1
|
Yin W, Xue H, Zhang Y, Li R, Liu M, Yue H, Ge D, Liu N. Steroid constituents of Solidago canadensis alleviate LPS-induced inflammation via AMPK regulated mitophagy/NLRP3 and NF-κB pathway. Eur J Pharmacol 2025; 998:177512. [PMID: 40113066 DOI: 10.1016/j.ejphar.2025.177512] [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: 10/18/2024] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Inflammation is a major risk factor for a variety of human diseases, such as sepsis, Inflammatory Bowel Disease (IBD) and also major cardiovascular disease including atherosclerosis. Solidago canadensis is used as a traditional medicine to treat inflammation-related diseases. However, the component with anti-inflammatory activity of Solidago canadensis is not clear. In this study, we aimed to search for new bioactive steroids from Solidago canadensis and investigate their anti-inflammatory activity both in vitro and in vivo. Lipopolysaccharides (LPS)-stimulated RAW264.7 cells, mouse bone marrow-derived macrophages (BMDMs) and peripheral blood mononuclear cells (PBMCs) were used to induce an inflammation response. Compound 10 outperformed other compounds for superior anti-inflammatory activity and significant inhibition of NLR family, pyrin domain containing 3 (NLRP3) inflammasome activation. Mechanistically, compound 10 induced mitophagy by activating AMP-activated protein kinas (AMPK) to suppress NLRP3 inflammasome activation. Inhibiting AMPK by inhibitor BML-275 significantly attenuated compound 10 induced mitophagy and subsequent the NLRP3 inflammasome. Besides, the NF-κB activation, key step in NLRP3 inflammasome priming, was also suppressed by compound 10 via activation of AMPK. In addition, the in vivo experiments showed that compound 10 could alleviate LPS-induced inflammatory and dextran sulfate sodium salt -induced colitis in C57BL/6 mice. Collectively, the present study, for the first time, shows that the steroids compound 10 exhibited anti-inflammatory effect via AMPK/mitophagy/NLRP3 as well as AMPK/NF-κB/NLRP3 signaling pathway, which strongly suggests the therapeutic potential of compound 10 in various inflammatory diseases.
Collapse
Affiliation(s)
- Wenying Yin
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Han Xue
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Yongqi Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Rongxian Li
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Mengjia Liu
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Hongwei Yue
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China.
| | - Di Ge
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Na Liu
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| |
Collapse
|
2
|
Ma C, Yu X, Zhang X, Su L, Jiang O, Cui R. Combination of radiotherapy and ICIs in advanced hepatocellular carcinoma: A systematic review of current evidence and future prospects (Review). Oncol Lett 2025; 30:342. [PMID: 40438865 PMCID: PMC12117537 DOI: 10.3892/ol.2025.15088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/24/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a global health concern because of its rising prevalence and high fatality rates. Conventional treatments for advanced HCC (aHCC) have limited success, emphasizing the need for novel treatment options. Radiotherapy (RT) treatments, such as stereotactic body radiation and proton therapy, improve local tumor management via precision targeting. Moreover, immune checkpoint inhibitors (ICIs) that target the programmed cell death protein 1(PD-1)/PD ligand 1 (PD-L1) and cytotoxic T lymphocyte associated protein 4 (CTLA-4) pathways have promise for systemic antitumor effectiveness. The combination of RT and ICIs takes advantage of their complementary mechanisms: RT kills immunogenic cells and controls the tumor microenvironment to increase antigen presentation, whereas ICIs enhance and maintain antitumor immune responses. This combination enhances tumor regression and immune response in aHCC, improving response rate and progression-free survival with manageable safety. The present review aimed to summarize the rationale for combining RT + ICIs in patients with aHCC and clinical outcomes, as well as ways to enhance this combination technique. The combination of these models is a promising technique for improving outcomes for patients with aHCC and warrants further investigation.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Xinlin Yu
- Department of Oncology, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Xialin Zhang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Lihong Su
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Ou Jiang
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Ran Cui
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| |
Collapse
|
3
|
Li YM, Li CX, Jureti R, Awuti G. Identification and Validation of Ferritinophagy-Related Biomarkers in Periodontitis. Int Dent J 2025; 75:1781-1797. [PMID: 40233623 PMCID: PMC12043013 DOI: 10.1016/j.identj.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/19/2025] [Accepted: 03/01/2025] [Indexed: 04/17/2025] Open
Abstract
OBJECTIVE While ferritinophagy is believed to play a significant role in the development of periodontitis, the exact mechanisms remain unclear. This study aimed to investigate the biomarkers associated with ferritinophagy in periodontitis using transcriptomic data. METHODS Two periodontitis-related datasets from Gene Expression Omnibus, GSE10334, and GSE16134, served as the training and validation cohorts, respectively. Additionally, 36 ferritinophagy-related genes (FRGs) were obtained from the GeneCards database. We compared the expression differences of FRGs between the periodontitis and control groups, identifying the different FRGs as candidates. Weighted gene coexpression network analysis (WGCNA) was applied to capture the key modules and modular genes related to periodontitis, utilizing the candidate FRG scores as trait. Then we intersected these with key module genes to identify differentially expressed FRGs. Hub genes were filtered using a protein-protein interaction network. Ultimately, biomarkers were acquired through machine learning, receiver operating characteristic curves, and expression levels. In addition, biomarker-associated immune cells and functional pathways were analysed to predict the upstream regulatory molecules. RESULTS In total, 18 candidate FRGs showed significant differences between the periodontitis and control groups, and from the protein-protein interaction network, eight hub genes were identified among the 175 differentially expressed FRGs by analysing 1096 differentially expressed genes and 4479 key modular genes. Eventually, ALDH2, diazepam binding inhibitor, HMGCR, OXCT1, and ACAT2 were identified as potential biomarkers through machine learning algorithms, receiver operating characteristic curve analysis, and gene expression assessments. In addition, resting dendritic cells, mast cells, and follicular helper T cells were positively correlated with the five biomarkers (Cor > 0.3 and P < .05). All five biomarkers are involved in the translation initiation pathway, including transcription factors like KLF5 and microRNAs such as hsa-miR-495-3p and hsa-miR-27a-3p. Reverse transcription-quantitative polymerase chain reaction analysis showed that all biomarkers were expressed at low levels in the periodontitis group. However, the differences in expression levels for OXCT1 and ACAT2 between groups were not statistically significant. CONCLUSIONS A total of five ferritinophagy-related biomarkers - ALDH2, diazepam binding inhibitor, HMGCR, OXCT1, and ACAT2 - were screened to explore new treatment options for periodontitis.
Collapse
Affiliation(s)
- Yi-Ming Li
- Department of Periodontology, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, National Clinical Medical Research Institute, Urumqi, China
| | - Chen-Xi Li
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, National Clinical Medical Research Institute, Urumqi, China; Dental Medicine Institute of Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Reyila Jureti
- Department of Periodontology, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, National Clinical Medical Research Institute, Urumqi, China
| | - Gulinuer Awuti
- Department of Periodontology, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, National Clinical Medical Research Institute, Urumqi, China
| |
Collapse
|
4
|
Yang H, Yang Y, Cui G, Xu Y, Zhao R, Le G, Xie Y, Li P. Dietary methionine restriction ameliorates atherosclerosis by remodeling the gut microbiota in apolipoprotein E-knockout mice. Food Funct 2025. [PMID: 40421996 DOI: 10.1039/d5fo00841g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Dietary methionine restriction (MR) has been shown to reduce the risk of atherosclerosis, but the specific regulatory effects and mechanisms remain unclear. This research intends to investigate the effects of MR on atherosclerosis in apolipoprotein E-knockout (ApoE-/-) mice fed a high-fat, high-cholesterol, high-choline diet and their mechanisms. ApoE-/- mice were fed a normal diet (0.86% methionine + 4.5% fat + 0% cholesterol + 0.2% choline), a high-fat, high-cholesterol, high-choline diet (0.86% methionine + 20% fat + 1% cholesterol + 1% choline), or a high-fat, high-cholesterol, high-choline + MR diet (0.17% methionine + 20% fat + 1% cholesterol + 1% choline) for 8 consecutive weeks. The results show that MR reduced body weight, fat mass, fat deposition in the liver and adipocytes, and plasma lipid levels; improved the morphological structure of the aorta; and reduced the aortic lesion area and lipid levels. In addition, MR downregulated aortic pro-inflammatory cytokine levels, upregulated aortic anti-inflammatory cytokine levels, and improved aortic oxidative stress. Moreover, metagenomic sequencing results suggested that MR improved the gut microbiota composition, particularly through increased relative abundance of short-chain fatty acid (SCFA)-producing bacteria, and changed the relative abundance of inflammation-, lipid metabolism-, and bile acid metabolism-related bacteria at the species level. Furthermore, MR promoted SCFA production and bile acid metabolism, and reduced cell adhesion molecules and foam cell formation in the aorta. Thus, our findings indicated that MR improved the gut microbiota composition, especially increased SCFA production, and ameliorated oxidative stress and inflammation in the aorta, thereby preventing atherosclerosis.
Collapse
Affiliation(s)
- Hao Yang
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
| | - Yuhui Yang
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
- Institute for Complexity Science, Henan University of Technology, Zhengzhou 450001, Henan, China
| | - Guifang Cui
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
| | - Yuncong Xu
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Renyong Zhao
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
| | - Guowei Le
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yanli Xie
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
| | - Peng Li
- Institute for Complexity Science, Henan University of Technology, Zhengzhou 450001, Henan, China
| |
Collapse
|
5
|
Shan W, Lan H, Wu Y, Xu Q, You M, Wu J. Fenofibrate maintains the integrity of the blood-brain barrier during cerebral ischemia-reperfusion injury by inhibiting Egr- 1. In Vitro Cell Dev Biol Anim 2025:10.1007/s11626-025-01044-z. [PMID: 40425900 DOI: 10.1007/s11626-025-01044-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/10/2025] [Indexed: 05/29/2025]
Abstract
Blood-brain barrier (BBB) damage and dysfunction are critical pathological features associated with cerebral ischemia-reperfusion injury in stroke. Fenofibrate, a lipid-regulating drug, has an unclear role in BBB function during stroke. This study investigates the effects of fenofibrate on BBB disruption and cerebrovascular endothelial cells induced by ischemia-reperfusion. Cerebral ischemia-reperfusion injury (CIRI) models were established using the middle cerebral artery occlusion (MCAO) method. Blood-brain barrier (BBB) integrity was assessed using Evans blue dye. The permeability of human brain microvascular endothelial cells (HBMVECs) was evaluated using fluorescein isothiocyanate (FITC)-dextran permeation assays and trans-endothelial electrical resistance (TEER) measurements. Additionally, real-time polymerase chain reaction (PCR), immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), and Western blot analysis were performed. We found that the administration of fenofibrate improved brain endothelial dysfunction by reducing the expression of vascular cell adhesion molecule- 1 (VCAM- 1) and E-selectin in MCAO mice. Furthermore, fenofibrate restored the expression of the tight junction protein occludin in the cortices of MCAO mice. Notably, fenofibrate alleviated BBB dysfunction in MCAO mice. In vitro studies demonstrated that fenofibrate ameliorated endothelial monolayer permeability under oxygen-glucose deprivation/reoxygenation (OGD/R) conditions and inhibited the expression of VCAM- 1 and E-selectin in HBMVECs. Moreover, fenofibrate restored occludin expression following OGD/R. We identified a novel mechanism whereby fenofibrate suppressed the elevation of Egr- 1 induced by OGD/R; however, overexpression of Egr- 1 abrogated the protective effects of fenofibrate on the upregulation of VCAM- 1 and E-selectin and the downregulation of occludin induced by OGD/R. Furthermore, overexpression of early growth response- 1 (Egr- 1) negated the protective effects of fenofibrate on endothelial monolayer permeability and trans-endothelial electrical resistance (TEER). Our findings suggest that fenofibrate may be a promising therapeutic agent for stroke treatment.
Collapse
Affiliation(s)
- Weifeng Shan
- Department of Anesthesiology, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, No. 15 Dazhong Street, Lishui, 323000, Zhejiang, China
| | - Haiyan Lan
- Department of Anesthesiology, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, No. 15 Dazhong Street, Lishui, 323000, Zhejiang, China
| | - Yini Wu
- Department of Anesthesiology, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, No. 15 Dazhong Street, Lishui, 323000, Zhejiang, China
| | - Qiaomin Xu
- Department of Anesthesiology, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, No. 15 Dazhong Street, Lishui, 323000, Zhejiang, China
| | - Minji You
- Department of Anesthesiology, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, No. 15 Dazhong Street, Lishui, 323000, Zhejiang, China
| | - Jimin Wu
- Department of Anesthesiology, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, No. 15 Dazhong Street, Lishui, 323000, Zhejiang, China.
| |
Collapse
|
6
|
Kan F, Wang D, Li S, Gao Y, Wang J. ITGA5 drives angiogenesis in diabetic retinopathy via TAK-1/NF-kB activation. Hum Cell 2025; 38:105. [PMID: 40410450 DOI: 10.1007/s13577-025-01233-8] [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: 09/01/2024] [Accepted: 05/11/2025] [Indexed: 05/25/2025]
Abstract
Diabetic retinopathy is a retinal damage, which causes vision impairment and blindness. Integrin Subunit Alpha 5 (ITGA5) regulates angiogenic response, but its roles in diabetic retinopathy remain unclear. In this work, diabetes mellitus was induced in rats by streptozotocin. ITGA5 interference was achieved by intravitreal delivery of adeno-associated virus. Upregulation of ITGA5 was found in diabetic rat retinal tissues. ITGA5 knockdown decreased the neovascularization, acellular capillary formation, and pericytes. The protein expression of vascular endothelial growth factor (VEGFA), vascular adhesion molecule-1(VCAM-1), and intercellular adhesion molecule-1 (ICAM-1) was reduced after ITGA5 interference. Besides, ITGA5 knockdown decreased the phosphorylation level of FAK, TAK-1, and p65. In vitro, rat retinal microvascular endothelial cells (RRMECs) were cultured under high glucose condition to stimulate diabetic environment. ITGA5 knockdown inhibited VEGFA secretion, tube formation, cell invasion, and migration. Upregulation of VCAM-1 and ICAM-1 that induced by high glucose was reversed by ITGA5 silencing. ITGA5 knockdown blocked the activation of TAK-1/NF-kB pathway in RRMECs. Additionally, in oxygen-induced retinopathy model, ITGA5 interference inhibited pathological neovascularization. These results demonstrate that ITGA5 contributes to the angiogenesis in diabetic retinopathy by activating TAK-1/NF-kB pathway.
Collapse
Affiliation(s)
- Feifei Kan
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Di Wang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Sijia Li
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Yi Gao
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Jianwen Wang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
7
|
Lai L, Wu C, Li X, Rong Y, Huang Y, Wang B. Urinary MCP-1 and VCAM-1 as non-invasive biomarkers for the diagnosis and activity assessment of lupus nephritis. PLoS One 2025; 20:e0323334. [PMID: 40388387 PMCID: PMC12088007 DOI: 10.1371/journal.pone.0323334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/05/2025] [Indexed: 05/21/2025] Open
Abstract
INTRODUCTION Accurate diagnosis of lupus nephritis (LN) and effective assessment of its disease activity are essential for optimal management. This study aimed to evaluate the potential of novel urinary biomarkers, MCP-1 and VCAM-1, in diagnosing and assessing LN activity, comparing their efficacy to traditional urinary biomarkers, and proposing a new standard for clinical application. METHODS A total of 55 LN patients who met the 1997 ACR diagnostic criteria for systemic lupus erythematosus (SLE) and 34 healthy controls (HCs) were included in this study. The LN patients were categorized into two groups based on their SLE disease activity indices (SLEDAI): the inactive lupus nephritis (NALN) group (SLEDAI 0-4, n = 32) and the active lupus nephritis (ALN) group (renal SLEDAI ≥ 4, n = 22). Additionally, the patients were further classified into mild (SLEDAI 5-9), moderate (SLEDAI 10-14), and severe (SLEDAI > 14) subgroups. All LN patients underwent testing for urinary MCP-1 (uMCP-1), urinary VCAM-1 (uVCAM-1), urinary α1-microglobulin (u-α1MG), urinary β2-microglobulin (u-β2MG), urinary IgG (u-IgG), and urinary albumin (u-ALB), as well as a percutaneous renal biopsy. RESULTS The levels of urinary MCP-1 and VCAM-1 (uMCP-1 and uVCAM-1) in the LN group were significantly elevated compared to the HCs (uMCP-1: P < 0.001; uVCAM-1: P < 0.01). Receiver operating characteristic (ROC) curve analysis revealed that the diagnostic efficacy of uMCP-1 and uVCAM-1 surpassed that of traditional biomarkers (uMCP-1: AUC = 0.79, P < 0.001; uVCAM-1: AUC = 0.77, P < 0.001). Multivariate logistic regression demonstrated a significant association between uMCP-1 and uVCAM-1 levels and the occurrence of LN (P < 0.001). Furthermore, these novel biomarkers exhibited stronger correlations with SLEDAI scores than traditional biomarkers (P < 0.001). Notably, patients with ALN had significantly higher levels of uMCP-1 and uVCAM-1 compared to those with NALN (uMCP-1: P < 0.01; uVCAM-1: P < 0.01). CONCLUSION The production of uMCP-1 and uVCAM-1 is closely associated with the onset and progression of LN (ISN/RPS: Class I - IV). These biomarkers may serve as valuable references for the diagnosis and prediction of LN and aid in the assessment of LN activity.
Collapse
Affiliation(s)
- Lichuan Lai
- Department of Laboratory, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Chunle Wu
- Department of Blood Transfusion, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Xiaohua Li
- Department of Rheumatology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yuxiang Rong
- Department of Laboratory, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Ying Huang
- Department of Laboratory, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Bangqin Wang
- Department of Rheumatology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| |
Collapse
|
8
|
Zhang B, Li Q, Wang W, Tian M, Xu D, Xie Y. PFOS and Its Substitute OBS Cause Endothelial Dysfunction to Promote Atherogenesis in ApoE -/- Mice. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:526-538. [PMID: 40400551 PMCID: PMC12090012 DOI: 10.1021/envhealth.4c00206] [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: 10/05/2024] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 05/23/2025]
Abstract
Perfluorooctanesulfonate (PFOS), an emerging contaminant with widespread concern, has been associated with the pathogenesis of atherosclerosis (AS). As a substitute for PFOS, sodium p-perfluorous nonenoxybenzenesulfonate (OBS) is extensively utilized in various applications and detected in human blood. However, its potential health risk in AS remain unclear. In this study, we investigated the comparative impacts of PFOS and OBS on endothelial dysfunction and atherogenesis. In the in vivo study, Apolipoprotein E knockout (ApoE-/-) mice were exposed to 0.4 or 4 mg/L PFOS/OBS for 12 weeks. We found that dyslipidemia developed more rapidly in the OBS-exposed mice than in the PFOS-exposed mice. PFOS exhibited a higher enrichment capacity in both blood and aortic tissues than OBS. Remarkably, OBS induced a more pronounced inflammatory response and caused a more significant disruption of the endothelial barrier in the aorta of ApoE-/- mice compared to PFOS. In vitro experiments showed that OBS, at the same exposure concentrations and durations as PFOS (0.1-20 μmol/L, 48 h), more effectively inhibited cell viability of human umbilical vein endothelial cells (HUVECs), caused higher levels of lactate dehydrogenase (LDH) release, and enhanced cell adhesion between HUVECs and monocytes. Both PFOS and OBS were found to activate the NF-κB signaling pathway and upregulate the expression of inflammatory factors. Notably, the use of OBS, but not PFOS, was shown to disrupt cell junctions and increase endothelial permeability by activating the MAPK/ERK signaling pathway. Our findings suggest that OBS may lead to endothelial dysfunction and have a greater impact on AS compared to PFOS, presenting significant health risks in cardiovascular diseases.
Collapse
Affiliation(s)
- Boxiang Zhang
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Qing Li
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Wensheng Wang
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Mingming Tian
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Dan Xu
- Institute
of Environmental Systems Biology, Environment Science and Engineering
College, Dalian Maritime University, Linghai Road 1, Dalian 116026, China
| | - Ying Xie
- The
Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian 116023, China
| |
Collapse
|
9
|
Bai W, Chen S, Liu T, Tang X, Xiong L, Li M, Guo R, He Q. Dual-action membrane-chimeric liposomes with self-reinforcing targeting for acute lung injury treatment. J Control Release 2025; 383:113820. [PMID: 40349780 DOI: 10.1016/j.jconrel.2025.113820] [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: 12/17/2024] [Revised: 05/01/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Acute lung injury (ALI) is a common acute and critical syndrome with high mortality. The uncontrollable feedback loop of inflammation is the primary cause of death in patients with ALI. Therefore, targeting the inflammatory site and breaking the inflammatory loop are key strategies for ALI treatment. Our work developed a myeloid cell membrane-chimeric liposome containing dexamethasone (DEX) and leukocyte adhesin-1 (LA-1), abbreviated as ML/LA@DEX NPs. During the preparation of ML/LA@DEX NPs, LA-1 could activate CD11b on the myeloid cell membrane, thereby improving the binding ability of ML/LA@DEX NPs to ICAM-1 on endothelial cells in pulmonary inflammatory lesions, and achieving self-reinforcing targeting of ALI. ML/LA@DEX NPs accumulated at the inflammatory lesions would competitively occupy the binding site of neutrophils to reduce their recruitment. Meanwhile, ML/LA@DEX NPs would release DEX to inhibit the cytokine storm. Through this two-pronged approach, ML/LA@DEX NPs effectively broke the positive feedback loop of inflammation and showed significant therapeutic effects on ALI, providing a new strategy for ALI treatment.
Collapse
Affiliation(s)
- Wenjing Bai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Shuang Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Tingting Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xian Tang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Lin Xiong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Rong Guo
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Melnik BC, Weiskirchen R, John SM, Stremmel W, Leitzmann C, Weiskirchen S, Schmitz G. White Adipocyte Stem Cell Expansion Through Infant Formula Feeding: New Insights into Epigenetic Programming Explaining the Early Protein Hypothesis of Obesity. Int J Mol Sci 2025; 26:4493. [PMID: 40429638 PMCID: PMC12110815 DOI: 10.3390/ijms26104493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/03/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Prolonged breastfeeding (BF), as opposed to artificial infant formula feeding (FF), has been shown to prevent the development of obesity later in life. The aim of our narrative review is to investigate the missing molecular link between postnatal protein overfeeding-often referred to as the "early protein hypothesis"-and the subsequent transcriptional and epigenetic changes that accelerate the expansion of adipocyte stem cells (ASCs) in the adipose vascular niche during postnatal white adipose tissue (WAT) development. To achieve this, we conducted a search on the Web of Science, Google Scholar, and PubMed databases from 2000 to 2025 and reviewed 750 papers. Our findings revealed that the overactivation of mechanistic target of rapamycin complex 1 (mTORC1) and S6 kinase 1 (S6K1), which inhibits wingless (Wnt) signaling due to protein overfeeding, serves as the primary pathway promoting ASC commitment and increasing preadipocyte numbers. Moreover, excessive protein intake, combined with the upregulation of the fat mass and obesity-associated gene (FTO) and a deficiency of breast milk-derived microRNAs from lactation, disrupts the proper regulation of FTO and Wnt pathway components. This disruption enhances ASC expansion in WAT while inhibiting brown adipose tissue development. While BF has been shown to have protective effects against obesity, the postnatal transcriptional and epigenetic changes induced by excessive protein intake from FF may predispose infants to early and excessive ASC commitment in WAT, thereby increasing the risk of obesity later in life.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Swen Malte John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany;
- Institute for Interdisciplinary Dermatological Prevention and Rehabilitation (iDerm), University of Osnabrück, D-49076 Osnabrück, Germany
| | | | - Claus Leitzmann
- Institut für Ernährungswissenschaft, Universität Gießen, D-35392 Gießen, Germany;
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Gerd Schmitz
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|
11
|
Crump KB, Kanelis E, Segarra-Queralt M, Pascuet-Fontanet A, Bermudez-Lekerika P, Alminnawi A, Geris L, Alexopoulos LG, Noailly J, Gantenbein B. TNF induces catabolism in human cartilaginous endplate cells in 3D agarose culture under dynamic compression. Sci Rep 2025; 15:15849. [PMID: 40328789 PMCID: PMC12056083 DOI: 10.1038/s41598-025-00538-w] [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: 02/18/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025] Open
Abstract
Intervertebral disc (IVD) degeneration is the leading cause of low back pain in young adults, and the cartilaginous endplate (CEP) is likely to play a key role in early IVD degeneration. To elucidate the effects of pro-inflammatory cytokines on the mechanobiology of the CEP, human CEP cells were seeded into 2% agarose, dynamically compressed up to 7%, and stimulated with tumor necrosis factor (TNF). It was hypothesized that dynamic compression would be sufficient to induce anabolism, while stimulation with TNF would induce catabolism. TNF was sufficient to induce a catabolic, time-dependent response in human CEP cells through downregulation of anabolic gene expression and increased secretion of pro-inflammatory proteins associated with herniated discs, bacteria inhibition, and pain. However, 7% strain or scaffold material, agarose, may not lead to full activation of integrins and downregulation of pro-inflammatory pathways, demonstrated in part through the unchanged gene expression of integrin subunits α5 and β1.
Collapse
Affiliation(s)
- Katherine B Crump
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012, Bern, Switzerland
| | - Exarchos Kanelis
- School of Mechanical Engineering, National Technical University of Athens, 15772, Zografou, Greece
- Protavio Ltd, 15341, Agia Paraskevi, Greece
| | | | | | - Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012, Bern, Switzerland
| | - Ahmad Alminnawi
- GIGA In Silico Medicine, University of Liège, Liège, 4000, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000, Leuven, Belgium
| | - Liesbet Geris
- GIGA In Silico Medicine, University of Liège, Liège, 4000, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000, Leuven, Belgium
| | - Leonidas G Alexopoulos
- School of Mechanical Engineering, National Technical University of Athens, 15772, Zografou, Greece
- Protavio Ltd, 15341, Agia Paraskevi, Greece
| | - Jérôme Noailly
- BCN Medtech, Universitat Pompeu Fabra, 08018, Barcelona, Spain
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland.
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Faculty of Medicine, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
12
|
Lv Z, Xie X. LINC00265 Serves as a Diagnostic and Prognostic Marker for Acute Ischemic Stroke and INHIBITS Disease Progression Through the miR- 155 - 5p/TRIM32 Axis. Mol Neurobiol 2025:10.1007/s12035-025-04973-8. [PMID: 40317416 DOI: 10.1007/s12035-025-04973-8] [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: 01/09/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Acute ischemic stroke (AIS) is a medical emergency stemming from a reduction in cerebral blood supply, leading to neuronal injury. LINC00265 may emerge as a highly promising biomarker for stroke. The aim of this investigation was to investigate the clinical and prognostic significance of LINC00265 in AIS, as well as to elucidate the molecular mechanisms through which LINC00265 influences AIS. Blood samples were collected from 131 AIS patients. qRT-PCR was employed to measure the expression levels of LINC00265. The diagnostic and prognostic value of LINC00265 were evaluated using ROC curve and Kaplan-Meier survival curves, respectively. To establish an in vitro model of AIS, HCMIEC/D3 cells were subjected to OGD/R treatment. Cell proliferation was assessed using CCK-8 assay. The concentrations of TNF-α, IL-6, IL-1β, IL-4, and IL-10 were measured using ELISA kit. The protein expression levels of VCAM-1 and ICAM-1 were detected by Western blot analysis. The expression of LINC00265 was found to be significantly downregulated in AIS patients and strongly correlated with disease severity. The reduced expression of LINC00265 exhibited considerable significance for both diagnosis and prognosis prediction of AIS. At the mechanistic level, LINC00265 mitigated OGD/R-induced cellular injury by modulating the miR-155-5p/TRIM32 axis. Therefore, overexpression of LINC00265 may potentially suppress disease progression in AIS by regulating miR-155-5p/TRIM32 axis.
Collapse
Affiliation(s)
- Zhicheng Lv
- Department of Neurosurgery, The First People's Hospital of Chenzhou, Chenzhou, 423000, China
| | - Xiaoming Xie
- Department of Neurosurgery Ward II, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), No. 818, Renmin Road, Changde, 415000, China.
| |
Collapse
|
13
|
Borda MG, Ramírez-Vélez R, Botero-Rodriguez F, Patricio-Baldera J, de Lucia C, Pola I, Barreto GE, Khalifa K, Bergland AK, Kivipelto M, Cederholm T, Zetterberg H, Ashton NJ, Ballard C, Siow R, Aarsland D. Anthocyanin supplementation in adults at risk for dementia: a randomized controlled trial on its cardiometabolic and anti-inflammatory biomarker effects. GeroScience 2025:10.1007/s11357-025-01669-8. [PMID: 40314845 DOI: 10.1007/s11357-025-01669-8] [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: 02/20/2025] [Accepted: 04/15/2025] [Indexed: 05/03/2025] Open
Abstract
Anthocyanins are dietary flavonoids shown to have a therapeutic capacity to mitigate inflammation and oxidative stress. The present secondary analyses from the "Anthocyanins in People at Risk for Dementia Study" were aimed at (I) determining the intervention's effect on blood-based markers of cardiovascular disease and inflammation and (II) evaluating whether baseline factors such as age, sex, inflammation, or cardiometabolic score may moderate the intervention's effect on inflammatory status. This study was an ancillary, 24-week randomized, double-blind, placebo-controlled Phase II trial. Sub-sample participants (n = 99), aged 60-80 years with mild cognitive impairment or cardiometabolic disorders, were randomized to receive either 320 mg/day of anthocyanins or placebo. The biomarkers analyzed included inflammatory biomarker assessment (IL - 6, IL - 8, IL - 10, IL - 1b, TNF - α, IFN - γ), and C-reactive protein (CRP), as well as albumin, thrombocytes, cholesterol, LDL, HDL, and triglycerides, which were longitudinally compared between both groups. Baseline characteristics were balanced between the groups. ANCOVA analyses reveal 24-week differences favoring the anthocyanin treatment in LDL cholesterol levels (ƞp2 = 0.078; p = 0.015), cardiometabolic score (ƞp2 = 0.073; p = 0.021), CRP levels (ƞp2 = 0.417; p = 0.0001), IL - 6 (ƞp2 = 0.085; p = 0.015), IL - 1b (ƞp2 = 0.058; p = 0.037), and Inflam z-score 5 (ƞp2 = 0.059, p = 0.004). Moderation analysis demonstrated that the inflammatory score at baseline was a significant predictor of the effect of the intervention on the CRP levels. Anthocyanin supplementation reduces CRP and cardiovascular disease biomarkers in individuals at risk of dementia, especially when there is increased inflammation at baseline. ClinicalTrials.gov study identifier: NCT03419039.
Collapse
Affiliation(s)
- Miguel German Borda
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway.
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación en Ciencias de La Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan Edo. de México, México.
| | - Robinson Ramírez-Vélez
- Navarrabiomed, IdiSNA, Hospital Universitario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Felipe Botero-Rodriguez
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- SynaptIA - Inteligencia artificial para la investigación en salud mental, Bogotá, Colombia
- Centro de Memoria y Cognición Intellectus, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Jonathan Patricio-Baldera
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Instituto de Investigación en Salud, Facultad de Ciencias de La Salud de La Universidad Autónoma de Santo Domingo, Santo Domingo, Dominican Republic
| | - Chiara de Lucia
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Ilaria Pola
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Khadija Khalifa
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
| | - Anne Katrine Bergland
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Tommy Cederholm
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, 62167, Uppsala, Sweden
- Theme Inflammation & Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK, Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicholas J Ashton
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Banner Alzheimer's Institute, University of Arizona, Phoenix, AZ, USA
| | - Clive Ballard
- Medical School, University of Exeter, University of Exeter, Exeter, UK
| | - Richard Siow
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Dag Aarsland
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| |
Collapse
|
14
|
El-Dessouki AM, Alzokaky AA, Raslan NA, Ibrahim S, Selim HMRM, Al-Karmalawy AA. Dabigatran attenuates methotrexate-induced hepatotoxicity by regulating coagulation, endothelial dysfunction, and the NF-kB/IL-1β/MCP-1 and TLR4/NLRP3 signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5129-5145. [PMID: 39527308 DOI: 10.1007/s00210-024-03567-w] [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: 08/08/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
This study examines Dabigatran's (Dab) capacity to mitigate methotrexate (MTX)-induced coagulation disorders and endothelial dysfunction, while exploring its effects on oxidative stress and inflammatory pathways (NF-kB/IL-1β/MCP-1, TLR4/NLRP3) in reducing hepatotoxicity. Rats were assigned to four groups: a control group receiving saline intraperitoneally (i.p.); an MTX group with a single MTX dose (20 mg/kg, i.p.) to induce hepatotoxicity; and two pretreatment groups receiving Dab orally at 15 mg/kg and 25 mg/kg for seven days before and 4 days after MTX administration. MTX-treated rats showed significant increases in liver enzymes (ALT, AST, ALP) and reductions in antioxidant enzymes (SOD, GSH), along with elevated coagulation parameters (tissue factor (TF), thrombin, fibrin, plasminogen activator inhibitor-1 (PAI-1)), leading to coagulation disorders. Endothelial dysfunction was evident with reduced eNOS expression, while inflammation increased through elevated iNOS, ICAM-1, and pro-inflammatory cytokines (MPO, NF-kB, TNF-α, IL-1β, MCP-1), alongside activation of the TLR4/NLRP3 inflammasome pathway and decreased IL-10 (p < 0.05). Immunohistochemistry revealed increased cytochrome c and caspase-3 expression, with histopathological damage. Dabigatran mitigated these effects, downregulating liver enzymes, modulating coagulation factors, restoring eNOS levels, and reducing histopathological and inflammatory markers. Dabigatran demonstrates significant therapeutic potential in alleviating methotrexate-induced hepatotoxicity through its antioxidant, anti-inflammatory, anticoagulant, and anti-apoptotic effects. Its regulation of coagulation parameters and endothelial function suggests a protective role against tissue damage, warranting further investigation.
Collapse
Affiliation(s)
- Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University (ACU), 6th of October City, 12566, Giza, Egypt.
| | - Amany A Alzokaky
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11651, Egypt
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Horus University, New Damietta, 34518, Egypt
| | - Nahed A Raslan
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11651, Egypt
- Clinical Pharmacy Department, College of Health Sciences and Nursing, Al-Rayan Colleges, AL-Madinah AL-Munawarah, Saudi Arabia
| | - Samar Ibrahim
- Pharmacy Practice and Clinical Pharmacy Department, Faculty of Pharmacy, Galala University-Ataka, Suez, Egypt
| | - Heba Mohammed Refat M Selim
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, 11597, Riyadh, Saudi Arabia
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad, 10023, Iraq.
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University, New Damietta, 34518, Egypt.
| |
Collapse
|
15
|
Simões‐Pires EN, Torrente D, Singh P, Strickland S, Norris EH. Synergistic effects of the Aβ/fibrinogen complex on synaptotoxicity, neuroinflammation, and blood-brain barrier damage in Alzheimer's disease models. Alzheimers Dement 2025; 21:e70119. [PMID: 40344319 PMCID: PMC12061846 DOI: 10.1002/alz.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/16/2024] [Accepted: 01/12/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is characterized by amyloid-beta (Aβ), hyperphosphorylated tau, chronic neuroinflammation, blood-brain barrier (BBB) damage, and synaptic dysfunction, leading to neuronal loss and cognitive deficits. Vascular proteins, including fibrinogen, extravasate into the brain, further contributing to damage and inflammation. Fibrinogen's interaction with Aβ is well-established, but how this interaction contributes to synaptic dysfunction in AD is unknown. METHODS Organotypic hippocampal cultures (OHC) were exposed to Aβ42 oligomers, fibrinogen, or Aβ42/fibrinogen complexes. Synaptotoxicity was analyzed by Western blot. Aβ42 oligomers, fibrinogen, or their complexes were intracerebroventricularly injected into mice. Histopathological AD markers, synaptotoxicity, neuroinflammation, and vascular markers were observed by Western blot and immunofluorescence. RESULTS Aβ42/fibrinogen complexes led to synaptic loss, tau181 phosphorylation, neuroinflammation, and BBB disruption, independent of Mac1/CD11b receptor signaling. Blocking Aβ42/fibrinogen complex formation prevented synaptotoxicity. DISCUSSION These findings indicate that the Aβ42/fibrinogen complex has a synergistic impact on hippocampal synaptotoxicity and neuroinflammation. HIGHLIGHTS Fibrinogen binds to the central region of Aβ, forming a plasmin-resistant complex. The Aβ/fibrinogen complex induces synaptotoxicity, inflammation, and BBB disruption. Synaptotoxicity induced by the complex is independent of Mac1 receptor signaling.
Collapse
Affiliation(s)
- Elisa Nicoloso Simões‐Pires
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Daniel Torrente
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Pradeep Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| |
Collapse
|
16
|
Zhang T, Li L, Huang S, Starodubtseva MN, Liu J. Anti-endothelial cell antibodies in pathogenesis of vasculitis. Front Immunol 2025; 16:1567293. [PMID: 40370444 PMCID: PMC12075250 DOI: 10.3389/fimmu.2025.1567293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025] Open
Abstract
Vasculitis is a group of syndromes characterized by inflammation, presence of autoantibodies and endothelial cells (ECs) damage, which lead to stenosis or occlusion of the vascular lumen. Anti-endothelial cell antibodies (AECAs) are a heterogeneous group of autoantibodies in vasculitis. AECAs bind to antigens and membrane-bound proteins of ECs, inducing inflammation, coagulation, and apoptosis. In this review, we discuss the pathological role of AECAs in different types of vasculitis. In addition, AECAs potentially induce alterations of ECs mechanical properties, and subsequently promotes angiogenic phenotypes in the occurrence of vasculitis.
Collapse
Affiliation(s)
- Tian Zhang
- Laboratory of Translational Medicine in Microvascular Regulation, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Laboraotry for Future Industry in Gene Editing in Vascular Endothelial Cells of Universities of Shandong Province, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Liqun Li
- Department of Clinical Laboratory, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Shengshi Huang
- Laboratory of Translational Medicine in Microvascular Regulation, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Laboraotry for Future Industry in Gene Editing in Vascular Endothelial Cells of Universities of Shandong Province, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Maria N. Starodubtseva
- Gomel State Medical University, Gomel, Belarus
- Institute of Radiobiology of NAS of Belarus, Gomel, Belarus
| | - Ju Liu
- Laboratory of Translational Medicine in Microvascular Regulation, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Laboraotry for Future Industry in Gene Editing in Vascular Endothelial Cells of Universities of Shandong Province, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
17
|
Zawada AM, Griesshaber B, Ottillinger B, Erlenkötter A, Crook N, Boyington S, Stauss-Grabo M, Kennedy JP, Lang T. Development and Investigation of a New Polysulfone Dialyzer with Increased Membrane Hydrophilicity. MEMBRANES 2025; 15:132. [PMID: 40422742 DOI: 10.3390/membranes15050132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 05/28/2025]
Abstract
Innovation in dialysis care is fundamental to improve well-being and outcomes of patients with end-stage kidney disease. The dialyzer is the core element of dialysis treatments, as it largely defines which substances are removed from the patient's body. Moreover, its large surface size is the major place of interaction of the patient's blood with artificial surfaces and thus may lead to undesired effects such as inflammation or coagulation. In the present article we summarize the development path for a new dialyzer, including in vitro and clinical evidence generation. We use the example of the novel FX CorAL dialyzer, which has recently entered European and US markets, to show which steps are needed to develop and characterize a new dialyzer. The FX CorAL dialyzer includes a new hydrophilic membrane, which features reduced protein adsorption, sustained performance, and an improved hemocompatibility profile, characterized in numerous in vitro and clinical studies. Safety evaluations revealed a favorable profile, with low incidences of adverse device effects. Insights gained from both in vitro and clinical studies contribute to the advancement of dialyzer development, ultimately leading to improved patient care.
Collapse
Affiliation(s)
- Adam M Zawada
- Product Development, Fresenius Medical Care Deutschland GmbH, 66606 Sankt Wendel, Germany
| | - Bettina Griesshaber
- Clinical Research, Global Medical Office, Fresenius Medical Care Deutschland GmbH, 61352 Bad Homburg, Germany
| | | | - Ansgar Erlenkötter
- Biosciences, Fresenius Medical Care Deutschland GmbH, 66606 Sankt Wendel, Germany
| | - Nathan Crook
- Applications Laboratory, Fresenius Medical Care, Ogden, UT 84404, USA
| | - Skyler Boyington
- Applications Laboratory, Fresenius Medical Care, Ogden, UT 84404, USA
| | - Manuela Stauss-Grabo
- Clinical Research, Global Medical Office, Fresenius Medical Care Deutschland GmbH, 61352 Bad Homburg, Germany
| | - James P Kennedy
- Product Development, Fresenius Medical Care, Ogden, UT 84404, USA
| | - Thomas Lang
- Clinical Research, Global Medical Office, Fresenius Medical Care Deutschland GmbH, 61352 Bad Homburg, Germany
| |
Collapse
|
18
|
Tang W, Zhu L, Shi L, Gu B. P-Selectin, Vascular Endothelial Cadherin, and Vascular Cell Adhesion Molecule-1 as Novel Biomarkers for ABO Hemolytic Disease of the Fetus and Newborn. Anal Cell Pathol (Amst) 2025; 2025:9411137. [PMID: 40331036 PMCID: PMC12055318 DOI: 10.1155/ancp/9411137] [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: 09/24/2024] [Accepted: 03/22/2025] [Indexed: 05/08/2025] Open
Abstract
Objective: This study aims to assess the potential of vascular endothelial injury markers, namely, P-selectin (PS), vascular endothelial cadherin (VE-Cad), and vascular cell adhesion molecule-1 (VCAM-1), as diagnostic and prognostic biomarkers for ABO hemolytic disease of the fetus and newborn (HDFN). Methods: A total of 218 pregnant women with ABO blood group incompatibility were recruited from the Third People's Hospital of Bengbu Affiliated to Bengbu Medical University. The serum levels of PS, VCAM-1, and VE-Cad were measured, and the participants were followed up until postpartum. The women were divided into an HDFN group and a control group based on the occurrence of ABO-HDFN. The correlations between the three vascular endothelial injury markers, pregnant anti-A/B antibody titers, and the occurrence and severity of HDFN were analyzed. Results: Compared to the control group, the levels of PS, VCAM-1, and VE-Cad were significantly elevated in the HDFN group. Additionally, these markers increased with higher IgG anti-A/B titers. For diagnosing HDFN, the area under the curve (AUC) for PS, VCAM-1, and VE-Cad were 0.826, 0.765, and 0.799, respectively. Moreover, the combined AUC of the three markers with IgG anti-A/B titers was 0.9. The levels of the three biomarkers were significantly negatively correlated with neonatal hemoglobin (Hb) and significantly positively correlated with reticulocyte percentage (Ret%), indirect bilirubin (IBIL), and lactate dehydrogenase (LDH). Univariate logistic regression indicated that increased levels of PS, VCAM-1, and VE-Cad were associated with a higher probability of ABO-HDFN. Multivariate logistic regression revealed that PS is an independent positive factor for HDFN. Conclusion: PS, VCAM-1, and VE-Cad provide experimental evidence for prenatal screening, diagnosis, early prevention and treatment of ABO-HDFN.
Collapse
Affiliation(s)
- Weichun Tang
- School of Medical Technology, Xinxiang Medical University, Xinxiang 453000, Henan, China
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Linlin Zhu
- School of Medical Technology, Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Liwei Shi
- School of Medical Technology, Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Biao Gu
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233030, Anhui, China
| |
Collapse
|
19
|
Bravo L, Martínez-López S, Sierra-Cinos JL, Mateos R, Sarriá B. Yerba Mate (Ilex paraguariensis St. Hill.) Tea May Have Cardiometabolic Beneficial Effects in Healthy and At-Risk Subjects: A Randomized, Controlled, Blind, Crossover Trial in Nonhabitual Consumers. Mol Nutr Food Res 2025:e70065. [PMID: 40263915 DOI: 10.1002/mnfr.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/09/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025]
Abstract
Yerba mate has been reported to have antihypertensive, hypocholesterolemic, antidiabetic, or antiobesity properties. Most evidences from human trials involved intakes of high amounts of mate by habitual consumers. Considering its increasing popularity, this study aimed at assessing the potential cardiometabolic effects of moderate intake of yerba mate by nonhabitual consumers. A randomized, crossover, controlled study was carried out in healthy and hypercholesterolemic subjects. Anthropometric parameters, blood pressure, blood lipids, glucose metabolism, inflammatory cytokines, chemokines, and different markers of endothelial function, as well as incretins, adipocytokines, and different hormones were measured at baseline and after 8 weeks consuming yerba mate or a decaffeinated isotonic drink (control). After daily consumption of three servings of mate tea, blood pressure, inflammatory cytokines, chemokines, and colony-stimulating factors decreased in all participants. LDL-C decreased in normocholesterolemic individuals, while the mate and control interventions elicited similar hypolipidemic action in the hypercholesterolemic group. Ghrelin and glucose-dependent insulinotropic polypeptide (GIP) significantly decreased after mate intake, while glucagon-like peptide 1 (GLP-1) and adipocytokines remained unchanged. Body fat percentage and tricipital skinfold decreased only in healthy subjects, with no effects on total body weight. In conclusion, yerba mate could exert cardiometabolic protective effects in healthy consumers and in subjects at moderate cardiovascular risk. Trial Registration: This trial was retrospectively registered in ClinicalTrials (NCT06729905).
Collapse
Affiliation(s)
- Laura Bravo
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Sara Martínez-López
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Department of Nutrition and Food Science, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Jose Luis Sierra-Cinos
- Department of Nutrition and Food Science, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
- Department of Health Science, School of Health Science, Universidad International Isabel I de Burgos (Ui1), Burgos, Spain
| | - Raquel Mateos
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Beatriz Sarriá
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Department of Nutrition and Food Science, School of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
20
|
Joo EY, Park JS, Shin HT, Yoo M, Kim SJ, Lee JE, Choi GS. Mesenchymal Stem Cell Therapy for Hutchinson-Gilford Progeria: Improvements in Arterial Stiffness and Bone Mineral Density in a Single Case. CHILDREN (BASEL, SWITZERLAND) 2025; 12:523. [PMID: 40310235 PMCID: PMC12025413 DOI: 10.3390/children12040523] [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: 02/12/2025] [Revised: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND/OBJECTIVES Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder that cause premature aging due to LMNA mutations and progerin accumulation. Although lonafarnib, an FDA-approved farnesyltransferase inhibitor, offers modest extension of life, the disease remains progressive. As progeria is associated with stem cell depletion and mesenchymal stem cell (MSC) therapy has shown efficacy in treating atherosclerosis, we aimed to evaluate its efficacy and safety in HGPS. METHODS A 7-year-old male with classic HGPS and preexisting severe cerebrovascular disease received four intravenous infusion of bone marrow-derived MSCs (2.5 × 10⁵ cells/kg) over 8 months. Growth, metabolic, cardiovascular, musculoskeletal, auditory, and inflammatory cytokines were monitored throughout the study. Prophylactic enoxaparin was administered to prevent vascular complications. RESULTS MSC therapy was associated with improved lean body mass (11.5%), bone mineral density (L-spine z-score: 0.55 → 2.03), reduced arterial stiffness (9.98% reductionin pulse wave velocity), joint range of motion, dentition, and decreased sICAM-1 levels. However, Cardiovascular deterioration continued, and the patient passed away 10 months after the fourth dose, likely due to progression of the underlying vascular disease. No severe adverse effects were attributed to MSC therapy. CONCLUSIONS MSC therapy may offer short-term benefits in arterial stiffness, bone health and inflammation in HGPS without notable safety concerns. Further studies are warranted to validate these findings, explore earlier intervention, and determine long-term efficacy and optimal dosing strategies.
Collapse
Affiliation(s)
- Eun-Young Joo
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Ji-Sun Park
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Hyun-Tae Shin
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
- Department of Dermatology, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Myungji Yoo
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Su-Jin Kim
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Ji-Eun Lee
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Gwang-Seong Choi
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Department of Dermatology, Inha University College of Medicine, Incheon 22212, Republic of Korea
| |
Collapse
|
21
|
Lu J, Yin Z, Qiu Y, Yang Y, Chen Z, Wu J, Wang Z. Exploring the Role of Inflammation and Metabolites in Bell's Palsy and Potential Treatment Strategies. Biomedicines 2025; 13:957. [PMID: 40299566 PMCID: PMC12024589 DOI: 10.3390/biomedicines13040957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
Introduction: Bell's palsy is a common acute peripheral neurological disorder causing unilateral facial paralysis. Its exact etiology remains unknown, but it is linked to inflammation, immune responses, infections, and ischemia. This study explores the potential causal relationship between Bell's palsy and peripheral blood inflammatory proteins, metabolites, and immune cell characteristics. Methods: Genetic data for Bell's palsy were obtained from the Finnish database (version R10) and IEU OpenGWAS. A two-sample Mendelian randomization (MR) approach was applied, analyzing 4907 plasma proteins, 731 immune cell traits, 91 inflammatory proteins, and 1400 metabolites. The Finnish dataset served as the discovery cohort, while the IEU OpenGWAS dataset acted as the validation cohort. Bioinformatics analyses included protein-protein interaction (PPI) networks, Gene Ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, colocalization, and Linkage Disequilibrium Score Regression (LDSC) to identify candidate proteins and explore potential therapeutic targets. Results: MR analysis identified 70 inflammatory proteins, 77 metabolites, and 26 immune cell traits as potentially causally associated with Bell's palsy. After external validation, BLVRB, HMOX2, TNFRSF12A, DEFB128, ITM2A, VEGF-A, and DDX58 remained significantly associated (p < 0.05). PPI network analysis led to 31 candidate proteins, and six core proteins (JAK2, IL27RA, OSM, CCL19, SELL, VCAM-1) were identified. Conclusions: Our study identifies causal relationships between inflammatory proteins, metabolites, immune cells, and Bell's palsy, highlighting that the JAK/STAT signaling pathway may be a potentially critical target for intervention in Bell's palsy, and that its modulation may provide new directions and opportunities for therapeutic strategies and drug discovery for the disease.
Collapse
Affiliation(s)
| | | | | | | | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; (J.L.); (Z.Y.); (Y.Q.); (Y.Y.); (Z.W.)
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; (J.L.); (Z.Y.); (Y.Q.); (Y.Y.); (Z.W.)
| | | |
Collapse
|
22
|
Figueiredo-Junior AT, Marques BCB, dos Santos DG, Gouveia WL, Meza RMP, Tinoco LW, Lima LM, Valenca SS, Lanzetti M. Mechanistic Advances in the Therapeutic Application of Bixin for Lung Inflammation In Vitro and In Vivo. Pharmaceuticals (Basel) 2025; 18:530. [PMID: 40283965 PMCID: PMC12030059 DOI: 10.3390/ph18040530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Nrf2 plays a key role in regulating the antioxidant response against oxidative stress. Therefore, it is imperative to examine the advantages of Nrf2 activation by new small molecules capable of inhibiting the Nrf2-Keap1 protein interaction that do not present electrophilic sites, since electrophilic compounds have intrinsic toxicity. The bixin pigment has been used as a form of treatment and prevention of several pathological conditions in animal models since it was described as an Nrf2 activator without electrophilic sites. This study aims to synthetize a soluble derivate KBx (potassium bixinate) and evaluate its ability to activate Nrf2/ARE in a model of exposure to cigarette smoke extract (CSE; in vitro) and intranasal LPS administration (in vivo). Methods: In the in vivo study, C57BL/6 mice were pretreated with 200 mg/kg of KBx (gavage) during 5 consecutive days and then challenged with 60 µg of LPS i.n. for 16 h. Bronchoalveolar lavage was collected to examine cytokines dosage. In the in vitro study, RAW 264.7 macrophages were exposed to CSE and post-treated with KBx to evaluate their ability to revert the redox imbalance caused by the stressor. Results: KBx was characterized using mass spectrometry (433.1778 m/z). KC levels were increased in the LPS group (p = 0.021), and KBx inhibited this (p = 0.001). IL-10 levels were decreased (p = 0.055) in the LPS group that was prevented when pretreated with KBx (p = 0.037). The in vitro study showed KBx to be a more potent derivate of bixin through its ability to intercept ROS formation with three-fold more potency, and it showed an anti-inflammatory propriety by reducing the nuclear translocation of p65 (p < 0.001). Conclusions: In conclusion, these data suggest that KBx was able to activate the Nrf2/ARE pathway and intercept ROS formation induced by CSE and LPS in both in vivo and in vitro studies.
Collapse
Affiliation(s)
- Alexsandro Tavares Figueiredo-Junior
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-971, RJ, Brazil
| | - Bruno Clemente Brandão Marques
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
| | - Douglas Galdino dos Santos
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
| | - Wesley Leandro Gouveia
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-971, RJ, Brazil
| | - Raysa Magali Pillpe Meza
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-971, RJ, Brazil
| | - Luzineide Wanderley Tinoco
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
- Instituto de Pesquisas de Produtos Naturais, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| | - Lídia Moreira Lima
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-971, RJ, Brazil
| | - Samuel Santos Valenca
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
| | - Manuella Lanzetti
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil; (A.T.F.-J.); (B.C.B.M.); (D.G.d.S.); (W.L.G.); (R.M.P.M.); (L.W.T.); (L.M.L.); (M.L.)
| |
Collapse
|
23
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025; 219:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75123, Sweden.
| |
Collapse
|
24
|
Ahsan F, Santoso B, Rahmawati NY, Alditia FN, Mufid AF, Sa'adi A, Dwiningsih SR, Tunjungseto A, Widyanugraha MYA. Soluble adhesion molecules in serum and peritoneal fluid are associated with pelvic pain in endometriosis. Int J Gynaecol Obstet 2025; 169:138-147. [PMID: 39513714 DOI: 10.1002/ijgo.16004] [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: 07/22/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
OBJECTIVE To investigate the association between soluble adhesion molecules (sE-cadherin, sE-selectin, sICAM-1, sVCAM-1) in serum and peritoneal fluid and pelvic pain in infertile women with endometriosis. METHODS This observational study included 86 infertile women undergoing diagnostic laparoscopy, 43 of whom were diagnosed with endometriosis. Pain intensity was evaluated using the visual analog scale (VAS). Levels of soluble adhesion molecules in serum and peritoneal fluid were quantified using ELISA. Statistical analysis included the Mann-Whitney U test for group comparisons, Spearman's rank correlation for associations with VAS scores, and receiver operating characteristic (ROC) curve analysis for diagnostic performance. RESULTS Serum sE-selectin levels were significantly higher in women with pelvic pain (P = 0.022) and correlated with VAS scores (r = 0.271, P = 0.012). Peritoneal sE-selectin and sICAM-1 levels were elevated in women with pelvic pain (P = 0.044 and P = 0.029, respectively) and showed positive correlations with VAS scores (r = 0.246, P = 0.022 and r = 0.310, P = 0.004, respectively). Comparing endometriosis and control groups, peritoneal sE-selectin and sICAM-1 levels were significantly higher in endometriosis cases with pelvic pain (P = 0.003 and P < 0.001, respectively). ROC analysis revealed the potential diagnostic value of serum sE-selectin (AUC = 0.698, P = 0.002), serum sICAM-1 (AUC = 0.721, P < 0.001), and serum sVCAM-1 (AUC = 0.750, P < 0.001) in distinguishing endometriosis from non-endometriosis cases. CONCLUSION Elevated levels of sE-selectin and sICAM-1 in serum and peritoneal fluid are associated with pelvic pain in women with endometriosis, suggesting their role in pain pathogenesis and potential as biomarkers for pain severity and disease diagnosis. Further research is warranted to explore the underlying mechanisms and validate these findings in larger cohorts.
Collapse
Affiliation(s)
- Fadhil Ahsan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Budi Santoso
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Nanda Yuli Rahmawati
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | - Alfin Firasy Mufid
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ashon Sa'adi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Sri Ratna Dwiningsih
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Arif Tunjungseto
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - M Y Ardianta Widyanugraha
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
25
|
Wang H, Huang N, Tan M, Zhang X, Chen J, Wei Q. Characteristics of cell adhesion molecules expression and environmental adaptation in yak lung tissue. Sci Rep 2025; 15:10914. [PMID: 40158021 PMCID: PMC11954989 DOI: 10.1038/s41598-025-95882-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Cell Adhesion Molecules (CAMs) play a crucial role in regulating immune responses and repairing damage caused by hypoxia. However, the relationship between the expression characteristics of CAMs in yak lung tissues and their adaptation to the plateau environment remains unclear. To address this question, we compared lung tissues from yaks and cattle at the same altitude. After digesting the lung tissues with trypsin or Type I collagenase for varying durations, we observed that fewer cells were isolated from yak tissues compared to cattle. RNA sequencing (RNA-seq) analysis revealed that the Differentially Expressed Genes (DEGs) in lung tissues of yaks and cattle were significantly enriched in cell adhesion-related pathways. Quantitative real-time PCR (qRT-PCR) further identified changes in the expression levels of five distinct types of CAMs. Among these, the cadherin family (CDH1, CDH2, CDH11, PCDH12, CD34) exhibited significantly higher expression in yaks than in cattle. These cadherins play a critical role in regulating lung inflammation and maintaining the alveolar-capillary barrier, thereby ensuring the structural stability of the lungs. Immunohistochemical staining demonstrated that the expression patterns of cell adhesion-related proteins (CDH1, CDH11, ITGB6, SELP, CD44) were largely consistent with the qRT-PCR results. In conclusion, compared to cattle, the enhanced cell adhesion capacity of yak lung tissues contributes to their superior adaptation to the harsh plateau environment.
Collapse
Affiliation(s)
- Huizhen Wang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Nating Huang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Minglu Tan
- Livestock and Veterinary Station of Huangyuan County, Xining, Qinghai, China
| | - Xun Zhang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Jiarui Chen
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Qing Wei
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China.
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China.
| |
Collapse
|
26
|
Zamanian MY, Maleki S, Oghenemaro EF, Singh M, Mohammadi M, Alkhayyat AH, Sapaev IB, Kaur P, Shirsalimi N, Nagarwal A. Omentin-1 as a promising biomarker and therapeutic target in hypertension and heart failure: a comprehensive review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04008-y. [PMID: 40126671 DOI: 10.1007/s00210-025-04008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/02/2025] [Indexed: 03/26/2025]
Abstract
Omentin-1, a novel adipocytokine predominantly secreted by visceral adipose tissue, has emerged as a significant factor in cardiovascular health, particularly regarding hypertension (HTN) and heart failure (HF). This manuscript investigates the multifaceted roles of omentin-1 in these conditions, emphasizing its protective effects on vascular function and its potential as both a biomarker and therapeutic target. Clinical studies indicate that reduced circulating levels of omentin-1 are associated with metabolic syndrome (MetS) and increased cardiovascular risk, while animal studies demonstrate its ability to ameliorate endothelial dysfunction and lower blood pressure. Omentin-1 exerts its beneficial effects through various signaling pathways, including AMP-activated protein kinase (AMPK) and protein kinase B (Akt), thereby promoting vasodilation, enhancing insulin sensitivity, and mitigating inflammation. In the context of HF, particularly heart failure with preserved ejection fraction (HFpEF), omentin-1 levels exhibit a negative correlation with diastolic dysfunction and inflammatory markers, suggesting its role in cardiac protection. Additionally, the manuscript discusses the implications of omentin-1 in managing obesity-related cardiovascular diseases and its potential utility as a prognostic marker for adverse outcomes in HF patients. Collectively, omentin-1 represents a promising avenue for research in cardiovascular health, with the potential to inform novel therapeutic strategies aimed at improving outcomes in patients with HTN and HF. Further research is necessary to elucidate the details of omentin-1 function and evaluate its potential in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Saba Maleki
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Guilan University of Medical Sciences (GUMS), Rasht, Guilan Province, Iran
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Mandeep Singh
- Directorate of Sports and Physical Education, University of Jammu, Jammu, India
| | - Maryam Mohammadi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Hussen Alkhayyat
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Ibrokhim B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan
- Scientific Researcher, University of Tashkent for Applied Sciences, Str. Gavhar 1, 100149, Tashkent, Uzbekistan
- Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Niyousha Shirsalimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Amritesh Nagarwal
- Department of Cardiology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| |
Collapse
|
27
|
Pickett JR, Wu Y, Ta HT. VCAM-1 as a common biomarker in inflammatory bowel disease and colorectal cancer: unveiling the dual anti-inflammatory and anti-cancer capacities of anti-VCAM-1 therapies. Cancer Metastasis Rev 2025; 44:40. [PMID: 40095109 PMCID: PMC11913972 DOI: 10.1007/s10555-025-10258-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Vascular cell adhesion molecule (VCAM)-1 has garnered significant research attention due to its potential as a disease biomarker and drug target across several inflammatory pathologies-including atherosclerosis, asthma, rheumatoid arthritis, and inflammatory bowel disease (IBD). The VCAM-1 protein has also been noted for its functional involvement in cancer metastasis and drug resistance to conventional chemotherapeutics. Although the anti-inflammatory and anti-cancer facets of VCAM-1 antagonisation have been examined separately, there is yet to be a review that explicitly addresses the functional interrelationship between these mechanisms. Furthermore, the pleiotropic mechanisms of anti-VCAM-1 therapies may present a useful paradigm for designing drug candidates with synergistic anti-inflammatory and anti-tumorigenic effects. The pathological overlap between inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CRC) serves as the quintessential disease model to observe this therapeutic duality. This review thereby details the adhesive mechanisms of VCAM-1 in colorectal disease-specifically, driving immune cell infiltration during IBD and tumour cell metastasis in CRC-and posits the potential of this receptor as a common drug target for both diseases. To explore this hypothesis, the current progress of novel VCAM-1-directed drug candidates in experimental models of IBD and CRC is also discussed.
Collapse
Affiliation(s)
- Jessica R Pickett
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia
| | - Yuao Wu
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia
| | - Hang Thu Ta
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia.
| |
Collapse
|
28
|
Huang W, Li L, Jiang Y, Lou Q, Gao J, Zhu C. Precision diagnostics for psychotic major depression: Construction and validation of a clinical indicator-based model. Clin Chim Acta 2025; 570:120204. [PMID: 39978458 DOI: 10.1016/j.cca.2025.120204] [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: 12/03/2024] [Revised: 01/15/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Differentiating psychotic major depression (PMD) from non-PMD (NPMD) is crucial as it influences treatment decisions, prognosis, and patient outcomes. This study aims to develop an efficient model for precision diagnostics of PMD based on clinical indicators. METHODS A total of 731 patients who visited our hospital with major depression (MD) were enrolled, including a discovery cohort and a validation cohort. We retrospectively analyzed the distribution differences of 20 clinical indicators in the discovery cohort. We included differential clinical indicators (DCIs) in the logistic regression algorithm analysis to establish a multi-panel detection model. The model's performance in distinguishing PMD from NPMD and in distinguishing bipolar MD from MD was validated in the validation cohort by the receiver operator characteristic curve (ROC), the area under the curve (AUC), sensitivity, and specificity. RESULTS We have constructed a six-DCIs diagnosis model (6MP) based on the discovery cohort. The AUC of 6MP for identifying PMD and NPMD was 0.826, and the sensitivity and specificity were 87.5 % and 70.59 %, respectively. In the validation cohort, the AUC for identifying PMD and NPMD was 0.781, and the sensitivity and specificity were 78.99 % and 67.31 %. The AUC for identifying bipolar MD and MD without psychotic symptoms was 0.716, and the sensitivity and specificity were 60.71 % and 76.92 %. CONCLUSIONS This study not only provides new tools for the precise diagnosis and treatment of PMD but also hopes to simplify the diagnostic process, improve the specificity of treatment, and thus bring more practical clinical benefits to patients.
Collapse
Affiliation(s)
- Weiquan Huang
- Laboratory Department of Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Lanqing Li
- Zhejiang Engineering Research Center for Intelligent Manufacturing of Clinical Diagnostic Equipment, Hangzhou 310020, China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang 310020, China
| | - Yan Jiang
- Laboratory Department of Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Qinqin Lou
- Zhejiang Engineering Research Center for Intelligent Manufacturing of Clinical Diagnostic Equipment, Hangzhou 310020, China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang 310020, China
| | - Junli Gao
- Zhejiang Engineering Research Center for Intelligent Manufacturing of Clinical Diagnostic Equipment, Hangzhou 310020, China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang 310020, China.
| | - Chunyan Zhu
- Laboratory Department of Pinghu Mental Health Center, Jiaxing 314200, China.
| |
Collapse
|
29
|
Beltran-Velasco AI, Clemente-Suárez VJ. Impact of Peripheral Inflammation on Blood-Brain Barrier Dysfunction and Its Role in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:2440. [PMID: 40141084 PMCID: PMC11942216 DOI: 10.3390/ijms26062440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
The blood-brain barrier (BBB) is essential for maintaining brain homeostasis by regulating molecular exchange between the systemic circulation and the central nervous system. However, its dysfunction, often driven by peripheral inflammatory processes, has been increasingly linked to the development and progression of neurodegenerative diseases such as Alzheimer's and Parkinson's. Emerging evidence suggests that the gut-brain axis plays a key role in BBB integrity, with intestinal dysbiosis and chronic inflammation contributing to barrier disruption through immune and metabolic pathways. Furthermore, the selective vulnerability of specific brain regions to BBB dysfunction appears to be influenced by regional differences in vascularization, metabolic activity, and permeability, making certain areas more susceptible to neurodegenerative processes. This review explored the molecular mechanisms linking peripheral inflammation, gut microbiota, and BBB dysfunction, emphasizing their role in neurodegeneration. A comprehensive literature review was conducted using Web of Science, PubMed, Scopus, Wiley, ScienceDirect, and Medline, covering publications from 2015 to 2025. The findings highlight a complex interplay between gut microbiota-derived metabolites, immune signaling, and BBB permeability, underscoring the need for targeted interventions such as microbiome modulation, anti-inflammatory therapies, and advanced drug delivery systems. The heterogeneity of the BBB across different brain regions necessitates the development of region-specific therapeutic strategies. Despite advancements, critical knowledge gaps persist regarding the precise mechanisms underlying BBB dysfunction. Future research should leverage cutting-edge methodologies such as single-cell transcriptomics and organ-on-chip models to translate preclinical findings into effective clinical applications. Addressing these challenges will be crucial for developing personalized therapeutic approaches to mitigate the impact of BBB dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ana Isabel Beltran-Velasco
- NBC Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain;
| | - Vicente Javier Clemente-Suárez
- Faculty of Medicine, Health and Sports, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| |
Collapse
|
30
|
Saba L, Cau R, Vergallo R, Kooi ME, Staub D, Faa G, Congiu T, Ntaios G, Wasserman BA, Benson J, Nardi V, Kawakami R, Lanzino G, Virmani R, Libby P. Carotid artery atherosclerosis: mechanisms of instability and clinical implications. Eur Heart J 2025; 46:904-921. [PMID: 39791527 DOI: 10.1093/eurheartj/ehae933] [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: 06/18/2024] [Revised: 08/25/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular disease remains a prominent cause of disability and premature death worldwide. Within this spectrum, carotid artery atherosclerosis is a complex and multifaceted condition, and a prominent precursor of acute ischaemic stroke and other cardiovascular events. The intricate interplay among inflammation, oxidative stress, endothelial dysfunction, lipid metabolism, and immune responses participates in the development of lesions, leading to luminal stenosis and potential plaque instability. Even non-stenotic plaques can precipitate a sudden cerebrovascular event, regardless of the degree of luminal encroachment. In this context, carotid imaging modalities have proved their efficacy in providing in vivo characterization of plaque features, contributing substantially to patient risk stratification and clinical management. This review emphasizes the importance of identifying high-risk individuals by use of current imaging modalities, biomarkers, and risk stratification tools. Such approaches inform early intervention and the implementation of personalized therapeutic strategies, ultimately enhancing patient outcomes in the realm of cardiovascular disease management.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, University of Cagliari, Via Università, 40, 09124 Cagliari, Italy
| | - Riccardo Cau
- Department of Radiology, University of Cagliari, Via Università, 40, 09124 Cagliari, Italy
| | - Rocco Vergallo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - M Eline Kooi
- Department of Radiology and Nuclear Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Daniel Staub
- Vascular Medicine/Angiology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Gavino Faa
- Department of Pathology, University of Cagliari, Cagliari, Italy
| | - Terenzio Congiu
- Department of Pathology, University of Cagliari, Cagliari, Italy
| | - George Ntaios
- Department of Internal Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Larissa 41132, Greece
| | - Bruce A Wasserman
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, MD, USA
| | - John Benson
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Valentina Nardi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Inc., Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Dabravolski SA, Churov AV, Ravani AL, Karimova AE, Luchinkin IG, Sukhorukov VN, Orekhov AN. The role of Epsins in atherosclerosis: From molecular mechanisms to therapeutic applications. Vascul Pharmacol 2025; 158:107457. [PMID: 39672315 DOI: 10.1016/j.vph.2024.107457] [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: 10/07/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
Atherosclerosis is a multifaceted disease characterised by chronic inflammation and vascular remodelling, leading to plaque formation and cardiovascular complications. Recent evidence highlights the critical role of epsins, a family of endocytic proteins, in the pathogenesis of atherosclerosis. This manuscript explores the multifarious functions of epsins in atherosclerosis, focusing on their involvement in angiogenesis, lymphangiogenesis, and the modulation of key signalling pathways. We discuss how epsins facilitate EndoMT through their interaction with the TGFβ signalling pathway, which contributes to vascular smooth muscle cell-like phenotypes and plaque instability. Additionally, we examine the therapeutic potential of targeting epsins, elucidating their interactions with crucial partners such as LDLR, LRP-1, and TLR 2/4, among others, in mediating lipid metabolism and inflammation. Furthermore, we highlight the promising prospects of epsin-targeting peptides and small interfering RNAs as therapeutic agents for atherosclerosis treatment. Despite these advancements, the research faces limitations, including a reliance on specific mouse models and a need for comprehensive studies on the long-term effects of epsin modulation. Therefore, future investigations should focus on elucidating the detailed mechanisms of epsin function and their implications in cardiovascular health, fostering collaborations to translate basic research into innovative therapeutic strategies. This work underscores the necessity for further exploration of epsins to unlock their full therapeutic potential in combating atherosclerosis and related cardiovascular diseases.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, Karmiel 2161002, Israel.
| | - Alexey V Churov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia; Pirogov Russian National Research Medical University, Russia Gerontology Clinical Research Centre, Moscow, Institute on Ageing Research, Russian Federation, 16 1st Leonova Street, 129226 Moscow, Russia
| | - Alessio L Ravani
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia
| | - Amina E Karimova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street, Building 4, 117418 Moscow, Russia
| | - Igor G Luchinkin
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia; Institute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, 119991 Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia
| |
Collapse
|
32
|
Yin M, Zheng X, Shi L. Targeting p38 MAPK: A potential bridge between ER stress and age-related bone loss. Cell Signal 2025; 127:111549. [PMID: 39638139 DOI: 10.1016/j.cellsig.2024.111549] [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: 08/26/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The endoplasmic reticulum (ER) is crucial in the development of numerous age-related bone disorders. Notably, ER stress can precipitate bone loss by orchestrating inflammatory responses, apoptosis, and autophagy through the activation of the p38 MAPK pathway. Age-related bone loss diseases pose a significant burden on society and healthcare as the global population ages. This review provides a comprehensive analysis of recent research advancements, delving into the critical role of ER stress-activated p38 MAPK in inflammation, apoptosis, and autophagy, as well as its impact on bone formation and bone resorption. This review elucidates the molecular mechanisms underlying the involvement of ER stress-activated p38 MAPK in osteoporosis, rheumatoid arthritis, periodontitis, and osteoarthritis and discusses the therapeutic potential of targeting p38 MAPK. Furthermore, this review provides a scientific foundation for new therapeutic strategies by highlighting prospective research directions.
Collapse
Affiliation(s)
- Meng Yin
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Zheng
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
33
|
Wang J, Shi H, Yang Y, Gong X. Crosstalk between ferroptosis and innate immune in diabetic kidney disease: mechanisms and therapeutic implications. Front Immunol 2025; 16:1505794. [PMID: 40092979 PMCID: PMC11906378 DOI: 10.3389/fimmu.2025.1505794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Diabetic kidney disease (DKD) is a prevalent complication of diabetes mellitus (DM), and its incidence is increasing alongside the number of diabetes cases. Effective treatment and long-term management of DKD present significant challenges; thus, a deeper understanding of its pathogenesis is essential to address this issue. Chronic inflammation and abnormal cell death in the kidney closely associate with DKD development. Recently, there has been considerable attention focused on immune cell infiltration into renal tissues and its inflammatory response's role in disease progression. Concurrently, ferroptosis-a novel form of cell death-has emerged as a critical factor in DKD pathogenesis, leading to increased glomerular filtration permeability, proteinuria, tubular injury, interstitial fibrosis, and other pathological processes. The cardiorenal benefits of SGLT2 inhibitors (SGLT2-i) in DKD patients have been demonstrated through numerous large clinical trials. Moreover, further exploratory experiments indicate these drugs may ameliorate serum and urinary markers of inflammation, such as TNF-α, and inhibit ferroptosis in DKD models. Consequently, investigating the interplay between ferroptosis and innate immune and inflammatory responses in DKD is essential for guiding future drug development. This review presents an overview of ferroptosis within the context of DKD, beginning with its core mechanisms and delving into its potential roles in DKD progression. We will also analyze how aberrant innate immune cells, molecules, and signaling pathways contribute to disease progression. Finally, we discuss the interactions between ferroptosis and immune responses, as well as targeted therapeutic agents, based on current evidence. By analyzing the interplay between ferroptosis and innate immunity alongside its inflammatory responses in DKD, we aim to provide insights for clinical management and drug development in this area.
Collapse
Affiliation(s)
- Jinyang Wang
- Department of Geriatric Integrative, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Haonan Shi
- School of Medicine, Shanghai University, Shanghai, China
| | - Ye Yang
- Department of Geriatric Integrative, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xueli Gong
- Department of Pathophysiology, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
34
|
Gusti Y, Liu W, Athar F, Cahill PA, Redmond EM. Endothelial Homeostasis Under the Influence of Alcohol-Relevance to Atherosclerotic Cardiovascular Disease. Nutrients 2025; 17:802. [PMID: 40077672 PMCID: PMC11901717 DOI: 10.3390/nu17050802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
Alcohol, in the form of ethyl alcohol or ethanol, is a widely consumed substance with significant implications for human health. Research studies indicate multifaceted effects of alcohol on the cardiovascular system with both protective and harmful effects on atherosclerotic cardiovascular disease (ASCVD), depending on the amount involved and the pattern of consumption. Among the critical components of the cardiovascular system are endothelial cells which line blood vessels. These cells are pivotal in maintaining vessel homeostasis, regulating blood flow, and preventing thrombosis. Their compromised function correlates with arterial disease progression and is predictive of cardiovascular events. Here we review research investigating how alcohol exposure affects the endothelium to gain insight into potential mechanisms mediating alcohol's influence on ASCVD underlying heart attacks and strokes. Studies highlight opposite effects of low versus high levels of alcohol on many endothelial functions. In general, low-to-moderate levels of alcohol (~5-25 mM) maintain the endothelium in a non-activated state supporting vascular homeostasis, while higher alcohol levels (≥50 mM) lead to endothelial dysfunction and promotes atherosclerosis. These biphasic endothelial effects of alcohol might underlie the varying impacts of different alcohol consumption patterns on ASCVD.
Collapse
Affiliation(s)
| | | | | | | | - Eileen M. Redmond
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642-8410, USA; (Y.G.); (W.L.); (F.A.); (P.A.C.)
| |
Collapse
|
35
|
Zhou Y, Hubscher CH. Biomarker expression level changes within rectal gut-associated lymphoid tissues in spinal cord-injured rats. Immunohorizons 2025; 9:vlaf002. [PMID: 40048710 PMCID: PMC11884801 DOI: 10.1093/immhor/vlaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Neurogenic bowel dysfunction (NBD) is common after spinal cord injury (SCI). Gut-associated lymphoid tissue (GALT), an organized structure within the mucosal immune system, is important for the maintenance of gut homeostasis and body health and serves as the first line barrier/defense against diet antigens, commensal microbiota, pathogens, and toxins in mucosal areas. The current study examined gene expression levels along six segments of anorectal tissue using real-time polymerase chain reaction (RT-PCR) in uninjured rats (28-day sham surgical controls) and at both 28- and 42-days post-T9 contusion injury. Consistent with our previous report of functional regional differences in the ano-rectum, we demonstrate the existence of GALTs located primarily within the segment at 3-4.5 cm from the rectal dentate line (termed rectal GALTs-rGALTs) in shams with upregulated gene expression levels of multiple biomarkers, including B cell and T cell-related genes, major histocompatibility complex (MHC) class II molecules, and germinal center (GC)-related genes, which was further confirmed by histologic examination. In the same rectal tissue segment following T9 SCI, inflammation-related genes were upregulated at 28 days post-injury (DPI) indicating that microbial infection and inflammation of rGALTs modified structure and function of rGALTs, while at 42 DPI rGALTs exhibited resolution of inflammation and impaired structure/function for extrafollicular B cell responses. Taken together, our data suggest that rGALTs exists in rat rectum for homeostasis of gut microbiota/barrier. SCI induces microbial infection and inflammation in rectal tissues containing rGALTs, which could contribute to development of SCI-related gut microbiome dysbiosis, NBD, and systemic diseases.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| | - Charles H Hubscher
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| |
Collapse
|
36
|
Naydenov S, Jekova M, Manov E, Runev N. Could Renal Vascular Variations Be Associated with Resistant Hypertension? A Single-Center Study. J Clin Med 2025; 14:1268. [PMID: 40004797 PMCID: PMC11855990 DOI: 10.3390/jcm14041268] [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: 12/22/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Anatomical variations in renal vessels are common in humans. However, the clinical significance of these congenital vascular abnormalities remains incompletely understood. This study aimed to assess the prevalence and characteristics of renal vascular variants in patients with arterial hypertension (HTN) and their association with the development of resistant HTN. Methods: We screened 3762 consecutive hypertensive patients hospitalized in our clinic and identified 128 (3.4%) with resistant HTN. These patients were matched with 128 hospitalized patients with controlled HTN. All participants underwent contrast-enhanced computed tomography imaging of the kidneys, including renovasography. Results: Anatomical renal vascular variations were identified in 64 (25%) of the 256 participants: 49 (38.3%) of the 128 patients with resistant HTN and 15 (11.7%) of the 128 patients with controlled HTN (p < 0.001). Among patients with renal vascular abnormalities, 76.6% had resistant HTN, while 23.4% had controlled HTN (p < 0.001). A regression analysis demonstrated a strong association between the presence of renal vascular variants and the development of resistant HTN, with an odds ratio of 4.67. Conclusions: Anatomical renal vascular variations were found to be common among hypertensive patients in this study and were strongly associated with the development of treatment-resistant HTN.
Collapse
Affiliation(s)
- Stefan Naydenov
- Department of Internal Diseases “Prof. St. Kirkovich”, Medical University of Sofia, 1431 Sofia, Bulgaria; (M.J.); (E.M.); (N.R.)
| | | | | | | |
Collapse
|
37
|
Shen Q, Yu Q, Chen T, Zhang L. Rosuvastatin mitigates blood-brain barrier disruption in sepsis-associated encephalopathy by restoring occludin levels. Eur J Med Res 2025; 30:103. [PMID: 39953583 PMCID: PMC11827257 DOI: 10.1186/s40001-025-02314-y] [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: 09/16/2024] [Accepted: 01/20/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) disruption is a key pathological feature of sepsis-associated encephalopathy (SAE). Rosuvastatin, a third-generation statin, exhibits diverse pharmacological functions beyond its lipid-lowering capacity. However, its potential neuroprotective role in SAE remains unclear. MATERIALS AND METHODS SAE models were established using the cecal ligation and puncture (CLP) method. BBB integrity was evaluated using NaF, and endothelial permeability was assessed by fluorescein isothiocyanate (FITC)-dextran assays. RESULTS Rosuvastatin significantly attenuated neuroinflammation in the brains of septic mice by reducing the expression of the pro-inflammatory cytokines interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), and tumor necrosis factor α (TNF-α). It also ameliorated vascular injury in the brain cortex of septic mice by decreasing the levels of vascular cell adhesion molecule-1 (VCAM-1) and E-selectin. Furthermore, Rosuvastatin preserved BBB integrity in septic mice by enhancing the expression of the tight junction protein occludin. In vitro studies demonstrated that Rosuvastatin alleviated endothelial permeability and increased transendothelial electrical resistance (TEER) in lipopolysaccharide (LPS)-stimulated human brain microvascular endothelial cells (HBMECs). Additionally, Rosuvastatin prevented the LPS-induced reduction of occludin and Krüppel-like factor 2 (KLF2) in HBMECs. Importantly, silencing KLF2 abrogated Rosuvastatin's protective effects on endothelial permeability and occludin expression. CONCLUSIONS These findings indicate that Rosuvastatin may be a promising therapeutic candidate for mitigating BBB dysfunction associated with SAE.
Collapse
Affiliation(s)
- Qin Shen
- Department of Critical Care Medicine, Chengdu Fifth People's Hospital, Geriatric Diseases Institute of Chengdu, Cancer Prevention and Treatment Institute of Chengdu, The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, No.33, Ma Shi Street, Chengdu, 611137, Sichuan, China
| | - Qian Yu
- Department of Cardiovascular Medicine, Chengdu Fifth People's Hospital, Geriatric Diseases Institute of Chengdu, Cancer Prevention and Treatment Institute of Chengdu, the Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Taojiang Chen
- Department of Critical Care Medicine, Chengdu Fifth People's Hospital, Geriatric Diseases Institute of Chengdu, Cancer Prevention and Treatment Institute of Chengdu, The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, No.33, Ma Shi Street, Chengdu, 611137, Sichuan, China
| | - Lijuan Zhang
- Department of Critical Care Medicine, Chengdu Fifth People's Hospital, Geriatric Diseases Institute of Chengdu, Cancer Prevention and Treatment Institute of Chengdu, The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, No.33, Ma Shi Street, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
38
|
Huang K, Pokhrel A, Echesabal-Chen J, Scott J, Bruce T, Jo H, Stamatikos A. Inhibiting MiR-33a-3p Expression Fails to Enhance ApoAI-Mediated Cholesterol Efflux in Pro-Inflammatory Endothelial Cells. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:329. [PMID: 40005445 PMCID: PMC11857470 DOI: 10.3390/medicina61020329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Atherosclerosis is an inflammatory condition that results in cholesterol accumulating within vessel wall cells. Atherosclerotic cardiovascular disease is the leading cause of mortality worldwide due to this disease being a major contributor to myocardial infarctions and cerebrovascular accidents. Research suggests that cholesterol accumulation occurring precisely within arterial endothelial cells triggers atherogenesis and exacerbates atherosclerosis. Furthermore, inflamed endothelium acts as a catalyst for atherosclerotic development. Therefore, enhancing cholesterol removal specifically in pro-inflammatory endothelial cells may be a potential treatment option for atherosclerosis. While we have previously shown that inhibiting the microRNA guide strand miR-33a-5p within pro-inflammatory endothelial cells increases both ABCA1 expression and apoAI-mediated cholesterol efflux, it is unknown whether inhibiting the miR-33a-3p passenger strand in pro-inflammatory endothelial cells causes similar atheroprotective effects. In this study, this is what we aimed to test. Materials and Methods: We used plasmid transfection to knockdown miR-33a-3p expression within cultured pro-inflammatory immortalized mouse aortic endothelial cells (iMAECs). We compared ABCA1 expression and apoAI-mediated cholesterol efflux within these cells to cultured pro-inflammatory iMAECs transfected with a control plasmid. Results: The knockdown of miR-33a-3p expression within pro-inflammatory iMAECs resulted in a significant increase in ABCA1 mRNA expression. However, the inhibition of miR-33a-3p did not significantly increase ABCA1 protein expression within pro-inflammatory iMAECs. Moreover, we failed to detect a significant increase in apoAI-mediated cholesterol efflux within pro-inflammatory iMAECs from miR-33a-3p knockdown. Conclusions: Our results indicative that the knockdown of miR-33a-3p alone does not enhance ABCA1-dependent cholesterol efflux within pro-inflammatory endothelial cells. To gain any atheroprotective benefit from inhibiting miR-33a-3p within pro-inflammatory endothelium, additional anti-atherogenic strategies would likely be needed in unison.
Collapse
Affiliation(s)
- Kun Huang
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (A.P.); (J.E.-C.)
| | - Achala Pokhrel
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (A.P.); (J.E.-C.)
| | - Jing Echesabal-Chen
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (A.P.); (J.E.-C.)
| | - Justin Scott
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (J.S.); (T.B.)
| | - Terri Bruce
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (J.S.); (T.B.)
| | - Hanjoong Jo
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA;
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (A.P.); (J.E.-C.)
| |
Collapse
|
39
|
Wang C, Cheng H, Dong X, Zhan Y, Liu Y, Wu N, Tang R, He H, Cao Y, Yang L, Ren J, Li X, Li P. Early assessment and treatment of ventricular remodeling in vivo via a targeted ultrasonic molecular probe loaded with oxygen and cholecystokinin. J Nanobiotechnology 2025; 23:104. [PMID: 39939853 PMCID: PMC11823180 DOI: 10.1186/s12951-025-03183-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/01/2025] [Indexed: 02/14/2025] Open
Abstract
Early identification and treatment of ventricular remodeling (VR) are crucial for delaying the progression of heart failure after myocardial infarction. This study aims to develop a dual-responsive phase-shift molecular probe loaded with a cholecystokinin octapeptide (CCK-8) and oxygen, which will provide a new integrated scheme for the assessment and treatment of VR. Biocompatible phospholipid shells were utilized to encapsulate CCK-8 and perfluoropentane (PFP), an efficient oxygen carrier. Surface modification involves reactive oxygen species (ROS)-responsive thioketal (TK) bonds and anti-ICAM-1 antibodies to create CCK-8 and oxygen-carrying phase-shift nanoparticles (PFP-O2-CCK8@lipid/TK-ICAM1 Ab nanoparticles, POC@L/TI NPs). These nanoparticles were designed for coronary artery endothelial cell targeting and responsiveness to dual stimuli. The results demonstrated that delayed myocardial contrast-enhanced echocardiography (DMCE) provided dynamic VR monitoring, with contrast intensity values showing a negative correlation with cardiac function parameter changes. POC@L/TI NPs significantly improved cardiac structural and functional parameters in rats with myocardial infarction and reperfusion and delayed the progression of heart failure by increasing tissue oxygenation, reducing the inflammatory response, inhibiting fibrotic scar formation and preventing myocardial cell apoptosis. This innovative approach combines supersaturated oxygen therapy with the multitarget therapeutic effect of CCK-8 and dynamic monitoring via DMCE to offer an integrated strategy for early detection and comprehensive VR treatment.
Collapse
Affiliation(s)
- Can Wang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Hongfeng Cheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
- Health Management (Physical Examination) Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Xiaoying Dong
- First Department of Respiratory and Critical Care Medicine/Pneumoconiosis Department, The Second Hospital of Heilongjiang Province, Harbin, P. R. China
| | - Yue Zhan
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Ying Liu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
- Department of Ultrasound, Chengdu Third People's Hospital, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, P. R. China
| | - Nianhong Wu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Rui Tang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Hongye He
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yuting Cao
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Liping Yang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Jianli Ren
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Xingsheng Li
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China.
| | - Pan Li
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University; Chongqing Key Laboratory of Ultrasound Molecular Imaging; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, P. R. China.
| |
Collapse
|
40
|
Csiha S, Hernyák M, Molnár Á, Lőrincz H, Katkó M, Paragh G, Bodor M, Harangi M, Sztanek F, Berta E. Alpha-Lipoic Acid Treatment Reduces the Levels of Advanced End Glycation Products in Type 2 Diabetes Patients with Neuropathy. Biomedicines 2025; 13:438. [PMID: 40002851 PMCID: PMC11852413 DOI: 10.3390/biomedicines13020438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Type 2 diabetes mellitus (T2DM) and its macro- and microvascular complications are major health concerns with multiple factors, like advanced end glycation products (AGEs), in the background. AGEs induce long-lasting functional modification of the proteins and collagen in the vascular wall and nerve tissue. We investigated the effect of alpha-lipoic acid (ALA) treatment on AGEs, soluble AGE receptor (sRAGE), the AGE/sRAGE ratio, and the parameters of endothelial dysfunction and their correlations. Methods: In our 6-month intervention study, 54 T2DM patients with neuropathy treated according to the actual therapeutic guidelines with unchanged oral antidiabetic drugs were included and treated by daily oral administration of 600 mg ALA. A total of 24 gender and age-matched T2DM patients without neuropathy served as controls. Results: In our work, we first demonstrated the attenuating effect of alpha lipoic acid therapy on AGEs in humans (11.89 (9.44-12.88) to 10.95 (9.81-12.82) AU/μg (p = 0.017)). sRAGE levels or the AGEs/sRAGE ratio were not affected by ALA treatment or by the presence of neuropathy. We found a correlation between the changes of AGEs and the improvement of current perception threshold and progranulin levels, and an inverse correlation with the change of asymmetric dimethylarginine. Conclusions: According to our results, ALA decreases AGEs, which may contribute to the clinically well-known beneficial effect in diabetic neuropathy and improvement of endothelial function.
Collapse
Affiliation(s)
- Sára Csiha
- Division of Endocrinology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Clinical Basics, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary;
- Doctoral School of Health Sciences, University of Debrecen, 4032 Debrecen, Hungary
| | - Marcell Hernyák
- Doctoral School of Health Sciences, University of Debrecen, 4032 Debrecen, Hungary
- Division of Metabolism, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Ágnes Molnár
- Division of Metabolism, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Hajnalka Lőrincz
- Division of Metabolism, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Mónika Katkó
- Division of Endocrinology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, 4032 Debrecen, Hungary
| | - György Paragh
- Division of Metabolism, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Miklós Bodor
- Division of Endocrinology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Clinical Basics, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary;
| | - Mariann Harangi
- Doctoral School of Health Sciences, University of Debrecen, 4032 Debrecen, Hungary
- Division of Metabolism, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Ferenc Sztanek
- Division of Metabolism, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Eszter Berta
- Department of Clinical Basics, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary;
- Division of Metabolism, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
41
|
Machoń NJ, Zdanowska N, Klimek-Trojan P, Owczarczyk-Saczonek A. Vascular Cell Adhesion Molecule 1 and E-Selectin as Potential Cardiovascular Risk Biomarkers in Psoriasis. Int J Mol Sci 2025; 26:792. [PMID: 39859506 PMCID: PMC11765541 DOI: 10.3390/ijms26020792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) and E-selectin are involved in different inflammatory diseases and may be potential cardiovascular risk biomarkers in psoriasis. They play an important role in regulating the recruitment and adhesion to endothelial cells during inflammation, affecting various conditions like vasculitis, atherosclerosis, and cardiovascular diseases. Positive outcomes have been observed when using Tumor Necrosis Factor Alpha (TNF-α) inhibitors and biological therapies that target selectins to control the functioning of endothelial cells and reduce inflammation in psoriasis and related conditions. Moreover, the effects of systemic treatments and ultraviolet B (UVB) phototherapy on VCAM-1 and E-selectin levels in psoriasis patients highlights the potential to impact the severity of psoriasis and activation of endothelial cells. In addition, various factors such as age, sex, metabolic syndrome, hyperglycemia, migraines, and tobacco smoking have been found to affect levels of VCAM-1 and E-selectin. This sheds light on understanding the complex relationship between endothelial activation and the development of diseases. Studies show the potential of using the levels of VCAM-1 and E-selectin as indicators of systemic treatment effectiveness and the progression of the disease. In summary, this review highlights the importance of VCAM-1 and E-selectin as potential biomarkers for assessing inflammation, disease severity and cardiovascular risk in individuals with psoriasis. The shared mechanisms of psoriasis and atherosclerosis, along with the effect of treatments on endothelial activation markers, provide significant insights for further research and approaches to manage inflammatory diseases in the future.
Collapse
Affiliation(s)
- Natalia Joanna Machoń
- School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland;
| | - Natalia Zdanowska
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, Collegium Medicum, University of Warmia and Mazury, Al. Wojska Polskiego 30, 10-229 Olsztyn, Poland
| | - Paulina Klimek-Trojan
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, Collegium Medicum, University of Warmia and Mazury, Al. Wojska Polskiego 30, 10-229 Olsztyn, Poland
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, Collegium Medicum, University of Warmia and Mazury, Al. Wojska Polskiego 30, 10-229 Olsztyn, Poland
| |
Collapse
|
42
|
Liu M, Chen R, Zheng Z, Xu S, Hou C, Ding Y, Zhang M, Bao M, He B, Li S. Mechanisms of inflammatory microenvironment formation in cardiometabolic diseases: molecular and cellular perspectives. Front Cardiovasc Med 2025; 11:1529903. [PMID: 39877020 PMCID: PMC11772298 DOI: 10.3389/fcvm.2024.1529903] [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: 11/18/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Cardiometabolic diseases (CMD) are leading causes of death and disability worldwide, with complex pathophysiological mechanisms in which inflammation plays a crucial role. This review aims to elucidate the molecular and cellular mechanisms within the inflammatory microenvironment of atherosclerosis, hypertension and diabetic cardiomyopathy. In atherosclerosis, oxidized low-density lipoprotein (ox-LDL) and pro-inflammatory cytokines such as Interleukin-6 (IL-6) and Tumor Necrosis Factor-alpha (TNF-α) activate immune cells contributing to foam cell formation and arterial wall thickening. Hypertension involves the activation of the renin-angiotensin system (RAS) alongside oxidative stress-induced endothelial dysfunction and local inflammation mediated by T cells. In diabetic cardiomyopathy, a high-glucose environment leads to the accumulation of advanced glycation end products (AGEs), activating the Receptor for Advanced Glycation Endproducts (RAGE) and triggering inflammatory responses that further damage cardiac and microvascular function. In summary, the inflammatory mechanisms in different types of metabolic cardiovascular diseases are complex and diverse; understanding these mechanisms deeply will aid in developing more effective individualized treatment strategies.
Collapse
Affiliation(s)
- Menghua Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rumeng Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiwei Zheng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Shuling Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chunyan Hou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yining Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mengling Zhang
- School of Stomatology, Changsha Medical University, Changsha, China
| | - Meihua Bao
- Hunan key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Binsheng He
- Hunan key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
43
|
Sun Y, Yee E, Fujiwara Y, Dickinson K, Guo Y, Sun Z, Hu J, Davila E, Schulick RD, Zhu Y. CD93 blockade promotes effector T-cell infiltration and facilitates adoptive cell therapy in solid tumors. J Immunother Cancer 2025; 13:e010554. [PMID: 39805660 PMCID: PMC11749055 DOI: 10.1136/jitc-2024-010554] [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: 09/11/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Adaptive cellular therapy (ACT), particularly chimeric antigen receptor (CAR)-T cell therapy, has been successful in the treatment of hemopoietic malignancies. However, poor trafficking of administered effector T cells to the tumor poses a great hurdle for this otherwise powerful therapeutic approach in solid cancers. Our previous study revealed that targeting CD93 normalizes tumor vascular functions to improve immune checkpoint blockade therapy. The objective of this study is to evaluate whether CD93 blockade improves ACT in solid cancers. METHODS Monoclonal antibodies (mAbs) against CD93 or IGFBP7 were administered in implanted mouse melanoma models to assess the effect of CD93 blockade on ACT. Different sources of effector T cells were used, including pre-activated CD8+OT-1, pmel-1 transgenic T cells, and CAR-T cells. Rip-OVA and Rip-TAG-OVA transgenic mice were used to evaluate the selective impact of CD93 blockade on effector T-cell infiltration in tumors. For mechanistic studies, vascular maturation was determined by immunofluorescent staining and flow cytometry was performed to examine tumor-infiltrating T lymphocytes. Neutralizing mAbs against adhesion molecules ICAM1 and VCAM1 were infused to assess their involvement. RESULTS Blockade of the CD93 pathway increases the expression of adhesion molecules on tumor vasculature to improve effector T-cell infiltration and function. T-cell transfer and CD93 blockade synergistically improve tumor vascular maturation, as well as inhibit tumor progression. Anti-CD93 selectively promotes effector T-cell infiltration in a tumorous setting where the CD93 pathway is upregulated. In a solid mouse tumor model, blockade of the CD93 pathway improves CAR-T therapy. CONCLUSIONS CD93 blockade normalizes tumor vasculature leading to improved effector T-cell infiltration and function in solid cancers. Our study advocates the application of CD93 blockade for ACT in solid cancers.
Collapse
Affiliation(s)
- Yi Sun
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Elliott Yee
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yuki Fujiwara
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kaitlyn Dickinson
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yujie Guo
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Zhiwei Sun
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Junyi Hu
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Eduardo Davila
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard D Schulick
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yuwen Zhu
- Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
44
|
Castro-Robles B, Cimas FJ, Arias-Salazar L, Ontañón J, Lozano J, López-López S, Andrés-Pretel F, Requena-Calleja MÁ, Mas A, Serrano-Heras G, Segura T, Solera J. Distinct response patterns of endothelial markers to the BNT162b2 mRNA COVID-19 booster vaccine are associated with the spike-specific IgG antibody production. Front Immunol 2025; 15:1471401. [PMID: 39835131 PMCID: PMC11743620 DOI: 10.3389/fimmu.2024.1471401] [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: 07/27/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Despite the efficacy and safety of SARS-CoV-2 vaccines, inflammatory and/or thrombotic episodes have been reported. Since the impact of COVID-19 vaccines on the endothelium remains uncertain, our objective was to assess endothelial activation status before and 90 days after the third dose of the BNT162b2 mRNA COVID-19 vaccine. Methods A prospective longitudinal study was conducted at University General Hospital of Albacete, involving 38 healthy health-care workers. Serum levels of endothelial markers (endocan and sVCAM-1) and spike S1-specific IgG antibodies were determined before and at 7, 15, 24 and 90days following vaccination. To analyze each participant´s individual response, we calculated relative increases/decreases (delta values) in endothelial markers and antibodies concentrations compared to their pre-vaccination levels. Results We identified two significantly distinct profiles of endothelial markers response, characterized by either increased or decreased serum levels of endocan and sVCAM. Incremental and decremental response groups did not differ in terms of age, sex, cardiovascular risk factors, previous SARS-CoV-2 infection and influenza vaccine co-administration. However, these responses were significantly associated with the relative spike-specific antibody production. Specifically, the greatest relative increase in antibodies was found in the decremental responders. Additionally, the higher delta antibody production was observed in non-previously infected individuals. Conclusion Administration of the BNT162b2 booster vaccine triggered a non-homogenous response of endothelial function markers among the study participants. Our findings improve the understanding of individual responses to the mRNA COVID-19 booster vaccine, which could be useful in assessing the need for booster doses, particularly in population at risk of vascular complications.
Collapse
Affiliation(s)
- Beatriz Castro-Robles
- Research Unit, General University Hospital of Albacete, Health Service of Castilla-La Mancha (SESCAM), Albacete, Spain
| | - Francisco J. Cimas
- Molecular Oncology Laboratory, Molecular Medicine Unit, Associated Unit of Biomedicine, University of Castilla-La Mancha-Spanish National Research Council (UCLM-CSIC), Albacete, Spain
- Mecenazgo COVID-19, Faculty of Medicine, University of Castilla-La Mancha, Albacete, Spain
| | - Lourdes Arias-Salazar
- Research Unit, General University Hospital of Albacete, Health Service of Castilla-La Mancha (SESCAM), Albacete, Spain
| | - Jesús Ontañón
- Immunology Unit, Clinical Analysis Department, General University Hospital of Albacete, Albacete, Spain
| | - Julia Lozano
- Microbiology Department, General University Hospital of Albacete, Albacete, Spain
| | - Susana López-López
- Research Unit, General University Hospital of Albacete, Health Service of Castilla-La Mancha (SESCAM), Albacete, Spain
| | - Fernando Andrés-Pretel
- Research Unit, General University Hospital of Albacete, Department of Statistics, Foundation of the National Paraplegics Hospital of Toledo, Albacete, Spain
| | | | - Antonio Mas
- Biomedicine Institute of UCLM (IB-UCLM), Faculty of Medicine, University of Castilla-La Mancha, Albacete, Spain
- Faculty of Pharmacy, University of Castilla-La Mancha, Albacete, Spain
- Associated Unit of Biomedicine UCLM-CSIC, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Gemma Serrano-Heras
- Research Unit, General University Hospital of Albacete, Health Service of Castilla-La Mancha (SESCAM), Albacete, Spain
| | - Tomás Segura
- Biomedicine Institute of UCLM (IB-UCLM), Faculty of Medicine, University of Castilla-La Mancha, Albacete, Spain
- Neurology Department, General University Hospital of Albacete, SESCAM, Albacete, Spain
| | - Javier Solera
- Faculty of Medicine, University of Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
45
|
Deng R, Huang G, Zhou J, Zeng K. PLASMA PROTEOME, METABOLOME MENDELIAN RANDOMIZATION IDENTIFIES SEPSIS THERAPEUTIC TARGETS. Shock 2025; 63:52-63. [PMID: 39194222 DOI: 10.1097/shk.0000000000002465] [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: 08/29/2024]
Abstract
ABSTRACT Background : The interrelation between the plasma proteome and plasma metabolome with sepsis presents a multifaceted dynamic that necessitates further research to elucidate the underlying causal mechanisms. Methods : Our investigation used public genome-wide association study data to explore the relationships among the plasma proteome, metabolome, and sepsis, considering different sepsis subgroup. Initially, two-sample Mendelian randomization established causal connections between the plasma proteome and metabolome with sepsis. Subsequently, multivariate and iterative Mendelian randomization analyses were performed to understand the complex interactions in plasma during sepsis. The validity of these findings was supported by thorough sensitivity analyses. Result : The study identified 25 plasma proteins that enhance risk and 34 that act as protective agents in sepsis. After P value adjustment (0.05/1306), ICAM5 emerged with a positive correlation to sepsis susceptibility ( P value = 2.14E-05, OR = 1.10, 95% CI = 1.05-1.15), with this significance preserved across three sepsis subgroup examined. Additionally, 29 plasma metabolites were recognized as risk factors, and 15 as protective factors for sepsis outcomes. After P value adjustment (0.05/997), elevated levels of 1,2,3-benzenetriol sulfate (2) was significantly associated with increased sepsis risk ( P value = 3.37E-05, OR = 1.18, 95% CI = 1.09-1.28). Further scrutiny revealed that this plasma metabolite notably augments the abundance of ICAM5 protein ( P value = 3.52E-04, OR = 1.11, 95% CI = 1.04-1.17), devoid of any detected heterogeneity, pleiotropy, or reverse causality. Mediated Mendelian randomization revealed ICAM5 mediated 11.9% of 1,2,3-benzenetriol sulfate (2)'s total effect on sepsis progression. Conclusion : This study details the causal link between the plasma proteome and metabolome with sepsis, highlighting the roles of ICAM5 and 1,2,3-benzenetriol sulfate (2) in sepsis progression, both independently and through crosstalk.
Collapse
Affiliation(s)
| | - Guiming Huang
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou City, Jiangxi Provence, China
| | - Juan Zhou
- Department of Thyroid and Breast Surgery, Ganzhou People's Hospital, Ganzhou City, Jiangxi Provence, China
| | - Kai Zeng
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
46
|
Rashki M, Ghasemzadeh Rahbardar M, Boskabady MH. Nutritional Advantages of Walnut ( Juglans regia L.) for Cardiovascular Diseases: A Comprehensive Review. Food Sci Nutr 2025; 13:e4526. [PMID: 39803290 PMCID: PMC11717060 DOI: 10.1002/fsn3.4526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/07/2024] [Accepted: 09/24/2024] [Indexed: 01/16/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain one of the leading causes of morbidity and mortality worldwide. In recent years, the potential role of dietary interventions in preventing and managing CVDs has gained significant attention. Among these dietary components, walnuts (Juglans regia L.) have emerged as a promising candidate due to their unique nutrient profile and potential cardiovascular benefits. This review aims to provide a comprehensive analysis of the existing literature on the role of walnuts in cardiovascular health. Using databases from Scopus, Google Scholar, and PubMed, the most relevant in vitro, in vivo, and clinical trial research has been collected from the time of inception until 2024. Several studies have shown that walnut consumption has a positive effect on a variety of cardiovascular risk factors. Walnut bioactive ingredients, including omega-3 fatty acids, antioxidants, fiber, and polyphenols, have been demonstrated to improve lipid profiles, blood pressure, endothelial function, inflammation, oxidative stress, and thrombosis. These processes all contribute to the possible cardioprotective properties of walnuts. Epidemiological and clinical research indicates that daily walnut consumption can reduce the risk of CVDs like coronary heart disease and stroke. Walnuts may aid in managing CVDs through mechanisms such as enhancing lipid profiles, reducing inflammation, and improving overall cardiovascular function. This review highlights the potential role of walnuts as a dietary strategy for the prevention and management of CVDs. Further understanding of the mechanisms and long-term effects of walnut consumption is crucial for optimizing their therapeutic potential and integrating them into clinical practice. Future research should focus on elucidating specific dose-response relationships and exploring the synergistic effects of walnuts in combination with other dietary and lifestyle interventions.
Collapse
Affiliation(s)
- Mostafa Rashki
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| | | | - Mohammad Hossein Boskabady
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Physiology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
47
|
Roudi HS, Safaei R, Dabbaghi MM, Fadaei MS, Saberifar M, Sakhaee K, Rahimi VB, Askari VR. Mechanistic Insights on Cardioprotective Properties of Ursolic Acid: Regulation of Mitochondrial and Non-mitochondrial Pathways. Curr Pharm Des 2025; 31:1037-1056. [PMID: 39710917 DOI: 10.2174/0113816128344497241120025757] [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: 07/15/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 12/24/2024]
Abstract
Ursolic acid, a natural pentacyclic triterpenoid compound, has been shown to have significant cardioprotective effects in various preclinical studies. This article reviews the various mechanisms by which ursolic acid achieves its cardioprotective effects, highlighting its potent anti-oxidant, anti-inflammatory, and anti- apoptotic properties. Ursolic acid upregulates anti-oxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GPx), effectively reducing oxidative stress, thereby decreasing reactive oxygen species (ROS) and improving lipid peroxidation levels. Furthermore, ursolic acid downregulates pro-inflammatory cytokines and inhibits key inflammatory pathways, such as nuclear factor kappa B (NF-κB), which results in its anti-inflammatory effects. These actions help in protecting cardiac tissues from acute and chronic inflammation. Ursolic acid also promotes mitochondrial function and energy metabolism by enhancing mitochondrial biogenesis and reducing dysfunction, which is critical during ischemia-reperfusion (I/R) injury. Additionally, ursolic acid influences multiple molecular pathways, including B-cell leukemia/lymphoma 2 protein (Bcl- 2)/Bcl-2 associated x-protein (Bax), miR-21/extracellular signal-regulated kinase (ERK), and phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), to reduce cardiomyocyte apoptosis. Collectively, these properties make ursolic acid a promising therapeutic agent for cardiovascular diseases (CVDs), warranting further research and clinical trials to harness its potential fully.
Collapse
Affiliation(s)
- Hesan Soleimani Roudi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rozhan Safaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mahdi Dabbaghi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Saleh Fadaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Saberifar
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Katayoun Sakhaee
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
48
|
Li Z, He Y, Zhang Q, Li B, Xiu R, Zhang H. Characterization of microcirculatory endothelial functions in a D-Galactose-induced aging model. Microvasc Res 2025; 157:104757. [PMID: 39490807 DOI: 10.1016/j.mvr.2024.104757] [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: 07/05/2024] [Revised: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Microcirculation health is critical to human health, and aging is an important factor affecting microcirculation health. Although D-Galactose has been widely used in aging research models, there is a lack of relevant studies on D-Galactose simulating microcirculatory aging. Here, we explored microcirculatory endothelial function in D-Galactose-induced aging mice. METHODS Intraperitoneal injection of 150 mg/(kg·d) of D-Galactose was given to cause senescence in mice. Aging was evaluated by SA-β-gal (senescence-associated β-galactosidase) staining. The auricular skin and hepatic microcirculation of mice were observed and detected by enzyme-linked immunosorbent assay (ELISA), immunohistochemistry (IHC) and microcirculation apparatus. The aging of microcirculation was analyzed from oxidative stress, endothelial impairment, inflammation, microvascular morphology and hemodynamics. RESULTS In aging mice, percentage of SA-β-gal positive area, oxidative stress products reactive oxygen species (ROS) and nitric oxide (NO), endothelial impairment marker syndecan-1 (SDC-1), stromal cell derived factor-1 (SDF-1), intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in the senescence-associated secretory phenotype (SASP) were all up-regulated. The tortuosity of microvessels increased in aging mice, the linear density did not change significantly, but the total length of narrow microvessels (TLNMV) increased and wide microvessels (TLWMV) decreased, speculate that vasomotor dysfunction may be present. Hemodynamically, both perfusion and velocity of blood flow were reduced in senescent mice, presumably due to endothelial dysfunction. CONCLUSION Microcirculatory endothelial dysfunction is induced by D-Galactose, leading to microcirculatory aging. In vivo, this is manifested by elevated levels of oxidative stress, impaired endothelial glycocalyx (eGC), and a greater production of chemokines and adhesive molecules. These changes cause vasomotor dysfunction and remodeling, ultimately leading to hemodynamic impairment.
Collapse
Affiliation(s)
- Zhuo Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yuhong He
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Qiuju Zhang
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Bingwei Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Ruijuan Xiu
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Honggang Zhang
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China.
| |
Collapse
|
49
|
Coquelet P, Da Cal S, El Hage G, Tastet O, Balthazard R, Chaumont H, Yuh SJ, Shedid D, Arbour N. Specific plasma biomarker signatures associated with patients undergoing surgery for back pain. Spine J 2025; 25:32-44. [PMID: 39276871 DOI: 10.1016/j.spinee.2024.09.002] [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: 04/15/2024] [Revised: 08/02/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND CONTEXT Intervertebral disc degeneration (IDD) affects numerous people worldwide. The role of inflammation is increasingly recognized but remains incompletely resolved. Peripheral molecules could access neovascularized degenerated discs and contribute to the ongoing pathology. PURPOSE To assess a large array of plasma molecules in patients with IDD to identify biomarkers associated with specific spinal pathologies and prognostic biomarkers for the surgery outcome. DESIGN Prospective observational study combining clinical data and plasma measures. PATIENT SAMPLE Plasma samples were collected just before surgery. Extensive clinical data (age, sex, smoking status, Modic score, glomerular filtration rate, etc.) were extracted from clinical files from 83 patients with IDD undergoing spine surgery. OUTCOME MEASURES Recovery 2 months postsurgery as assessed by the treating neurosurgeon. METHODS Over 40 biological molecules were measured in patients' plasma using multiplex assays. Statistical analyses were performed to identify associations between biological and clinical characteristics (age, sex, Body Mass Index (BMI), smoking status, herniated disc, radiculopathy, myelopathy, stenosis, MODIC score, etc.) and plasma levels of biological molecules. RESULTS Plasma levels of Neurofilament Light chain (NfL) were significantly elevated in patients with myelopathy and spinal stenosis compared to herniated disc. Plasma levels of C- reactive protein (CRP), Neurofilament Light chain (NfL), and Serum Amyloid A (SAA) were negatively associated, while CCL22 levels were positively associated with an efficient recovery 2 months postsurgery. CONCLUSIONS Our results show that CRP and CCL22 plasma levels combined with the age of the IDD patient can predict the 2-month postsurgery recovery (Area Under the Curve [AUC]=0.883). Moreover, NfL could become a valuable monitoring tool for patients with spinal cord injuries.
Collapse
Affiliation(s)
- Perrine Coquelet
- Department of Neurosciences, Université de Montréal, Montréal, Quebec, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Sandra Da Cal
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Gilles El Hage
- Neurosurgery Service, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Quebec, Canada
| | - Olivier Tastet
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Renaud Balthazard
- Department of Neurosciences, Université de Montréal, Montréal, Quebec, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Hugo Chaumont
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Sung-Joo Yuh
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada; Neurosurgery Service, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Quebec, Canada; Department of Surgery, Division of Neurosurgery, Université de Montréal, Montréal, Quebec, Canada
| | - Daniel Shedid
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada; Neurosurgery Service, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Quebec, Canada; Department of Surgery, Division of Neurosurgery, Université de Montréal, Montréal, Quebec, Canada
| | - Nathalie Arbour
- Department of Neurosciences, Université de Montréal, Montréal, Quebec, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada.
| |
Collapse
|
50
|
Perrone P, Ortega-Luna R, Manna C, Álvarez-Ribelles Á, Collado-Diaz V. Increased Adhesiveness of Blood Cells Induced by Mercury Chloride: Protective Effect of Hydroxytyrosol. Antioxidants (Basel) 2024; 13:1576. [PMID: 39765902 PMCID: PMC11673208 DOI: 10.3390/antiox13121576] [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: 11/11/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Mercury (Hg) is a highly toxic environmental contaminant that can harm human health, ultimately leading to endothelial dysfunction. Hg toxicity is partially mediated by the exposure of the cell membrane's surface of erythrocytes (RBCs) to phosphatidylserine (PS). In the context of these challenges, hydroxytyrosol, a phenolic compound of olive oil, has the ability to mitigate the toxic effects of Hg. This study aims to analyze the effect of Hg on the adhesion of RBCs and polymorphonuclear cells (PMNs) to the vascular endothelium and the potential protective effect of hydroxytyrosol, as these interactions are crucial in the development of cardiovascular diseases (CVDs). RBCs, PMNs, and human vein endothelial cells (HUVECs) were treated with increasing concentrations of HgCl2 and, in some cases, with hydroxytyrosol, and their adhesion to HUVECs and the expression of adhesion molecules were subsequently analyzed. Our results demonstrate that HgCl2 significantly increases the adhesion of both RBCs (2.72 ± 0.48 S.E.M., p-value < 0.02) and PMNs (11.19 ± 1.96 S.E.M., p-value < 0.05) to HUVECs and that their adhesiveness is significantly reduced following treatment with hydroxytyrosol (RBCs, 1.2 ± 1.18 S.E.M., p-value < 0.02 and PMNs, 4.04 ± 1.35 S.E.M., p-value < 0.06). Interestingly, HgCl2 does not alter the expression of adhesion molecules on either HUVECs or RBCs, suggesting that reduced exposure to PS is a key factor in hydroxytyrosol protection against HgCl2-induced RBC adhesion to the endothelium. On the other hand, HgCl2 induces increased expression of several PMN adhesion molecules (CD11b 215.4 ± 30.83 S.E.M. p-value < 0.01), while hydroxytyrosol inhibits their expression (e.g., CD11b 149 ± 14.35 S.E.M., p-value < 0.03), which would seem to be the mechanism by which hydroxytyrosol restricts PMN-endothelium interactions. These results provide new insights into the molecular mechanisms through which hydroxytyrosol mitigates the harmful effects of Hg on cardiovascular health, highlighting its potential as a therapeutic agent that can reduce the cardiovascular risk related to heavy metal exposure.
Collapse
Affiliation(s)
- Pasquale Perrone
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (R.O.-L.); (V.C.-D.)
| | - Caterina Manna
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Ángeles Álvarez-Ribelles
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (R.O.-L.); (V.C.-D.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Victor Collado-Diaz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (R.O.-L.); (V.C.-D.)
| |
Collapse
|