
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- ABC Collaborative Laboratory, Association for Integrated Aging and Rejuvenation Solutions (ABC CoLAB), Loulé, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Inês M Araújo
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- ABC Collaborative Laboratory, Association for Integrated Aging and Rejuvenation Solutions (ABC CoLAB), Loulé, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon, Portugal
| |
Collapse
|
34
|
Xie G, Xu Z, Li F, Kong M, Wang P, Shao Y. Aerobic Exercise Ameliorates Cognitive Disorder and Declined Oxidative Stress via Modulating the Nrf2 Signaling Pathway in D-galactose Induced Aging Mouse Model. Neurochem Res 2024; 49:2408-2422. [PMID: 38839706 DOI: 10.1007/s11064-024-04164-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 03/03/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
The aim of this research was to explore the potential of treadmill exercise in preventing brain aging and neurodegenerative diseases caused by oxidative stress, by studying its effects on D-galactose-induced mice and the mechanisms involved. The results showed that C57BL/6 mice induced with D-gal exhibited cognitive impairment and oxidative stress damage, which was ameliorated by treadmill exercise. The Morris water maze also showed that exercise improved cognitive performance in aging mice and alleviated hippocampal and mitochondrial damage. The study also found that treadmill exercise increased the expression of nuclear factor Nrf2, p-GSK3β, HO-1, NQO1, BDNF, and Bcl-2 proteins while decreasing the expression of Bax. Furthermore, there was a substantial increase in the levels of CAT, GSH-PX and SOD in the serum, along with a decrease in MDA levels. The outcomes propose that aerobic exercise has the potential to hinder oxidative stress and cell death in mitochondria through the modulation of the Nrf2/GSK3β signaling pathway, thus improving cognitive impairment observed in the aging model induced by D-galactose. It appears that treadmill exercise could potentially serve as an effective therapeutic approach to mitigating brain aging and neurodegenerative diseases triggered by oxidative stress.
Collapse
Affiliation(s)
- Guangjing Xie
- Institute of Geriatrics, Hubei University of Chinese Medicine, Wuhan, Hubei, China
- College of Physical Education and Health, Hubei University of Chinese Medicine, Wuhan, Hubei, China
- Hubei Shizhen Labortary, Wuhan, Hubei, China
| | - Zixuan Xu
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Feizhou Li
- College of Clinical Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Meng Kong
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Ping Wang
- Institute of Geriatrics, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
- Hubei Shizhen Labortary, Wuhan, Hubei, China.
| | - Yuping Shao
- College of Physical Education and Health, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
- Hubei Shizhen Labortary, Wuhan, Hubei, China.
| |
Collapse
|
35
|
Malla R, Kumari S, Ganji SP, Srilatha M, Nellipudi HR, Nagaraju GP. Reactive oxygen species of tumor microenvironment: Harnessing for immunogenic cell death. Biochim Biophys Acta Rev Cancer 2024; 1879:189154. [PMID: 39019409 DOI: 10.1016/j.bbcan.2024.189154] [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: 03/01/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
The tumor microenvironment (TME) is a dynamic and complex system that undergoes continuous changes in its network architecture, notably affecting redox homeostasis. These alterations collectively shape a diverse ecosystem actively supporting tumor progression by influencing the cellular and molecular components of the TME. Despite the remarkable clinical advancements in cancer immunotherapy, its spectrum of clinical utility is limited by the altered TME and inadequate tumor immunogenicity. Recent studies have revealed that some conventional and targeted therapy strategies can augment the efficacy of immunotherapy even in patients with less immunogenic solid tumors. These strategies provoke immunogenic cell death (ICD) through the ROS-dependent liberation of damage-associated molecular patterns (DAMPs). These DAMPs recognize and bind with Pattern Recognition Receptors (PRRs) on immune cells, activating and maturing defense cells, ultimately leading to a robust antitumor immune response. The present review underscores the pivotal role of redox homeostasis in orchestrating the transition of TME from a cold to a hot phenotype and the ROS-ICD axis in immune response induction. Additionally, it provides up-to-date insights into strategies that leverage ROS generation to induce ICD. The comprehensive analysis aims to develop ROS-based effective cancer immunotherapies for less immunogenic tumors.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Seema Kumari
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Swapna Priya Ganji
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India
| | | | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
36
|
Dhyani N, Tian C, Gao L, Rudebush TL, Zucker IH. Nrf2-Keap1 in Cardiovascular Disease: Which Is the Cart and Which the Horse? Physiology (Bethesda) 2024; 39:0. [PMID: 38687468 PMCID: PMC11460534 DOI: 10.1152/physiol.00015.2024] [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: 03/11/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024] Open
Abstract
High levels of oxidant stress in the form of reactive oxidant species are prevalent in the circulation and tissues in various types of cardiovascular disease including heart failure, hypertension, peripheral arterial disease, and stroke. Here we review the role of nuclear factor erythroid 2-related factor 2 (Nrf2), an important and widespread antioxidant and anti-inflammatory transcription factor that may contribute to the pathogenesis and maintenance of cardiovascular diseases. We review studies showing that downregulation of Nrf2 exacerbates heart failure, hypertension, and autonomic function. Finally, we discuss the potential for using Nrf2 modulation as a therapeutic strategy for cardiovascular diseases and autonomic dysfunction.
Collapse
Affiliation(s)
- Neha Dhyani
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Changhai Tian
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Lie Gao
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Tara L Rudebush
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| |
Collapse
|
37
|
Giardina C, Kuo A, Nito K, Kurkcu S. Early onset colorectal cancer: Cancer promotion in young tissue. Biochem Pharmacol 2024; 226:116393. [PMID: 38942088 DOI: 10.1016/j.bcp.2024.116393] [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: 01/23/2024] [Revised: 05/30/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
The incidence of colorectal cancer (CRC) in patients under 50 has been increasing over the past several decades. The factors underlying the increase in early onset colorectal cancer (EOCRC) are not entirely clear, although several genetic and clinical differences with late onset colorectal cancer (LOCRC) have been noted. EOCRC cases are often diagnosed at a more advanced stage, raising the possibility that these cancers progress more rapidly than LOCRC cases. The impact of age on cancer progression is an intriguing topic and numerous lines of research have found that a young tissue environment is often more promotional. In fact, a less hospitable promotional tissue environment in older individuals may offset the increased cancer risk associated with the increased mutational load associated with age. Here we address how youthful aspects of angiogenesis, the tumor immune response, and the oxidative stress response may contribute to the rapid progression of EOCRC. Understanding the factors promoting EOCRC may provide insight into why EOCRC cases are increasing.
Collapse
Affiliation(s)
- Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA.
| | - Alan Kuo
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Klea Nito
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Shan Kurkcu
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
38
|
Smyth SP, Nixon B, Skerrett-Byrne DA, Burke ND, Bromfield EG. Building an Understanding of Proteostasis in Reproductive Cells: The Impact of Reactive Carbonyl Species on Protein Fate. Antioxid Redox Signal 2024; 41:296-321. [PMID: 38115641 DOI: 10.1089/ars.2023.0314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Significance: Stringent regulation of protein homeostasis pathways, under both physiological and pathological conditions, is necessary for the maintenance of proteome fidelity and optimal cell functioning. However, when challenged by endogenous or exogenous stressors, these proteostasis pathways can become dysregulated with detrimental consequences for protein fate, cell survival, and overall organism health. Most notably, there are numerous somatic pathologies associated with a loss of proteostatic regulation, including neurodegenerative disorders, type 2 diabetes, and some cancers. Recent Advances: Lipid oxidation-derived reactive carbonyl species (RCS), such as 4-hydroxynonenal (4HNE) and malondialdehyde, are relatively underappreciated purveyors of proteostatic dysregulation, which elicit their effects via the nonenzymatic post-translational modification of proteins. Emerging evidence suggests that a subset of germline proteins can serve as substrates for 4HNE modification. Among these, prevalent targets include succinate dehydrogenase, heat shock protein A2 and A-kinase anchor protein 4, all of which are intrinsically associated with fertility. Critical Issues: Despite growing knowledge in this field, the RCS adductomes of spermatozoa and oocytes are yet to be comprehensively investigated. Furthermore, the manner by which RCS-mediated adduction impacts protein fate and drives cellular responses, such as protein aggregation, requires further examination in the germline. Given that RCS-protein adduction has been attributed a role in infertility, there has been sparked research investment into strategies to prevent lipid peroxidation in germ cells. Future Directions: An increased depth of knowledge regarding the mechanisms and substrates of RCS-mediated protein modification in reproductive cells may reveal important targets for the development of novel therapies to improve fertility and pregnancy outcomes for future generations.
Collapse
Affiliation(s)
- Shannon P Smyth
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Bio21 Institute, School of BioSciences, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Brett Nixon
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
| | - David A Skerrett-Byrne
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Nathan D Burke
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Bio21 Institute, School of BioSciences, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth G Bromfield
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Bio21 Institute, School of BioSciences, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
39
|
Mayer C, Riera-Ponsati L, Kauppinen S, Klitgaard H, Erler JT, Hansen SN. Targeting the NRF2 pathway for disease modification in neurodegenerative diseases: mechanisms and therapeutic implications. Front Pharmacol 2024; 15:1437939. [PMID: 39119604 PMCID: PMC11306042 DOI: 10.3389/fphar.2024.1437939] [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: 05/24/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Neurodegenerative diseases constitute a global health issue and a major economic burden. They significantly impair both cognitive and motor functions, and their prevalence is expected to rise due to ageing societies and continuous population growth. Conventional therapies provide symptomatic relief, nevertheless, disease-modifying treatments that reduce or halt neuron death and malfunction are still largely unavailable. Amongst the common hallmarks of neurodegenerative diseases are protein aggregation, oxidative stress, neuroinflammation and mitochondrial dysfunction. Transcription factor nuclear factor-erythroid 2-related factor 2 (NRF2) constitutes a central regulator of cellular defense mechanisms, including the regulation of antioxidant, anti-inflammatory and mitochondrial pathways, making it a highly attractive therapeutic target for disease modification in neurodegenerative disorders. Here, we describe the role of NRF2 in the common hallmarks of neurodegeneration, review the current pharmacological interventions and their challenges in activating the NRF2 pathway, and present alternative therapeutic approaches for disease modification.
Collapse
Affiliation(s)
| | - Lluís Riera-Ponsati
- NEUmiRNA Therapeutics, Copenhagen, Denmark
- Center for RNA Medicine, Aalborg University, Copenhagen, Denmark
| | - Sakari Kauppinen
- NEUmiRNA Therapeutics, Copenhagen, Denmark
- Center for RNA Medicine, Aalborg University, Copenhagen, Denmark
| | | | | | | |
Collapse
|
40
|
Dada T, Mahalingam K, Bhartiya S. Reversing Aging and Improving Health Span in Glaucoma Patients: The Next Frontier? J Curr Glaucoma Pract 2024; 18:87-93. [PMID: 39575133 PMCID: PMC11576344 DOI: 10.5005/jp-journals-10078-1451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024] Open
Abstract
How to cite this article: Dada T, Mahalingam K, Bhartiya S. Reversing Aging and Improving Health Span in Glaucoma Patients: The Next Frontier? J Curr Glaucoma Pract 2024;18(3):87-93.
Collapse
Affiliation(s)
- Tanuj Dada
- Department of Ophthalmology, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi, India
| | - Karthikeyan Mahalingam
- Department of Ophthalmology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Shibal Bhartiya
- Department of Ophthalmology and Community Outreach, Marengo Asia Hospitals, Gurugram and Faridabad, Haryana, India; Mayo Clinic, Jacksonville, Florida, United States
| |
Collapse
|
41
|
Miler M, Živanović J, Ajdžanović V, Milenkovic D, Cesar T, Filipović MR, Milošević V. Lemon extract reduces the hepatic oxidative stress and persulfidation levels by upregulating the Nrf2 and Trx1 expression in old rats. Biofactors 2024; 50:756-771. [PMID: 38194360 DOI: 10.1002/biof.2038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/22/2023] [Indexed: 01/10/2024]
Abstract
Citrus flavanones are recognized as promising bioactives within the concept of healthy aging. Thus, the present study investigated the effects of a nutritionally relevant dose of lemon extract (LE) on liver redox regulation and persulfidation levels in 24-month-old Wistar rats. LE (40 mg/kg b.m.) was administered orally once daily for 4 weeks. Control groups received either vehicle (sunflower oil) or remained intact. The applied methodology considered qPCR, Western blot, protein persulfidation levels evaluation, histochemistry in line with immunofluorescence, liver biochemical assays (glutathione, total -SH groups and malonaldehyde; MDA), liver enzymes in serum and in silico analysis to explore the potential interaction/binding between the proteins studied in the paper. Our results showed that LE increased glutathione peroxidase (GPx), reductase (GR), glutamate-cysteine ligase catalytic and modifier subunit, respectively, as well as Nrf2 gene expressions, but decreased the expression of superoxide dismutase 2 (SOD2). Upon LE application, protein expression showed upregulation of NRF2, SOD2, GPx, GR, and thioredoxin 1 (Trx1). LE significantly decreased the protein persulfidation levels and concentration of MDA, a marker of oxidative damage in the cell. Histological analysis showed a normal liver histoarchitecture without pathological changes, aligning with the normal serum level of hepatic enzymes. Obtained results showed that LE, by modulating hepatic redox regulators Nrf2 and Trx1, diminishes oxidative stress and alters the persulfidation levels, suggesting a considerable beneficial antioxidant potential of lemon flavanones in the old-aged liver.
Collapse
Affiliation(s)
- Marko Miler
- Department of Cytology, Institute for Biological Research "Siniša Stanković" (IBISS)- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jasmina Živanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" (IBISS)- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" (IBISS)- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Dragan Milenkovic
- Division of Cardiovascular Medicine, University of California Davis, Davis, California, USA
| | - Thais Cesar
- Graduate Program in Food, Nutrition and Food Engineering, Sao Paulo State University (UNESP), Araraquara, Brasil
| | - Miloš R Filipović
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Verica Milošević
- Department of Anatomy, Faculty of Medicine, University of Niš, Niš, Serbia
| |
Collapse
|
42
|
Tsuji A, Kotani E, Inoue YH. Sesamin Exerts an Antioxidative Effect by Activating the Nrf2 Transcription Factor in the Glial Cells of the Central Nervous System in Drosophila Larvae. Antioxidants (Basel) 2024; 13:787. [PMID: 39061856 PMCID: PMC11274309 DOI: 10.3390/antiox13070787] [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: 05/17/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Sesame seeds are abundant in sesamin, which exerts health-promoting effects such as extending the lifespan of adult Drosophila and suppressing oxidative stress by activating the Nrf2 transcription factor. Here, we investigated whether sesamin activated Nrf2 in larval tissues and induced the expression of Nrf2 target genes. In the sesamin-fed larvae, Nrf2 was activated in the central nervous system (CNS), gut, and salivary glands. The ectopic expression of Keap1 in glial cells inhibited sesamin-induced Nrf2 activation in the whole CNS more than in the neurons, indicating that sesamin activates Nrf2 in glia efficiently. We labeled the astrocytes as well as cortex and surface glia with fluorescence to identify the glial cell types in which Nrf2 was activated; we observed their activation in both cell types. These data suggest that sesamin may stimulate the expression of antioxidative genes in glial cells. Among the 17 candidate Nrf2 targets, the mRNA levels of Cyp6a2 and Cyp6g1 in cytochrome P450 were elevated in the CNS, gut, and salivary glands of the sesamin-fed larvae. However, this elevation did not lead to resistance against imidacloprid, which is detoxified by these enzymes. Our results suggest that sesamin may exert similar health-promoting effects on the human CNS and digestive tissues.
Collapse
Affiliation(s)
| | | | - Yoshihiro H. Inoue
- Biomedical Research Center, Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Sakyo, Kyoto 606-0962, Japan; (A.T.); (E.K.)
| |
Collapse
|
43
|
Bhargavan B, Chhunchha B, Kubo E, Singh DP. DNA methylation as an epigenetic mechanism in the regulation of LEDGF expression and biological response in aging and oxidative stress. Cell Death Discov 2024; 10:296. [PMID: 38909054 PMCID: PMC11193803 DOI: 10.1038/s41420-024-02076-2] [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: 03/22/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024] Open
Abstract
The physiological quantum of stress-inducible transcriptional protein, Lens Epithelium-Derived Growth Factor (LEDGF), is vital for the maintenance of cellular physiology. Erratic epigenetic reprogramming in response to oxidative stress or with advancing age is found to be a major cause in the gene silencing, leading to pathobiologies. Using aging human (h) eye lens/lens epithelial cells (LECs) coupled with redox-active Peroxiredoxin 6 (Prdx6)-deficient (Prdx6-/-) mLECs as model systems, herein, we showed that in aging/oxidative stress, the human LEDGF gene was regulated by unique methylation patterns of CGs nucleotides within and around the Sp1 binding site(s) of CpG island of the LEDGF promoter (-170 to -27nts). The process caused the repression of LEDGF and its target, Hsp27, resulting in reactive oxygen species (ROS) amplification and cellular insults. This phenomenon was opposed to the unmethylated promoter in LECs. Clinically, we observed that the loss of LEDGF in the Prdx6-/- mLECs or aging lenses/LECs, correlating with increased expression of DNMT1, DNMT3a, and DNMT3b along with the methyl CpG binding protein 2 (MeCP2). Upon oxidative stress, the expression of these molecules was increased with the dramatic reduction in LEDGF expression. While demethylating agent, 5-Aza deoxycytidine (5-AzaC) transposed the aberrant methylation status, and revived LEDGF and Hsp27 expression. Mechanistically, the chloramphenicol acetyltransferase (CAT) reporter gene driven by the LEDGF promoter (-170/ + 35) and ChIP assays uncovered that 5-AzaC acted on GC/Sp1 sites to release LEDGF transcription. The data argued, for the first time, that de novo methylation of CGs around and within Sp1 sites of the CpG island directly disrupted Sp1 activity, which ensued in LEDGF repression and its biological functions. The findings should improve our understanding of cellular insults-associated with aberrant DNMTs-mediated LEDGF's activity, and can offer strategies for therapeutic intervention to halt aging/oxidative stress-induced abnormalities.
Collapse
Affiliation(s)
- Biju Bhargavan
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bhavana Chhunchha
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Ishikawa, 9200293, Japan
| | - Dhirendra P Singh
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
44
|
Zhong R, Shen L, Fan Y, Luo Q, Hong R, Sun X, Zhou X, Wan J. Anti-aging mechanism and effect of treatment with raw and wine-steamed Polygonatum sibiricum on D-galactose-induced aging in mice by inhibiting oxidative stress and modulating gut microbiota. Front Pharmacol 2024; 15:1335786. [PMID: 38774211 PMCID: PMC11106437 DOI: 10.3389/fphar.2024.1335786] [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: 11/09/2023] [Accepted: 04/04/2024] [Indexed: 05/24/2024] Open
Abstract
Background Polygonatum sibiricum (PS) is a traditional Chinese medicine (TCM) first recorded in Mingyi Bielu. The book documents that PS can nourish five internal organs, be taken for a long time, relax the body and prolong lifespan. Presently, PS is widely used in TCM to prevent premature graying of hair. Based on TCM theory and clinical trials, the wine steaming processed product from PS provides a better effect. However, no published study has elucidated the anti-aging mechanism. Purpose The study aim was to investigate the anti-aging mechanism of PS and its wine steaming processed product in mice, specifically focusing on the effect of D-galactose (D-gal) surrounding the intestinal flora and the Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2-antioxidant response elements (Keap1/Nrf2/ARE) pathway. Methods The chemical components in Raw PS (RPS) and Wine-steamed PS (WPS) were identified by ultra-performance liquid chromatography-hybrid quadrupole-Orbitrap high-resolution mass spectrometry (UPLC-Q-Orbitrap HRMS). An aging model using Kunming mice was established through intraperitoneally injected D-gal. Concentrations of RPS and WPS at 5, 10, or 15 g/kg/day levels were administered intragastrically, respectively. The body weight, liver and spleen indexes, superoxide dismutase (SOD), glutathione peroxidase (GSH-PX), and malondialdehyde (MDA) activities in serum and brain tissue were recorded. Hematoxylin and eosin (HE) stained brain tissue was histopathologically examined. The expressions of Keap1, Nrf2 and heme oxygenase 1 (HO-1) in the brain tissue at the mRNA and protein levels were respectively detected by reverse transcription-polymerase chain reaction (RT-PCR) and western blot (WB). Moreover, an Illumina Hiseq platform was used for 16S ribosomal RNA (16S rRNA) high-throughput sequencing to evaluate the proportions of intestinal flora in aging mice. Results The proportions of saccharides, flavonoids, and triterpene acids were different between RPS and WPS. In the aging model mice, WPS outperformed RPS in improving body weight and mental state by increasing the spleen index, SOD and GSH-PX activities, decreasing the liver index and MDA activities, and restoring the histopathological morphology in D-gal-induced aging mice. At the mRNA levels, RPS and WPS significantly reduced the expression of Keap1 and increased the expressions of Nrf2 and HO-1. The trend in protein expressions was similar to that of the mRNA results, and WPS had a stronger effect than RPS. Fecal microbiota analysis showed that RPS and WPS restored intestinal microbiota proportions to normal levels. Conclusion The results demonstrated that PS and its WPS had a positive effect in relieving oxidative stress in aging mice. WPS outperformed RPS, which might be related to the activation of the Keap1/Nrf2/ARE pathway and regulation of intestinal flora.
Collapse
Affiliation(s)
- Ruixue Zhong
- Sichuan Provincial Orthopedic Hospital, Chengdu, China
| | - Ling Shen
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Yilin Fan
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Qiaomei Luo
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Ran Hong
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Xiaoli Sun
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Xia Zhou
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jun Wan
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
45
|
Taylor AL, Dubuisson O, Pandey P, Zunica ERM, Vandanmagsar B, Dantas WS, Johnson A, Axelrod CL, Kirwan JP. Restricting bioenergetic efficiency enhances longevity and mitochondrial redox capacity in Drosophila melanogaster. Aging Cell 2024; 23:e14107. [PMID: 38343281 PMCID: PMC11113268 DOI: 10.1111/acel.14107] [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: 08/21/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 05/24/2024] Open
Abstract
Mitochondria are essential for survival and as such, impairments in organelle homeostasis significantly accelerate age-related morbidity and mortality. Here, we determined the contribution of bioenergetic efficiency to life span and health span in Drosophila melanogaster utilizing the mitochondrial uncoupler BAM15. Life span was determined in flies fed a normal diet (ND) or high fat diet (HFD) supplemented with vehicle or BAM15. Locomotor function was determined by negative geotaxis assay in middle-aged flies fed vehicle or BAM15 under ND or HFD conditions. Redox capacity (high-resolution respirometry/fluorometry), citrate synthase (enzyme activity), mtDNA content (qPCR), gene expression (qPCR), and protein expression (western blot) were assessed in flight muscle homogenates of middle-aged flies fed vehicle or BAM15 ND. The molar ratio of H2O2 and O2 (H2O2:O2) in a defined respiratory state was calculated as a measure of redox balance. BAM15 extended life span by 9% on ND and 25% on HFD and improved locomotor activity by 125% on ND and 53% on HFD. Additionally, BAM15 enhanced oxidative phosphorylation capacity supported by pyruvate + malate, proline, and glycerol 3-phosphate. Concurrently, BAM15 enhanced the mitochondrial H2O2 production rate, reverse electron flow from mitochondrial glycerol-3-phosphate dehydrogenase (mGPDH) to Complex I, mGPDH, and Complex I without altering the H2O2:O2 ratio. BAM15 upregulated transcriptional signatures associated with mitochondrial function and fitness as well as antioxidant defense. BAM15-mediated restriction of bioenergetic efficiency prolongs life span and health span in Drosophila fed a ND or HFD. Improvements in life span and health span in ND were supported by synergistic enhancement of muscular redox capacity.
Collapse
Affiliation(s)
- Analisa L. Taylor
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Olga Dubuisson
- Department of Biological SciencesLouisiana State UniversityBaton RougeLouisianaUSA
| | - Pritika Pandey
- Department of Biological SciencesLouisiana State UniversityBaton RougeLouisianaUSA
| | - Elizabeth R. M. Zunica
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Bolormaa Vandanmagsar
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Wagner S. Dantas
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Alyssa Johnson
- Department of Biological SciencesLouisiana State UniversityBaton RougeLouisianaUSA
| | - Christopher L. Axelrod
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - John P. Kirwan
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| |
Collapse
|
46
|
Carrow KP, Hamilton HL, Hopps MP, Li Y, Qiao B, Payne NC, Thompson MP, Zhang X, Magassa A, Fattah M, Agarwal S, Vincent MP, Buyanova M, Bertin PA, Mazitschek R, Olvera de la Cruz M, Johnson DA, Johnson JA, Gianneschi NC. Inhibiting the Keap1/Nrf2 Protein-Protein Interaction with Protein-Like Polymers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311467. [PMID: 38241649 PMCID: PMC11257647 DOI: 10.1002/adma.202311467] [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: 10/31/2023] [Revised: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Successful and selective inhibition of the cytosolic protein-protein interaction (PPI) between nuclear factor erythroid 2-related factor 2 (Nrf2) and Kelch-like ECH-associating protein 1 (Keap1) can enhance the antioxidant response, with the potential for a therapeutic effect in a range of settings including in neurodegenerative disease (ND). Small molecule inhibitors have been developed, yet many have off-target effects, or are otherwise limited by poor cellular permeability. Peptide-based strategies have also been attempted to enhance specificity, yet face challenges due to susceptibility to degradation and lack of cellular penetration. Herein, these barriers are overcome utilizing a polymer-based proteomimetics. The protein-like polymer (PLP) consists of a synthetic, lipophilic polymer backbone displaying water soluble Keap1-binding peptides on each monomer unit forming a brush polymer architecture. The PLPs are capable of engaging Keap1 and displacing the cellular protective transcription factor Nrf2, which then translocates to the nucleus, activating the antioxidant response element (ARE). PLPs exhibit increased Keap1 binding affinity by several orders of magnitude compared to free peptides, maintain serum stability, are cell-penetrant, and selectively activate the ARE pathway in cells, including in primary cortical neuronal cultures. Keap1/Nrf2-inhibitory PLPs have the potential to impact the treatment of disease states associated with dysregulation of oxidative stress, such as NDs.
Collapse
Affiliation(s)
- Kendal P Carrow
- Department of Biomedical Engineering, McCormick School of Engineering, Medical Scientist Training Program, Feinberg School of Medicine, International Institute for Nanotechnology, Northwestern University, Evanston, 60208, IL, USA
| | - Haylee L Hamilton
- School of Pharmacy, University of Wisconsin, Madison, 57305, WI, USA
| | - Madeline P Hopps
- International Institute for Nanotechnology, Department of Chemistry, Chemistry of Life Processes Institute, Northwestern University, Evanston, 60208, IL, USA
| | - Yang Li
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, 60208, IL, USA
| | - Baofu Qiao
- Department of Natural Sciences, Baruch College, City University of New York, New York, 10010, NY, USA
| | - N Connor Payne
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, 02138, MA, USA
| | - Matthew P Thompson
- International Institute for Nanotechnology, Department of Chemistry, Chemistry of Life Processes Institute, Northwestern University, Evanston, 60208, IL, USA
| | - Xiaoyu Zhang
- International Institute for Nanotechnology, Department of Chemistry, Chemistry of Life Processes Institute, Northwestern University, Evanston, 60208, IL, USA
| | - Assa Magassa
- International Institute for Nanotechnology, Department of Chemistry, Chemistry of Life Processes Institute, Northwestern University, Evanston, 60208, IL, USA
| | - Mara Fattah
- International Institute for Nanotechnology, Department of Chemistry, Chemistry of Life Processes Institute, Northwestern University, Evanston, 60208, IL, USA
| | - Shivangi Agarwal
- Grove Biopharma, Inc, 1375 W. Fulton St., Ste. 650, Chicago, 60558, IL, USA
| | - Michael P Vincent
- Grove Biopharma, Inc, 1375 W. Fulton St., Ste. 650, Chicago, 60558, IL, USA
| | - Marina Buyanova
- Grove Biopharma, Inc, 1375 W. Fulton St., Ste. 650, Chicago, 60558, IL, USA
| | - Paul A Bertin
- Grove Biopharma, Inc, 1375 W. Fulton St., Ste. 650, Chicago, 60558, IL, USA
| | - Ralph Mazitschek
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, 02142, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Monica Olvera de la Cruz
- Department of Materials Science & Engineering, Robert R. McCormick School of Engineering and Applied Science, Center for Computation and Theory of Soft Materials, Northwestern University, Evanston, 60208, IL, USA
| | - Delinda A Johnson
- School of Pharmacy, University of Wisconsin, Madison, 57305, WI, USA
| | - Jeffrey A Johnson
- School of Pharmacy, University of Wisconsin, Madison, 57305, WI, USA
| | - Nathan C Gianneschi
- Departments of Chemistry, Materials Science & Engineering, Biomedical Engineering, Pharmacology, Simpson Querrey Institute, Chemistry of Life Processes Institute, Lurie Cancer Center, International Institute for Nanotechnology, Northwestern University, Evanston, 60208, IL, USA
| |
Collapse
|
47
|
Xu X, Shan M, Chu C, Bie S, Wang H, Cai S. Polysaccharides from Polygonatum kingianum Collett & Hemsl ameliorated fatigue by regulating NRF2/HO-1/NQO1 and AMPK/PGC-1α/TFAM signaling pathways, and gut microbiota. Int J Biol Macromol 2024; 266:131440. [PMID: 38593898 DOI: 10.1016/j.ijbiomac.2024.131440] [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: 01/23/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
Polygonatum kingianum Coll & Hemsl is an important Chinese medicine used for enhancing physical function and anti-fatigue, and polysaccharides (PKPs) are considered as the main bioactive components. However, the mechanisms through which PKPs exert their anti-fatigue effects are not fully understood. This study aimed more comprehensively to explore the anti-fatigue mechanisms of PKPs, focusing on metabolism, protein expression, and gut flora, by using exhaustive swimming experiments in mice. Results showed a significant increase in the exhaustive swimming time of the mice treated with PKPs, especially in the high-dose group (200 mg/kg/day). Further studies showed that PKPs remarkably improves several fatigue-related physiological indices. Additionally, 16S rRNA sequence analysis showed that PKPs increased antioxidant bacteria (e.g., g_norank_f_Muribaculaceae) and the production of short-chain fatty acids (SCFAs), while reducing the abundance of harmful bacteria (e.g., g_Escherichia-Shigella and g_Helicobacter). PKPs also mitigated oxidative stress through activating the NRF2/HO-1 signaling pathway, and promoted energy metabolism by upregulating the expression of AMPK/PGC-1α/TFAM signaling pathway proteins. This research may offer theoretical support for incorporating PKPs as a novel dietary supplement in functional foods targeting anti-fatigue properties.
Collapse
Affiliation(s)
- Xingrui Xu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China; Yunnan Engineering Research Center for Fruit & Vegetable Products, Kunming, Yunnan Province 650500, People's Republic of China; International Green Food Processing Research and Development Center of Kunming City, Kunming, Yunnan Province 650500, People's Republic of China
| | - Meimei Shan
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China; Yunnan Engineering Research Center for Fruit & Vegetable Products, Kunming, Yunnan Province 650500, People's Republic of China; International Green Food Processing Research and Development Center of Kunming City, Kunming, Yunnan Province 650500, People's Republic of China
| | - Chuanqi Chu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China; Yunnan Engineering Research Center for Fruit & Vegetable Products, Kunming, Yunnan Province 650500, People's Republic of China; International Green Food Processing Research and Development Center of Kunming City, Kunming, Yunnan Province 650500, People's Republic of China
| | - Shenke Bie
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China; Yunnan Engineering Research Center for Fruit & Vegetable Products, Kunming, Yunnan Province 650500, People's Republic of China; International Green Food Processing Research and Development Center of Kunming City, Kunming, Yunnan Province 650500, People's Republic of China
| | - He Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province 212100, People's Republic of China
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China; Yunnan Engineering Research Center for Fruit & Vegetable Products, Kunming, Yunnan Province 650500, People's Republic of China; International Green Food Processing Research and Development Center of Kunming City, Kunming, Yunnan Province 650500, People's Republic of China.
| |
Collapse
|
48
|
Zytner P, Kutschbach A, Gong W, Ohse VA, Taudte L, Kipp AP, Klotz LO, Priebs J, Steinbrenner H. Selenium-Enriched E. coli Bacteria Mitigate the Age-Associated Degeneration of Cholinergic Neurons in C. elegans. Antioxidants (Basel) 2024; 13:492. [PMID: 38671939 PMCID: PMC11047679 DOI: 10.3390/antiox13040492] [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: 03/27/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Selenium (Se) is an essential trace element for humans and animals, but high-dose supplementation with Se compounds, most notably selenite, may exert cytotoxic and other adverse effects. On the other hand, bacteria, including Escherichia coli (E. coli), are capable of reducing selenite to red elemental Se that may serve as a safer Se source. Here, we examined how a diet of Se-enriched E. coli bacteria affected vital parameters and age-associated neurodegeneration in the model organism Caenorhabditis elegans (C. elegans). The growth of E. coli OP50 for 48 h in medium supplemented with 1 mM sodium selenite resulted in reddening of the bacterial culture, accompanied by Se accumulation in the bacteria. Compared to nematodes supplied with the standard E. coli OP50 diet, the worms fed on Se-enriched bacteria were smaller and slimmer, even though their food intake was not diminished. Nevertheless, given the choice, the nematodes preferred the standard diet. The fecundity of the worms was not affected by the Se-enriched bacteria, even though the production of progeny was somewhat delayed. The levels of the Se-binding protein SEMO-1, which serves as a Se buffer in C. elegans, were elevated in the group fed on Se-enriched bacteria. The occurrence of knots and ruptures within the axons of cholinergic neurons was lowered in aged nematodes provided with Se-enriched bacteria. In conclusion, C. elegans fed on Se-enriched E. coli showed less age-associated neurodegeneration, as compared to nematodes supplied with the standard diet.
Collapse
Affiliation(s)
- Palina Zytner
- Institute of Nutritional Sciences, Nutrigenomics Section, Friedrich Schiller University Jena, D-07743 Jena, Germany; (P.Z.); (A.K.); (W.G.); (V.A.O.); (L.-O.K.)
| | - Anne Kutschbach
- Institute of Nutritional Sciences, Nutrigenomics Section, Friedrich Schiller University Jena, D-07743 Jena, Germany; (P.Z.); (A.K.); (W.G.); (V.A.O.); (L.-O.K.)
| | - Weiye Gong
- Institute of Nutritional Sciences, Nutrigenomics Section, Friedrich Schiller University Jena, D-07743 Jena, Germany; (P.Z.); (A.K.); (W.G.); (V.A.O.); (L.-O.K.)
| | - Verena Alexia Ohse
- Institute of Nutritional Sciences, Nutrigenomics Section, Friedrich Schiller University Jena, D-07743 Jena, Germany; (P.Z.); (A.K.); (W.G.); (V.A.O.); (L.-O.K.)
| | - Laura Taudte
- Institute of Nutritional Sciences, Department of Nutritional Physiology, Friedrich Schiller University Jena, D-07743 Jena, Germany; (L.T.); (A.P.K.)
| | - Anna Patricia Kipp
- Institute of Nutritional Sciences, Department of Nutritional Physiology, Friedrich Schiller University Jena, D-07743 Jena, Germany; (L.T.); (A.P.K.)
| | - Lars-Oliver Klotz
- Institute of Nutritional Sciences, Nutrigenomics Section, Friedrich Schiller University Jena, D-07743 Jena, Germany; (P.Z.); (A.K.); (W.G.); (V.A.O.); (L.-O.K.)
| | - Josephine Priebs
- Institute of Nutritional Sciences, Nutrigenomics Section, Friedrich Schiller University Jena, D-07743 Jena, Germany; (P.Z.); (A.K.); (W.G.); (V.A.O.); (L.-O.K.)
| | - Holger Steinbrenner
- Institute of Nutritional Sciences, Nutrigenomics Section, Friedrich Schiller University Jena, D-07743 Jena, Germany; (P.Z.); (A.K.); (W.G.); (V.A.O.); (L.-O.K.)
| |
Collapse
|
49
|
Song W, Zhang L, Cui X, Wang R, Ma J, Xu Y, Jin Y, Wang D, Lu Z. Nobiletin alleviates cisplatin-induced ototoxicity via activating autophagy and inhibiting NRF2/GPX4-mediated ferroptosis. Sci Rep 2024; 14:7889. [PMID: 38570541 PMCID: PMC10991266 DOI: 10.1038/s41598-024-55614-4] [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: 10/15/2023] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Nobiletin, a citrus polymethoxy flavonoid with antiapoptotic and antioxidative properties, could safeguard against cisplatin-induced nephrotoxicity and neurotoxicity. Cisplatin, as the pioneer of anti-cancer drug, the severe ototoxicity limits its clinical applications, while the effect of nobiletin on cisplatin-induced ototoxicity has not been identified. The current study investigated the alleviating effect of nobiletin on cisplatin-induced ototoxicity and the underlying mechanisms. Apoptosis and ROS formation were evaluated using the CCK-8 assay, Western blotting, and immunofluorescence, indicating that nobiletin attenuated cisplatin-induced apoptosis and oxidative stress. LC3B and SQSTM1/p62 were determined by Western blotting, qPCR, and immunofluorescence, indicating that nobiletin significantly activated autophagy. Nobiletin promoted the nuclear translocation of NRF2 and the transcription of its target genes, including Hmox1, Nqo1, and ferroptosis markers (Gpx4, Slc7a11, Fth, and Ftl), thereby inhibiting ferroptosis. Furthermore, RNA sequencing analysis verified that autophagy, ferroptosis, and the NRF2 signaling pathway served as crucial points for the protection of nobiletin against ototoxicity caused by cisplatin. Collectively, these results indicated, for the first time, that nobiletin alleviated cisplatin-elicited ototoxicity through suppressing apoptosis and oxidative stress, which were attributed to the activation of autophagy and the inhibition of NRF2/GPX4-mediated ferroptosis. Our study suggested that nobiletin could be a prospective agent for preventing cisplatin-induced hearing loss.
Collapse
Affiliation(s)
- Wenao Song
- Department of Clinical Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China
| | - Li Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Xiaolin Cui
- Department of Clinical Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China
| | - Rongrong Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China
| | - Jingyu Ma
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yue Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China
| | - Yan Jin
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Dawei Wang
- Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China.
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
50
|
Zurawska G, Jończy A, Niklewicz M, Sas Z, Rumieńczyk I, Kulecka M, Piwocka K, Rygiel TP, Mikula M, Mleczko-Sanecka K. Iron-triggered signaling via ETS1 and the p38/JNK MAPK pathway regulates Bmp6 expression. Am J Hematol 2024; 99:543-554. [PMID: 38293789 DOI: 10.1002/ajh.27223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/13/2023] [Accepted: 01/01/2024] [Indexed: 02/01/2024]
Abstract
BMP6 is an iron-sensing cytokine whose transcription in liver sinusoidal endothelial cells (LSECs) is enhanced by high iron levels, a step that precedes the induction of the iron-regulatory hormone hepcidin. While several reports suggested a cell-autonomous induction of Bmp6 by iron-triggered signals, likely via sensing of oxidative stress by the transcription factor NRF2, other studies proposed the dominant role of a paracrine yet unidentified signal released by iron-loaded hepatocytes. To further explore the mechanisms of Bmp6 transcriptional regulation, we used female mice aged 10-11 months, which are characterized by hepatocytic but not LSEC iron accumulation, and no evidence of systemic iron overload. We found that LSECs of aged mice exhibit increased Bmp6 mRNA levels as compared to young controls, but do not show a transcriptional signature characteristic of activated NFR2-mediated signaling in FACS-sorted LSECs. We further observed that primary murine LSECs derived from both wild-type and NRF2 knock-out mice induce Bmp6 expression in response to iron exposure. By analyzing transcriptomic data of FACS-sorted LSECs from aged versus young mice, as well as early after iron citrate injections, we identified ETS1 as a candidate transcription factor involved in Bmp6 transcriptional regulation. By performing siRNA-mediated knockdown, small-molecule treatments, and chromatin immunoprecipitation in primary LSECs, we show that Bmp6 transcription is regulated by iron via ETS1 and p38/JNK MAP kinase-mediated signaling, at least in part independently of NRF2. Thereby, these findings identify the new components of LSEC iron sensing machinery broadly associated with cellular stress responses.
Collapse
Affiliation(s)
- Gabriela Zurawska
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Aneta Jończy
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Marta Niklewicz
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Zuzanna Sas
- Medical University of Warsaw, Warsaw, Poland
| | - Izabela Rumieńczyk
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maria Kulecka
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Tomasz P Rygiel
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Michal Mikula
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | |
Collapse
|