1
|
Wu Y, Pei J, Xu Y, Yu F, Xu S. Selenium: 48-year journey of global clinical trials. Mol Cell Biochem 2025; 480:3253-3265. [PMID: 39755855 DOI: 10.1007/s11010-024-05202-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: 10/22/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
Selenium, an essential trace mineral for health, has seen a rise in clinical trials over the past nearly 5 decades. Our aim here is to provide a comprehensive and concise overview of selenium clinical trials from 1976 to 2023. Overall, the evolution of selenium clinical trials over 48 years has advanced through phases of emergence, prosperity, and either stability or transition. The USA plays pivotal roles in establishing large research clusters and fostering strong collaborative ties of selenium clinical trials. Low-selenium levels are noted in a higher proportion of selenium observational trials, while selenium intervention trials are delineated by nine key functional classifications. The emphasis in intervention trials is that selenium product development should be on conducting clinical trials in diseases with higher efficacy, such as those involving antioxidant and endocrine and metabolic disease. Moreover, inorganic forms such as sodium selenite and semi-organic forms like selenized yeast were recognized as primary sources of selenium, while nano-selenium has emerged as a new selenium source in clinical treatments. Selenium is mainly consumed through tablets and oral administration, with a recommended upper limit of 200 µg per day for managing most diseases. In addition, genes encoding selenoproteins or factors of relevance for selenium metabolism, inflammation, and immunity, which have a higher number of records in all trials, are poised to steer future investigations into functional mechanisms of selenium. We believe this review will offer fresh perspectives on selenium clinical trials and identify potential avenues for future selenium research.
Collapse
Affiliation(s)
- Yikun Wu
- Guizhou University Medical College, Guizhou University, Guiyang, 550025, China
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Jun Pei
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400015, China
| | - Yuangao Xu
- Clinic for Kidney and Hypertension Diseases, Hannover Medical School, 30625, Hannover, Germany
| | - Fuxun Yu
- NHC Key Laboratory of Pulmonary Immunological Disease, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Shuxiong Xu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| |
Collapse
|
2
|
Xia Y, Chen K, Wang Y, Jiang Q, Du Y, Luo D, Li X, Li S. Importance of Selenoprotein O in Regulating Hmgb1: A New Direction for Modulating ROS-Dependent NETs Formation to Aggravate the Progression of Acute Liver Inflammation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9382-9397. [PMID: 40189811 DOI: 10.1021/acs.jafc.5c01956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Selenoproteins (Sels) are a class of essential biomolecules that play critical roles in cellular homeostasis. SelO was identified as the preferential source of selenium in the liver, implying its potential as a key regulatory factor in hepatic pathophysiology. Bioinformatics analysis of data from GEO data sets revealed marked downregulation of SelO in liver injury. However, its function and regulatory mechanisms in the liver remain unclear. To address this, we investigated the effect of SelO ablation on acute liver inflammation, focusing on its association with inflammation and neutrophil extracellular traps (NETs) formation. Wild-type (WT) and SelO-knockout mice were used to establish a lipopolysaccharide (LPS) exposure model and a coculture model (AML12 cells and neutrophils) in vitro. Our findings revealed that LPS stimulation significantly reduced SelO expression in the WT mouse liver. SelO deletion promoted the expression of Hmgb1 and marker cytokines for chemokines, NETs generation, pyroptosis and inflammation, and induced an imbalance in redox homeostasis. Immunofluorescence, SYTOX staining, and scanning electron microscopy confirmed that SelO silencing promoted reactive oxygen species (ROS)-dependent NETs formation. Moreover, the coculture model demonstrated that excessive NETs formation exacerbated SelO-ablation-induced hepatic inflammation. Importantly, we confirmed the significant involvement of the Hmgb1/ROS axis in the development of acute liver inflammation in the absence of SelO. Our results demonstrated that SelO ablation promoted neutrophil recruitment and enhanced ROS-dependent NETs formation by increasing Hmgb1 expression levels, thereby aggravating LPS-induced pyroptosis and inflammation. This study not only uncovered the crucial biological functions of SelO, but also shed light on its regulatory implications in the inflammatory process.
Collapse
Affiliation(s)
- Yu Xia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Animal Science College, Hebei North University, Zhangjiakou 075000, China
| | - Kai Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yidan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qihang Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yongzhen Du
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Dongliu Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiang Li
- National Selenium-Rich Product Quality Supervision and Inspection Center, Enshi 445000, China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
3
|
Ma SY, Liu YM, Wang J. Potential bidirectional regulatory effects of botanical drug metabolites on tumors and cardiovascular diseases based on the PI3K/Akt/mTOR pathway. Front Pharmacol 2025; 16:1467894. [PMID: 40196368 PMCID: PMC11973345 DOI: 10.3389/fphar.2025.1467894] [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: 07/21/2024] [Accepted: 03/11/2025] [Indexed: 04/09/2025] Open
Abstract
Pharmacological interventions targeting the phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway are predominantly employed as anticancer therapies, yet they are frequently associated with significant cardiac toxicity. Additionally, the PI3K/Akt/mTOR pathway plays a crucial role in the treatment of cardiovascular diseases, highlighting its dual significance in both oncology and cardiology. Therefore, the PI3K/Akt/mTOR pathway has become an ideal signaling pathway for studying cardioprotection, anticancer effects, and their associated cardiac toxicity. Botanical drugs have emerged as a significant source for developing therapeutic agents with anticancer and cardioprotective effects, often exhibiting bidirectional protective properties. Consequently, this study investigates the bidirectional regulatory influence of botanical drug metabolites in oncology and cardiology via the PI3K/Akt/mTOR pathway. The research indicated that the PI3K/Akt/mTOR signaling pathway plays a critical regulatory role in the pathogenesis of both tumors and cardiovascular diseases. The botanical drug metabolites Ruscogenin, Sulforaphane, Naringenin, Kaempferol, Poncirin, and Puerarin can improve cancer by inhibiting the phosphorylation levels within the PI3K/Akt/mTOR signaling cascade. Moreover, they also provide cardioprotective effects in cardiac injury conditions by activating the phosphorylation levels of the PI3K/Akt/mTOR pathway. Therefore, the phosphorylation dynamics of key components in the PI3K/Akt/mTOR pathway, particularly the phosphorylation of Akt, along with the functional implications of different phosphorylation sites, may offer new therapeutic strategies and insights for cancer treatment and the mitigation of cardiotoxicity associated with cancer therapies.
Collapse
Affiliation(s)
| | | | - Jie Wang
- Guang’anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| |
Collapse
|
4
|
de Jager C, Soliman E, Theus MH. Interrogating mediators of single-cell transcriptional changes in the acute damaged cerebral cortex: Insights into endothelial-astrocyte interactions. Mol Cell Neurosci 2025; 133:104003. [PMID: 40090391 DOI: 10.1016/j.mcn.2025.104003] [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: 11/26/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025] Open
Abstract
Traumatic brain injury (TBI) induces complex cellular and molecular changes, challenging recovery and therapeutic development. Although molecular pathways have been implicated in TBI pathology, the cellular specificity of these mechanisms remains underexplored. Here, we investigate the role of endothelial cell (EC) EphA4, a receptor tyrosine kinase receptor involved in axonal guidance, in modulating cell-specific transcriptomic changes within the damaged cerebral cortex. Utilizing single-cell RNA sequencing (scRNA-seq) in an experimental TBI model, we mapped transcriptional changes across various cell types, with a focus on astrocytes and ECs. Our analysis reveals that EC-specific knockout (KO) of EphA4 triggers significant alterations in astrocyte gene expression and shifts predominate subclusters. We identified six distinct astrocyte clusters (C0-C5) in the damaged cortex including as C0-Mobp/Plp1+; C1-Slc1a3/Clu+; C2-Hbb-bs/Hba-a1/Ndrg2+; C3-GFAP/Lcn2+; C4-Gli3/Mertk+, and C5-Cox8a+. We validate a new Sox9+ cluster expressing Mertk and Gas, which mediates efferocytosis to facilitate apoptotic cell clearance and anti-inflammatory responses. Transcriptomic and CellChat analyses of EC-KO cells highlights upregulation of neuroprotective pathways, including increased amyloid precursor protein (APP) and Gas6. Key pathways predicted to be modulated in astrocytes from EC-KO mice include oxidative phosphorylation and FOXO signaling, mitochondrial dysfunction and ephrin B signaling. Concurrently, metabolic and signaling pathways in endothelial cells-such as ceramide and sphingosine phosphate metabolism and NGF-stimulated transcription-indicate an adaptive response to a metabolically demanding post-injury hypoxic environment. These findings elucidate potential interplay between astrocytic and endothelial responses as well as transcriptional networks underlying cortical tissue damage.
Collapse
Affiliation(s)
- Caroline de Jager
- Translational Biology Medicine and Health Graduate Program, Blacksburg, VA 24061, USA
| | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA; Center for Engineered Health, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
5
|
De Bartolo A, Angelone T, Rocca C. Elucidating emerging signaling pathways driving endothelial dysfunction in cardiovascular aging. Vascul Pharmacol 2025; 158:107462. [PMID: 39805379 DOI: 10.1016/j.vph.2025.107462] [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: 10/28/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
The risk for developing cardiovascular diseases dramatically increases in older individuals, and aging vasculature plays a crucial role in determining their morbidity and mortality. Aging disrupts endothelial balance between vasodilators and vasoconstrictors, impairing function and promoting pathological vascular remodeling. In this Review, we discuss the impact of key and emerging molecular pathways that transduce aberrant inflammatory signals (i.e., chronic low-grade inflammation-inflammaging), oxidative stress, and mitochondrial dysfunction in aging vascular compartment. We focus on the interplay between these events, which contribute to generating a vicious cycle driving the progressive alterations in vascular structure and function during cardiovascular aging. We also discuss the primary role of senescent endothelial cells and vascular smooth muscle cells, and the potential link between vascular and myeloid cells, in impairing plaque stability and promoting the progression of atherosclerosis. The aim of this summary is to provide potential novel insights into targeting these processes for therapeutic benefit.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
6
|
Sun T, Wang F, Qian M, Wang J, Guo M. Glycyrrhizin Alleviates the Damage Caused by Zearalenone and Protects the Glandular Stomach of Chickens. Animals (Basel) 2025; 15:489. [PMID: 40002971 PMCID: PMC11851901 DOI: 10.3390/ani15040489] [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: 12/17/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Zearalenone (ZEA) is a kind of mycotoxin that widely contaminates food and feed and poses a threat to poultry farming. As a natural extract, glycyrrhizin acid (GA) has antioxidant, antibacterial, and anti-inflammatory effects. Although studies have revealed the toxic effects of ZEA on the liver, the mechanism by which GA reduces ZEA's toxic protective glandular stomach remains unclear. In order to study the therapeutic effect of GA on tissue damage caused by ZEA, we conducted in vivo and in vitro experiments to compare the expression of inflammation, oxidative stress, apoptosis, and necrosis. The results showed that ZEA can induce inflammation in tissues and cells, inducing apoptosis and necrosis. In addition, GA can alleviate the toxic effects caused by ZEA and protect cells. Dietary GA significantly increased the antioxidant capacity of glandulae and inhibited the overexpression of NFκB/IκB-α and its mediated inflammatory response. Moreover, GA decreased the expression of pro-apoptotic factors and necrosis factors, thereby alleviating apoptosis and necrosis of chicken glandular stomach cells. At present, the mechanism of ZEA damage to livers and lungs has been confirmed by studies. However, there have been no studies on GA alleviating the damage caused by ZEA to the glandular stomach. Therefore, the purpose of this study was to explore the mechanism of GA alleviating the damage caused by ZEA in the glandular stomach through in vivo and in vitro experimental comparison. The results may provide some reference for the solution of feed contamination.
Collapse
Affiliation(s)
| | | | | | | | - Mengyao Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (T.S.); (F.W.); (M.Q.); (J.W.)
| |
Collapse
|
7
|
Swetha K, Indumathi MC, Kishan R, Siddappa S, Chen CH, Marathe GK. Selenium Mitigates Caerulein and LPS-induced Severe Acute Pancreatitis by Inhibiting MAPK, NF-κB, and STAT3 Signaling via the Nrf2/HO-1 Pathway. Biol Trace Elem Res 2025:10.1007/s12011-025-04531-2. [PMID: 39907886 DOI: 10.1007/s12011-025-04531-2] [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: 08/29/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025]
Abstract
Severe acute pancreatitis (SAP) leads to systemic inflammation, resulting in multiorgan damage. Acute lung injury and acute respiratory distress syndrome develop in one-third of SAP patients, with a high mortality rate of 60% due to secondary complications. Patients with pancreatitis often have selenium deficiency, and selenium supplements may provide beneficial effects. This study examined the protective role of selenium in a model of SAP induced by caerulein + lipopolysaccharide (cae + LPS). Mice were administered selenium (1 mg/kg) before being challenged with caerulein (6 injections of 50 μg/kg) and LPS (10 mg/kg). At 3 h after the last caerulein injection, blood was collected for estimating pancreatic enzymes and cytokine levels, and the mice were euthanized. We performed morphological and histological studies, measured levels of protease and oxidative stress markers and conducted western blot, ELISA, and RT-qPCR analyses. We examined lung tissue histologically and estimated myeloperoxidase levels. Selenium pretreatment significantly reduced pancreatic enzyme levels such as amylase, lipase, and proteases (specifically MMPs) and reversed tissue injury in the pancreas and lungs caused by cae + LPS. In addition, selenium-treated mice showed decreased levels of inflammatory markers and chemokines. Examination of the downstream inflammatory pathways confirmed the protective effect of selenium, which mediates its anti-inflammatory and antioxidant action by inhibiting the major inflammatory signaling pathways (MAPKs, NF-κB, and STAT3) and activating the phosphorylation of Nrf2 via Nrf2/HO-1 pathways. These findings suggest that selenium may be a potential therapeutic option for treating SAP-associated secondary complications.
Collapse
Affiliation(s)
- Kamatam Swetha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri Mysore, 570006, India
| | | | - Raju Kishan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri Mysore, 570006, India
| | - Shiva Siddappa
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education and Research, Mysore, 570015, India
| | - Chu-Huang Chen
- Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX, 77030, USA
| | - Gopal K Marathe
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri Mysore, 570006, India.
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri Mysore, 570006, India.
| |
Collapse
|
8
|
Yang J, Geng Y, Zhao B, Liu T, Luo JL, Gao XJ. Green tea polyphenols alleviate TBBPA-induced gastric inflammation and apoptosis by modulating the ROS-PERK/IRE-1/ATF6 pathway in mouse models. Food Funct 2024; 15:10179-10189. [PMID: 39301672 DOI: 10.1039/d4fo03012e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Green tea polyphenols (GTP), an important phytochemical in the daily human diet, bind to various cellular receptors and exert anti-inflammatory and antioxidant benefits. The environmental contaminant tetrabromobisphenol A (TBBPA) enters the digestive system through multiple pathways, resulting in oxidative stress (OS), gastroenteritis, and mucosal injury. The aim of this study was to explore the molecular mechanisms of TBBPA-induced gastritis in mice treated with GTP in vivo and in an in vitro model. The results showed that exposure to TBBPA increased reactive oxygen species (ROS) levels, activated oxidative stress (OS) induced endoplasmic reticulum stress (ERS), and the expression of endoplasmic reticulum stress-related factors (e.g., GRP78, PERK, IRE-1, ATF-6, etc.) increased. The inflammatory pathway NF-κB was activated, and the pro-inflammatory factors TNF-α, IL-1β, and IL-6 increased, while triggering a cascade reaction mediated by caspase-3. However, the addition of GTP could inhibit OS, restore the balance of endoplasmic reticulum homeostasis, and improve the inflammatory infiltration and apoptosis of gastric mucosal epithelial cells. Therefore, GTP alleviated ERS, reduced inflammation and apoptosis, and restored the gastric mucosal barrier by alleviating TBBPA-induced OS in mouse gastric tissues and GES-1 cells. This provides basic information for exploring the antioxidant mechanism of GTP and further investigating the toxic effects of TBBPA on mouse gastric mucosa.
Collapse
Affiliation(s)
- Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China.
| | - Yuan Geng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China.
| | - Bing Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China.
| | - Tianjing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China.
| | - Ji-Long Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China.
| | - Xue-Jiao Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China.
| |
Collapse
|
9
|
Wang YC, Shueng PW, Hu CY, Tung FI, Chen MH, Liu TY. Hyaluronic acid-based injectable formulation developed to mitigate metastasis and radiation-induced skin fibrosis in breast cancer treatment. Carbohydr Polym 2024; 336:122136. [PMID: 38670762 DOI: 10.1016/j.carbpol.2024.122136] [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: 12/07/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024]
Abstract
The standard treatment for early-stage breast cancer involves breast-conserving surgery followed by adjuvant radiotherapy. However, approximately 20 % of patients experience distant metastasis, and adjuvant radiotherapy often leads to radiation-induced skin fibrosis (RISF). In this study, we develop an on-site injectable formulation composed of selenocystamine (SeCA) and hyaluronic acid (HyA), referred to as SeCA cross-linked HyA (SCH) agent, and investigate its potential to mitigate metastasis and prevent RISF associated with breast cancer therapy. SCH agents are synthesized using the nanoprecipitation method to modulate cell-cell tight junctions and tissue inflammation. The toxicity assessments reveal that SCH agents with a higher Se content (Se payload 17.4 μg/mL) are well tolerated by L929 cells compared to SeCA (Se payload 3.2 μg/mL). In vitro, SCH agents significantly enhance cell-cell tight junctions and effectively mitigate migration and invasion of breast cancer cells (4T1). In vivo, SCH agents mitigate distant lung metastasis. Furthermore, in animal models, SCH agents reduce RISF and promote wound repair. These findings highlight the potential of SCH agents as a novel therapeutic formulation for effectively mitigating metastasis and reducing RISF. This holds great promise for improving clinical outcomes in breast cancer patients undergoing adjuvant radiotherapy.
Collapse
Affiliation(s)
- Yu-Chi Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chan-Yu Hu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Fu-I Tung
- Department of Orthopaedics, Yang-Ming Branch, Taipei City Hospital, Taipei 111024, Taiwan; Department of Health and Welfare, College of City Management, University of Taipei, Taipei 111036, Taiwan
| | - Ming-Hong Chen
- Division of Neurosurgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan; Department of Electrical Engineering, Yuan Ze University, Taoyuan City 320315, Taiwan
| | - Tse-Ying Liu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan.
| |
Collapse
|
10
|
Sun X, Xu S, Liu T, Wu J, Yang J, Gao XJ. Zinc supplementation alleviates oxidative stress to inhibit chronic gastritis via the ROS/NF-κB pathway in a mouse model. Food Funct 2024; 15:7136-7147. [PMID: 38887927 DOI: 10.1039/d4fo01142b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Zinc (Zn) is an important trace element; it is involved in the regulation and maintenance of many physiological functions in organisms and has anti-inflammatory and antioxidant properties. Chronic gastritis is closely associated with damage to the gastric mucosa, which is detrimental to the health of humans and animals. There are few studies on the effects of zinc on, for example, gastric mucosal damage, oxidative stress, inflammation and cell death in mice. Therefore, we established in vivo and in vitro models of inflammatory injury and investigated the effects of zinc supplementation in C57BL/6 mice and Ges-1 cells and examined the expression of factors associated with oxidative stress, inflammation and cell death. In this study, the results of in vivo and in vitro experiments showed that reactive oxygen species (ROS) levels increased after sodium salicylate exposure. Malondialdehyde levels increased, the activity of the antioxidant enzymes catalase and superoxide dismutase decreased, and the activity of glutathione decreased. The NF-κB signaling pathway was activated, the levels of proinflammatory factors (TNF-α, IL-1β, and IL-6) increased, and the expression of cell death-related factors (Bax, Bcl-2, Caspase3, Caspase7, Caspase9, RIP1, RIP3, and MLKL) increased. Zinc supplementation attenuated the level of oxidative stress and reduced the level of inflammation and cell death. Our study indicated that sodium salicylate induced the production of large amounts of reactive oxygen species and activated the NF-κB pathway, leading to inflammatory damage and cell death in the mouse stomach. Zinc supplementation modulated the ROS/NF-κB pathway, reduced the level of oxidative stress, and attenuated inflammation and cell death in the mouse stomach and Ges-1 cells.
Collapse
Affiliation(s)
- Xiaoran Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Shuang Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Tianjing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Jiawei Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Xue-Jiao Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
11
|
Opriș-Belinski D, Cobilinschi CO, Caraiola S, Ungureanu R, Cotae AM, Grințescu IM, Cobilinschi C, Andrei AC, Țincu R, Ene R, Mirea L. Trace Element Deficiency in Systemic Sclerosis-Too Much Effort for Some Traces? Nutrients 2024; 16:2053. [PMID: 38999801 PMCID: PMC11242991 DOI: 10.3390/nu16132053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Trace elements are essential for several physiological processes. To date, various data have suggested that inadequate levels of trace elements may be involved in the pathogenesis of different chronic diseases, including immune-mediated ones, or may develop during their course. Systemic sclerosis (SSc) is a complex autoimmune multisystemic disease, primarily characterized by microvascular dysregulation, the widespread activation of the immune system and tissue fibrosis. According to the latest reports regarding the pathogenesis of SSc, the main pathophysiological processes-inflammation, vasculopathy and fibrosis-may include various trace element derangements. The present literature review aims to update the available data regarding iron, zinc, copper and selenium status in SSc as well as to underline the possible implications of these trace elements in the complexity of the pathogenic process of the disease. We observe that the status of trace elements in SSc plays a crucial role in numerous pathogenic processes, emphasizing the necessity for proper monitoring and supplementation. The reported data are heterogenous and scarce, and future studies are needed in order to draw clearer conclusions about their complete spectrum.
Collapse
Affiliation(s)
- Daniela Opriș-Belinski
- Department of Internal Medicine Rheumatology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Rheumatology and Internal Medicine, Sfânta Maria Clinical Hospital, 011172 Bucharest, Romania
| | - Claudia Oana Cobilinschi
- Department of Internal Medicine Rheumatology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Rheumatology and Internal Medicine, Sfânta Maria Clinical Hospital, 011172 Bucharest, Romania
| | - Simona Caraiola
- Department of Internal Medicine Rheumatology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Rheumatology and Internal Medicine, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Raluca Ungureanu
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | - Ana-Maria Cotae
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | - Ioana Marina Grințescu
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | - Cristian Cobilinschi
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | - Andrei Cosmin Andrei
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | - Radu Țincu
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
- Department of Clinical Toxicology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Răzvan Ene
- Department of Orthopedics, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Liliana Mirea
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
12
|
Lv H, Wang J, Geng Y, Xu T, Han F, Gao XJ, Guo MY. Green tea polyphenols inhibit TBBPA-induced lung injury via enhancing antioxidant capacity and modulating the NF-κB pathway in mice. Food Funct 2024; 15:3411-3419. [PMID: 38470815 DOI: 10.1039/d4fo00480a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Tetrabromobisphenol A (TBBPA) is a global pollutant. When TBBPA is absorbed by the body through various routes, it can have a wide range of harmful effects on the body. Green tea polyphenols (GTPs) can act as antioxidants, resisting the toxic effects of TBBPA on animals. The effects and mechanisms of GTP and TBBPA on oxidative stress, inflammation and apoptosis in the mouse lung are unknown. Therefore, we established in vivo and in vitro models of TBBPA exposure and GTP antagonism using C57 mice and A549 cells and examined the expression of factors related to oxidative stress, autophagy, inflammation and apoptosis. The results of the study showed that the increase in reactive oxygen species (ROS) levels after TBBPA exposure decreased the expression of autophagy-related factors Beclin1, LC3-II, ATG3, ATG5, ATG7 and ATG12 and increased the expression of p62; oxidative stress inhibits autophagy levels. The increased expression of the pro-inflammatory factors IL-1β, IL-6 and TNF-α decreased the expression of the anti-inflammatory factor IL-10 and activation of the NF-κB p65/TNF-α pathway. The increased expression of Bax, caspase-3, caspase-7 and caspase-9 and the decreased expression of Bcl-2 activate apoptosis-related pathways. The addition of GTP attenuated oxidative stress levels, restored autophagy inhibition and reduced the inflammation and apoptosis levels. Our results suggest that GTP can attenuate the toxic effects of TBBPA by modulating ROS, reducing oxidative stress levels, increasing autophagy and attenuating inflammation and apoptosis in mouse lung and A549 cells. These results provide fundamental information for exploring the antioxidant mechanism of GTP and further for studying the toxic effects of TBBPA.
Collapse
Affiliation(s)
- Hongli Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Jingjing Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Yuan Geng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Tianchao Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Fuxin Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Xue-Jiao Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Meng-Yao Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
13
|
Wei C, Xu T, Geng Y, Yang J, Lv H, Guo MY. High-fat diet disrupts the gut microbiome, leading to inflammation, damage to tight junctions, and apoptosis and necrosis in Nyctereutes procyonoides intestines. Microbiol Spectr 2024; 12:e0418223. [PMID: 38376358 PMCID: PMC10986597 DOI: 10.1128/spectrum.04182-23] [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: 12/12/2023] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
Given the burgeoning Nyctereutes procyonoides breeding industry and its growing scale, it is imperative to investigate the impact of high-fat diets on the health of these animals. This study involved 30 male Nyctereutes procyonoides of comparable weights (3 kg ±0.5), randomly assigned to either a control group or a high-fat diet group (n = 15 each). The latter group was fed a mixture of lard and basal diet in a 2:5 ratio, establishing a high-fat diet model in Nyctereutes procyonoides. This diet induced diarrhea and histopathological changes in the Nyctereutes procyonoides. Analysis of the small intestine contents using 16S rRNA sequencing revealed a high-fat diet-induced disruption in the gut microbiota. Specifically, Escherichia-Shigella emerged as the biomarker in the high-fat diet group (P = 0.049), while Vagococcus was prevalent in the control group (P = 0.049), indicating a significant increase in harmful bacteria in the high-fat diet group. Furthermore, this disrupted gut flora correlated with inflammation and oxidative stress, as evidenced by marked increases in TNF-α (P < 0.01), IL-1β (P < 0.05), and IL-6 (P < 0.05) levels, measured via q-PCR, Western blot, and oxidative stress assays. In addition, q-PCR analysis revealed significant upregulation of apoptosis and necrosis markers, including Bax, Caspase3, Caspase9, Caspase12, RIPK3, and RIPK1 (P < 0.01 to P < 0.001), and a concurrent downregulation of the anti-apoptotic gene Bcl-2 (P < 0.01) in the high-fat diet group, consistent with protein expression trends. These findings suggest that a high-fat diet alters the gut microbiome toward a more harmful bacterial composition, escalating inflammatory responses and intestinal tissue permeability, culminating in intestinal cell apoptosis and necrosis.IMPORTANCEThis study examines the impact of high-fat diets on Nyctereutes procyonoides. Our research established a Nyctereutes procyonoides model on a high-fat diet, revealing significant health impacts, such as diarrhea, histological anomalies, and alterations in the gut microbiota. These findings emphasize the importance of preventing health issues and promoting sustainable industry growth. They highlight the significant impact of diet on gut microbiota and overall animal health.
Collapse
Affiliation(s)
- Chengwei Wei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Dongbeinongda Animal Hospital Ltd., Harbin, China
| | - Tianchao Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Geng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hongli Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Meng-yao Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|