1
|
Yu Y, Yu T, Liu K, Li Y, Luan Y, Yang T, Li W, Cong H, Wu X. Perimenopausal depression: Targeting inflammation and oxidative stress (Review). Mol Med Rep 2025; 31:161. [PMID: 40211702 DOI: 10.3892/mmr.2025.13526] [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: 01/15/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025] Open
Abstract
Depressive disorder is a highly disabling condition that affects more than 300 million individuals worldwide, with women affected at a higher rate than men. With the aging of the population, the incidence of perimenopausal depression has risen markedly, seriously jeopardizing women's physical and mental health. Symptoms of perimenopausal depression include feelings of depression, stress, anxiety and endocrine dysfunctions, particularly hypogonadism and senescence. During perimenopause, estrogen and progesterone levels fluctuate erratically, adding to the risk of developing depression associated with perimenopause. As a result of these hormonal changes, proinflammatory mediators are produced and oxidative stress is induced, which finally leads to progressive neuronal damage. The present study mainly reviewed roles of neuroinflammation in perimenopausal depression and explained potential anti‑inflammatory and anti‑oxidative stress mechanisms for clinically effective therapeutic treatment.
Collapse
Affiliation(s)
- Yang Yu
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Tianyang Yu
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Kaili Liu
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yushuai Li
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yifeng Luan
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Tianyi Yang
- Department of Respiratory Medicine, Heilongjiang Academy of Sciences of Traditional Chinese Medicine, Harbin, Heilongjiang 150036, P.R. China
| | - Wenzhong Li
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Huifang Cong
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Xiuhong Wu
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
2
|
Kim DU, Kweon B, Oh JY, Noh GR, Lim Y, Yu J, Kim MJ, Kim DG, Park SJ, Bae GS. Curcumin ameliorates cerulein‑induced chronic pancreatitis through Nrf‑2/HO‑1 signaling. Mol Med Rep 2025; 31:136. [PMID: 40145554 PMCID: PMC11963747 DOI: 10.3892/mmr.2025.13501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic pancreatitis (CP) is an invasive inflammatory disorder characterized by endocrine and exocrine dysfunction. There are currently no effective drugs for the treatment of CP. The present study investigated whether curcumin improves cerulein‑induced CP fibrosis in a mouse model and pancreatic stellate cells (PSCs). The CP mouse model was established by intraperitoneally injecting cerulein (50 µg/kg) for 3 weeks (six times at 1 h intervals/day; 4 days/week). To investigate the effects of curcumin, dimethyl sulfoxide or curcumin was injected intraperitoneally 1 h before the first daily injection of cerulein. To determine the severity of CP, the pancreas was harvested 24 h after the last cerulein injection for histological examination and assessment of PSC activation and collagen deposition. Additionally, levels of the nuclear factor erythroid 2‑related factor 2 (Nrf2) and heme oxygenase‑1 (HO‑1) were evaluated to determine the mechanism underlying the anti‑fibrotic effect of curcumin in PSCs. Curcumin improved pancreatic injury associated with CP by inhibiting PSC activation and collagen deposition. Moreover, curcumin increased HO‑1 expression levels via the activation of Nrf2 in PSCs, which suppressed the activation of PSCs. In conclusion, the present results suggest that curcumin can ameliorate pancreatic fibrosis induced by repetitive cerulein challenges via the induction of HO‑1 and is a beneficial agent for the treatment of CP.
Collapse
Affiliation(s)
- Dong-Uk Kim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Bitna Kweon
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jin-Young Oh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Gyeong-Ran Noh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yebin Lim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jihyun Yu
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Myoung-Jin Kim
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Dong-Gu Kim
- Department of Herbology, College of Korean Medicine, Dong-Eui University, Busan, Gyeongnam 47887, Republic of Korea
| | - Sung-Joo Park
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Gi-Sang Bae
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan-daero, Iksan 54538, Republic of Korea
| |
Collapse
|
3
|
Ren XM, Wang J, Zhao F, Zhang P, Zhang Z, Yang Z, He H, Xu Z, Huang B, Pan X. 6:2 fluorotelomer sulfonate as a safer alternative to PFOS: Comparative cytotoxicity and oxidative stress mechanisms in pancreatic β-cells (INS-1 model). Toxicol In Vitro 2025; 105:106034. [PMID: 39978700 DOI: 10.1016/j.tiv.2025.106034] [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: 12/12/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Previous studies suggest that 6:2 fluorotelomer sulfonate (6:2 FTSA) exhibits lower hepatotoxicity and reduced reproductive and developmental toxicity compared to perfluorooctane sulfonate (PFOS), indicating it may offer a safer alternative. This study aimed to investigate whether 6:2 FTSA is safer than PFOS in terms of its cytotoxic effects on pancreatic β-cells. Using rat insulinoma cells (INS-1) as a model of pancreatic β-cells, we compared the effects of 6:2 FTSA and PFOS in both their acid (6:2 FTSA-H, PFOS-H) and potassium salt forms (6:2 FTSA-K, PFOS-K) on cell viability through Cell Counting Kit-8 (CCK-8) assays, Trypan Blue staining, and apoptosis assays. Results indicated that 6:2 FTSA was less toxic to INS-1 cells than PFOS (6:2 FTSA-H < PFOS-H; 6:2 FTSA-K < PFOS-K), the LOECs of 6:2 FTSA-H, 6:2 FTSA-K, PFOS-H, and PFOS-K were 150 μM, 150 μM, 20 μM, and 10 μM under FBS free conditions, respectively. To further explore whether these compounds induce cell death via oxidative stress, we measured intracellular reactive oxygen species (ROS) levels, superoxide dismutase (SOD) activity, and malondialdehyde (MDA) levels. All four compounds induced oxidative stress in INS-1 cells, with oxidative stress levels corresponding to cytotoxicity, suggesting β-cell death may occur via an oxidative stress mechanism. In conclusion, this study supports the notion that 6:2 FTSA is a safer alternative to PFOS, particularly regarding risks related to pancreatic β-cell cytotoxic effects. While the in vitro experiments in this study provide valuable preliminary information on the compounds' effects on cells and their mechanisms, they cannot fully capture the complexity of the in vivo environment. Therefore, future research should include in vivo experiments to validate the findings from the in vitro studies and comprehensively evaluate the actual effects of the compounds in living organisms.
Collapse
Affiliation(s)
- Xiao-Min Ren
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Jianying Wang
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Fenqing Zhao
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Pingping Zhang
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Zhenghuan Zhang
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Zhongneng Yang
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Huan He
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Zhixiang Xu
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Bin Huang
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China
| | - Xuejun Pan
- Faculty of Environmental Science and Engineering. Kunming University of Science and Technology, Kunming 650500, China.
| |
Collapse
|
4
|
Chen Y, Gong Y, Li D, Feng W, Chen Y, Zhao T, Yang L, Mao G, Wu X. Dinotefuran exposure induces immunotoxicity in zebrafish embryos. Comp Biochem Physiol C Toxicol Pharmacol 2025; 295:110206. [PMID: 40222697 DOI: 10.1016/j.cbpc.2025.110206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/19/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
As a typical neonicotinoid insecticide, dinotefuran (DIN) has immunotoxicity, but its immunotoxicity effects on aquatic organisms and its molecular mechanisms are not known. In this study, zebrafish embryos were used as model to reveal its immunotoxicity and its mechanism from the perspective of inflammatory response. Zebrafish larvae were exposed to DIN at environmentally relevant concentrations (0, 2, 200, and 2000 mg/L) for 120 h, followed by comprehensive analyses of immune cell populations, immune marker activities, oxidative stress levels, and gene expression profiles. The results showed that the number of neutrophils and macrophages, two types of innate immune cells, was significantly reduced, and the activities of the immune markers, lysozyme (LYS), immunoglobulin M (IgM), and complement protein C3, were significantly inhibited. The level of oxidative stress in zebrafish larvae was significantly elevated and antioxidant enzyme activities were markedly inhibited in a dose-dependent manner after exposure to DIN. In this case, pro-inflammatory cytokines such as TNF-α, IL-6 and IL-1β were also abnormally expressed, and the expression levels of the genes related to the NF-κB pathway and the JNK-STAT pathway, tlr4a, myd88, nf-κb p65, jak1, jak2, and stat3, were elevated. The expression levels of genes related to the antioxidant signaling pathway Nrf2-keap1 signaling pathway were suppressed. Taken together, our results suggest that DIN exposure causes damage to the immune system of zebrafish embryos-larvae, generating oxidative stress and inflammatory responses that lead to immunotoxicity.
Collapse
Affiliation(s)
- Yunshuo Chen
- School of the Environment and Safety Engineering, School of Emergency Management, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China
| | - Yacan Gong
- School of the Environment and Safety Engineering, School of Emergency Management, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China
| | - Dan Li
- School of the Environment and Safety Engineering, School of Emergency Management, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China
| | - Weiwei Feng
- School of the Environment and Safety Engineering, School of Emergency Management, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China; Institute of environmental health and ecological safety, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China
| | - Yao Chen
- School of the Environment and Safety Engineering, School of Emergency Management, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China; Institute of environmental health and ecological safety, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China
| | - Ting Zhao
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China
| | - Liuqing Yang
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China
| | - Guanghua Mao
- School of the Environment and Safety Engineering, School of Emergency Management, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China.
| | - Xiangyang Wu
- School of the Environment and Safety Engineering, School of Emergency Management, Jiangsu University, 301 Xuefu Rd, Zhenjiang 212013, China.
| |
Collapse
|
5
|
Yu T, Ding C, Peng J, Liang G, Tang Y, Zhao J, Li Z. SIRT7-mediated NRF2 deacetylation promotes antioxidant response and protects against chemodrug-induced liver injury. Cell Death Dis 2025; 16:232. [PMID: 40169535 PMCID: PMC11961749 DOI: 10.1038/s41419-025-07549-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025]
Abstract
NRF2 has been recognized as a central hub that neutralizes ROS and restores intracellular redox balance. In addition to KEAP1 mediated ubiquitin-proteasome degradation, post-translational modifications of NRF2 are critical for regulating its nuclear translocation and activation but precise mechanisms underly this regulation remain elusive. In this study, we found that SIRT7 was sufficient and essential for NRF2 nuclear localization and activation. Knockdown of SIRT7 significantly impaired intercellular ROS homeostasis and increased apoptosis in response to oxidative stress including chemodrug treatment. SIRT7 interacted with NRF2 and induced its deacetylation, by which inhibited binding of NRF2 to KEAP1, enhanced NRF2 protein stability and promoted its nuclear translocation. SIRT7 induced NRF2 deacetylation at K443 and K518 sites. Lysine-arginine mutations of these sites (2KR NRF2) significantly reduced KEAP1/NRF2 binding, increased NRF2 nuclear translocation and target gene expression, decreased intercellular ROS level, whereas lysine-glutamine (2KQ) mutant showed similar subcellular localization and functions with WT. Knockdown SIRT7 in hepatocyte exacerbated Oxaliplatin (Oxa) induced hepatic injury and inflammation. While AAV8-NRF2-mediated hepatic NRF2 overexpression or NRF2 agonist significantly prevented Oxa-induced elevation of ALT levels, sinusoidal dilatation and inflammation in SIRT7HKO mice. Our data thus uncovered previously unidentified role of SIRT7 in modulating NRF2 nuclear localization and activation via deacetylation. Activating SIRT7 might offer protection against chemodrug-induced liver injury.
Collapse
Affiliation(s)
- Tingzi Yu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, Hunan, China
| | - Cong Ding
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, Hunan, China
| | - Jinying Peng
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, Hunan, China
| | - Gaoshuang Liang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, Hunan, China
| | - Yongyi Tang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, Hunan, China
| | - Jinqiu Zhao
- Department of infectious disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Department of Cancer center, University of Hawaii at Manoa, Honolulu, HI, USA.
| | - Zhuan Li
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, Hunan, China.
| |
Collapse
|
6
|
Xue GM, Xue JF, Pan H, Feng H, Lei JW, Chen H, Feng WS, Li GS, Wang Y. Phenylpropanoid dimers from Chrysanthemum indicum with antioxidant effects via activating the Nrf2 signaling pathway. PHYTOCHEMISTRY 2025; 232:114372. [PMID: 39725186 DOI: 10.1016/j.phytochem.2024.114372] [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: 07/28/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Five phenylpropanoid dimers chrysanthephenyls A-E, with the 1-O-1'-type (1) and 1-O-2'-type (2-5) ether bond linking modes, were isolated from the aerial parts of Chrysanthemum indicum. Among them, chrysanthephenyls D and E were two pairs of enantiomers, and chrysanthephenyl E was further resolved into its enantiomers (+)-chrysanthephenyl E and (-)-chrysanthephenyl E via chiral-phase HPLC. Their structures were characterized on the basic analysis of the spectroscopic techniques (IR, HRESIMS, 1D and 2D NMR), and their absolute configurations were determined by DP4+ NMR analysis and ECD calculation method. The antioxidant activity of chrysanthephenyls A-E was determined by measuring their free radical scavenging effects using the trolox equivalent antioxidant activity (TEAC) assay, and chrysanthephenyls D and E showed 17 and 15 times better activity respectively than that of the positive control trolox. Moreover, a mechanistic study revealed that the potential antioxidant activity of chrysanthephenyl D decreased the level of reactive oxygen species (ROS) mediated via activating nuclear factor E2-related factor 2 (Nrf2) and its downstream oxidases.
Collapse
Affiliation(s)
- Gui-Min Xue
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Jin-Feng Xue
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Hao Pan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Hao Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jing-Wei Lei
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Hui Chen
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Wei-Sheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Guo-Sheng Li
- The Affiliated Taizhou Second People's Hospital of Yangzhou University, Taizhou, 225500, China.
| | - Yan Wang
- Henan Province Hospital of Traditional Chinese Medicine, No. 6, Dongfeng Road, Jinshui District, Zhengzhou City, Henan Province, 450002, China.
| |
Collapse
|
7
|
Sabeti Akbar-Abad M, Farkhondeh T, Majidpour M, Samini F, Aschner M, Alemzadeh E, Samarghandian S. The Therapeutic Role of Saffron and Its Components Mediated Through Nrf2 in Diabetes and Related Pathologies. J Med Food 2025; 28:309-324. [PMID: 40172359 DOI: 10.1089/jmf.2024.k.0135] [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: 04/04/2025] Open
Abstract
Today, diabetes is considered a growing global epidemic. In the diabetic environment, a large amount of reactive oxygen species are produced. This type of active oxygen causes severe damage to cell membranes, proteins, and DNA. Therefore, finding a solution to deal with and reduce this type of reactive oxygen is very important. One of the most effective ways to deal with oxidative damage and inflammation is the modulation of the nuclear factor erythroid 2 (Nrf2) signaling pathway. One of the useful natural substances that can be used for treatment in the signaling system is saffron. In this article, research evaluating the medicinal effects of saffron and its compounds and their mechanisms of action, especially the Nrf2 signaling pathway, have been investigated and studied. The results show that saffron and its components have the potential to treat diabetes due to their unique properties.
Collapse
Affiliation(s)
| | - Tahereh Farkhondeh
- Geriatric Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahdi Majidpour
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fariborz Samini
- Department of Neurosurgery, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Effat Alemzadeh
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Department of Physiology, School of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
8
|
Vörösházi J, Mackei M, Sebők C, Tráj P, Márton RA, Neogrády Z, Mátis G. Protective effects of baicalin against deoxynivalenol-induced oxidative and inflammatory damage in chicken-derived hepatic 3D cell cultures. Sci Rep 2025; 15:11180. [PMID: 40169826 PMCID: PMC11962109 DOI: 10.1038/s41598-025-95868-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
Deoxynivalenol (DON) is a trichothecene mycotoxin often contaminating grains used in poultry feed production and causing several adverse effects in farm animals. Therefore, it is important to investigate compounds that can be potential candidates to mitigate these effects, such as baicalin. The effects of DON and baicalin were investigated in chicken-derived 3D hepatic cell cultures, and cell viability, LDH activity, oxidative parameters (NRF-2, 8-OHdG) and inflammatory parameters (IL-6, IL-8, IFN-γ) were monitored for 24 and 48 h. Our results suggest that DON reduced cellular metabolic activity but did not prove to be cytotoxic, and baicalin was able to attenuate this adverse effect. The change in extracellular LDH activity suggests that after 48 h the cells have already started to respond to the adverse effects of the toxin and protective mechanisms were induced. Based on the measured oxidative parameters, baicalin showed antioxidant activity, but after longer exposure, our results indicate a prooxidant effect. Baicalin also had an anti-inflammatory effect based on the amount of IL-6 and IL-8, while DON exerted a dose-and time-dependent pleiotropic activity. These results suggest that DON may have an impact on cellular inflammation and oxidative homeostasis, and that baicalin could be able to alleviate these adverse effects.
Collapse
Affiliation(s)
- Júlia Vörösházi
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary.
| | - Máté Mackei
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Csilla Sebők
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Patrik Tráj
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Rege Anna Márton
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Zsuzsanna Neogrády
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Gábor Mátis
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, 1078, Hungary
| |
Collapse
|
9
|
Špaková I, Smolko L, Sabolová G, Badovská Z, Kalinová K, Madreiter-Sokolowski C, Graier WF, Mareková M, Vašková J, Rabajdová M. Selective targeting of genes regulated by zinc finger proteins in endometriosis and endometrioid adenocarcinoma by zinc niflumato complex with neocuproine. Sci Rep 2025; 15:10126. [PMID: 40128272 PMCID: PMC11933352 DOI: 10.1038/s41598-025-94249-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: 06/11/2024] [Accepted: 03/12/2025] [Indexed: 03/26/2025] Open
Abstract
Inadequate angiogenesis of endometriotic implants stimulated by the inflammatory microenvironment in the uterine region leads to the development of gynecological diseases, which significantly reduce the fertility and vitality of young women. Angiogenic processes are controlled by factors whose activities are regulated at the gene level by reactive oxygen species (ROS), hypoxia-induced factors (HIFs), and zinc-finger proteins (ZnFs) or posttranscriptionally via non-coding RNAs. The cooperation of these factors is responsible for the manifestation of pathological stimuli in the form of endometriosis of the body of the uterus, ovaries, or peritoneum, from which endometrioid carcinoma can develop. Molecules that can control gene expression by their intercalation to target DNA sequence, such as [Zn(neo)(nif)2], could prevent the hyperactivation of pro-angiogenic pathways (decrease HIF-1α, VEGF-A, TGF-β1, COX2, and ANG2/ANG1), reduce the formation of ROS, and reduce the risk of uterine neoplasticity. The NSAID-metal complex [Zn(neo)(nif)2] shows an ability to intercalate into ZNF3-7 target DNA sequence at a higher rate, which could explain its effect on genes regulated by this transcription factor. In addition, [Zn(neo)(nif)2] affects ROS production and Ca2+ level, possibly pointing to mitochondrial dysfunction as a potential cause for the described apoptosis.
Collapse
Affiliation(s)
- Ivana Špaková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Lukáš Smolko
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Gabriela Sabolová
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Zuzana Badovská
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Katarína Kalinová
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Corina Madreiter-Sokolowski
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Mária Mareková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Janka Vašková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia.
| |
Collapse
|
10
|
Lauenstein J, van de Weyer S, Alsaleh R, Wiedmer C, Buettner A, Kersch C, Schmitz-Spanke S. Exploring the Activation of the Keap1-Nrf2-ARE Pathway by PAHs in Children's Toys. Contact Dermatitis 2025. [PMID: 40088110 DOI: 10.1111/cod.14792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/03/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Children are particularly susceptible to environmental pollutants. This study assessed the skin sensitisation risk associated with polycyclic aromatic hydrocarbons (PAHs), prevalent in toys. OBJECTIVES To evaluate the skin sensitisation potential of PAHs using the KeratinoSens assay. METHODS Individual PAHs (acenaphthylene, anthracene, benzo[a]anthracene, benzo[a]pyrene (B[a]P), benzo[b]fluoranthene (B[b]F), benzo[e]pyrene, benzo[g,h,i]perylene, benzo[k]fluoranthene (B[k]F), chrysene, fluoranthene, fluorene, naphthalene, phenanthrene, pyrene and triphenylene) and ternary mixtures containing B[a]P were assessed for their ability to activate the Keap1-Nrf2-ARE pathway in human keratinocytes. The concentration addition model and additive index were used to predict and analyse mixture effects. RESULTS Among the individual PAHs, B[k]F demonstrated the most potent activation of the pathway, exhibiting a 34-fold higher potency relative to B[a]P. B[b]F, chrysene and B[a]P also exhibited significant activation, while the remaining PAHs displayed negligible or weak activation. Notably, PAH mixtures exhibited synergistic effects, except for those composed solely of potent sensitizers. CONCLUSIONS This study provides the first assessment of the skin sensitization potential of these PAHs. The findings suggest that B[k]F, B[b]F and chrysene may pose a higher risk of skin sensitisation than previously thought. Additionally, the synergistic effects observed in mixtures highlight the importance of considering combined exposures when assessing PAH exposure risk.
Collapse
Affiliation(s)
- Jonas Lauenstein
- Institute and Outpatient Clinic of Occupational, Social, and Environmental Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Simon van de Weyer
- Institute and Outpatient Clinic of Occupational, Social, and Environmental Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Rasha Alsaleh
- Institute and Outpatient Clinic of Occupational, Social, and Environmental Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Wiedmer
- Department 1-Agriculture, Ecotrophology and Landscape Development, Anhalt University of Applied Sciences, Bernburg, Germany
| | - Andrea Buettner
- Chair of Aroma and Smell Research, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Fraunhofer Institute for Process Engineering and Packaging IVV, Freising, Germany
| | - Christian Kersch
- Institute and Outpatient Clinic of Occupational, Social, and Environmental Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Simone Schmitz-Spanke
- Institute and Outpatient Clinic of Occupational, Social, and Environmental Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
11
|
Elmorsy EA, Elsisi HA, Alkhamiss AS, Alsoqih NS, Khodeir MM, Alsalloom AA, Almeman AA, Elghandour SR, Nadwa EH, Khalifa AK, Khaled BEA, Ramadan A, Kamal MM, Alsaeed TS, Alharbi MS, Abdel-Moneim AMH, Ellethy AT, Saber S. Activation of SIRT1 by SRT1720 alleviates dyslipidemia, improves insulin sensitivity and exhibits liver-protective effects in diabetic rats on a high-fat diet: New insights into the SIRT1/Nrf2/NFκB signaling pathway. Eur J Pharm Sci 2025; 206:107002. [PMID: 39778687 DOI: 10.1016/j.ejps.2025.107002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/29/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Insulin resistance and diabetes are associated with non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) conditions, which are distinguished by metabolic dysfunction, oxidative stress and inflammation. Sirtuin 1 (SIRT1), a NAD+-dependent deacetylase, is fundamental in regulating metabolic pathways, reducing inflammation, and improving antioxidant defenses. This is the first study to investigate the effects of SRT1720, a SIRT1 activator, in diabetic rats on a high-fat diet. SRT1720 significantly lowered fasting blood glucose and insulin levels and enhanced glucose tolerance and HOMA-IR and QUICKI scores, indicating increased insulin sensitivity. The treatment also reduced total cholesterol, triglycerides, and LDL levels, showing amelioration of dyslipidemia. Moreover, SRT1720 lowered markers of liver fibrosis, including TGF-β, TIMP-1, Col1a1, and hydroxyproline, and decreased inflammation by reducing NFκB activity and pro-inflammatory cytokines (TNF-α and IL-6). Furthermore, SRT1720 augmented Nrf2 activity and HO-1 levels. Consequently, the SRT1720's protective role improved liver function and histology and prolonged rats' survival. These functions were suppressed by the co-administration of the SIRT1 inhibitor EX527, confirming that the beneficial effects of SRT1720 are SIRT1-dependent. Correlation analyses uncovered that increased SIRT1 activity was strongly associated with decreased oxidative stress, inflammation, insulin resistance, and fibrosis markers. To conclude, our results find that SRT1720 represents a promising therapeutic strategy for managing Type 2 diabetes in NAFLD or NASH patients possibly through the modulation of the SIRT1/Nrf2/NFκB signaling pathwa. SRT1720 could potentially halt or reverse the progression of these conditions and associated complications and merits further investigations.
Collapse
Affiliation(s)
- Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Hossam A Elsisi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Abdullah S Alkhamiss
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Norah Suliman Alsoqih
- Department of Pediatrics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Mostafa M Khodeir
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Pathology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Abdulaziz A Alsalloom
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Ahmad A Almeman
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Sahar R Elghandour
- Department of Anatomy and Histology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Eman Hassan Nadwa
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Amira Karam Khalifa
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt; Department of Medical Pharmacology, Nahda Faculty of Medicine, Beni Suef, 62521, Egypt.
| | - Bahaa Eldin Ali Khaled
- Department of Anatomy, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Asmaa Ramadan
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Manal M Kamal
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt; Department of Physiology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Thamir Saad Alsaeed
- Department of Biology and Immunology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Mariam S Alharbi
- Department of Medicine, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | | | - Abousree T Ellethy
- Department of Basic Oral Sciences and Dental Education, Biochemistry Division, College of Dentistry, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Sameh Saber
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| |
Collapse
|
12
|
Lee SE, Park HJ, Han DH, Lim ES, Lee HB, Yoon JW, Park CO, Kim SH, Oh SH, Lee DG, Pyeon DB, Kim EY, Park SP. Paralichthys olivaceus egg extract improves porcine oocyte quality by decreasing oxidative stress. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2025; 67:325-341. [PMID: 40264539 PMCID: PMC12010222 DOI: 10.5187/jast.2024.e26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 04/24/2025]
Abstract
This study aimed to assess the influence of Paralichthys olivaceus egg extract (POEE) treatment on the maturation and development of porcine oocytes subjected to oxidative stress during in vitro maturation (IVM). POEE, notably rich in vitamin B9 (folic acid [FA]), was assessed alongside FA for antioxidant activity across various concentrations. In the 650 ppm POEE (650 POEE) group, there was a significant rise in glutathione (GSH) levels and an improved developmental rate in porcine oocytes experiencing oxidative stress during IVM. Treatment with 0.3 FA exhibited substantial reduction in ROS activity. Both 650 POEE and 0.3 FA groups demonstrated inhibited abnormal spindle organization and chromosomal misalignment, with increased blastocyst formation and decreased apoptotic cells. Treatment with 650 POEE elevated mRNA expression of development-related genes (SOX2, NANOG, and POU5F1). In conclusion, POEE effectively mitigates oxidative stress, enhances embryonic quality, and improves developmental potential in porcine oocytes on IVM.
Collapse
Affiliation(s)
- Seung-Eun Lee
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Hyo-Jin Park
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Dong-Hun Han
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Eun-Seo Lim
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Han-Bi Lee
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Jae-Wook Yoon
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Chan-Oh Park
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - So-Hee Kim
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Seung-Hwan Oh
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Do-Geon Lee
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Da-Bin Pyeon
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Eun-Young Kim
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
- Mirae Cell Bio, Seoul 04795,
Korea
| | - Se-Pill Park
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Mirae Cell Bio, Seoul 04795,
Korea
- Department of Bio Medical Informatic,
College of Applied Life Sciences, Jeju National University,
Jeju 63243, Korea
| |
Collapse
|
13
|
Arlen MT, Patterson SJ, Page MK, Liu R, Caruana V, Wilson ET, Laporte SA, Goniewicz ML, Harris CS, Eidelman DH, Baglole CJ. Cannabis vaping elicits transcriptomic and metabolomic changes involved in inflammatory, oxidative stress, and cancer pathways in human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2025; 328:L478-L496. [PMID: 39823205 DOI: 10.1152/ajplung.00131.2024] [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: 04/16/2024] [Revised: 05/27/2024] [Accepted: 01/02/2025] [Indexed: 01/19/2025] Open
Abstract
The increasing shift from cannabis smoking to cannabis vaping is largely driven by the perception that vaping to form an aerosol represents a safer alternative to smoking and is a form of consumption appealing to youth. Herein, we compared the chemical composition and receptor-mediated activity of cannabis smoke extract (CaSE) to cannabis vaping extract (CaVE) along with the biological response in human bronchial epithelial cells. Chemical analysis using HPLC and GC/MS revealed that cannabis vaping aerosol contained fewer toxicants than smoke; CaSE and CaVE contained teratogens, carcinogens, and respiratory toxicants. A bioluminescence resonance energy transfer (BRET)-based biosensor detected the receptor-mediated activity of the extracts, primarily driven by Δ9-tetrahydrocannabinol (Δ9-THC) concentration. RNA-sequencing showed both CaSE and CaVE induced similar transcriptional responses, significantly upregulating genes within pathways related to inflammation, cancer, and cellular stress. This was paralleled by downregulation of pathways related to lipid synthesis and metabolism from both CaSE and CaVE. Targeted metabolomics revealed significant changes in metabolites involved in lipid and membrane metabolism, energy production, nucleotide/DNA/RNA pathways, and oxidative stress response, suggesting potential impairment of lung epithelial cell repair and function. In addition, the upregulation of 5-hydroxymethylcytosine (5hmC) indicates epigenetic changes potentially contributing to inflammation, oxidative stress, and an increased risk of cancer. These findings challenge the notion that cannabis vaping is risk-free, highlighting an urgent need for comprehensive research into its respiratory health effects. This comparison of cannabis consumption methods offers insights that could inform public health policies and raise consumer awareness regarding the potential risks of inhaling cannabis aerosol.NEW & NOTEWORTHY Cannabis use is increasing worldwide amid broad acceptance and legalization. The prevalence of traditional smoking is diminishing in favor of vaping dry flower. This is the first study to provide initial evidence that cannabis aerosol contains carcinogenic, teratogenic, and respiratory toxicants that induce transcriptional responses in epithelial cells analogous to those from cannabis smoke, suggesting potential adverse pulmonary effects.
Collapse
Affiliation(s)
- Maddison T Arlen
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Stephanie J Patterson
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Michelle K Page
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States
| | - Rui Liu
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Vincenza Caruana
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Emily T Wilson
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Stéphane A Laporte
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Maciej L Goniewicz
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States
| | - Cory S Harris
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - David H Eidelman
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Carolyn J Baglole
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Sun H, Yue T, Hou Y, Li T, Li Z, Liu H, Zhang P. Dietary geniposide supplementation could enhance hepatic lipid metabolism, immunity, antioxidant capacity, and ammonia stress resistance in turbot ( Scophthalmus maximus). ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:458-468. [PMID: 40092351 PMCID: PMC11909448 DOI: 10.1016/j.aninu.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 03/19/2025]
Abstract
This study aimed to evaluate the effects of dietary geniposide supplementation on growth performance, lipid metabolism, health status, and ammonia stress resistance in turbot (Scophthalmus maximus). Four hundred fifty fish were randomly allocated into 5 treatments with triplicate tanks (30 fish per tank). They were hand-fed to apparent satiety for 56 d with a basal diet (GP0) or diets containing 100, 200, 400, and 800 mg/kg geniposide (termed as GP100, GP200, GP400, GP800, respectively). After the conclusion of the feeding trial, the fish were exposed to ammonia stress for 96 h. The results showed that the growth performance were not affected by geniposide (P > 0.05). Dietary supplementation with geniposide decreased crude lipid in viscera without liver, subcutaneous adipose tissue (SAT), and the liver, as well as triglyceride concentrations in plasma, the liver and SAT (P < 0.05). Dietary supplementation with 400 and 800 mg/kg geniposide significantly down-regulated lipogenesis-related gene expression, as well as fatty acid uptake-related gene expression, while significantly up-regulated triglyceride secretion-related gene expression in the liver compared with the control group (P < 0.05). The GP800 group exhibited a significant reduction in plasma malondialdehyde contents compared with the control group, while both the GP200 and GP800 groups showed a significant increase in plasma complement C3 activities (P < 0.05). Furthermore, there was a notable enhancement in plasma lysozyme and total superoxide dismutase levels in the geniposide supplemented groups compared to the control group (P < 0.05). Additionally, a significant decrease in the mRNA level of pro-inflammatory cytokine and a remarkable increase in the mRNA expression of anti-inflammatory cytokines were discovered in geniposide supplemented groups relative to the control group (P < 0.05). Cumulative survival rates after ammonia stress in the GP400 and GP800 groups were statistically higher than that in the control group (P < 0.05). In conclusion, dietary geniposide supplementation could reduce lipid deposition in turbot by regulating lipid metabolism and transportation, and remarkably enhance immunity, antioxidant ability, and resistance to ammonia stress in turbot. Based on the quadratic regression analyses, the optimal concentrations of geniposide were estimated to be 545.21 to 668.41 mg/kg feed.
Collapse
Affiliation(s)
- Haoran Sun
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Tongtong Yue
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Yuqing Hou
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Tao Li
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Zhi Li
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Shijiazhuang 050024, China
- Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang 050024, China
| | - Haiyan Liu
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Shijiazhuang 050024, China
- Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang 050024, China
| | - Peiyu Zhang
- Laboratory of Aquatic Animal Nutrition and Ecology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Shijiazhuang 050024, China
- Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang 050024, China
| |
Collapse
|
15
|
Reilly NA, Dekkers KF, Molenaar J, Arumugam S, Kuipers TB, Ariyurek Y, Hoeksema MA, Jukema JW, Heijmans BT. EPA Induces an Anti-Inflammatory Transcriptome in T Cells, Implicating a Triglyceride-Independent Pathway in Cardiovascular Risk Reduction. JACC Basic Transl Sci 2025; 10:383-395. [PMID: 40139879 PMCID: PMC12013851 DOI: 10.1016/j.jacbts.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 03/29/2025]
Abstract
Twice-daily intake of purified eicosapentaenoic acid (EPA) reduces atherosclerotic cardiovascular disease risk in patients with high triglycerides, but its exact mechanism remains unclear. We exposed non-activated CD4+ T cells to 100μM EPA, oleic acid, palmitic acid, or control, and conducted RNA and ATAC-sequencing after 48 hours. EPA exposure downregulated immune response-related genes like HLA-DRA, CD69, and IL2RA, and upregulated oxidative stress prevention genes like NQO1. Transcription factor footprinting showed decreased GATA3 and PU.1, and increased REV-ERB. These effects were specific to EPA, suggesting it induces an anti-inflammatory transcriptomic landscape in CD4+ T cells, contributing to its observed cardiovascular benefits.
Collapse
Affiliation(s)
- Nathalie A Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Koen F Dekkers
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeroen Molenaar
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Sinthuja Arumugam
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas B Kuipers
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Yavuz Ariyurek
- Leiden Genome Technology Center, Department of Human Genetics, Leiden University Medical Center, the Netherlands
| | - Marten A Hoeksema
- Department of Medical Biochemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
16
|
Singh AA, Katiyar S, Song M. Phytochemicals Targeting BDNF Signaling for Treating Neurological Disorders. Brain Sci 2025; 15:252. [PMID: 40149774 PMCID: PMC11939912 DOI: 10.3390/brainsci15030252] [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: 01/23/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Neurological disorders are defined by a deterioration or disruption of the nervous system's structure and function. These diseases, which include multiple sclerosis, Alzheimer's disease, Parkinson's disease, Huntington's disease, and schizophrenia, are caused by intricate pathological processes that include excitotoxicity, neuroinflammation, oxidative stress, genetic mutations, and compromised neurotrophic signaling. Although current pharmaceutical treatments relieve symptoms, their long-term efficacy is limited due to adverse side effects and weak neuroprotective properties. However, when combined with other neuroprotective drugs or adjunct therapy, they may offer additional benefits and improve treatment outcomes. Phytochemicals have emerged as attractive therapeutic agents due to their ability to regulate essential neurotrophic pathways, especially the brain-derived neurotrophic factor (BDNF) signaling cascade. BDNF is an important target for neurodegenerative disease (ND) treatment since it regulates neuronal survival, synaptic plasticity, neurogenesis, and neuroprotection. This review emphasizes the molecular pathways through which various phytochemicals-such as flavonoids, terpenoids, alkaloids, and phenolic compounds-stimulate BDNF expression and modulate its downstream signaling pathways, including GSK-3β, MAPK/ERK, PI3K/Akt/mTOR, CREB, and Wnt/β-catenin. This paper also highlights how phytochemical combinations may interact to enhance BDNF activity, offering new therapeutic options for ND treatment. Despite their potential for neuroprotection, phytochemicals face challenges related to pharmacokinetics, blood-brain barrier (BBB) permeability, and absorption, highlighting the need for further research into combination therapies and improved formulations. Clinical assessment and mechanistic understanding of BDNF-targeted phytotherapy should be the main goals of future studies. The therapeutic efficacy of natural compounds in regulating neurotrophic signaling is highlighted in this review, providing a viable approach to the prevention and treatment of NDs.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Shweta Katiyar
- Department of Botany, SBN Government PG College, Barwani 451551, MP, India;
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
17
|
Goodin DS. The epidemiology, pathology and pathogenesis of MS: Therapeutic implications. Neurotherapeutics 2025:e00539. [PMID: 40021419 DOI: 10.1016/j.neurot.2025.e00539] [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: 11/02/2024] [Revised: 12/30/2024] [Accepted: 01/22/2025] [Indexed: 03/03/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic, and potentially disabling, inflammatory disease of the central nervous system (CNS). MS is generally characterized by recurrent, and self-limited, episodes of neurological dysfunction, which occur unpredictably and often result in multifocal tissue injury within the CNS. Currently, women are affected two to three times as often as men although this may not have been the case during earlier Time-Periods. The pathogenesis of MS is known to involve both critical genetic and environmental mechanisms. Nevertheless, in addition to these two mechanisms, disease-pathogenesis also involves a "truly" random event. Indeed, it is this random mechanism, which is responsible for the currently-observed (and increasing) excess of women among patients with MS. This review summarizes the current state of knowledge regarding the pathogenesis of MS (includong its epidemiology, pathology, and genetics) and considers the therapeutic implications that these pathogenetic mechanisms have both for our currently available therapies as well as for the possible therapeutic approaches to the management of this potentially disabling condition in the future.
Collapse
Affiliation(s)
- Douglas S Goodin
- University of California, San Francisco and the San Francisco VA Medical Center, 675 Nelson Rising Lane, Suite #221D, San Francisco, CA 94158, USA.
| |
Collapse
|
18
|
Wayal V, Wang SD, Hsieh CC. Novel bioactive peptides alleviate Western diet-induced MAFLD in C57BL/6J mice by inhibiting NLRP3 inflammasome activation and pyroptosis via TLR4/NF-κB and Keap1/Nrf2/HO-1 signaling pathways. Int Immunopharmacol 2025; 148:114177. [PMID: 39874846 DOI: 10.1016/j.intimp.2025.114177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
Metabolic-associated fatty liver disease (MAFLD) has emerged as a leading chronic liver disease. This condition is characterized by an abnormal accumulation of fat within liver and can progress from simple steatosis to more severe stages involving chronic inflammation and oxidative stress. In this study, we investigated the potential therapeutic effects and underlying mechanism of novel bioactive peptides (EWYF and EWFY) on Western diet-induced MAFLD in C57BL/6J mice. Mice fed a normal chow diet (ND group) and Western diet (WD and treatment groups) for 8 weeks. Treatment groups received EWYF and EWFY peptides in low (10 mg/kg/day) and high (50 mg/kg/day) doses were divided into four groups: EWYF10, EWYF50, EWFY10, and EWFY50. Western diet-induced body weight gain and increased liver weight along with visceral adiposity, which were markedly reversed by bioactive peptides in a dose-dependent manner. Additionally, bioactive peptides significantly reduced hepatic steatosis, liver injury and proinflammatory response. Western diet-induced glucose tolerance and insulin resistance, whereas bioactive peptides significantly improved glucose tolerance and insulin sensitivity. Persistent intake of Western diet triggered chronic inflammation and severe oxidative stress, which were significantly alleviated by bioactive peptides treatment via inhibiting NOD-like receptor protein 3 (NLRP3) inflammasome activation and mitigated pyroptosis by modulating TLR4/NF-κB and Keap1/Nrf2/HO-1 signaling pathways. Furthermore, molecular docking studies suggest that EWYF and EWFY act as fructokinase antagonists and TLR4 inhibitors, which potentially alleviates Western diet-induced MAFLD. Collectively, these findings highlight EWYF and EWFY as promising candidates for MAFLD treatment due to their potent antioxidant and anti-inflammatory properties via specific molecular inhibition.
Collapse
Affiliation(s)
- Vipul Wayal
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407224 Taiwan
| | - Shulhn-Der Wang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404328 Taiwan; Green Abiotechnology Co. Ltd, Taichung 429010 Taiwan
| | - Chang-Chi Hsieh
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407224 Taiwan.
| |
Collapse
|
19
|
Liu R, Gao L, Zhang X, Ge P, Wang L, Zhou K, Yang C, Wang L, Song L. The Regulation of γ-Aminobutyric Acid on Antioxidative Defense Response of Pacific Oyster upon High-Temperature Stress. Antioxidants (Basel) 2025; 14:222. [PMID: 40002408 PMCID: PMC11852102 DOI: 10.3390/antiox14020222] [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: 12/23/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Recent studies have found that high temperatures cause oxidative stress and even mass mortality in Pacific oysters (Crassostrea gigas). The role of γ-aminobutyric acid (GABA) in improving antioxidative defense in aquatic animals is increasingly of interest. In the present study, the oxidative stress of Pacific oysters to high-temperature stress was examined, and the regulation of GABA on the antioxidative defense was further investigated. Following 6 h of exposure to 28 °C seawater, a significant increase in the mRNA expression levels of nuclear factor-E2-related factor 2 (Nrf2), superoxide dismutase (SOD), and catalase (CAT), as well as the activities of SOD and CAT, was observed in the gill, compared to those at 0 h. An increase of glutamate decarboxylase (GAD), GABA receptor (GABAAR-α and GABABR-B) mRNA levels, and GABA contents were also detected after 28 °C exposure compared to those at 0 h. Furthermore, the activities and mRNA expression levels of SOD and CAT were significantly upregulated after GABA treatment, while decreased after either GAD inhibitor or GABA receptor inhibitor treatment under high-temperature stress. Meanwhile, the enhanced effects of GABA on antioxidant enzyme activities were reduced when Nrf2 was inhibited by ML385, accompanied by an increase in MDA content. After high-temperature stress, compared with the GABA treatment group, the activities and mRNA expression levels of SOD and CAT were significantly upregulated by GSK-3β inhibitor treatment. Meanwhile, the elevation of antioxidant enzyme activities by GABA was attenuated by the AKT inhibitor treatment. Collectively, GABA first activated GABA receptors under high-temperature stress and then increased the activities of SOD and CAT and reduced MDA content by AKT/GSK-3β and Nrf2 pathways to protect the oysters against oxidative damage upon stress. The present results offer new insights for understanding the regulation mechanisms of antioxidative defense by the neuroendocrine system in molluscs.
Collapse
Affiliation(s)
- Ranyang Liu
- College of Life Sciences, Liaoning Normal University, Dalian 116029, China; (R.L.); (L.S.)
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Lei Gao
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Xueshu Zhang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Pingan Ge
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Ling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Keli Zhou
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Chuanyan Yang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- College of Life Sciences, Liaoning Normal University, Dalian 116029, China; (R.L.); (L.S.)
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Linsheng Song
- College of Life Sciences, Liaoning Normal University, Dalian 116029, China; (R.L.); (L.S.)
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (L.G.); (X.Z.); (P.G.); (L.W.); (K.Z.); (C.Y.)
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| |
Collapse
|
20
|
Ma X, Zhao H, Song JK, Zhang Z, Gao CJ, Luo Y, Ding XJ, Xue TT, Zhang Y, Zhang MJ, Zhou M, Wang RP, Kuai L, Li B. Retracing from Outcomes to Causes: NRF2-Driven GSTA4 Transcriptional Regulation Controls Chronic Inflammation and Oxidative Stress in Atopic Dermatitis Recurrence. J Invest Dermatol 2025; 145:334-345.e11. [PMID: 38879155 DOI: 10.1016/j.jid.2024.05.018] [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: 01/31/2024] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 07/14/2024]
Abstract
Atopic dermatitis (AD), a chronic and recurrent inflammatory skin disorder, presents a high incidence and imposes a substantial economic burden. Preventing its recurrence remains a significant challenge in dermatological therapy owing to poorly understood underlying mechanisms. In our study, we adopted a strategy of tracing the mechanisms of recurrence from clinical outcomes. We developed a mouse model of recurrent AD and applied clinically validated treatment regimens. Transcriptomic analyses revealed a pronounced enrichment in the glutathione metabolic pathway in the treated group. Through integrated bioinformatics and in vivo validation, we identified glutathione S-transferase alpha 4 (GSTA4) as a pivotal mediator in AD recurrence. Immunohistochemical analysis demonstrated decreased GSTA4 expression in lesions from patients with AD. Functionally, in vitro overexpression of GSTA4 significantly curtailed AD-like inflammatory responses and ROS production. Moreover, we discovered that NRF2 transcriptional activity regulates GSTA4 expression and function. Our treatment notably augmented NRF2-mediated GSTA4 transcription, yielding pronounced anti-inflammatory and ROS-neutralizing effects. Conclusively, our findings implicate GSTA4 as a critical factor in the recurrence of AD, particularly in the context of oxidative stress and chronic inflammation. Targeting the NRF2-GSTA4 axis emerges as a promising anti-inflammatory and antioxidative strategy for preventing AD recurrence.
Collapse
Affiliation(s)
- Xin Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hang Zhao
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jian-Kun Song
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China; Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Zhan Zhang
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Chun-Jie Gao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China; Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Ying Luo
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Jie Ding
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ting-Ting Xue
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ying Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China; Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Meng-Jie Zhang
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Mi Zhou
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Rui-Ping Wang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China; Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Le Kuai
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China.
| | - Bin Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
21
|
Kim E, Cha D, Jang SJ, Cho J, Moh SH, Lee S. Redox control of NRF2 signaling in oocytes harnessing Porphyra derivatives as a toggle. Free Radic Biol Med 2025; 227:680-693. [PMID: 39674422 DOI: 10.1016/j.freeradbiomed.2024.12.033] [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] [Received: 10/28/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
This study investigated the potential of Porphyra derivatives (PD), including Porphyra334, to activate the nuclear factor erythroid 2-related factor 2 (NRF2) pathway in porcine oocytes to enhance oocyte competency and intracellular networks. Conventional methods for manipulating mitochondrial functions and antioxidant pathways often rely upon genetic modifications that are impractical for direct application in humans. We hypothesized that PD serves as a natural regulator of the NRF2 pathway without requiring genetic intervention. To test this hypothesis, brusatol (Bru), a direct NRF2 inhibitor, was used to evaluate the specific role of PD in NRF2-mediated processes. The results demonstrated that PD significantly improved oocyte maturation, blastocyst formation, and mitochondrial function, including subsequent lipid metabolism. PD activates NRF2 and its downstream antioxidant response elements (AREs), whereas Bru inhibits these effects. Co-treatment with PD and Bru resulted in the partial recovery of NRF2 activity. These findings suggest that PD functions as a toggle for NRF2 activation, potentially offering a non-genetic strategy for enhancing oocyte quality and embryo development by modulating antioxidant mechanisms and mitochondrial functions. This study provides new avenues for investigating natural compounds in the context of reproductive biology and assisted reproductive technologies (ARTs).
Collapse
Affiliation(s)
- Euihyun Kim
- Plant Cell Research Institute of BIO-FD&C Co. Ltd., Incheon, 21990, Republic of Korea
| | - Dabin Cha
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Sung Joo Jang
- Plant Cell Research Institute of BIO-FD&C Co. Ltd., Incheon, 21990, Republic of Korea
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang Hyun Moh
- Plant Cell Research Institute of BIO-FD&C Co. Ltd., Incheon, 21990, Republic of Korea
| | - Sanghoon Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
22
|
Ping K, Yang R, Chen H, Xie S, Xiang Y, Li M, Lu Y, Dong J. Gypenoside XLIX Activates the Sirt1/Nrf2 Signaling Pathway to Inhibit NLRP3 Inflammasome Activation to Alleviate Septic Acute Lung Injury. Inflammation 2025; 48:42-60. [PMID: 38717633 DOI: 10.1007/s10753-024-02041-2] [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: 02/24/2024] [Revised: 04/21/2024] [Accepted: 04/29/2024] [Indexed: 02/09/2025]
Abstract
Currently, treatment options for acute lung injury (ALI) are limited. Gypenoside XLIX (Gyp-XLIX) is known for its anti-inflammatory properties, but there is a lack of extensive research on its effects against ALI. This study induced ALI in mice through cecal ligation and puncture surgery and investigated the biological activity and potential mechanisms of Gypenoside XLIX (40 mg/kg) by intraperitoneal injection. The in vitro ALI model was established using mouse lung epithelial (MLE-12) cells stimulated with lipopolysaccharide (LPS) and adenosine triphosphate (ATP). Various methods, including Hematoxylin and Eosin (H&E) staining, biochemical assay kits, Quantitative Polymerase Chain Reaction (qPCR) analysis, Western blotting, Terminal deoxynucleotidyl transferase dUTP Nick End Labeling (TUNEL) assay, immunofluorescence, and flow cytometry, were employed for this research. The results indicated that pretreatment with Gypenoside XLIX significantly alleviated pathological damage in mouse lung tissues and reduced the expression levels of inflammatory factors. Additionally, Gypenoside XLIX inhibited ROS levels and NLRP3 inflammasome, possibly mediated by the Sirt1/Nrf2 signaling pathway. Moreover, Gypenoside XLIX significantly inhibited sepsis-induced lung cell apoptosis and excessive autophagy of mitochondria. Specifically, it suppressed mitochondrial pathway apoptosis and the Pink1/Parkin pathway of mitochondrial autophagy. These findings reveal the multifaceted effects of Gypenoside XLIX in anti-inflammatory, antioxidative, and inhibition of cell apoptosis and autophagy. This provides strong support for its therapeutic potential in sepsis-related lung injuries.
Collapse
Affiliation(s)
- Kaixin Ping
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
- Institute of Neuroscience, Neurosurgery Department, The First People's Hospital of Lianyungang, Lianyungang, 222005, China
| | - Rongrong Yang
- Department of Oncology, The Second People's Hospital of Lianyungang (The Oncology Hospital of Lianyungang), Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Huizhen Chen
- Institute of Neuroscience, Neurosurgery Department, The First People's Hospital of Lianyungang, Lianyungang, 222005, China
| | - Shaocheng Xie
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yannan Xiang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
- Department of Oncology, The Second People's Hospital of Lianyungang (The Oncology Hospital of Lianyungang), Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Mengxin Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
- Institute of Neuroscience, Neurosurgery Department, The First People's Hospital of Lianyungang, Lianyungang, 222005, China
| | - Yingzhi Lu
- Department of Oncology, The Second People's Hospital of Lianyungang (The Oncology Hospital of Lianyungang), Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222000, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
23
|
Zayed M, Jeong BH. Adipose-Derived Mesenchymal Stem Cell Secretome Attenuates Prion Protein Peptide (106-126)-Induced Oxidative Stress via Nrf2 Activation. Stem Cell Rev Rep 2025; 21:589-592. [PMID: 39480613 DOI: 10.1007/s12015-024-10811-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Affiliation(s)
- Mohammed Zayed
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, 54531, Republic of Korea
- Department of Bioactive Material Sciences and Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju, 54896, Republic of Korea
- Department of Surgery, College of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, 54531, Republic of Korea.
- Department of Bioactive Material Sciences and Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
24
|
Furugaki K, Fujimura T, Sakaguchi N, Watanabe Y, Uchibori K, Miyauchi E, Hayashi H, Katayama R, Yoshiura S. Combined blockade of GPX4 and activated EGFR/HER3 bypass pathways inhibits the development of ALK-inhibitor-induced tolerant persister cells in ALK-fusion-positive lung cancer. Mol Oncol 2025; 19:519-539. [PMID: 39369284 PMCID: PMC11793004 DOI: 10.1002/1878-0261.13746] [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: 07/24/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Cancers can develop resistance to treatment with ALK tyrosine kinase inhibitors (ALK-TKIs) via emergence of a subpopulation of drug-tolerant persister (DTP) cells that can survive initial drug treatment long enough to acquire genetic aberrations. DTP cells are thus a potential therapeutic target. We generated alectinib-induced DTP cells from a patient-derived ALK+ non-small-cell lung cancer (NSCLC) cell line and screened 3114 agents in the anticancer compounds library (TargetMol). We identified phospholipid hydroperoxide glutathione peroxidase GPX4 as being involved in promoting the survival of DTP cells. GPX4 was found to be upregulated in DTP cells and to promote cell survival by suppressing reactive oxygen species (ROS) accumulation; GPX4 inhibitors blocked this upregulation and facilitated ROS-mediated cell death. Activated bypass signals involving epidermal growth factor receptor (EGFR)/receptor tyrosine-protein kinase erbB-3 (HER3) were also identified in DTP cells, and co-treatment with EGFR-TKI plus ALK-TKI enhanced ROS levels. Triple combination with an ALK-TKI plus a bypass pathway inhibitor plus a GPX4 inhibitor suppressed cell growth and induced intracellular ROS accumulation more greatly than did treatment with each agent alone. The combined inhibition of ALK plus inhibition of activated bypass signals plus inhibition of GPX4 may be a potent therapeutic strategy for patients with ALK+ NSCLC to prevent the development of resistance to ALK-TKIs and lead to tumor eradication.
Collapse
Affiliation(s)
- Koh Furugaki
- Product Research DepartmentChugai Pharmaceutical Co., LtdYokohamaJapan
| | - Takaaki Fujimura
- Product Research DepartmentChugai Pharmaceutical Co., LtdYokohamaJapan
| | - Narumi Sakaguchi
- Biological Technology DepartmentChugai Pharmaceutical Co., LtdYokohamaJapan
| | | | - Ken Uchibori
- Department of Thoracic Medical OncologyThe Cancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Eisaku Miyauchi
- Department of Respiratory MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Hidetoshi Hayashi
- Department of Medical OncologyKindai University Faculty of MedicineSayamaJapan
| | - Ryohei Katayama
- Division of Experimental ChemotherapyCancer Chemotherapy Center, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Shigeki Yoshiura
- Product Research DepartmentChugai Pharmaceutical Co., LtdYokohamaJapan
| |
Collapse
|
25
|
Adrar N, Gulsunoglu-Konuskan Z, Ceylan FD, Capanoglu E. Overview and trends in electrochemical sensors, biosensors and cellular antioxidant assays for oxidant and antioxidant determination in food. Talanta 2025; 283:127058. [PMID: 39509903 DOI: 10.1016/j.talanta.2024.127058] [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: 05/30/2024] [Revised: 08/14/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024]
Abstract
Screening and quantifying antioxidants from food samples, their antioxidant activity, as well as the assessment of food oxidation is critical, not only for ensuring food quality and safety, but also to understand and relate these parameters to the shelf life, sensory attributes, and health aspects of food products. For this purpose, several methods have been developed and used for decades, which regardless of their effectiveness, present a certain number of drawbacks mainly related to extensive sample preparation and technical complexity, time requirements, and the use of hazardous chemicals. Electrochemical sensors and biosensors are gaining popularity in food analysis due to their high sensitivity, specificity, rapid response times, and potential for miniaturisation and portability. Furthermore, other modern methods using whole living cells such as the cellular antioxidant activity assay, the antioxidant power 1 assay, and the catalase-like assays, may interpret more realistic antioxidant results rather than just reporting the ability to scavenge free radicals in isolated systems with extrapolation to reality. This paper provides an overview of electrochemical sensors, biosensors, and cellular antioxidant assays, and reviews the latest advancements and emerging trends in these techniques for determining oxidants and antioxidants in complex food matrices. The performances of different strategies are described for each of these approaches to provide insights into the extent to which these methods can be exploited in the field and inspire new research to fill the current gaps.
Collapse
Affiliation(s)
- Nabil Adrar
- Istanbul Technical University, Faculty of Chemical and Metallurgical Engineering, Department of Food Engineering, 34469, Maslak, Istanbul, Turkey.
| | - Zehra Gulsunoglu-Konuskan
- Istanbul Aydin University, Faculty of Health Science, Nutrition and Dietetics Department, 34295, Kucukcekmece, Istanbul, Turkey
| | - Fatma Duygu Ceylan
- Istanbul Technical University, Faculty of Chemical and Metallurgical Engineering, Department of Food Engineering, 34469, Maslak, Istanbul, Turkey
| | - Esra Capanoglu
- Istanbul Technical University, Faculty of Chemical and Metallurgical Engineering, Department of Food Engineering, 34469, Maslak, Istanbul, Turkey.
| |
Collapse
|
26
|
Elemam NM, Nader MA, Abdelmageed ME. Ameliorative impact of sacubitril/valsartan on paraquat-induced acute lung injury: role of Nrf2 and TLR4/NF-κB signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03785-w. [PMID: 39869189 DOI: 10.1007/s00210-025-03785-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025]
Abstract
Herbicides such as paraquat (PQ) are frequently utilized particularly in developing nations. The present research concentrated on the pulmonary lesions triggered by PQ and the beneficial effect of the angiotensin receptor neprilysin inhibitor (ARNI), sacubitril/valsartan, against such pulmonary damage. Five groups of rats were established: control, ARNI, PQ (10 mg/kg), ARNI 68 + PQ, and ARNI 34 + PQ. Following euthanasia, lungs were isolated and subjected to a histopathological test, and the ELISA technique was used to evaluate oxidative stress biomarkers, toll-like receptor 4 (TLR4), nuclear factor erythroid 2-related factor 2 (Nrf2), phosphatidylinositol-3-kinase (PI3K), phosphorylated protein kinase B (p-AKT), and inflammatory markers: nuclear factor kappa B p65 subunit (NF-κB p65), tumor necrosis factor α (TNFα), and interleukin 1beta (IL-1β). In conjunction with abnormally high levels of malondialdehyde (MDA) and inducible nitric oxide synthase (iNOS), the PQ group also displayed low levels of reduced glutathione (GSH) and total antioxidant capacity (TAC). Additionally, TLR4, PI3K, and p-AKT were significantly elevated together with unusually low level of Nrf2. Moreover, inflammatory biomarkers, NF-κB p65, TNFα, and IL-1β, were abnormally elevated. Meanwhile, ARNI-treated groups reversed all alterations precipitated by PQ in a dose-dependent manner. ARNI could mitigate pulmonary damage triggered by PQ via potential antioxidant anti-inflammatory qualities.
Collapse
Affiliation(s)
- Nourhane M Elemam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa, 7731168, Egypt.
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa, 7731168, Egypt
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa, 7731168, Egypt
| |
Collapse
|
27
|
Alves I, Araújo EMQ, Dalgaard LT, Singh S, Børsheim E, Carvalho E. Protective Effects of Sulforaphane Preventing Inflammation and Oxidative Stress to Enhance Metabolic Health: A Narrative Review. Nutrients 2025; 17:428. [PMID: 39940284 PMCID: PMC11821257 DOI: 10.3390/nu17030428] [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: 12/06/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 02/14/2025] Open
Abstract
The worldwide obesity epidemic has led to a drastic increase in diabetes and cardiovascular disease in younger generations. Further, maintaining metabolic health during aging is frequently a challenge due to poor diets and decreased mobility. In this setting, bioactive nutrients that are naturally occurring antioxidants, such as sulforaphane (SFN), are of high nutritional interest. SFN, a bioactive compound that is present in cruciferous vegetables, is a molecule that protects cells from cytotoxic damage and mitigates oxidative stress, protecting against disease. It exerts its action through the activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2). Many studies have been performed in animals and humans to evaluate its effects on cancer, brain health, and neurodegenerative disorders. However, fewer clinical studies have been performed to evaluate its effects on insulin resistance and the development of type 2 diabetes mellitus (T2DM) across the lifespan. Given that, in some parts of the world, particularly in Europe, the population is growing older at a significant rate, it is crucial to promote healthy habits (healthy foods, dietary pattern, precision nutrition, and physical activity) from an early stage in life and across the lifespan to avoid debilitating health conditions occurring during adulthood and aging. Thus, in this narrative review, we discuss the protective effects of SFN supplementation on inflammatory and oxidative stress pathways and relate them to metabolic disease.
Collapse
Affiliation(s)
- Inês Alves
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal;
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA;
| | - Edilene Maria Queiroz Araújo
- Nutritional Genomics and Metabolic Dysfunctions Research and Extension Center, Department of Life Sciences, State University of Bahia, Salvador 41195001, BA, Brazil;
| | - Louise T. Dalgaard
- Department of Science and Environment, Roskilde University, Universitetsvej 1, DK-4000 Roskilde, Denmark;
| | - Sharda Singh
- Division of Hematology & Oncology, Department of Internal Medicine, Texas Tech University Medical Sciences Center, Lubbock, TX 79430, USA;
| | - Elisabet Børsheim
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA;
- Department of Pediatrics & Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
- Arkansas Children’s Nutrition Center, Little Rock, AR 72202, USA
| | - Eugenia Carvalho
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinar Research, University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
28
|
He Y, Liu Y, Zhang M. The beneficial effects of curcumin on aging and age-related diseases: from oxidative stress to antioxidant mechanisms, brain health and apoptosis. Front Aging Neurosci 2025; 17:1533963. [PMID: 39906716 PMCID: PMC11788355 DOI: 10.3389/fnagi.2025.1533963] [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: 11/25/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025] Open
Abstract
Aging and age-related disease are among the most common and challenging issues worldwide. During the aging process, the accumulation of oxidative stress, DNA damage, telomere dysfunction, and other related changes lead to cellular dysfunction and the development of diseases such as neurodegenerative and cardiovascular conditions. Curcumin is a widely-used dietary supplement against various diseases such as cancer, diabetes, cardiovascular diseases and aging. This agent mediates its effects through several mechanisms, including the reduction of reactive oxygen species (ROS) and oxidative stress-induced damage, as well as the modulation of subcellular signaling pathways such as AMPK, AKT/mTOR, and NF-κB. These pathways are involved in cellular senescence and inflammation, and their modulation can improve cell function and help prevent disease. In cancer, Curcumin can induce apoptosis in a variety of different tumor cell lines. Curcumin also activates redox reactions within cells inducing ROS production that leads to the upregulation of apoptosis receptors on the tumor cell membrane. Curcumin can also upregulate the expression and activity of p53 that inhibits tumor cell proliferation and increases apoptosis. Furthermore, curcumin has a potent inhibitory effect on the activity of nuclear factor kappa B (NF-κB) and cyclooxygenase-2 (COX-2), which are involved in the overexpression of antiapoptosis genes such as Bcl-2. It can also attenuate the regulation of antiapoptosis phosphoinositide 3-kinases (PI3K) signaling and increase the expression of mitogen-activated protein kinases (MAPKs) to induce endogenous production of ROS. Therefore, herein, we aim to summarize how curcumin affect different epigenetic processes (such as apoptosis and oxidative stress) in order to change aging-related mechanisms. Furthermore, we discuss its roles in age-related diseases, such as Alzheimer, Parkinson, osteoporosis, and cardiovascular diseases.
Collapse
Affiliation(s)
- Ying He
- Department of Biological and Food Engineering, Lyuliang University, Lishi, Shanxi, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Yongqing Liu
- Department of Biological and Food Engineering, Lyuliang University, Lishi, Shanxi, China
| | - Min Zhang
- Key Laboratory of Agro-Products Primary Processing, Academy of Agricultural Planning and Engineering, MARA, Beijing, China
| |
Collapse
|
29
|
Długosz A, Wróblewski M, Błaszak B, Szulc J. The Role of Nutrition, Oxidative Stress, and Trace Elements in the Pathophysiology of Autism Spectrum Disorders. Int J Mol Sci 2025; 26:808. [PMID: 39859522 PMCID: PMC11765825 DOI: 10.3390/ijms26020808] [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: 12/27/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social communication and interaction, alongside repetitive behaviors, and atypical sensory-motor patterns. The growing prevalence of ASD has driven substantial advancements in research aimed at understanding its etiology, preventing its onset, and mitigating its impact. This ongoing effort necessitates continuous updates to the body of knowledge and the identification of previously unexplored factors. The present study addresses this need by examining the roles of nutrition, oxidative stress, and trace elements in the pathophysiology of ASD. In this review, an overview is provided of the key dietary recommendations for individuals with ASD, including gluten-free and casein-free (GFCF) diets, ketogenic diets (KDs), and other nutritional interventions. Furthermore, it explores the involvement of oxidative stress in ASD and highlights the significance of trace elements in maintaining neuropsychiatric health. The impact of these factors on molecular and cellular mechanisms was discussed, alongside therapeutic strategies and their efficacy in managing ASD.
Collapse
Affiliation(s)
- Anna Długosz
- Department of Food Industry Technology and Engineering, Faculty of Chemical Technology and Engineering, Bydgoszcz University of Science and Technology, 3 Seminaryjna St., 85-326 Bydgoszcz, Poland; (B.B.); (J.S.)
| | - Marcin Wróblewski
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Błażej Błaszak
- Department of Food Industry Technology and Engineering, Faculty of Chemical Technology and Engineering, Bydgoszcz University of Science and Technology, 3 Seminaryjna St., 85-326 Bydgoszcz, Poland; (B.B.); (J.S.)
| | - Joanna Szulc
- Department of Food Industry Technology and Engineering, Faculty of Chemical Technology and Engineering, Bydgoszcz University of Science and Technology, 3 Seminaryjna St., 85-326 Bydgoszcz, Poland; (B.B.); (J.S.)
| |
Collapse
|
30
|
Tran QA, Tran GV, Velic S, Xiong HM, Kaur J, Moosavi Z, Nguyen P, Duong N, Luu VT, Singh G, Bui T, Rose M, Ho L. Effects of Astragaloside IV and Formononetin on Oxidative Stress and Mitochondrial Biogenesis in Hepatocytes. Int J Mol Sci 2025; 26:774. [PMID: 39859490 PMCID: PMC11765978 DOI: 10.3390/ijms26020774] [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: 12/06/2024] [Revised: 01/07/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Over-accumulation of reactive oxygen species (ROS) causes hepatocyte dysfunction and apoptosis that might lead to the progression of liver damage. Sirtuin-3 (SIRT3), the main NAD+-dependent deacetylase located in mitochondria, has a critical role in regulation of mitochondrial function and ROS production as well as in the mitochondrial antioxidant mechanism. This study explores the roles of astragaloside IV (AST-IV) and formononetin (FMR) in connection with SIRT3 for potential antioxidative effects. It was shown that the condition of combined pre- and post-treatment with AST-IV or FMR at all concentrations statistically increased and rescued cell proliferation. ROS levels were not affected by pre-or post-treatment individually with AST-IV or pre-treatment with FMR; however, post-treatment with FMR resulted in significant increases in ROS in all groups. Significant decreases in ROS levels were seen when pre- and post-treatment with AST-IV were combined at 5 and 10 μM, or FMR at 5 and 20 μM. In the condition of combined pre- and post-treatment with 10 μM AST-IV, there was a significant increase in SOD activity, and the transcriptional levels of Sod2, Cat, and GPX1 in all treatment groups, which is indicative of reactive oxygen species detoxification. Furthermore, AST-IV and FMR activated PGC-1α and AMPK as well as SIRT3 expression in AML12 hepatocytes exposed to t-BHP-induced oxidative stress, especially at high concentrations of FMR. This study presents a novel mechanism whereby AST-IV and FMR yield an antioxidant effect through induction of SIRT3 protein expression and activation of an antioxidant mechanism as well as mitochondrial biogenesis and mitochondrial content and potential. The findings suggest these agents can be used as SIRT3 modulators in treating oxidative-injury hepatocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Linh Ho
- College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| |
Collapse
|
31
|
He J, Hewett SJ. Nrf2 Regulates Basal Glutathione Production in Astrocytes. Int J Mol Sci 2025; 26:687. [PMID: 39859401 PMCID: PMC11765531 DOI: 10.3390/ijms26020687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Astrocytes produce and export glutathione (GSH), an important thiol antioxidant essential for protecting neural cells from oxidative stress and maintaining optimal brain health. While it has been established that oxidative stress increases GSH production in astrocytes, with Nrf2 acting as a critical transcription factor regulating key components of the GSH synthetic pathway, the role of Nrf2 in controlling constitutive GSH synthetic and release mechanisms remains incompletely investigated. Our data show that naïve primary mouse astrocytes cultured from the cerebral cortices of Nrf2 knockout (Nrf2-/-) pups have significantly less intracellular and extracellular GSH levels when compared to astrocytes cultured from Nrf2 wild-type (Nrf2+/+) pups. Key components of the GSH synthetic pathway, including xCT (the substrate-specific light chain of the substrate-importing transporter, system xc-), glutamate-cysteine ligase [catalytic (GCLc) and modifying (GCLm) subunits], were affected. To wit: qRT-PCR analysis demonstrates that naïve Nrf2-/- astrocytes have significantly lower basal mRNA levels of xCT and both GCL subunits compared to naïve Nrf2+/+ astrocytes. No change in mRNA levels of glutathione synthetase (GS) or the GSH exporting transporter, Mrp1, was found. Western blot analysis reveals reduced protein levels of both subunits of GCL, while (seleno)cystine uptake into Nrf2-/- astrocytes was reduced compared to Nrf2+/+ astrocytes, confirming decreased system xc- activity. These findings suggest that Nrf2 regulates the basal production of GSH in astrocytes through constitutive transcriptional regulation of GCL and xCT.
Collapse
Affiliation(s)
| | - Sandra J. Hewett
- Program in Neuroscience, Department of Biology, Syracuse University, Syracuse, NY 13210, USA;
| |
Collapse
|
32
|
Luan J, Yu M, Gu Q, Zhou X, Shao Y, Chen T, Zhang J, Zhu Z, Song N, Yang J. Fatty acid synthase inhibition improves hypertension-induced erectile dysfunction by suppressing oxidative stress and NLRP3 inflammasome-dependent pyroptosis through activating the Nrf2/HO-1 pathway. Front Immunol 2025; 15:1532021. [PMID: 39877365 PMCID: PMC11772187 DOI: 10.3389/fimmu.2024.1532021] [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: 11/22/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Background Erectile dysfunction (ED) is a prevalent male sexual disorder, commonly associated with hypertension, though the underlying mechanisms remain poorly understood. Objective This study aims to explore the role of Fatty acid synthase (Fasn) in hypertension-induced ED and evaluate the therapeutic potential of the Fasn inhibitor C75. Materials and methods Erectile function was assessed by determining the intracavernous pressure/mean arterial pressure (ICP/MAP) ratio, followed by the collection of cavernous tissue for transcriptomic and non-targeted metabolomic analyses. In vitro, a concentration of 10-6 M angiotensin II (Ang II) was applied to rat aortic endothelial cells (RAOECs) to establish a model of hypertension. In vivo, spontaneously hypertensive rats (SHR) were randomly divided into two groups. The SHR+C75 group received intraperitoneal injections of C75 at a dose of 2 mg/kg once a week. After five weeks of treatment, the erectile function of the rats was assessed, and penile tissues were harvested for further analysis. Molecular and protein expression were assessed using Western blotting, qRT-PCR, immunofluorescence staining, and immunohistochemistry. Results The SHR exhibited ED, indicated by reduced maximum ICP/MAP ratios. Histologically, corpus cavernosum tissue of SHR showed elevated fibrosis and endothelial dysfunction. Additionally, increased expression of the NLRP3 inflammasome, Caspase-1, GSDMD, and the pro-inflammatory cytokines IL-1β and IL-18 was observed. Multi-omics analysis revealed significant enrichment in lipid metabolic pathways, with Fasn identified as a hub gene. In vitro, siFasn and C75 enhanced antioxidant markers Nrf2 and HO-1, reduced ROS accumulation, and suppressed NLRP3 and GSDMD levels. In vivo, C75 treatment restored endothelial function and reversed erectile dysfunction, accompanied by decreased oxidative stress and pyroptosis in the penile corpus cavernosum. Conclusion These findings suggest that Fasn inhibition may offer a promising therapeutic strategy for hypertension-induced ED by alleviating oxidative stress and suppressing NLRP3 inflammasome-dependent endothelial cell pyroptosis via activation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Jiaochen Luan
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengchi Yu
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Gu
- Department of Urology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan Zhou
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunqiang Shao
- Department of Urology, People’s Hospital of Xinjiang Kizilsu Kirgiz Autonomous Prefecture, Kizilsu Kirgiz Autonomous Prefecture, China
| | - Tong Chen
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayi Zhang
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Zhu
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ninghong Song
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Yang
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
33
|
Jiang Y, He P, Sheng K, Peng Y, Wu H, Qian S, Ji W, Guo X, Shan X. The protective roles of eugenol on type 1 diabetes mellitus through NRF2-mediated oxidative stress pathway. eLife 2025; 13:RP96600. [PMID: 39792010 PMCID: PMC11723580 DOI: 10.7554/elife.96600] [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] [Indexed: 01/30/2025] Open
Abstract
Type 1 diabetes mellitus (T1DM), known as insulin-dependent diabetes mellitus, is characterized by persistent hyperglycemia resulting from damage to the pancreatic β cells and an absolute deficiency of insulin, leading to multi-organ involvement and a poor prognosis. The progression of T1DM is significantly influenced by oxidative stress and apoptosis. The natural compound eugenol (EUG) possesses anti-inflammatory, anti-oxidant, and anti-apoptotic properties. However, the potential effects of EUG on T1DM had not been investigated. In this study, we established the streptozotocin (STZ)-induced T1DM mouse model in vivo and STZ-induced pancreatic β cell MIN6 cell model in vitro to investigate the protective effects of EUG on T1DM, and tried to elucidate its potential mechanism. Our findings demonstrated that the intervention of EUG could effectively induce the activation of nuclear factor E2-related factor 2 (NRF2), leading to an up-regulation in the expressions of downstream proteins NQO1 and HMOX1, which are regulated by NRF2. Moreover, this intervention exhibited a significant amelioration in pancreatic β cell damage associated with T1DM, accompanied by an elevation in insulin secretion and a reduction in the expression levels of apoptosis and oxidative stress-related markers. Furthermore, ML385, an NRF2 inhibitor, reversed these effects of EUG. The present study suggested that EUG exerted protective effects on pancreatic β cells in T1DM by attenuating apoptosis and oxidative stress through the activation of the NRF2 signaling pathway. Consequently, EUG holds great promise as a potential therapeutic candidate for T1DM.
Collapse
Affiliation(s)
- Yalan Jiang
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Pingping He
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ke Sheng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yongmiao Peng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Huilan Wu
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Songwei Qian
- Department of Genaral Surgery, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s HospitalQuzhouChina
- Department of General Surgery, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Weiping Ji
- Department of Genaral Surgery, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s HospitalQuzhouChina
- Department of General Surgery, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiaoling Guo
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiaoou Shan
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
34
|
Hamidizad Z, Kadkhodaee M, Kianian F, Ranjbaran M, Heidari F, Seifi B. Neuroprotective Effects of Sodium Nitroprusside on CKD-Induced Cognitive Dysfunction in Rats: Role of CBS and Nrf2/HO-1 Pathway. Neuromolecular Med 2025; 27:8. [PMID: 39775152 DOI: 10.1007/s12017-024-08828-8] [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: 11/11/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Chronic kidney disease (CKD) is a conceivable new risk factor for cognitive disorder and dementia. Uremic toxicity, oxidative stress, and peripheral-central inflammation have been considered important mediators of CKD-induced nervous disorders. Nitric oxide (NO) is a retrograde neurotransmitter in synapses, and has vital roles in intracellular signaling in neurons. This research aims to determine the effectiveness of NO in CKD-induced cognitive deficits by considering the nuclear factor-erythroid factor 2-related factor 2 (Nrf2)/ heme oxygenase-1 (HO-1) signaling pathway and the important roles of cystathionine beta-synthase (CBS, H2S producing enzyme). Forty rats were divided into four experimental groups: sham, five-sixth (5/6) nephrectomy (5/6Nx, CKD), CKD + NO donor (Sodium nitroprusside, SNP), CKD + SNP and a CBS inhibitor (amino-oxy acetic acid, AOAA). To assess the neurocognitive abilities, eleven weeks after 5/6Nx, behavioral tests (Novel object recognition test, Passive avoidance test, and Barnes maze test) were done. Twelfth week after 5/6Nx, blood urea nitrogen (BUN) and serum creatinine (sCr) levels, as well as the nuclear factor-erythroid factor 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1) expression levels and neuronal injury in the hippocampus and prefrontal cortex were assessed. As predicted, the levels of BUN and sCr (both P < 0.001) and neuronal injury in the hippocampus (P < 0.001 for CA1; CA3; DG) and prefrontal cortex (P < 0.001) increased in CKD rats as well as 5/6Nx induced reduction of Nrf2 (both P < 0.001) /HO-1(P < 0.001; P < 0.01 respectively) pathway activity in the hippocampus and prefrontal cortex in CKD rats. Moreover, CKD leads to cognitive disorder and memory loss (Novel object recognition test (NOR) (P < 0.001), Passive avoidance test (PA) (P < 0.001) and Barnes maze (BA) (Escape latency (P < 0.001); Error (P < 0.001)). SNP treatment significantly improved Nrf2 (both P < 0.001) /HO-1 (P < 0.001; P < 0.05 respectively) pathways and neuronal injury (P < 0.001 for CA1; CA3; DG) in the hippocampus and prefrontal cortex in CKD rats as well as enhanced learning and memory ability in CKD rats. However, ameliorating effects of SNP on cognitive disorder (NOR (P < 0.05), PA (P < 0.001) and BA (Escape latency (P < 0.05); Error (P < 0.001)) and Nrf2 (P < 0.01; P < 0.001 in the hippocampus and prefrontal cortex respectively) /HO-1 (P < 0.05 in both) signaling pathway activity were nullified by CBS inhibitor and H2S reduction. In conclusion, this study demonstrated that NO improved CKD-induced cognitive impairment and neuronal death which is may be depended to CBS activity and endogenous H2S levels.
Collapse
Affiliation(s)
- Zeinab Hamidizad
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Mehri Kadkhodaee
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Farzaneh Kianian
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Mina Ranjbaran
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Fatemeh Heidari
- Department of Anatomy, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Behjat Seifi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran.
| |
Collapse
|
35
|
Xu L, Li W, Chen Y, Liu S, Liu G, Luo W, Cao G, Wang S. Metformin Regulates Cardiac Ferroptosis to Reduce Metabolic Syndrome-Induced Cardiac Dysfunction. Appl Biochem Biotechnol 2025; 197:179-193. [PMID: 39106027 DOI: 10.1007/s12010-024-05038-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
High-fat diet-induced metabolic syndrome (MetS) is closely associated with cardiac dysfunction. Recent research studies have indicated a potential association between MetS and ferroptosis. Furthermore, metformin can alleviate MetS-induced cardiac ferroptosis. Metformin is a classic biguanide anti-diabetic drug that has protective effects on cardiovascular diseases, which extend beyond its indirect glycemic control. This study aimed to assess whether MetS mediates cardiac ferroptosis, thereby causing oxidative stress and mitochondrial dysfunction. The results revealed that metformin can mitigate cardiac reactive oxygen species and mitochondrial damage, thereby preserving cardiac function. Mechanistic analysis revealed that metformin upregulates the expression of cardiac Nrf2. Moreover, Nrf2 downregulation compromises the cardio-protective effects of metformin. In summary, this study indicated that MetS promotes cardiac ferroptosis, and metformin plays a preventive and therapeutic role, partially through modulation of Nrf2 expression.
Collapse
Affiliation(s)
- Liancheng Xu
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
- Fujian Medical University, Fuzhou, 350108, China
| | - Wenwen Li
- Department of Nephrology, Suqian First Hospital, Suqian, 223800, China
| | - Yu Chen
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Shan Liu
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Guodong Liu
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Weihuan Luo
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Guanyi Cao
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China
| | - Shiping Wang
- Department of General Surgery, Suqian First Hospital, No.120 Suzhi Street, Suqian, 223800, Jiangsu Province, China.
| |
Collapse
|
36
|
Musapoor S, Davoodian N, Kadivar A, Ahmadi E, Nazari H. Media Supplementation With Gamma-Oryzanol Improves the Outcome of Ovine Oocyte Maturation In Vitro. Vet Med Sci 2025; 11:e70134. [PMID: 39688528 DOI: 10.1002/vms3.70134] [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: 05/17/2024] [Revised: 09/29/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND The process of maturing ovine oocyte in vitro has not yet been raised with acceptable results. OBJECTIVE This study was designed to evaluate the γ-oryzanol effect as a supplement of maturation media on the development of ovine oocytes to blastocyst. METHODS Aspirated from ovine ovaries, morphologically normal cumulus-oocyte complexes (COCs) were matured in media supplemented with or without 5 µM γ-oryzanol. Matured oocytes were divided into two parts: one evaluated for their nuclear maturation, the level of GSH and ROS, mitochondrial membrane potential (MMP) and the pattern of transcription in oocytes and respective cumulus cells (CCs), and another subjected to fertilisation and culture to assess the development of oocytes to the blastocyst. RESULTS γ-Oryzanol improved the proportion of cleaved embryos and total blastocysts in the treated group, which was linked to improved MMP, higher levels of intracellular GSH and lower levels of ROS. A lower proportion of MI and GVBD was recorded for treated oocytes in comparison with control, although the proportion of MII oocytes was not different between groups. The treated oocytes and CCs showed downregulation of genes related to apoptosis (BAX and CASP-9) and upregulation of genes related to antioxidative status (NRF2, CAT and SOD). In conclusion, our results demonstrated the improved developmental outcome of supplemented oocytes so that the antioxidant response and higher enzymatic activity were maintained, and the generation of ROS was turned off; therefore, a novel alternative for counteracting oxidative stress in ovine oocytes undergoing maturation was offered by γ-oryzanol through an antioxidative pathway.
Collapse
Affiliation(s)
- Saeed Musapoor
- Research Institute of Animal Embryo Technology, Shahrekord University, Shahrekord, Iran
| | - Najmeh Davoodian
- Research Institute of Animal Embryo Technology, Shahrekord University, Shahrekord, Iran
| | - Ali Kadivar
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Ebrahim Ahmadi
- Research Institute of Animal Embryo Technology, Shahrekord University, Shahrekord, Iran
| | - Hassan Nazari
- Research Institute of Animal Embryo Technology, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
37
|
Xu M, Liu L, Fan Z, Niu L, Ning W, Cheng H, Li M, Huo W, Zhou P, Deng H, Chen W, Che L. Effect of different dietary oil sources on the performance, egg quality and antioxidant capacity during the late laying period. Poult Sci 2025; 104:104615. [PMID: 39637658 PMCID: PMC11664395 DOI: 10.1016/j.psj.2024.104615] [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: 08/09/2024] [Revised: 11/25/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024] Open
Abstract
This study investigated the effects of different dietary ratios of linseed and soybean oils on the performance, egg quality, and antioxidant capacity of late-phase laying hens. A total of 360 70-week-old Jinghong laying hens were randomly assigned to four groups of six replicates each, with 15 chickens per replicate. Diets with linseed oil to soybean oil ratios of 3:0 (T1), 2:1 (T2), 1:2 (T3), and 0:3 (T4) were fed for 4 weeks. No significant differences in egg weight, feed intake of laying hens, egg production, or feed-to-egg ratio (P > 0.05) were observed among the groups. Compared with the T4 group, the T2 group had a significantly higher number of 8-10 mm follicles. Moreover, albumen height and Haugh units were significantly higher in the T3 group than in the T4 group (P < 0.05), although significant differences were not observed among the T1, T2, and T3 groups. With an increase in linseed oil addition to the feed, the content of n-3 polyunsaturated fatty acids in chicken eggs significantly increased (P < 0.05). Compared to the T4 group, the addition of linseed oil to the diet significantly reduced the blood malondialdehyde content and increased the blood glutathione peroxidase (GSH-PX) and superoxide dismutase enzyme activity. The GSH-PX activity and total antioxidant capacity in the oviducts of the T3 group were significantly higher than those of the T4 group (P < 0.05). The protein expression levels of Nrf2, HO-1, and NQO-1 in the oviduct tissues were significantly higher in the T3 group than in the T4 group (P < 0.05). This study showed that a linseed oil to soybean oil ratio of 1:2 in the T3 group enhanced egg quality by reducing oxidative stress and improving the oviduct microenvironment.
Collapse
Affiliation(s)
- Mengmeng Xu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No.6 North Longzihu Road, Zhengdong New District, Zhengzhou, 450046, Henan, China
| | - Le Liu
- College of Livestock Husbandry and Veterinary Engineering, Henan Agricultural University, No. 15 Longzi Lake University Campus, Zhengzhou, 450046, China
| | - Zongze Fan
- College of Livestock Husbandry and Veterinary Engineering, Henan Agricultural University, No. 15 Longzi Lake University Campus, Zhengzhou, 450046, China
| | - Lizhu Niu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No.6 North Longzihu Road, Zhengdong New District, Zhengzhou, 450046, Henan, China
| | - Wenxi Ning
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No.6 North Longzihu Road, Zhengdong New District, Zhengzhou, 450046, Henan, China
| | - He Cheng
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No.6 North Longzihu Road, Zhengdong New District, Zhengzhou, 450046, Henan, China
| | - Mengyun Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No.6 North Longzihu Road, Zhengdong New District, Zhengzhou, 450046, Henan, China
| | - Wenying Huo
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No.6 North Longzihu Road, Zhengdong New District, Zhengzhou, 450046, Henan, China
| | - Pan Zhou
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China
| | - Hongyu Deng
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No.6 North Longzihu Road, Zhengdong New District, Zhengzhou, 450046, Henan, China
| | - Wen Chen
- College of Livestock Husbandry and Veterinary Engineering, Henan Agricultural University, No. 15 Longzi Lake University Campus, Zhengzhou, 450046, China
| | - Long Che
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No.6 North Longzihu Road, Zhengdong New District, Zhengzhou, 450046, Henan, China.
| |
Collapse
|
38
|
Umrao A, Pahuja M, Chatterjee NS. Safety and efficacy of omaveloxolone v/s placebo for the treatment of Friedreich's ataxia in patients aged more than 16 years: a systematic review. Orphanet J Rare Dis 2024; 19:495. [PMID: 39736600 DOI: 10.1186/s13023-024-03474-6] [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: 06/05/2024] [Accepted: 11/20/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Friedreich's ataxia (FA) is a rare genetic disorder caused by silencing of the frataxin gene (FXN), which leads to multiorgan damage. Nrf2 is a regulator of FXN, which is a modulator of oxidative stress in animals and humans. Omaveloxolone (Omav) is an Nrf2 activator and has been reported to have antioxidative potential in various disease conditions. The present review was conducted to determine the use of Omav, the only FDA-approved treatment for FA. METHODS Three electronic databases, Cochrane, PubMed and Google Scholar, were searched with terms such as 'Omaveloxolone', 'Friedreich ataxia', 'genetic diseases', 'autosomal recessive', and 'rare disorders' using various advanced search filters. Articles were screened, extracted, and assessed for quality, and a qualitative synthesis of the data was performed. The study protocol was registered in PROSPERO (CRD42024531449). RESULTS A total of 201 records were found, with very few published research articles on the topic. Only two randomized clinical trials published in a series of three research articles were included in the current systematic review. Peak load exercise and modified Friedreich's Ataxia Rating Scale (mFARS) values were considered the major outcome measures for determining the efficacy of 150 mg Omav capsules/day in FA. Exploratory outcome measures, such as low-contrast letter visual acuity test, exercise test, T25-FW, 9-HPT, health-related quality of life, and biochemical tests, were also assessed along with adverse events in all the studies. CONCLUSION Although, the quality of the articles demonstrated low bias. However, the short duration, small sample size, and missing data, including the values of different measures of mFARS scores in patients, limit the generalizability of the results. Further studies with longer durations and in severe patients with foot deformities are needed to clearly define the efficacy of Omav in FA and to determine the optimal drug for FA patients in India.
Collapse
Affiliation(s)
- Ankita Umrao
- Discovery Research Division, Indian Council of Medical Research (ICMR) Headquarters, V. Ramalingaswami Bhawan, Ansari Nagar, P.O. Box 4911, New Delhi, 110029, India
| | - Monika Pahuja
- Discovery Research Division, Indian Council of Medical Research (ICMR) Headquarters, V. Ramalingaswami Bhawan, Ansari Nagar, P.O. Box 4911, New Delhi, 110029, India.
| | - Nabendu Sekhar Chatterjee
- Discovery Research Division, Indian Council of Medical Research (ICMR) Headquarters, V. Ramalingaswami Bhawan, Ansari Nagar, P.O. Box 4911, New Delhi, 110029, India
| |
Collapse
|
39
|
Zhang XJ, Pu YK, Yang PY, Wang MR, Zhang RH, Li XL, Xiao WL. Isolicoflavonol ameliorates acute liver injury via inhibiting NLRP3 inflammasome activation through boosting Nrf2 signaling in vitro and in vivo. Int Immunopharmacol 2024; 143:113233. [PMID: 39366075 DOI: 10.1016/j.intimp.2024.113233] [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: 07/30/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND NOD like receptor pyrin domain containing 3 (NLRP3) inflammasome is involved in innate immunity, and related to liver injury. However, no inflammasome inhibitors are clinically available until now. Our previous research suggests that isolicoflavonol (ILF), isolated from Macaranga indica, is a potent NLRP3 inflammasome inhibitor, but its mechanism is unclear. METHODS Fluorescent imaging and Western blot assay were used to ascertain the effects of ILF on pyroptosis and NLRP3 inflammasome activation in macrophages. Next, Nrf2 signal pathway, its downstream gene transcription and expression were further investigated. ML385, a Nrf2 inhibitor, was used to verify whether ILF targets Nrf2 signaling. A carbon tetrachloride induced liver injury model was introduced to evaluate the liver protection activity of ILF in mice. RESULTS This work revealed that ILF inhibited macrophage LDH release and IL-1β secretion in a dose-dependent manner. ILF had no significant cytotoxic effect on macrophage, it reduced pyroptosis and Gasdermin D N-terminal fragment formation. Moreover, ILF inhibited IL-1β maturation and Caspase-1 cleavage, but did not affect NLRP3, pro-Caspase-1, pro-IL-1β and ASC expression. ILF decreased ASC speck rate and reduced ASC oligomer formation. ILF decreased aggregated JC-1 formation restoring mitochondria membrane potential. In addition, ILF increased Nrf2 expression, extended Nrf2 lifespan and upregulated Nrf2 signaling pathway in macrophages whether the NLRP3 inflammasome was activated or not. Besides, ILF increased Nrf2 nuclear translocation, maintained a high proportion of Nrf2 in the nucleus, and upregulated ARE-related gene transcription and expression. Furthermore, Nrf2 signal inhibition attenuated compound ILF-mediated inhibition of pyroptosis, inflammasome activation and upregulation of Nrf2 signaling. ILF in a liver injury mouse model inhibited NLRP3 inflammasome activation and enhanced Nrf2 signaling. CONCLUSION Our study verified that ILF ameliorates liver injury via inhibiting NLRP3 inflammasome activation through boosting Nrf2 signaling, and highlighted that ILF is a potent anti-inflammatory drug for inflammasome-related liver diseases.
Collapse
Affiliation(s)
- Xing-Jie Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Yu-Kun Pu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Peng-Yun Yang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Meng-Ru Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Rui-Han Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China.
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China; Southwest United Graduate School, Kunming 650500, Yunnan, China.
| |
Collapse
|
40
|
Kan HL, Wang SS, Liao CL, Tsai WR, Wang CC, Tung CW. An Integrated Testing Strategy and Online Tool for Assessing Skin Sensitization of Agrochemical Formulations. TOXICS 2024; 12:936. [PMID: 39771151 PMCID: PMC11728478 DOI: 10.3390/toxics12120936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
Non-animal assessment of skin sensitization is a global trend. Recently, scientific efforts have been focused on the integration of multiple evidence for decision making with the publication of OECD Guideline No. 497 for defined approaches to skin sensitization. The integrated testing strategy (ITS) methods reported by the guideline integrates in chemico, in vitro, and in silico testing to assess both hazard and potency of skin sensitization. The incorporation of in silico methods achieved comparable performance with fewer experiments compared to the traditional two-out-of-three (2o3) method. However, the direct application of current ITSs to agrochemicals can be problematic due to the lack of agrochemicals in the training data of the incorporated in silico methods. To address the issue, we present ITS-SkinSensPred 2.0 for agrochemicals and agrochemical formulations using a reconfigured in silico model SkinSensPred for pesticides. Compared to ITSv2, the proposed ITS-SkinSensPred 2.0 achieved an 11% and 16% improvement in the accuracy and correct classification rate for hazard identification and potency classification, respectively. In addition, an online ITS tool was implemented and available on the SkinSensDB website. The tool is expected to be useful for evaluating skin sensitization of substances.
Collapse
Affiliation(s)
- Hung-Lin Kan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; (H.-L.K.); (S.-S.W.)
| | - Shan-Shan Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; (H.-L.K.); (S.-S.W.)
- Ph.D. Program in Environmental and Occupational Medicine, College of Medicine, Kaohsiung Medical University and National Health Research Institutes, Kaohsiung 80756, Taiwan
| | - Chun-Lin Liao
- Agricultural Chemicals Research Institute, Ministry of Agriculture, Taichung 41358, Taiwan; (C.-L.L.); (W.-R.T.)
| | - Wei-Ren Tsai
- Agricultural Chemicals Research Institute, Ministry of Agriculture, Taichung 41358, Taiwan; (C.-L.L.); (W.-R.T.)
| | - Chia-Chi Wang
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; (H.-L.K.); (S.-S.W.)
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei 10675, Taiwan
| |
Collapse
|
41
|
Zhang Z, Gao J, Jia L, Kong S, Zhai M, Wang S, Li W, Wang S, Su Y, Li W, Zhu C, Wang W, Lu Y, Li W. Excessive glutathione intake contributes to chemotherapy resistance in breast cancer: a propensity score matching analysis. World J Surg Oncol 2024; 22:345. [PMID: 39709466 DOI: 10.1186/s12957-024-03626-9] [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: 11/02/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND We aim to explore the impact of excessive glutathione (GSH) intake on chemotherapy sensitivity in breast cancer. METHODS Clinicopathological data were collected from 460 breast cancer patients who underwent adjuvant chemotherapy from January 2016 to December 2019 from Zhengzhou University People's Hospital. The clinicopathological characteristics following GSH treatment were collected and compared with those in Non-GSH group after 1:2 propensity score matching (PSM). Intracellular GSH levels and the expression of antioxidant enzymes (NRF2, GPX4 and SOD1) were evaluated in tumor tissues in 51 patients receiving neoadjuvant chemotherapy. RESULTS The recurrence rate after adjuvant chemotherapy was significantly higher in the GSH group (n = 28, 31.8%) than that in the Non-GSH group (n = 39, 22.2%; P = 0.010). Additionally, patients in the HGSH group (high GSH intake, ≥ 16 days) exhibited an elevated recurrence rate compared to that in the LGSH group (low GSH intake, < 16 days) (n = 15 (46.8%) vs. n = 52 (22.4%); P = 0.003). Cox regression revealed that High GSH intake, Ki67 ≥ 30%, Triple negative and Lymphovascular invasion were independent risk factors of progression after adjuvant chemotherapy. Among patients receiving neoadjuvant chemotherapy, intracellular GSH levels and the expression levels of antioxidant enzymes (NRF2, GPX4 and SOD1) in the resistant patients were substantially higher (P < 0.001). CONCLUSIONS Excessive GSH intake may contribute to chemotherapy resistance in breast cancer, and the levels of intracellular GSH and antioxidant enzymes are elevated in resistant patients after neoadjuvant chemotherapy, indicating that the standardization of GSH intake may assist in reducing chemotherapy resistance.
Collapse
Affiliation(s)
- Zhiyuan Zhang
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Jiaru Gao
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Linjiao Jia
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Shuxin Kong
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Maosen Zhai
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Shuai Wang
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Wenwen Li
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Shoukai Wang
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Yuqing Su
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Wanyue Li
- Department of Radiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Changzheng Zhu
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanxiang Lu
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China.
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China.
| | - Wentao Li
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China.
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China.
| |
Collapse
|
42
|
Liu Y, Wang C, Li M, Zhu Y, Liu K, Liu Y, Luo M, Zhang C. Natural ingredients in the regulation of abnormal lipid peroxidation: a potential therapy for pulmonary diseases. Front Pharmacol 2024; 15:1507194. [PMID: 39759448 PMCID: PMC11695318 DOI: 10.3389/fphar.2024.1507194] [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: 10/07/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Pulmonary diseases are a major category of diseases that pose a threat to human health. The most common drugs currently used to treat lung diseases are still chemical drugs, but this may lead to drug resistance and damage to healthy organs in the body. Therefore, developing new drugs is an urgent task. Lipid peroxidation is caused by the disruption of redox homeostasis, accumulation of reactive oxygen species (ROS), depletion of glutathione (GSH), and inactivation of glutathione peroxidase 4 (GPX4). Lipid peroxidation is closely related to the occurrence and progression of respiratory diseases, including acute lung injury, asthma, pulmonary fibrosis, pulmonary hypertension, chronic obstructive pulmonary disease, and lung cancer. Natural ingredients have high safety, high availability, and low cost, and can regulate lipid peroxidation through multiple pathways and targets, making them valuable new drugs. This article aims to summarize the pharmacology and mechanism of natural ingredients targeting lipid peroxidation in the treatment of lung diseases. The reviewed data indicate that natural ingredients are a promising anti-lipid peroxidation drug, mainly alleviating lipid peroxidation through the cystine/glutamate antiporter (System Xc -)/GSH/GPX4 axis, Nrf2 pathway, and ROS pathway. In the future, it will still be necessary to further study the mechanisms of natural products in treating pulmonary diseases through lipid peroxidation and conduct multi-center, large-sample clinical trials to promote the development of new drugs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
43
|
Kovács P, Schwarcz S, Nyerges P, Bíró TI, Ujlaki G, Bai P, Mikó E. Anticarcinogenic effects of ursodeoxycholic acid in pancreatic adenocarcinoma cell models. Front Cell Dev Biol 2024; 12:1487685. [PMID: 39723238 PMCID: PMC11668698 DOI: 10.3389/fcell.2024.1487685] [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: 08/28/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Changes to the composition of the microbiome in neoplasia, is termed oncobiosis, may affect tumor behavior through the changes to the secretion of bacterial metabolites. In this study we show, that ursodeoxycholic acid (UDCA), a bacterial metabolite, has cytostatic properties in pancreatic adenocarcinoma cell (PDAC) models. UDCA in concentrations corresponding to the human serum reference range suppressed PDAC cell proliferation. UDCA inhibited the expression of epithelial mesenchymal transition (EMT)-related markers and invasion capacity of PDAC cells. UDCA treatment increased oxidative/nitrosative stress by reducing the expression of nuclear factor, erythroid 2-like 2 (NRF2), inducing inducible nitric oxide synthase (iNOS) and nitrotyrosine levels and enhancing lipid peroxidation. Furthermore, UDCA reduced the expression of cancer stem cell markers and the proportion of cancer stem cells. Suppression of oxidative stress by antioxidants, blunted the UDCA-induced reduction in cancer stemness. Finally, we showed that UDCA induced mitochondrial oxidative metabolism. UDCA did not modulate the effectiveness of chemotherapy agents used in the chemotherapy treatment of pancreatic adenocarcinoma. The antineoplastic effects of UDCA, observed here, may contribute to the induction of cytostasis in PDAC cell models by providing a more oxidative/nitrosative environment.
Collapse
Affiliation(s)
- Patrik Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szandra Schwarcz
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Petra Nyerges
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tímea Ingrid Bíró
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
44
|
Hossain R, Noonong K, Nuinoon M, Majima HJ, Eawsakul K, Sompol P, Rahman MA, Tangpong J. Network Pharmacology, Molecular Docking, and In Vitro Insights into the Potential of Mitragyna speciosa for Alzheimer's Disease. Int J Mol Sci 2024; 25:13201. [PMID: 39684911 DOI: 10.3390/ijms252313201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Mitragyna speciosa Korth. Havil (MS) has a traditional use in relieving pain, managing hypertension, treating cough, and diarrhea, and as a morphine substitute in addiction recovery. Its potential in addressing Alzheimer's disease (AD), a neurodegenerative condition with no effective treatments, is under investigation. This study aims to explore MS mechanisms in treating AD through network pharmacology, molecular docking, and in vitro studies. Using network pharmacology, we identified 19 MS components that may affect 60 AD-related targets. The compound-target network highlighted significant interactions among 60 nodes and 470 edges, with an average node degree of 15.7. The KEGG enrichment analysis revealed Alzheimer's disease (hsa05010) as a relevant pathway. We connected 20 targets to tau and β-amyloid proteins through gene expression data from the AlzData database. Docking studies demonstrated high binding affinities of MS compounds like acetylursolic acid, beta-sitosterol, isomitraphylline, and speciophylline to AD-related proteins, such as AKT1, GSK3B, NFκB1, and BACE1. In vitro studies showed that ethanolic (EE), distilled water (DWE), and pressurized hot water (PHWE) extracts of MS-treated 100 μM H2O2-induced SH-SY5Y cells significantly reduced oxidative damage. This research underscores the multi-component, multi-target, and multi-pathway effects of MS on AD, providing insights for future research and potential clinical applications.
Collapse
Affiliation(s)
- Rahni Hossain
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat 80160, Thailand
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Kunwadee Noonong
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Manit Nuinoon
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
- Hematology and Transfusion Science Research Center, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Hideyuki J Majima
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Komgrit Eawsakul
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Pradoldej Sompol
- Department of Pharmacology & Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Jitbanjong Tangpong
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
45
|
Eroglu B, Isales C, Eroglu A. Age and duration of obesity modulate the inflammatory response and expression of neuroprotective factors in mammalian female brain. Aging Cell 2024; 23:e14313. [PMID: 39230054 PMCID: PMC11634740 DOI: 10.1111/acel.14313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/09/2024] [Accepted: 07/27/2024] [Indexed: 09/05/2024] Open
Abstract
Obesity has become a global epidemic and is associated with comorbidities, including diabetes, cardiovascular, and neurodegenerative diseases, among others. While appreciable insight has been gained into the mechanisms of obesity-associated comorbidities, effects of age, and duration of obesity on the female brain remain obscure. To address this gap, adolescent and mature adult female mice were subjected to a high-fat diet (HFD) for 13 or 26 weeks, whereas age-matched controls were fed a standard diet. Subsequently, the expression of inflammatory cytokines, neurotrophic/neuroprotective factors, and markers of microgliosis and astrogliosis were analyzed in the hypothalamus, hippocampus, and cerebral cortex, along with inflammation in visceral adipose tissue. HFD led to a typical obese phenotype in all groups independent of age and duration of HFD. However, the intermediate duration of obesity induced a limited inflammatory response in adolescent females' hypothalamus while the hippocampus, cerebral cortex, and visceral adipose tissue remained unaffected. In contrast, the prolonged duration of obesity resulted in inflammation in all three brain regions and visceral adipose tissue along with upregulation of microgliosis/astrogliosis and suppression of neurotrophic/neuroprotective factors in all brain regions, denoting the duration of obesity as a critical risk factor for neurodegenerative diseases. Importantly, when female mice were older (i.e., mature adult), even the intermediate duration of obesity induced similar adverse effects in all brain regions. Taken together, our findings suggest that (1) both age and duration of obesity have a significant impact on obesity-associated comorbidities and (2) early interventions to end obesity are critical to preserving brain health.
Collapse
Affiliation(s)
- Binnur Eroglu
- Department of Neuroscience and Regenerative MedicineMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
| | - Carlos Isales
- Department of Neuroscience and Regenerative MedicineMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
- Department of MedicineMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
| | - Ali Eroglu
- Department of Neuroscience and Regenerative MedicineMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
- Department of Obstetrics and GynecologyMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
46
|
Yao C, Li Z, Sun K, Zhang Y, Shou S, Jin H. Mitochondrial dysfunction in acute kidney injury. Ren Fail 2024; 46:2393262. [PMID: 39192578 PMCID: PMC11360640 DOI: 10.1080/0886022x.2024.2393262] [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: 06/13/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Acute kidney injury (AKI) is a systemic clinical syndrome increasing morbidity and mortality worldwide in recent years. Renal tubular epithelial cells (TECs) death caused by mitochondrial dysfunction is one of the pathogeneses. The imbalance of mitochondrial quality control is the main cause of mitochondrial dysfunction. Mitochondrial quality control plays a crucial role in AKI. Mitochondrial quality control mechanisms are involved in regulating mitochondrial integrity and function, including antioxidant defense, mitochondrial quality control, mitochondrial DNA (mtDNA) repair, mitochondrial dynamics, mitophagy, and mitochondrial biogenesis. Currently, many studies have used mitochondrial dysfunction as a targeted therapeutic strategy for AKI. Therefore, this review aims to present the latest research advancements on mitochondrial dysfunction in AKI, providing a valuable reference and theoretical foundation for clinical prevention and treatment of this condition, ultimately enhancing patient prognosis.
Collapse
Affiliation(s)
- Congcong Yao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Ziwei Li
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Keke Sun
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Zhang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Songtao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Heng Jin
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
47
|
Zheng M, Chavda VP, Vaghela DA, Bezbaruah R, Gogoi NR, Patel K, Kulkarni M, Shen B, Singla RK. Plant-derived exosomes in therapeutic nanomedicine, paving the path toward precision medicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156087. [PMID: 39388922 DOI: 10.1016/j.phymed.2024.156087] [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: 04/06/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Plant-derived exosomes (PDEs), are nanoscale vesicles secreted by multivesicular bodies, play pivotal roles in critical biological processes, including gene regulation, cell communication, and immune defense against pathogens. Recognized for their potential health-promoting properties, PDEs are emerging as innovative components in functional nutrition, poised to enhance dietary health benefits. PURPOSE To describe the efficacy of PDEs in nanoform and their application as precision therapy in many disorders. STUDY DESIGN The design of this review was carried out in PICO format using randomized clinical trials and research articles based on in vivo and in vitro studies. METHODS All the relevant clinical and research studies conducted on plant-derived nanovesicle application and efficacy were included, as retrieved from PubMed and Cochrane, after using specific search terms. This review was performed to determine PDEs' efficacy as nanomedicine and precision therapy. Sub-group analysis and primary data were included to determine the relationship with PDEs. RESULT PDEs are extracted from plant materials using sophisticated techniques like precipitation, size exclusion, immunoaffinity capture, and ultracentrifugation, encapsulating vital molecules such as lipids, proteins, and predominantly microRNAs. Although their nutritional impact may be minimal in small quantities, the broader application of PDEs in biomedicine, particularly as vehicles for drug delivery, underscores their significance. They offer a promising strategy to improve the bioavailability and efficacy of therapeutic agents carrying nano-bioactive substances that exhibit anti-inflammatory, antioxidant, cardioprotective, and anti-cancer activities. CONCLUSION PDEs enhance the therapeutic potency of plant-derived phytochemicals, supporting their use in disease prevention and therapy. This comprehensive review explores the multifaceted aspects of PDEs, including their isolation methods, biochemical composition, health implications, and potential to advance medical and nutritional interventions.
Collapse
Affiliation(s)
- Min Zheng
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, 610218, China
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M College of Pharmacy, Ahmedabad 380009, Gujrat, India.
| | - Dixa A Vaghela
- Pharmacy section, L.M College of Pharmacy Ahmedabad 380009, Gujrat, India
| | - Rajashri Bezbaruah
- Department of Pharmacology, Dibrugarh University, Dibrugarh 786004, Assam
| | - Niva Rani Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh 786004, Assam
| | - Kaushika Patel
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, LJ University, Ahmedabad 382210, Gujarat, India
| | - Mangesh Kulkarni
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, LJ University, Ahmedabad 382210, Gujarat, India; Department of Pharmaceutics, Gandhinagar Institute of Pharmacy, Gandhinagar University, Moti Bhoyan, Khatraj-Kalol Road 382721, Gujarat, India
| | - Bairong Shen
- Institutes for Systems Genetics, West China Tianfu Hospital, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rajeev K Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| |
Collapse
|
48
|
Hassanein EHM, Althagafy HS, Mansour SMA, Omar ZMM, Hussein Hassanein MM, Abd El-Ghafar OAM. Vinpocetine attenuates 5-fluorouracil-induced intestinal injury: role of the Keap1/Nrf2/HO-1, NF-κB/TLR4/SOCS3 and RIPK1/RIPK3/MLKL signals. Immunopharmacol Immunotoxicol 2024; 46:884-892. [PMID: 39439043 DOI: 10.1080/08923973.2024.2415111] [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: 03/04/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES 5-Fluorouracil (5-FU) is a chemotherapy drug commonly prescribed in cancer management. Unfortunately, intestinal mucositis restricts 5-FU clinical use. Vinpocetine (VNP) is a synthetic alkaloid that is derived from vincamine. Our study was conducted to elucidate the intestinal protective effects of VNP on 5-FU intestinal injury in rats and explore the underlying mechanisms. MATERIALS AND METHODS 5-FU was injected i.p. for five days, while VNP was given P.O (5 and 10 mg/kg). RESULTS VNP effectively mitigates oxidative stress by a significant increase in GSH and SOD and decreasing MDA content mediated by Nrf2, HO-1 upregulation, and significant Keap1 downregulation. VNP mitigated inflammatory perturbations by decreasing MPO, TNF-α, IL-1β, and IL-6 facilitated by downregulating NF-κB and TLR4 and upregulating SOCS3 levels. In addition, the RIPK1, RIPK3, MLKL, and caspase-8 expression levels were significantly decreased, evidenced improvement of intestinal necroptosis by VNP. CONCLUSION Hence, VNP potently prevents intestinal injury induced by 5-FU by modulating Keap1/Nrf2/HO-1, NF-κB/TLR4/SOCS3, and RIPK1/RIPK3/MLKL signals.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Sherif M A Mansour
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Assiut, Egypt
| | - Zainab M M Omar
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Assiut, Egypt
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | | | - Omnia A M Abd El-Ghafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| |
Collapse
|
49
|
Kang JH, Kim DH, Yoo J, Shin JH, Kim JH, Lee JW, Shin SH. Sinapine suppresses ROS-induced C2C12 myoblast cell death through MAPK and autophagy pathways. Food Sci Biotechnol 2024; 33:3629-3637. [PMID: 39493388 PMCID: PMC11525351 DOI: 10.1007/s10068-024-01718-6] [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: 06/06/2024] [Revised: 09/05/2024] [Accepted: 09/18/2024] [Indexed: 11/05/2024] Open
Abstract
Oxidative stress in skeletal muscle can lead to muscle atrophy through reactive oxygen species (ROS)-induced damage and cell death. tert-Butyl hydroperoxide (TBHP), an exogenous ROS generator, induces oxidative stress and cell death in various cells. Sinapine from cruciferous plants possesses beneficial effects, but its role in protecting skeletal muscle cells against ROS-induced cell death remains unclear. This study demonstrates that sinapine pretreatment significantly reduced TBHP-induced cell death and ROS accumulation in a dose-dependent manner. TBHP activated mitogen-activated protein kinase (MAPK) pathways including Akt, p38, and JNK, and triggered autophagy. Sinapine suppressed the phosphorylation of Akt, MEK3/6, p38, MEK4, and JNK, and modulated key autophagy markers. Notably, the co-treatment of MAPK inhibitors attenuated TBHP-induced cell death and LC3B-II accumulation. These findings suggest that sinapine is a promising phytochemical for mitigating oxidative stress-mediated muscle injury, offering potential therapeutic strategies for maintaining skeletal muscle homeostasis and addressing muscle-related pathologies.
Collapse
Affiliation(s)
- Jung Hyun Kang
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Dong Hwan Kim
- Department of Bio & Medical Bigdata (BK4 Program), Gyeongsang National University, Jinju, 52828 South Korea
| | - Jin Yoo
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Jun Hong Shin
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Ju Hyun Kim
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Ji Won Lee
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
| | - Seung Ho Shin
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828 South Korea
- Department of Bio & Medical Bigdata (BK4 Program), Gyeongsang National University, Jinju, 52828 South Korea
| |
Collapse
|
50
|
Ferrara F, Valacchi G. Role of microbiota in the GUT-SKIN AXIS responses to outdoor stressors. Free Radic Biol Med 2024; 225:894-909. [PMID: 39505118 DOI: 10.1016/j.freeradbiomed.2024.11.003] [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] [Received: 08/09/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Beside the respiratory tract, the skin and the gut represent the first defensive lines of our body against the external insults displaying many important biochemical features able to maintain the epithelial barrier integrity and to regulate the tissue immune responses. The human microbiome is essential in maintaining the tissue homeostasis and its dysregulation may lead to tissue conditions including inflammatory pathologies. Among all external insults, air pollutants have been shown to cause oxidative stress damage within the target tissues via an OxInflammatory response. Dysregulation of the gut microbiome (dysbiosis) by outdoor stressors, including air pollutants, may promote the exacerbation of the skin tissue damage via the interplay between the gut-skin axis. The intent of this review is to highlight the ability of exogenous stressors to modulate the human gut-skin axis via a redox regulated mechanism affecting the microbiome and therefore contributing to the development and aggravation of gut and skin conditions.
Collapse
Affiliation(s)
- Francesca Ferrara
- Department of Chemical, Pharmaceuticals and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121, Ferrara, Italy; Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC, 28081, USA; Kyung Hee University, Department of Food and Nutrition, Seoul, South Korea.
| |
Collapse
|