1
|
Lin Y, Wang J, Bu F, Zhang R, Wang J, Wang Y, Huang M, Huang Y, Zheng L, Wang Q, Hu X. Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis. Gut Microbes 2025; 17:2452229. [PMID: 39840620 DOI: 10.1080/19490976.2025.2452229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.
Collapse
Affiliation(s)
- Yuling Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Bu
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Wang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiumei Hu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Song CY, Huang HZ, Yan TT, Cui CX, Wu HY, Chen J, Peng JH, Chen NY, Tang J, Pan SL. Downregulation of miR-27a-3p induces endothelial injury and senescence and its significance in the development of coronary heart disease. Cell Signal 2025; 131:111759. [PMID: 40147550 DOI: 10.1016/j.cellsig.2025.111759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
miR-27a-3p is a multifunctional miRNA that plays a critical role in the process of angiogenesis. However, its specific effect on coronary heart disease (CHD), particularly on the regulation of downstream molecules and the resulting impact on endothelial cell injury, has not yet been fully elucidated. This study aimed to explore the relationship between miR-27a-3p and CHD and its underlying mechanical molecular pathways in CHD patients and modeled endothelial cells with techniques such as RT-qPCR, RNA sequencing and bioinformatics. Consequently, the expression of miR-27a-3p was significantly decreased in CHD patients. In endothelial cells, overexpression of miR-27a-3p was observed to decrease malonaldehyde, gamma H2A histone family member X and interleukin 6 while increased superoxide dismutase, thus reduced endothelial injury and senescence. RNA sequencing and bioinformatics revealed glutamate ionotropic receptor NMDA type subunit 2D (GRIN2D) as a target gene of miR-27a-3p, and dual luciferase assays confirmed the direct binding of miR-27a-3p to the 3'UTR of GRIN2D. Subsequent validation experiments demonstrated that miR-27a-3p inhibited the protein expression of GRIN2D and PKC and suppressed the activation of the MAPK/ERK signaling pathway by reduced downstream MEK and ERK phosphorylation, leading to enhanced endothelial apoptosis. In conclusion, miR-27a-3p played a crucial role in regulating endothelial cell dysfunction which may trigger coronary atherosclerosis and CHD by targeting GRIN2D in the PKC/MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Chong-Yang Song
- Department of Pathophysiology, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Hai-Zhen Huang
- Department of Pathophysiology, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Ting-Ting Yan
- Department of General Geriatrics, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Chen-Xi Cui
- Department of Pathophysiology, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China; Department of pathology, the First People's Hospital of Nanning, the Fifth Affiliated Hospital of Guangxi Medical University, 89 Qixing Road, Nanning 530022, Guangxi, China
| | - Hua-Yu Wu
- Experimental Center for Medicine, the First People's Hospital of Nanning, the Fifth Affiliated Hospital of Guangxi Medical University, 89 Qixing Road, Nanning 530022, Guangxi, China
| | - Jing Chen
- Department of Pathophysiology, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China; Biobank, Department of Scientific Research, the First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning 530022, Guangxi, China
| | - Jun-Hua Peng
- Department of Pathophysiology, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Ning-Yuan Chen
- Department of Pathophysiology, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Jun Tang
- Department of Pathophysiology, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China.
| | - Shang-Ling Pan
- Department of Pathophysiology, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China.
| |
Collapse
|
3
|
Mahmood NMS, Mahmud AMR, Maulood IM. Vascular actions of Ang 1-7 and Ang 1-8 through EDRFs and EDHFs in non-diabetes and diabetes mellitus. Nitric Oxide 2025; 156:9-26. [PMID: 40032212 DOI: 10.1016/j.niox.2025.02.003] [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: 10/14/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in regulating vascular homeostasis, while angiotensin 1-8 (Ang 1-8) traditionally dominates as a vasoconstrictor factor. However, the discovery of angiotensin 1-7 (Ang 1-7) and Ang 1-8 has revealed counter-regulatory mechanisms mediated through endothelial-derived relaxing factors (EDRFs) and endothelial-derived hyperpolarizing factors (EDHFs). This review delves into the vascular actions of Ang 1-7 and Ang 1-8 in both non-diabetes mellitus (non-DM) and diabetes mellitus (DM) conditions, highlighting their effects on vascular endothelial cell (VECs) function as well. In a non-DM vasculature context, Ang 1-8 demonstrate dual effect including vasoconstriction and vasodilation, respectively. Additionally, Ang 1-7 induces vasodilation upon nitric oxide (NO) production as a prominent EDRFs in distinct mechanisms. Further research elucidating the precise mechanisms underlying the vascular actions of Ang 1-7 and Ang 1-8 in DM will facilitate the development of tailored therapeutic interventions aimed at preserving vascular health and preventing cardiovascular complications.
Collapse
Affiliation(s)
- Nazar M Shareef Mahmood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq.
| | - Almas M R Mahmud
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Ismail M Maulood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
4
|
Gao J, Pan H, Guo X, Huang Y, Luo JY. Endothelial Krüppel-like factor 2/4: Regulation and function in cardiovascular diseases. Cell Signal 2025; 130:111699. [PMID: 40023301 DOI: 10.1016/j.cellsig.2025.111699] [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: 12/02/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
This review presents an overview of the regulation, function, disease-relevance and pharmacological regulation of the critical endothelial transcription factors KLF2/4 in vasculature. The regulatory mechanisms of KLF2/4 expression and activity in vascular endothelium in response to hemodynamic forces and biochemical stimuli are depicted. The functional effects mediated by direct or indirect target genes of KLF2/4 in endothelial cells are systematically summarized. The contributory roles that dysregulated KLF2/4 play in relevant cardiovascular pathologies, such as atherosclerotic vascular lesions, pulmonary arterial hypertension and vascular complications of diabetes were reviewed. Moreover, this review also discusses the pharmacological regulation of KLF2/4 by drugs used in clinics and therapeutic possibility by directly targeting these two transcription factors for treating atherosclerotic cardiovascular diseases. Finally, prospective opinions on the gaps in disclosing novel vascular function mediated by KLF2/4 and future research needs are expressed.
Collapse
Affiliation(s)
- Jing Gao
- Department of Cardiology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Hongjie Pan
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Jiang-Yun Luo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Cheng R, Cheng X, Jiang D, Xiong J, Ding Y, Liu J, Zhao H, Feng H, Wu D, Zhang W. Spectrum-effect relationship of the cardiovascular-protective effect of with Chrysanthemi Flos by UPLC-MS/MS and component knock-out method. Food Chem Toxicol 2025; 200:115372. [PMID: 40054725 DOI: 10.1016/j.fct.2025.115372] [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: 12/09/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Chrysanthemi Flos (CF), as one of the important 'dual-use' plants, possesses great pharmacological research and development potential. This work aimed to find the pharmacodynamic material basis of CF in cardiovascular-protection by spectrum-effect relationship and component knock-out method. The fingerprint was established by ultra-high performance liquid chromatography and 25 peaks were picked out as common peaks. The common peaks were identified by ultra-performance liquid chromatography-quadrupole-orbitrap-mass spectrometry including twelve flavonoids, nine phenylpropanoids, three organic acids, and one nucleoside. The cardiovascular-protective effect of CF was determined by angiotensin II-induced injury model of human umbilical vein endothelial cells. Grey relation analysis, partial least squares regression analysis and Pearson's correlation analysis were performed to assess the relationship between the cardiovascular-protective effect and ingredients. Spectrum-effect relationship and component knock-out method revealed that P11 (luteolin-7-O-β-D-glucoside), P14 (3,4-O-dicaffeoylquinic acid), P16 (1,5-O-dicaffeoylquinic acid), and P17 (3,5-O-dicaffeoylquinic acid) were the pharmacological material basis for the cardiovascular-protective effect of CF. This work preliminarily elucidated the pharmacodynamic material basis of cardiovascular-protective effect of CF, which could be used to considerable methods and insight for the fundamental research of the pharmacodynamic material basis of Traditional Chinese Medicine.
Collapse
Affiliation(s)
- Ranran Cheng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Xiang Cheng
- Bozhou Vocational and Technical College, Bozhou, 236800, China.
| | - Dongliang Jiang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Junwei Xiong
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Yangfei Ding
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Juan Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Hongsu Zhao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Hangmin Feng
- Anhui Provincial Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei, 230012, China.
| | - Deling Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Provincial Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei, 230012, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, China.
| | - Wei Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Provincial Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei, 230012, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, China; Joint Research Center for Chinese Herbal Medicine of Anhui of IHM, Hefei, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
6
|
Briones A, Hernanz R, García‐Redondo A, Rodríguez C, Blanco‐Colio L, Val‐Blasco A, Alonso M, Salaices M. Role of Inflammatory and Proresolving Mediators in Endothelial Dysfunction. Basic Clin Pharmacol Toxicol 2025; 136:e70026. [PMID: 40159875 PMCID: PMC11955787 DOI: 10.1111/bcpt.70026] [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: 11/24/2024] [Revised: 02/20/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Excessive local inflammation is a common mechanism in many cardiovascular diseases (CVDs) such as hypertension, atherosclerosis and aortic aneurysms. In endothelial cells, inflammatory cytokines such as interferons, tumour necrosis factor alpha or interleukins increase oxidative stress and contractile prostanoids and the expression of adhesion molecules that reduce nitric oxide (NO) availability and bind leucocytes, thereby impairing endothelial function. Despite this evidence, anti-inflammatory therapies are not yet indicated for the treatment of most CVD. Resolution of inflammation is mediated by a family of specialized pro-resolving mediators (SPMs) that act on cognate G protein-coupled receptors to limit immune cell infiltration and initiate tissue repair. SPMs, generated from omega-3 and omega-6 polyunsaturated fatty acids, belong to four major families: lipoxins, resolvins, protectins and maresins. SPM receptors are expressed in immune and vascular cells where they regulate important processes such as phagocytosis and polarization, production of cytokines, NO and prostacyclin, and modulation of smooth muscle cell phenotype. Growing evidence in animal models demonstrates that activation of SPM receptors can protect vascular function and structure and provide beneficial effects in various CVD. We will review recent advances in the role of inflammation and SPMs in vascular (dys)function in hypertension, atherosclerosis, and aortic aneurysms.
Collapse
Affiliation(s)
- Ana M. Briones
- Department of Pharmacology, Faculty of MedicineUniversidad Autónoma de MadridMadridSpain
- Hospital La Paz Institute for Health Research (IdiPaz)MadridSpain
- CIBER Cardiovascular (CIBERCV)MadridSpain
| | - Raquel Hernanz
- Hospital La Paz Institute for Health Research (IdiPaz)MadridSpain
- CIBER Cardiovascular (CIBERCV)MadridSpain
- Department of Basic Health SciencesUniversidad Rey Juan CarlosMadridSpain
| | - Ana B. García‐Redondo
- Hospital La Paz Institute for Health Research (IdiPaz)MadridSpain
- CIBER Cardiovascular (CIBERCV)MadridSpain
- Department of Physiology, Faculty of MedicineUniversidad Autónoma de MadridMadridSpain
| | - Cristina Rodríguez
- CIBER Cardiovascular (CIBERCV)MadridSpain
- Institut de Recerca Sant Pau (IR SANT PAU)BarcelonaSpain
| | - Luis M. Blanco‐Colio
- CIBER Cardiovascular (CIBERCV)MadridSpain
- Laboratory for Vascular BiologyIIS‐Fundación Jiménez DíazMadridSpain
| | - Almudena Val‐Blasco
- Hospital La Paz Institute for Health Research (IdiPaz)MadridSpain
- CIBER Cardiovascular (CIBERCV)MadridSpain
| | - María J. Alonso
- Hospital La Paz Institute for Health Research (IdiPaz)MadridSpain
- CIBER Cardiovascular (CIBERCV)MadridSpain
- Department of Basic Health SciencesUniversidad Rey Juan CarlosMadridSpain
| | - Mercedes Salaices
- Department of Pharmacology, Faculty of MedicineUniversidad Autónoma de MadridMadridSpain
- Hospital La Paz Institute for Health Research (IdiPaz)MadridSpain
- CIBER Cardiovascular (CIBERCV)MadridSpain
| |
Collapse
|
7
|
Lau C, Primus CP, Shabbir A, Chhetri I, Ono M, Masucci M, Bin Noorany Aubdool MA, Amarin J, Hamers AJ, Khan Z, Kumar NA, Montalvo Moreira SA, Nuredini G, Osman M, Whitear C, Godec T, Kapil V, Massimo G, Khambata RS, Rathod KS, Ahluwalia A. Accelerating inflammatory resolution in humans to improve endothelial function and vascular health: Targeting the non-canonical pathway for NO. Redox Biol 2025; 82:103592. [PMID: 40209616 PMCID: PMC12005330 DOI: 10.1016/j.redox.2025.103592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Chronic cardiovascular diseases (CVD) are characterised by low-grade systemic inflammation in part due to reduced nitric oxide (NO) bioavailability associated with endothelial dysfunction. Bioavailability of NO can be enhanced by activation of the non-canonical pathway, through increased dietary inorganic nitrate consumption with the potential to attenuate inflammation. METHODS We sought to determine whether dietary inorganic nitrate influences the inflammatory response in models of localised (cantharidin-induced blisters) and systemic inflammation (typhoid vaccine), in healthy male volunteers and conducted two clinical trials; Blister-NITRATE and Typhoid-NITRATE respectively. RESULTS We show that dietary nitrate attenuates endothelial dysfunction following typhoid vaccine administration and accelerates resolution of cantharidin-induced blisters. Both phenomena were associated with an increased level of pro-resolving mediators consequent to a reduction in the expression and activity of pro-inflammatory monocytes. Moreover, we show that leukocytes of the monocyte lineage express the nitrite reductase XOR, that may drive localised nitrite reduction to elevate NO (and cGMP) to drive the protective phenotype. CONCLUSIONS Inorganic nitrate improves endothelial function in the setting of systemic inflammation. Whilst the immediate inflammatory response appeared unaffected by inorganic nitrate treatment, during the resolution phase of the acute inflammatory response lower levels of pro-inflammatory classical inflammatory and intermediate monocytes and attenuated levels of inflammatory cytokines and chemokines were evident. We propose that this reflects a pro-resolution phenotype that may be of potential therapeutic benefit in patients with established CVD. CLINICAL TRIAL REGISTRATION URL: https://www. CLINICALTRIALS gov; unique identifiers NCT02715635, NCT03183830.
Collapse
Affiliation(s)
- Clement Lau
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Christopher P Primus
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Asad Shabbir
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ismita Chhetri
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mutsumi Ono
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Masucci
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muhammad Aadil Bin Noorany Aubdool
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Amarin
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alexander Jp Hamers
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Zara Khan
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nitin Ajit Kumar
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Gani Nuredini
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Miski Osman
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Charlotte Whitear
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tom Godec
- Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vikas Kapil
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gianmichele Massimo
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rayomand S Khambata
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Krishnaraj S Rathod
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Cardiology, Barts Heart Centre, 2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Amrita Ahluwalia
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
8
|
Xia H, Pan Z, Hong Y, Zhao Q, Fan W. A retrospective study on the correlation between antibody levels and endothelial function in SLE patients: An analysis based on ultrasound and serum biomarkers. Mol Immunol 2025; 181:66-74. [PMID: 40073698 DOI: 10.1016/j.molimm.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/09/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) was a complex autoimmune disease characterized by a spectrum of clinical and immunological manifestations, with cardiovascular disease (CVD) being a leading cause of morbidity and mortality. Endothelial dysfunction was critical in the pathogenesis of atherosclerosis and other cardiovascular complications in SLE. This study aimed to investigate the correlation between autoantibody levels and endothelial function in SLE patients using ultrasound and serum biomarkers. METHODS A retrospective case-control study was conducted with 317 SLE patients treated from December 2021 to December 2023. Patients were categorized based on Flow-Mediated Dilation (FMD) values into an abnormal endothelial function group (n = 191) and a normal function group (n = 126). Serum biomarkers, including soluble thrombomodulin (sTM), von Willebrand factor (vWF), and soluble vascular cell adhesion molecule-1 (sVCAM-1), were assessed. Autoantibody levels were measured using enzyme-linked immunosorbent assays for anti-double stranded DNA (anti-dsDNA), anti-Smith, and anticardiolipin antibodies levels. Statistical analyses, including correlation and logistic regression, were performed to determine associations between antibody levels and endothelial function. RESULTS Higher levels of Anti-Smith were significantly associated with poorer endothelial function, while higher Anti-dsDNA levels were positive correlated with endothelial function (Anti-Smith: coefficient = -0.168, Std_Error = 0.027, t_value = -6.228, P < 0.001; Anti-dsDNA: coefficient = 0.140, Std_Error = 0.022, t_value = 6.345, P < 0.001). These results underscore the importance of antibody levels in assessing endothelial health. CONCLUSION This study highlights the intricate relationship between specific autoantibodies and endothelial dysfunction in SLE patients. Elevated sVCAM-1 and Anti-Smith levels were associated with a higher risk of endothelial impairment, whereas Anti-dsDNA antibodies showed a positively correlated better endothelial function.
Collapse
Affiliation(s)
- Huan Xia
- Department of Medical Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan Province 571100, China.
| | - Zaixing Pan
- Department of Medical Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan Province 571100, China
| | - Yun Hong
- Department of Medical Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan Province 571100, China
| | - Qingzhu Zhao
- Department of Medical Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan Province 571100, China
| | - Weili Fan
- Department of Medical Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan Province 571100, China
| |
Collapse
|
9
|
Peng P, Ding S, Liang M, Zheng W, Kang Y, Liu W, Shi H, Gao C. A self-sacrificing anti-inflammatory coating promotes simultaneous cardiovascular repair and reendothelialization of implanted devices. Bioact Mater 2025; 47:502-512. [PMID: 40026826 PMCID: PMC11872464 DOI: 10.1016/j.bioactmat.2025.01.037] [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: 11/25/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
During interventional surgeries of implantable cardiovascular devices in addressing cardiovascular diseases (CVD), the inevitable tissue damage will trigger host inflammation and vascular lumen injury, leading to delayed re-endothelization and intimal hyperplasia. Endowing cardiovascular implants with anti-inflammatory and endothelialization functions is conducive to the target site, offering significant tissue repair and regeneration benefits. Herein, inspired by the snake's molting process, a ShedWise device was developed by using the poly(propylene fumarate) polyurethane (PPFU) as the foundational material, which was clicked with hyperbranched polylysine (HBPL) and followed by conjugation with pro-endothelial functional Arg-Glu-Asp-Val peptide (REDV), and finally coated with a "self-sacrificing" layer having reactive oxygen species (ROS)-scavenging ability and degradability. During the acute inflammation in the initial stage of implantation, the ROS-responsive hyperbranched poly(acrylate-capped thioketone-containing ethylene glycol (HBPAK) coating effectively modulated the level of environmental inflammation and resisted initial protein adsorption, showcasing robust tissue protection. As the coating gradually "sacrificed", the exposed hyperbranched HBPL-REDV layer recruited specifically endothelial cells and promoted surface endothelialization. In a rat vascular injury model, the ShedWise demonstrated remarkable efficiency in reducing vascular restenosis, protecting the injured tissue, and fostering re-endothelization of the target site. This innovative design will introduce a novel strategy for surface engineering of cardiovascular implants and other medical devices.
Collapse
Affiliation(s)
- Pai Peng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Shili Ding
- Department of Hand Surgery, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310003, China
| | - Min Liang
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yongyuan Kang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Wenxing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Haifei Shi
- Department of Hand Surgery, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310003, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
- The State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
10
|
Ji XM, Dong XX, Li JP, Tai GJ, Qiu S, Wei W, Silumbwe CW, Damdinjav D, Otieno JN, Li XX, Xu M. Fisetin Clears Senescent Cells Through the Pi3k-Akt-Bcl-2/Bcl-xl Pathway to Alleviate Diabetic Aortic Aging. Phytother Res 2025. [PMID: 40259678 DOI: 10.1002/ptr.8507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/18/2025] [Accepted: 03/29/2025] [Indexed: 04/23/2025]
Abstract
Vascular aging is a major contributor to age-related cardiovascular diseases (CVDs) and type 2 diabetes mellitus (T2DM) induced early arterial aging and excessive senescent cells (SCs) burden in vessels. Inhibiting cellular senescence or eliminating SCs could effectively improve aging-related CVDs. Fisetin, a flavonoid extracted from cotinus coggygria scop, has shown potential in alleviating aging by clearing SCs. This study investigated the unexplored mechanisms and efficacy of fisetin in alleviating T2DM-related aortic aging. The T2DM mouse model was induced using a high-fat diet and low-dose streptozotocin injection. Chronic fisetin treatment's protective effects against aortic aging were assessed via senescence-associated beta-galactosidase (SA-β-Gal) staining, histopathology, and vasomotor function. RNA-sequencing and western blotting identified relevant signaling pathways and protein expression. Fisetin's effects on SCs and senescence-associated secretory phenotype (SASP) factors were evaluated through cell viability, apoptosis, and co-culture assays. Docking simulations suggested fisetin as a potential Phosphoinositide 3-kinase (Pi3k) inhibitor. In vivo, chronic fisetin treatment reduced aortic SCs burden, alleviating T2DM-related and natural aortic aging. In vitro, fisetin selectively induced apoptosis of senescent endothelial cells via regulating the Pi3k-Protein Kinase B (Akt)-B-cell lymphoma (Bcl)-2/Bcl-xl pathway and suppressed SASP and its detrimental effects. Furthermore, fisetin combined with metformin therapy showed superior anti-aging effects on T2DM-related aortic aging compared to metformin monotherapy. In conclusion, chronic fisetin treatment alleviates T2DM-related aortic aging via clearing the SCs burden and abrogating the SASP factors. Fisetin combined with metformin therapy might be a potential therapeutic strategy for T2DM-related CVDs.
Collapse
Affiliation(s)
- Xiao-Man Ji
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin-Xin Dong
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jia-Peng Li
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guang-Jie Tai
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shu Qiu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wei Wei
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ceaser Wankumbu Silumbwe
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Davaadagva Damdinjav
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Joseph Nicolao Otieno
- Institute of Traditional Medicine, Muhimbili University of Health and Allied Sciencea, Dar es Salaam, Tanzania
| | - Xiao-Xue Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
Islam K, Islam R, Nguyen I, Malik H, Pirzadah H, Shrestha B, Lentz IB, Shekoohi S, Kaye AD. Diabetes Mellitus and Associated Vascular Disease: Pathogenesis, Complications, and Evolving Treatments. Adv Ther 2025:10.1007/s12325-025-03185-9. [PMID: 40252164 DOI: 10.1007/s12325-025-03185-9] [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: 08/14/2024] [Accepted: 03/19/2025] [Indexed: 04/21/2025]
Abstract
Diabetes mellitus is a metabolic disorder, characterized by elevated blood sugar levels (hyperglycemia) and insulin dysregulation. This disease is associated with morbidity and mortality, including significant potential vascular complications. High levels of hyperglycemia lead to not only elevated levels of reactive oxygen species but also advanced glycation end products, which are detrimental to the vascular endothelium and reduce protective compounds such as nitric oxide and prostacyclin. This damage contributes to the development of both macrovascular and microvascular complications. The present investigation explores the pathophysiological mechanisms of diabetic vascular complications and evaluates current management strategies, including lifestyle modifications, pharmacological treatments, and emerging therapies. The review underscores the importance of ongoing progress in diabetes management and patient education to lead to optimal patient-health outcomes and quality of life for individuals with diabetes mellitus.
Collapse
Affiliation(s)
- Kazi Islam
- Central State University, 1400 Brush Row Road, Wilberforce, OH, 45384, USA
| | - Rahib Islam
- LSU Health Sciences Center New Orleans School of Medicine, 1901 Gravier Street, New Orleans, LA, 70112, USA
| | - Ivan Nguyen
- LSU Health Sciences Center New Orleans School of Medicine, 1901 Gravier Street, New Orleans, LA, 70112, USA
| | - Hassan Malik
- LSU Health Sciences Center New Orleans School of Medicine, 1901 Gravier Street, New Orleans, LA, 70112, USA
| | - Humza Pirzadah
- LSU Health Sciences Center New Orleans School of Medicine, 1901 Gravier Street, New Orleans, LA, 70112, USA
| | - Barsha Shrestha
- LSU Health Sciences Center New Orleans School of Medicine, 1901 Gravier Street, New Orleans, LA, 70112, USA
| | - Isabella B Lentz
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA.
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
- Department of Pharmacology, Toxicology, and Neurosciences, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| |
Collapse
|
12
|
Zhang C, Wang Y, Huang F, Zhang Y, Huang M, Liu H, Liu Y, Wang Q, Liu C, Angwa L, Gao Y, Sun D, Jiang Y. Novel mechanism of fluoride induced cardiovascular system injury by regulating p53/miR200c-3p during endothelial dysfunction. ENVIRONMENTAL RESEARCH 2025; 271:121102. [PMID: 39952459 DOI: 10.1016/j.envres.2025.121102] [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: 12/03/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND The impairment of the cardiovascular system by fluoride has attracted public health concern, and its toxic effects on ECs have garnered extensive research attention. However, epidemiological clues of fluoride induced cardiovascular injury are limited. The function of ECs is crucial for the early diagnosis of CVD, yet mechanisms through which fluoride disrupts endothelial function are still unclear. PURPOSE To investigate the relationship between fluoride exposure and hypertension in population by epidemiological investigation. To explore the potential mechanism of functional injury of ECs induced by fluoride. RESULT Epidemiological studies have shown that the risk of hypertension in study population increased with the increased of urinary fluoride concentration [OR = 1.565, 95%CI (1.143, 2.142)]. In rat model with fluorosis alongside a model of fluoride induced ECs injury, NaF led to anti-adhesion of ECs and barrier dysfunction. Notably, the expression levels of eNOS and NO were found to be decreased, while the expression levels of ACE, vWF, ICAM-1, VCAM-1 and ET-1 were elevated. Our findings also indicated that NaF induced oxidative stress in ECs, evidenced by significant increased in ROS and MDA levels and decreased protein expression of GPx4 and SOD activity. It was further found that NaF activated the p53/miR-200c-3p signaling axis via ROS, leading to endothelial dysfunction. CONCLUSION This study found that fluoride exposure was a risk factor for hypertension. In addition, fluoride could cause ECs dysfunction by inducing oxidative stress and activating p53/miR-200c-3p. These findings were helpful to further understand the mechanism of fluoride induced cardiovascular system injury and provide a theoretical basis for fluoride induced cardiovascular system injury.
Collapse
Affiliation(s)
- Chao Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Yue Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China
| | - Fengya Huang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China
| | - Yaoyuan Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China
| | - Mingyue Huang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China
| | - Hui Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Yunzhu Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Qiaoyu Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Chang Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China
| | - Linet Angwa
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; United State University-Africa, Department of Phamaceutics, Pharmacy Practice, and Public Health, People's Republic of China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China.
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China.
| | - Yuting Jiang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, People's Republic of China; NHC Key Laboratory of Etiology and Epidemiology(Harbin Medical University), People's Republic of China; Joint Key Laboratory of Endemic Diseases, Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University, People's Republic of China; Center for Chronic Disease Prevention and Control, Harbin Medical University, Harbin, People's Republic of China.
| |
Collapse
|
13
|
Wang Y, Chen T, Peng F, Wang S, Zhao F, Wang X, Xie L. Activating TRPV1 by evodiamine attenuates atherosclerosis by inhibiting endothelial microparticle release. Int Immunopharmacol 2025; 155:114657. [PMID: 40239331 DOI: 10.1016/j.intimp.2025.114657] [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: 09/08/2024] [Revised: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Endothelial microparticles (EMPs) were known as the biomarker of endothelial dysfunction, which could initiate atherosclerosis(AS). The traditional chinese medicine Evodiae and its main alkaloid component, evodiamine, exerts anti-atherosclerosis effects through transient receptor potential vanilloid type 1(TRPV1). However, it is unclear whether the anti-atherosclerosis effect of evodiamine is related to the generation of EMPs. The present study aims to investigate the anti-atherosclerosis effect of evodiamine and the underlying mechanism of the formation and release of EMPs. To establish the AS mice model, ApoE-/- mice were fed a high-fat diet for eight weeks. Histopathology results were evaluated using hematoxylin and eosin staining. The quantity of EMPs was examined by flow cytometry. Serum lipid and cytokines levels were assessed by ELISA kits, and protein expression was determined by Western blotting. We found that evodiamine decreased the plasma EMPs levels, reduced the lipid levels, IL-6 and ET-1 levels, and reduced the size of atherosclerotic lesions in ApoE-/- mice significantly. Moreover, evodiamine significantly down-regulated the expression levels of ROCK, which was involved in the release of EMPs. In contrast, pre-treatment with capsazepine (the blocker of TRPV1) abrogated these effects of evodiamine. In vitro, lipopolysaccharide was used to induce the release of EMPs in the human aortic endothelial cells(HAECs). Consistently silencing the expression of TRPV1 in the cells through siRNA interference resulted in an elevation of EMPs levels and the expression of ROCK. In conclusion, activating TRPV1 by evodiamine may inhibit the activation of ROCK and then decrease the release of EMPs, relieve the inflammation of the endothelial cells, and finally attenuate the development of atherosclerosis.
Collapse
Affiliation(s)
- Yan Wang
- Department Of Physiology, Guizhou University Of Traditional Chinese Medicine, Dongqing Road, Huaxi University town, Guiyang 550025, China.
| | - Tianqi Chen
- Department Of Physiology, Guizhou University Of Traditional Chinese Medicine, Dongqing Road, Huaxi University town, Guiyang 550025, China
| | - Fang Peng
- Department Of Physiology, Guizhou University Of Traditional Chinese Medicine, Dongqing Road, Huaxi University town, Guiyang 550025, China
| | - Shuoshi Wang
- Clinical Medical laboratory, The second affiliated hospital of Guizhou university of traditional chinese medicine, 72 Dongxin Road, Nanming District, Guiyang 550003, China
| | - Fuyang Zhao
- Faculty of Pharmacy, Guizhou university of traditional chinese medicine, Guiyang 550025,China
| | - Xiaoli Wang
- Faculty of Pharmacy, Guizhou university of traditional chinese medicine, Guiyang 550025,China
| | - Linchong Xie
- Faculty of Pharmacy, Guizhou university of traditional chinese medicine, Guiyang 550025,China
| |
Collapse
|
14
|
Benedicto I, Hamczyk MR, Dorado B, Andrés V. Vascular cell types in progeria: victims or villains? Trends Mol Med 2025:S1471-4914(25)00056-5. [PMID: 40240194 DOI: 10.1016/j.molmed.2025.03.005] [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: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare genetic disease caused by progerin, a broadly expressed mutant variant of lamin A protein that accelerates aging and leads to premature death typically in adolescence. Progerin affects many organs and reproduces many characteristics of physiological aging, with the main cause of death in HGPS being atherosclerotic cardiovascular disease (CVD). Due to the rarity of HGPS, advances in understanding the disease and progress toward new therapeutic approaches are crucially dependent on preclinical models. We discuss recent research developments from a variety of HGPS experimental systems, with a special focus on in vivo studies of the role of vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) that are key players in atherosclerosis.
Collapse
Affiliation(s)
- Ignacio Benedicto
- Centro de Investigaciones Biológicas Margarita Salas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| | - Magda R Hamczyk
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, 8000 Aarhus C, Denmark
| | - Beatriz Dorado
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| |
Collapse
|
15
|
Peng H, Du Z, Li J, Wang W, Li Z, Ru S. The sprouting angiogenesis and vascular dysfunction triggered by bisphenol S and tetrabromobisphenol S through disrupting vascular endothelial-cadherin in zebrafish. ENVIRONMENTAL RESEARCH 2025:121632. [PMID: 40246265 DOI: 10.1016/j.envres.2025.121632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Exogenous chemical toxicants may be important inducers of pathological angiogenesis diseases. However, few studies have investigated the associations between pathological angiogenesis diseases and chemical toxicant exposures, and the specific mechanism by which chemical toxicants induce sprouting angiogenesis is unclear. In this study, zebrafish were exposed to bisphenol S (BPS, 1∼100 μg/L) and tetrabromobisphenol S (TBBPS, 0.1 and 10 μg/L) from the embryonic stage to the larval stage to investigate how pollutants interfere with angiogenesis and the function of ectopic sprouting vessels. The results showed that BPS and TBBPS promoted ectopic sprouting angiogenesis in different types of vascular plexuses, including the posterior cardinal vein (PCV) and superficial choroidal vessels (SOVs), at different developmental time points. Proteomic analyses of eGFP-positive endothelial cells (ECs) isolated from Tg(flk1: eGFP) zebrafish revealed that both BPS and TBBPS induced ectopic angiogenesis by acting on vascular endothelial-cadherin (VE-cadherin) and activating downstream proangiogenic signaling. In ectopic sprouting vessels induced by BPS and TBBPS, increased endothelial permeability resulted in white blood cell recruitment. Human oxidized lipids also tended to deposit in these ectopic vessels following BPS and TBBPS exposure. These findings suggest that chemical toxicant-induced ectopic angiogenesis is an important cause of vascular dysfunction and related diseases.
Collapse
Affiliation(s)
- Hongyuan Peng
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Zehui Du
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Jiali Li
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China.
| | - Ze Li
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
16
|
Zaman S, Wasfy JH, Kapil V, Ziaeian B, Parsonage WA, Sriswasdi S, Chico TJA, Capodanno D, Colleran R, Sutton NR, Song L, Karam N, Sofat R, Fraccaro C, Chamié D, Alasnag M, Warisawa T, Gonzalo N, Jomaa W, Mehta SR, Cook EES, Sundström J, Nicholls SJ, Shaw LJ, Patel MR, Al-Lamee RK. The Lancet Commission on rethinking coronary artery disease: moving from ischaemia to atheroma. Lancet 2025; 405:1264-1312. [PMID: 40179933 DOI: 10.1016/s0140-6736(25)00055-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/01/2025] [Accepted: 01/09/2025] [Indexed: 04/05/2025]
Affiliation(s)
- Sarah Zaman
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Jason H Wasfy
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Vikas Kapil
- William Harvey Research Institute, Centre for Cardiovascular Medicine and Devices, NIHR Barts Biomedical Research Centre, Queen Mary University of London, St Bartholomew's Hospital, London, UK
| | - Boback Ziaeian
- Division of Cardiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| | - William A Parsonage
- Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, QLD, Australia; Department of Cardiology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Sira Sriswasdi
- Center of Excellence in Computational Molecular Biology, Chulalongkorn University, Pathum Wan, Bangkok, Thailand; Faculty of Medicine, Chulalongkorn University, Pathum Wan, Bangkok, Thailand
| | - Timothy J A Chico
- School of Medicine and Population Health, University of Sheffield, Sheffield, UK; British Heart Foundation Data Science Centre, Health Data Research UK, London, UK
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero Universitaria Policlinico, University of Catania, Catania, Italy
| | - Róisín Colleran
- Department of Cardiology and Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Nadia R Sutton
- Department of Internal Medicine, and Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lei Song
- Department of Cardiology, National Clinical Research Centre for Cardiovascular Diseases, Fuwai Hospital, Beijing, China; Peking Union Medical College (Chinese Academy of Medical Sciences), Beijing, China
| | - Nicole Karam
- Cardiology Department, European Hospital Georges Pompidou, Paris City University, Paris, France
| | - Reecha Sofat
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Chiara Fraccaro
- Division of Cardiology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Daniel Chamié
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Mirvat Alasnag
- Cardiac Center, King Fahd Armed Forces Hospital, Jeddah, Saudi Arabia
| | | | - Nieves Gonzalo
- Cardiology Department, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Walid Jomaa
- Cardiology B Department, Fattouma Bourguiba University Hospital, University of Monastir, Monastir, Tunisia
| | - Shamir R Mehta
- Population Health Research Institute, Hamilton Health Sciences, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Elizabeth E S Cook
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Johan Sundström
- Uppsala University, Uppsala, Sweden; The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | | | - Leslee J Shaw
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manesh R Patel
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Rasha K Al-Lamee
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
17
|
Zhang C, Zhang L, Chen L, Zheng Y, Zhang S, Guo S, Hu S. Mitigating effects of hydroxysafflor yellow a on atherosclerotic inflammatory responses based on flavonoid macromolecule compound: Inhibition of Piezo1-YAP/JNK protein pathway. Int J Biol Macromol 2025; 309:142961. [PMID: 40220830 DOI: 10.1016/j.ijbiomac.2025.142961] [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: 03/09/2025] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/14/2025]
Abstract
Atherosclerosis (AS) is an important cardiovascular disease caused by inflammation. The inhibitory effect of HysA on the Piezo1-YAP/JNK signaling pathway in a mouse model of AS was studied to clarify its anti-inflammatory effects. The study evaluated HysA's ability to bind Piezo1 through molecular docking analysis. C57BL/6 mice were used to establish AS model, and cell treatment and animal experiments were carried out. Inflammatory markers and lipid accumulation were evaluated using quantitative real-time PCR (qRT-PCR), Westernblot, enzyme-linked immunosorbent assay (ELISA), immunofluorescence, and oil red O staining techniques. Weight changes in mice were recorded to monitor disease progression. The molecular docking results show that HysA has a good binding affinity with Piezo1. In a mouse model of AS, treatment with HysA significantly reduced levels of inflammatory cytokines and inhibited the activation of YAP and JNK signaling pathways. The HysA treatment group showed lower oil red O staining levels, indicating that it effectively mitigated lipid deposition. Therefore, HysA significantly alleviates the inflammatory response of atherosclerosis by inhibiting the Piezo1-YAP/JNK signaling pathway, suggesting its potential application in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Chunxiao Zhang
- Traditional Chinese Medicine Department, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing City 312000, Zhejiang Province, China
| | - Lei Zhang
- Internal Medicine Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan City 250011, Shandong Province, China
| | - Lanyong Chen
- Clinical College, Zhejiang Chinese Medical University, Hangzhou City 310000, Zhejiang Province, China
| | - Yuhui Zheng
- Traditional Chinese Medicine Department, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing City 312000, Zhejiang Province, China
| | - Shengyang Zhang
- Traditional Chinese Medicine Department, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing City 312000, Zhejiang Province, China
| | - Shuren Guo
- Clinical College, Zhejiang Chinese Medical University, Hangzhou City 310000, Zhejiang Province, China
| | - Songfeng Hu
- Traditional Chinese Medicine Department, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing City 312000, Zhejiang Province, China.
| |
Collapse
|
18
|
Attia KAM, Abdel-Monem AH, Abdel-Raoof AM, Eissa AS. Development and validation of a highly sensitive HPLC method for quantifying cardiovascular drugs in human plasma using dual detection. Sci Rep 2025; 15:12229. [PMID: 40210736 PMCID: PMC11986036 DOI: 10.1038/s41598-025-94907-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
Cardiovascular diseases are the major cause of global mortality, and often require the concomitant use of a number of drugs to prevent and reduce these deaths. The challenge is to find effective and accurate methods for analyzing these drugs in plasma. This research introduces an innovative, sustainable HPLC-FLD method for the concurrent determination of bisoprolol (BIS), amlodipine besylate (AML), telmisartan (TEL), and atorvastatin (ATV) within human plasma. Chromatographic separation was achieved using an isocratic elution mode on a Thermo Hypersil BDS C18 column (150 × 4.6 mm, 5.0 μm), while the mobile phase comprised of ethanol and 0.03 M potassium phosphate buffer (pH 5.2) in a 40:60 ratio, with a flow rate of 0.6 mL/min. The eluate was analyzed using UV detection within the wavelength range of 210-260 nm to confirm effective separation of the four cardiovascular drugs. For enhanced specificity, a fluorescence detector was set to 227ex/298em for BIS, 294ex/365em for TEL, 274ex/378em for ATV, and 361ex/442em for amlodipine. The method was validated following the International Council for Harmonisation (ICH) guidelines. Linearity was established within the ranges of 5-100 ng/mL for BIS and AML, 0.1-5 ng/mL for TEL, and 10-200 ng/mL for ATV, ensuring accuracy and precision. The significant of the current work represented in introduction of a highly sensitive, and selective analytical method, utilizing an economical sample preparation strategy, for the simultaneous determination of four different cardiovascular drugs (bisoprolol, amlodipine, telmisartan, and atorvastatin) in spiked human plasma. The extraction of sample was carried by liquid-liquid extraction (LLE) and analyzed by LC-fluorescence detector. The chromatographic run was short (less than10 min) which is a greet economical value.
Collapse
Affiliation(s)
- Khalid A M Attia
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, 11751, Egypt
| | - Ahmed H Abdel-Monem
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, 11751, Egypt
| | - Ahmed M Abdel-Raoof
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, 11751, Egypt
| | - Amr S Eissa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt.
| |
Collapse
|
19
|
Al-Kuraishy HM, Sulaiman GM, Mohsin MH, Mohammed HA, Dawood RA, Albuhadily AK, Al-Gareeb AI, Albukhaty S, Abomughaid MM. Targeting of AMPK/MTOR signaling in the management of atherosclerosis: Outmost leveraging. Int J Biol Macromol 2025; 309:142933. [PMID: 40203916 DOI: 10.1016/j.ijbiomac.2025.142933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Atherosclerosis (AS) is a chronic vascular disorder that is characterized by the thickening and narrowing of arteries due to the development of atherosclerotic plaques. The traditional risk factors involved in AS are obesity, type 2 diabetes (T2D), dyslipidemia, hypertension, and smoking. Furthermore, non-traditional risk factors for AS, such as inflammation, sleep disturbances, physical inactivity, air pollution, and alterations of gut microbiota, gained attention in relation to the pathogenesis of AS. Interestingly, the pathogenesis of AS, is complex and related to different abnormalities of cellular and sub-cellular signaling pathways. It has been illustrated that AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (MTOR) pathways are involved in AS pathogenesis. Mounting evidence indicated that AMPK plays a critical role in attenuating the development of AS by activating autophagy, which is impaired during atherogenesis. AMPK has a vasculoprotective effect by reducing lipid accumulation, inflammatory cell proliferation, and the release of pro-inflammatory cytokines, as well as decreasing inflammatory cell adhesion to the vascular endothelium. AMPK activation by metformin inhibits the migration of vascular smooth muscle cells (VSMCs) and AS development. However, the MTOR pathway contributes to AS by inhibiting autophagy, highlighting autophagy as a crucial link between the AMPK and MTOR pathways in AS pathogenesis. The MTOR is a key inducer of endothelial dysfunction and is involved in the development of AS. Therefore, both the AMPK and MTOR pathways play a crucial role in the pathogenesis of AS. However, the exact role of AMPK and MTOR pathways in the pathogenesis of AS is not fully clarified. Therefore, this review aims to discuss the potential role of the AMPK/MTOR signaling pathway in AS, and how AMPK activators and MTOR inhibitors influence the development and progression of AS. In conclusion, AMPK activators and MTOR inhibitors have vasculoprotective effects against the development and progression of AS.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ghassan M Sulaiman
- Department of Applied Sciences, University of Technology, Baghdad, Iraq.
| | - Mayyadah H Mohsin
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Retaj A Dawood
- Department of Biology, College of Science, Al-Mustaqbal University, Hilla 51001, Iraq
| | - Ali K Albuhadily
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir ibn Hayyan Medical University, Al-Ameer Qu, PO.Box13 Kufa, Najaf, Iraq
| | | | - Mosleh M Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 255, Bisha 67714, Saudi Arabia
| |
Collapse
|
20
|
Huo Z, Wu S, Li L, Zhang J, Liu Y, Han X, Chen S, Wang G, Li Y, Zhang S, Borné Y, Geng T, Huang Z. Long-Term Systolic Blood Pressure Time in Target Range and Cardiovascular Disease in Individuals With Hypertension. JACC. ADVANCES 2025:101702. [PMID: 40243985 DOI: 10.1016/j.jacadv.2025.101702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Previous post hoc analyses of clinical trials have suggested that a higher percentage of systolic blood pressure time in target range (SBP-TTR) was associated with a decreased risk of cardiovascular disease (CVD). However, there is limited evidence from large prospective cohort studies on the relationship between long-term SBP-TTR and the risk of CVD. OBJECTIVES The purpose of this study was to assess the association between long-term SBP-TTR and the risk of CVD in adults with hypertension. METHODS This study included 26,929 hypertensive patients (mean age 52.8 ± 10.7 years; 83.1% males), free of CVD at baseline from the Kailuan Study. Blood pressure was measured biennially from 2006 to 2012, and the SBP target range was defined as 120 to 140 mm Hg. SBP-TTR was calculated by the Rosendaal linear interpolation method. Incident CVD events were ascertained via the linkage of electronic health record data. RESULTS During a median follow-up of 8.6 years, 2,565 CVD, including 472 myocardial infarction and 2,151 stroke cases were documented. Comparing the high SBP-TTR (75%-100%) with the low SBP-TTR (0%-25%) group, the multivariable HRs were 0.67 (95% CI: 0.59-0.76) for CVD, 0.76 (95% CI: 0.56-1.02) for myocardial infarction, and 0.64 (95% CI: 0.56-0.74) for stroke. In addition, the association between SBP-TTR and CVD risk was stronger among individuals aged <65 years than their older counterparts (P for interaction <0.001). CONCLUSIONS A greater percentage of SBP-TTR was associated with a lower risk of CVD among patients with hypertension. These findings underscore the importance of maintaining SBP within the target range for the prevention of CVD among hypertensive individuals.
Collapse
Affiliation(s)
- Zhenyu Huo
- School of Public Health, North China University of Science and Technology, Tangshan, China; Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Liuxin Li
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Jingdi Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China; Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xu Han
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Guodong Wang
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Yun Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Shunming Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Yan Borné
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Tingting Geng
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhe Huang
- Department of Cardiology, Kailuan General Hospital, Tangshan, China.
| |
Collapse
|
21
|
Zhou Z, Li T, Qin H, Wang X, He S, Fan Z, Ye Q, Du Y. Acacetin as a natural cardiovascular therapeutic: mechanisms and preclinical evidence. Front Pharmacol 2025; 16:1493981. [PMID: 40255574 PMCID: PMC12006078 DOI: 10.3389/fphar.2025.1493981] [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: 09/10/2024] [Accepted: 02/06/2025] [Indexed: 04/22/2025] Open
Abstract
Globally, cardiovascular disease (CVD) has emerged as a leading cause of mortality and morbidity. As the world's population ages, CVD incidence is on the rise, and extensive attention has been drawn to optimizing the therapeutic regimens. Acacetin, a natural flavonoid derived from various plants, has been demonstrated to have a wide spectrum of pharmacological properties, such as antioxidant, anti-inflammatory, anti-bacterial, and anti-tumor activities, as well as protective effects on diverse tissues and organs. Recently, increasing numbers of studies (mostly preclinical) have indicated that acacetin has potential cardiovascular protective effects and might become a novel therapeutic strategy for CVDs. The importance of acacetin in CVD treatment necessitates a systematic and comprehensive review of its protective effects on the cardiovascular system and the underlying mechanisms involved. Here, we first provide an overview of some basic properties of acacetin. Subsequently, the protective effects of acacetin on multiple CVDs, like arrhythmias, cardiac ischemia/reperfusion injury, atherosclerosis, myocardial hypertrophy and fibrosis, drug-induced cardiotoxicity, diabetic cardiomyopathy, hypertension, and cardiac senescence, are discussed in detail. The underlying mechanisms by which acacetin exhibits cardiovascular protection appear to involve suppressing oxidative stress, reducing inflammation, preventing cardiomyocyte apoptosis and endothelial cell injury, as well as regulating mitochondrial autophagy and lipid metabolism. Meanwhile, several critical signaling pathways have also been found to mediate the protection of acacetin against CVDs, including phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin (PI3K/Akt/mTOR), sirtuin 1/AMP-activated protein kinase/peroxisome proliferator-activated receptor-γ coactivator-1α (Sirt1/AMPK/PGC-1α), transforming growth factor-β1/small mothers against decapentaplegic 3 (TGF-β1/Smad3), protein kinase B/endothelial nitric oxide synthase (Akt/eNOS), and others. Finally, we highlight the existing problems associated with acacetin that need to be addressed, such as the requirement for clinical evidence and enhanced bioavailability, as well as its potential as a promising cardiovascular drug candidate.
Collapse
Affiliation(s)
- Zihe Zhou
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological key Laboratory of Sichuan Province, Institute of Cardiovascular Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Helin Qin
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological key Laboratory of Sichuan Province, Institute of Cardiovascular Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Shanshan He
- Department of Basic Medicine, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yanfei Du
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological key Laboratory of Sichuan Province, Institute of Cardiovascular Medicine, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
22
|
Li J, Hou D, Li J, Li R, Sun M. Association between the atherogenic index of plasma and the systemic immuno-inflammatory index using NHANES data from 2005 to 2018. Sci Rep 2025; 15:11245. [PMID: 40175471 PMCID: PMC11965486 DOI: 10.1038/s41598-025-96090-8] [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: 12/23/2024] [Accepted: 03/26/2025] [Indexed: 04/04/2025] Open
Abstract
The atherogenic index of plasma (AIP) is used to evaluate the risk of atherosclerosis, while the systemic immune-inflammation index (SII) measures inflammation. The AIP and SII are indicators used to predict diseases in various areas. This study aims to explore the relationship between AIP and SII. A cross-sectional study design was used to recruit 70,190 participants from the National Health and Nutrition Examination Survey (NHANES) conducted between 2005 and 2018, excluding AIP missing data, SII missing data, participants under 20 years of age, and participants with missing covariates to eventually include 8163 participants. We used weighted multiple linear regression analysis, trend test, smooth curve fitting and threshold effect analysis to examine the relationship between AIP and SII. Among the 8163 participants included in the study, the mean (± SD) age was 48.412 ± 16.842 years. The mean SII (± SD) for all participants was 519.910 ± 316.974. In a model adjusted for all covariates (Model 3), AIP showed a significant positive correlation with SII [β (95% CI) 32.497 (5.425, 59.569), P = 0.021]. The smooth curve fitting results of AIP and SII are an "inverted U-shape" non-linear relationship, and the inflection point is at AIP = 0.82. This positive association between AIP and SII was found only in females and participants under 50. Specifically, for females, the positive correlation between AIP and SII was linear [β (95% CI) 80.791 (44.625, 116.958); P < 0.001]. In participants under 50, the positive correlation between AIP and SII was [β (95% CI) 34.198 (3.087, 65.310); P = 0.034], and there was also an "inverted U-shape" non-linear relationship with an inflection point of AIP = 0.549. For participants aged 20-50 years and males, the smooth curve showed a "down-flat-down" non-linear relationship. There is a significant positive correlation between AIP and SII. A positive association between AIP and SII was observed exclusively in females and among participants under 50. Furthermore, AIP and SII demonstrated a nonlinear relationship that resembles an "inverted U-shape". These findings offer new insights into the prevention, treatment, and management of cardiovascular disease. However, further comprehensive cohort studies are necessary to validate the relationship between AIP and SII.
Collapse
Affiliation(s)
- Jiayu Li
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Dan Hou
- PLA Northern Theater Command General Hospital, Shenyang, Liaoning, China.
| | - Jiarong Li
- Shaoguan University, Shaoguan, Guangdong, China
| | - Rongcai Li
- Guangzhou Institute of Technology, Guangzhou, Guangdong, China
| | - Ming Sun
- PLA Northern Theater Command General Hospital, Shenyang, Liaoning, China
| |
Collapse
|
23
|
You LL, Luo XB, Zhou WQ, Zhang RC, Li ZH, Xu JX, Ran J, Xu J. Aerobic exercise modulates aortic chondrogenesis and calcification via 5-methoxytryptophan and P38MAPK in atherosclerotic rats. Exp Gerontol 2025; 202:112722. [PMID: 40024450 DOI: 10.1016/j.exger.2025.112722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND 5-Methoxytryptophan (5-MTP), a new endothelial factor with vasoprotective and anti-inflammatory effects, reduces aortic chondrogenesis and calcification during atherosclerosis. The aim of this study was to investigate the effects of aerobic exercise on aortic chondrogenesis and calcification during atherosclerosis in rats. To investigate the effect of aerobic exercise on the expression of 5-MTP/P38 MAPK signaling pathway. To lay a theoretical foundation for the therapeutic effect of exercise in rat atherosclerosis model. METHODS Establishment of a rat model of atherosclerosis using a high-fat diet combined with intraperitoneal injection of vitamin D3 (VD3). The aerobic exercise group underwent moderate-intensity aerobic exercise on an exercise treadmill for 8 weeks, while the atherosclerosis model group and the control group did not exercise. After exercise, blood and aortic samples were collected from all rats to evaluate the levels of serum triglyceride (TG), cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDLC), aortic chondrogenesis, calcification, 5-MTP level, collagen II, P38MAPK, pp38 MAPK, and IL-6 protein content. RESULTS (1)8 weeks of aerobic exercise significantly reduced aortic chondrogenesis, area of calcification, serum LDL-C, TC levels, atherosclerotic index and serum IL-6 levels in rats (p < 0.01), and lowered TG levels (p < 0.05);(2)8 weeks of aerobic exercise significantly increased aortic 5-MTP levels (p < 0.01) and decreased the content of aortic pp38MAPK, collagen II and IL-6 proteins significantly (p < 0.01). The pp38MAPK/P38MAPK ratio was also decreased (p < 0.01). CONCLUSION 8 weeks of aerobic exercise training improved dyslipidemia and reduced aortic chondrogenesis and calcification formation in AS rats. The mechanism may be related to increasing aortic 5-MTP levels and inhibiting the P38MAPK/ IL-6 signaling pathway.
Collapse
Affiliation(s)
| | - Xiao-Bing Luo
- Sichuan Orthopedic Hospital, Chengdu, Sichuan 610041, China.
| | - Wen-Qi Zhou
- Sichuan Orthopedic Hospital, Chengdu, Sichuan 610041, China.
| | - Rui-Chi Zhang
- Chengdu Sport University, School of Sports Medicine and Health, Chengdu, Sichuan 610041, China
| | - Zhong-Hao Li
- Chengdu Sport University, School of Sports Medicine and Health, Chengdu, Sichuan 610041, China
| | - Jia-Xin Xu
- Chengdu Sport University, School of Sports Medicine and Health, Chengdu, Sichuan 610041, China
| | - Jing Ran
- Sichuan Orthopedic Hospital, Chengdu, Sichuan 610041, China
| | - Jie Xu
- Sichuan Orthopedic Hospital, Chengdu, Sichuan 610041, China
| |
Collapse
|
24
|
Sun L, Leng R, Liu M, Su M, He Q, Zhang Z, Liu Z, Wang Z, Jiang H, Wang L, Guo S, Xu Y, Huo Y, Miller CL, Banach M, Huang Y, Evans PC, Pelisek J, Camici GG, Berk BC, Offermanns S, Ge J, Xu S, Weng J. Endothelial MICU1 protects against vascular inflammation and atherosclerosis by inhibiting mitochondrial calcium uptake. J Clin Invest 2025; 135:e181928. [PMID: 40166941 PMCID: PMC11957702 DOI: 10.1172/jci181928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 01/31/2025] [Indexed: 04/02/2025] Open
Abstract
Mitochondrial dysfunction fuels vascular inflammation and atherosclerosis. Mitochondrial calcium uptake 1 (MICU1) maintains mitochondrial Ca2+ homeostasis. However, the role of MICU1 in vascular inflammation and atherosclerosis remains unknown. Here, we report that endothelial MICU1 prevents vascular inflammation and atherosclerosis by maintaining mitochondrial homeostasis. We observed that vascular inflammation was aggravated in endothelial cell-specific Micu1 knockout mice (Micu1ECKO) and attenuated in endothelial cell-specific Micu1 transgenic mice (Micu1ECTg). Furthermore, hypercholesterolemic Micu1ECKO mice also showed accelerated development of atherosclerosis, while Micu1ECTg mice were protected against atherosclerosis. Mechanistically, MICU1 depletion increased mitochondrial Ca2+ influx, thereby decreasing the expression of the mitochondrial deacetylase sirtuin 3 (SIRT3) and the ensuing deacetylation of superoxide dismutase 2 (SOD2), leading to the burst of mitochondrial reactive oxygen species (mROS). Of clinical relevance, we observed decreased MICU1 expression in the endothelial layer covering human atherosclerotic plaques and in human aortic endothelial cells exposed to serum from patients with coronary artery diseases (CAD). Two-sample Wald ratio Mendelian randomization further revealed that increased expression of MICU1 was associated with decreased risk of CAD and coronary artery bypass grafting (CABG). Our findings support MICU1 as an endogenous endothelial resilience factor that protects against vascular inflammation and atherosclerosis by maintaining mitochondrial Ca2+ homeostasis.
Collapse
Affiliation(s)
- Lu Sun
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ruixue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Monan Liu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Meiming Su
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Qingze He
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhidan Zhang
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhenghong Liu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhihua Wang
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hui Jiang
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Shuai Guo
- School of Basic Medical Sciences, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiming Xu
- School of Basic Medical Sciences, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuqing Huo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Clint L. Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Paul C. Evans
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jaroslav Pelisek
- Department of Vascular Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Bradford C. Berk
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, Anhui, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, Anhui, China
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
25
|
Abolfazli S, Karav S, Johnston TP, Sahebkar A. Regulatory effects of resveratrol on nitric oxide signaling in cardiovascular diseases. Pharmacol Rep 2025; 77:355-374. [PMID: 39832074 DOI: 10.1007/s43440-025-00694-w] [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: 10/22/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Cardiovascular illnesses are multifactorial disorders and represent the primary reasons for death worldwide, according to the World Health Organization. As a signaling molecule, nitric oxide (NO) is extremely permeable across cellular membranes owing to its unique molecular features, like its small molecular size, lipophilicity, and free radical properties. Some of the biological effects of NO are vasodilation, inhibition in the growth of vascular smooth muscle cells, and functional regulation of cardiac cells. Several therapeutic approaches have been tested to increase the production of NO or some downstream NO signaling pathways. The health benefits of red wine are typically attributed to the polyphenolic phytoalexin, resveratrol (3,5,4'-trihydroxy-trans-stilbene), which is found in several plant species. Resveratrol has beneficial cardiovascular properties, some of which are mediated through endothelial nitric oxide synthase production (eNOS). Resveratrol promotes NO generation from eNOS through various methods, including upregulation of eNOS expression, activation in the enzymatic activity of eNOS, and reversal of eNOS uncoupling. Additionally, by reducing of oxidative stress, resveratrol inhibits the formation of superoxide and inactivation NO, increasing NO bioavailability. This review discusses the scientific literature on resveratrol's beneficial impact on NO signaling and how this effect improves the function of vascular endothelium.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
26
|
Xie X, Huang M, Ma S, Xin Q, Wang Y, Hu L, Zhao H, Li P, Liu M, Yuan R, Miao Y, Zhu Y, Cong W. The role of long non-coding RNAs in cardiovascular diseases: A comprehensive review. Noncoding RNA Res 2025; 11:158-187. [PMID: 39896344 PMCID: PMC11783329 DOI: 10.1016/j.ncrna.2024.12.009] [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: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide, posing significant challenges to healthcare systems. Despite advances in medical interventions, the molecular mechanisms underlying CVDs are not yet fully understood. For decades, protein-coding genes have been the focus of CVD research. However, recent advances in genomics have highlighted the importance of long non-coding RNAs (lncRNAs) in cardiovascular health and disease. Changes in lncRNA expression specific to tissues may result from various internal or external factors, leading to tissue damage, organ dysfunction, and disease. In this review, we provide a comprehensive discussion of the regulatory mechanisms underlying lncRNAs and their roles in the pathogenesis and progression of CVDs, such as coronary heart disease, atherosclerosis, heart failure, arrhythmias, cardiomyopathies, and diabetic cardiomyopathy, to explore their potential as therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuena Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meiwen Huang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuying Wang
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lantian Hu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Liu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Weihong Cong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
27
|
Lan Q, Chen J, Yang Y. Chromofungin mitigates free fatty acids-induced endothelial inflammation via inhibition of NOD-like receptor thermal protein domain-associated protein 3 mediated by adenosine 5'-monophosphate-activated protein kinase. Biotechnol Appl Biochem 2025; 72:460-468. [PMID: 39358914 DOI: 10.1002/bab.2676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
Free fatty acids (FFAs) have emerged as significant risk factors for atherosclerosis (AS). Prolonged exposure to FFAs induces vascular endothelial injury, including inflammatory responses and oxidative stress, which are central events in AS. Chromofungin (CHR), a peptide derived from chromogranin A (CGA), has been implicated in various biological functions. However, its physiological roles in endothelial biology and its involvement in the pathological development of AS have not been previously reported. In the present study, we investigated the underlying mechanisms through which CHR exerts its beneficial effects on FFA-challenged human aortic endothelial cells (HAECs). We found that treatment with CHR ameliorated the FFA-induced reduction in cell viability and increase in lactate dehydrogenase (LDH) release. Additionally, CHR mitigated oxidative stress by reducing mitochondrial reactive oxygen species (ROS) levels and increasing superoxide dismutase (SOD) activity. Furthermore, exposure to FFAs increased NADPH oxidase (NOX) 4 expression at both the mRNA and protein levels, which were attenuated by CHR in a dose-dependent manner. Notably, CHR reduced the levels of nucleotide-binding domain and leucine-rich repeat-containing (NLR) family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD (ASC), and cleaved caspase-1 (p10), key components of the NLRP3 inflammasome complex, as well as interleukin 1β (IL-1β) and interleukin-18 (IL-18) expression. Mechanistically, it was demonstrated that FFAs reduced the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), which were rescued by CHR in a dose-dependent manner. Conversely, inhibition of AMPK with its specific inhibitor compound C abolished the protective effects of CHR against FFA-induced activation of the NLRP3 inflammasome in HAECs. Based on these findings, we conclude that CHR may serve as a promising agent for maintaining normal endothelial cell function and treating AS.
Collapse
Affiliation(s)
- Qing Lan
- Department of Cardiology, Deyang People's Hospital, Deyang, Sichuan, China
| | - Jian Chen
- Department of Cardiology, Deyang People's Hospital, Deyang, Sichuan, China
| | - Yongqiang Yang
- Department of Cardiology, Deyang People's Hospital, Deyang, Sichuan, China
| |
Collapse
|
28
|
Ritorto G, Ussia S, Mollace R, Serra M, Tavernese A, Palma E, Muscoli C, Mollace V, Macrì R. The Pivotal Role of Thiamine Supplementation in Counteracting Cardiometabolic Dysfunctions Associated with Thiamine Deficiency. Int J Mol Sci 2025; 26:3090. [PMID: 40243711 PMCID: PMC11988323 DOI: 10.3390/ijms26073090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The isolation, structural elucidation, and synthesis of pure thiamin were achieved in 1936, marking a milestone in vitamin research. As an organic compound soluble in water, thiamin is essential for carbohydrate metabolism in plants and animals, and in its active form-as part of the thiamin pyrophosphate coenzyme-performs these functions. Thereby, thiamin represents an essential vitamin to human health and is involved in several pathways that regulate several pathophysiological mechanisms. Cardiovascular disease is significantly impacted by thiamine imbalance and its supplementation offers substantial improvements to the associated conditions. In this comprehensive review, we aimed to examine the dual role of thiamine deficiency and accumulation, focusing on an analysis of the causes of thiamine deficiency. We detailed the effects of thiamine deficiency on metabolism and on cardiovascular risk and heart failure, explaining the molecular mechanisms involved in metabolic dysfunction, and highlighting the role of B1 vitamin supplementation in diabetes mellitus management and atherosclerosis development and progression. Indeed, B1 supplementation counteracts oxidative stress and inflammation, significantly ameliorating glycemic and lipemic profiles. Additionally, we reported the beneficial effects of thiamine in counteracting cardiotoxicity induced by cancer therapy. Although preclinical data strongly support the benefits of thiamine, clinical trial findings are in contrast and contradictory, hampered by limitations such as small sample sizes and inadequate follow-up. Further research is needed to investigate thiamine's potential benefits, overcoming current study limitations and evaluating its use as a supplemental therapy alongside standard treatments in different high-cardiovascular-risk conditions.
Collapse
Affiliation(s)
- Giovanna Ritorto
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (G.R.); (S.U.); (C.M.); (V.M.); (R.M.)
| | - Sara Ussia
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (G.R.); (S.U.); (C.M.); (V.M.); (R.M.)
| | - Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (G.R.); (S.U.); (C.M.); (V.M.); (R.M.)
- Department of Experimental Medicine, University “Tor Vergata” of Rome, 00133 Rome, Italy
| | - Maria Serra
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (G.R.); (S.U.); (C.M.); (V.M.); (R.M.)
| | - Annamaria Tavernese
- Department of Medicine and Surgery, University Campus Bio-Medico of Rome, 00128 Rome, Italy;
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (G.R.); (S.U.); (C.M.); (V.M.); (R.M.)
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (G.R.); (S.U.); (C.M.); (V.M.); (R.M.)
- Renato Dulbecco Institute, Lamezia Terme, 88046 Catanzaro, Italy
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (G.R.); (S.U.); (C.M.); (V.M.); (R.M.)
| |
Collapse
|
29
|
Cai Z, Zhang Y, He L, Cui M, Zhang W, E L, Yang H, Ling Q, Hoffmann PR, He J, Gou S, Liu F, Huang Z. Methylseleninic Acid Elevating the Nrf2-GPX4 Axis Relieves Endothelial Dysfunction and Ferroptosis Induced by Arsenic Exposure. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7445-7455. [PMID: 40071728 DOI: 10.1021/acs.jafc.4c12515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Chronic exposure to arsenic (As), a ubiquitous contaminant, poses deleterious health risks to humans, including cardiovascular disease. Recent studies have implicated ferroptosis, in which the essential micronutrient selenium (Se) plays a crucial role, in several As-induced pathological processes. However, whether Se can counteract As-induced endothelial dysfunction through ferroptosis remains unclear. Herein, methylseleninic acid (MSA), a methylselenium metabolite, was used as a Se supplement to investigate the underlying effect and mechanism of Se in As-induced endothelial dysfunction involving ferroptosis in vivo and in vitro. As exposure induced endothelial dysfunction in mice, as indicated by increased aortic permeability, increased number of circulating endothelial cells, and endothelial mitochondria exhibiting ferroptosis-related alterations. Additionally, As induced ferroptosis-related cell death in mouse aortic endothelial cells, accompanied by impaired redox homeostasis, relatively low Se status, and decreased expressions of selenoproteome, including GPX4. Notably, these were attenuated by MSA via activation of Nrf2 and upregulation of three GPX4 isoforms, which were further abrogated by the Nrf2 antagonist ML385. Finally, MSA exhibited ameliorative effects on endothelial ferroptosis and dysfunction in the aortas of As-exposed mice. These results demonstrate that As causes endothelial ferroptosis and dysfunction by affecting the Se-Nrf2/GPX4 axis, which can be relieved by MSA. This study provides novel evidence implicating Se in As-induced endothelial dysfunction by mitigating ferroptosis.
Collapse
Affiliation(s)
- Zhihui Cai
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Yutian Zhang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Leting He
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Miao Cui
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Weijie Zhang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Lingling E
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, First Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Hui Yang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Qinjie Ling
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, United States
| | - Jingjun He
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Shan Gou
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Fei Liu
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Zhi Huang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong 510632, China
| |
Collapse
|
30
|
Yang L, Li X, Ni L, Lin Y. Treatment of endothelial cell dysfunction in atherosclerosis: a new perspective integrating traditional and modern approaches. Front Physiol 2025; 16:1555118. [PMID: 40206381 PMCID: PMC11979162 DOI: 10.3389/fphys.2025.1555118] [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: 01/03/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Atherosclerosis (AS), a prime causative factor in cardiovascular disease, originates from endothelial cell dysfunction (ECD). Comprising a vital part of the vascular endothelium, endothelial cells play a crucial role in maintaining vascular homeostasis, optimizing redox balance, and regulating inflammatory responses. More evidence shows that ECD not only serves as an early harbinger of AS but also exhibits a strong association with disease progression. In recent years, the treatment strategies for ECD have been continuously evolving, encompassing interventions ranging from lifestyle modifications to traditional pharmacotherapy aimed at reducing risk factors, which also have demonstrated the ability to improve endothelial cell function. Additionally, novel strategies such as promising biotherapy and gene therapy have drawn attention. These methods have demonstrated enormous potential and promising prospects in improving endothelial function and reversing AS. However, it is essential to remain cognizant that the current treatments still present significant challenges regarding therapeutic efficacy, long-term safety, and ethical issues. This article aims to provide a systematic review of these treatment methods, analyze the mechanisms and efficacy of various therapeutic strategies, with the goal of offering insights and guidance for clinical practice, and further advancing the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
31
|
Wei Y, Chen Z, Zhao Q, Wang K, Zhu X, Zhou G, Yu Z. Apelin-13 Inhibits the Adhesion of Monocytes to Endothelial Cells via the Gfi1/NF-κB Signaling Pathway. Biotechnol Appl Biochem 2025:e2746. [PMID: 40123119 DOI: 10.1002/bab.2746] [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: 11/20/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025]
Abstract
Oxidized low-density lipoprotein (ox-LDL)-induced endothelial dysregulation and the abnormal interaction between monocytes have been considered key risk factors for atherosclerosis. The study investigated the effects of apelin-13 on ox-LDL-induced endothelial dysfunction in human aortic endothelial cells (HAECs). Cells were stimulated with ox-LDL (100 mg/L), either alone or in combination with apelin-13 at concentrations of 3 and 6 µM. Multiple techniques, including real-time PCR, Western blot analysis, enzyme-linked immunosorbent assay (ELISA), cell attachment assays, and luciferase activity assays, were employed. Our results showed that ox-LDL reduced the expression of the G-protein-coupled apelin receptor (APJ) in HAECs. However, treatment with apelin-13 reduced the expression of lectin-like ox-LDL receptor 1 (LOX-1) against ox-LDL and inhibited the expression of pro-inflammatory cytokines in HAECs. Real-time PCR and ELISA assay demonstrated that apelin-13 also inhibited the expression of cell adhesion molecules intercellular cell adhesion molecule-1 (ICAM-1) and E-selectin. The calcein AM staining method displayed that apelin-13 mitigated ox-LDL-induced attachment of THP-1 monocytes to HAECs. Furthermore, apelin-13 prevented the reduction of growth factor independence-1 (Gfi1) and the activation of NF-κB in HAECs, as evidenced by the luciferase activity assay. Knockdown of Gfi1 counteracted the inhibitory effects of apelin-13 on the attachment of THP-1 monocytes to HAECs, suggesting that the protective effects of apelin-13 in endothelial dysfunction are mediated by the Gfi1/ NF-κB signaling pathway. These findings suggest that apelin-13 may have therapeutic potential in preventing atherosclerosis by improving endothelial function and reducing inflammation.
Collapse
Affiliation(s)
- Yujuan Wei
- Department of Medicine, Central South University, Xiangya School of Medicine, Changsha, Hunan, China
| | - Zhihong Chen
- Department of Cardiology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, Hunan, China
| | - Qi Zhao
- Department of Cardiology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, Hunan, China
| | - Kaili Wang
- Department of Cardiology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, Hunan, China
| | - Xiaojing Zhu
- Department of Cardiology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, Hunan, China
| | - Guobing Zhou
- Department of Cardiology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, Hunan, China
| | - Zhengke Yu
- Department of Cardiology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, Hunan, China
| |
Collapse
|
32
|
Hanford KML, Dzobo KE, Versloot M, Peter J, Kroon J. Protocol to generate a 3D atherogenesis-on-chip model for studying endothelial-macrophage crosstalk in atherogenesis. STAR Protoc 2025; 6:103559. [PMID: 39799579 PMCID: PMC11773240 DOI: 10.1016/j.xpro.2024.103559] [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: 09/13/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/15/2025] Open
Abstract
The endothelium is the gatekeeper of vessel health, and its dysfunction is pivotal in driving atherogenesis. Here, we present a protocol to replicate endothelial-macrophage crosstalk during atherogenesis, called the "atherogenesis-on-chip" model, based on the Emulate dual-channel perfusion system. We describe a model for studying endothelial-macrophage interactions during atherogenesis in human aortic endothelial cells and human macrophages using qPCR and secretome analysis, fluorescence microscopy, and flow cytometry. This protocol could be adapted toward more complex plaque microenvironment or other disease settings.
Collapse
Affiliation(s)
- Katie M L Hanford
- Department of Experimental Vascular Medicine, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands
| | - Kim E Dzobo
- Department of Experimental Vascular Medicine, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands
| | - Miranda Versloot
- Department of Experimental Vascular Medicine, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jorge Peter
- Department of Experimental Vascular Medicine, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, VIB, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), 3000 Leuven, Belgium.
| |
Collapse
|
33
|
Jiang R, Wang T, Han K, Peng P, Zhang G, Wang H, Zhao L, Liang H, Lv X, Du Y. Impact of anti-inflammatory diets on cardiovascular disease risk factors: a systematic review and meta-analysis. Front Nutr 2025; 12:1549831. [PMID: 40181944 PMCID: PMC11965126 DOI: 10.3389/fnut.2025.1549831] [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: 12/22/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Chronic inflammation, via multiple pathways, influences blood pressure and lipid profiles, serving as a significant risk factor for the onset of cardiovascular disease (CVD). Anti-inflammatory dietary patterns may ameliorate CVD risk factors through the modulation of inflammatory mediators and metabolic factors, potentially leading to improved cardiovascular outcomes. Current findings regarding the relationship between dietary habits and CVD risk factors, such as blood pressure and lipid levels, exhibit considerable variability. We performed a comprehensive systematic review and meta-analysis to explore the possible association between anti-inflammatory dietary patterns (such as the Mediterranean diet, DASH diet, Nordic diet, Ketogenic diet, and Vegetarian diet) and CVD risk factors. Methods We conducted a comprehensive search across five databases: PubMed, Web of Science, Cochrane Library, Embase, and China National Knowledge Infrastructure (CNKI). Ultimately, we identified 18 eligible randomized controlled trials (including randomized crossover trials), which were subjected to meta-analysis utilizing RevMan 5 and Stata 18. Results A comprehensive meta-analysis of these studies conducted based on random effects model indicated that, in comparison to an Omnivorous diet, interventions centered on anti-inflammatory diets were linked to significant reductions in Systolic Blood Pressure (SBP) (MD: -3.99, 95% CI: -6.01 to -1.97; p = 0.0001), Diastolic Blood Pressure (DBP) (MD: -1.81, 95% CI: -2.73 to -0.88; p = 0.0001), Low Density Lipoprotein Cholesterol (LDL-C) (SMD: -0.23, 95% CI: -0.39 to -0.07; p = 0.004), Total Cholesterol (TC) (SMD: -0.31, 95% CI: -0.43 to -0.18; p < 0.00001) and High-sensitivity C-reactive Protein (hs-CRP) (SMD: -0.16, 95% CI: -0.31 to -0.00; p = 0.04). No notable correlations were identified between High Density Lipoprotein Cholesterol (HDL-C) and Triglycerides (TG). Discussion The findings indicate that anti-inflammatory diets may lower serum hs-CRP levels and positively influence the reduction of CVD risk factors, such as blood pressure and lipid profiles, thereby contributing to the prevention and progression of cardiovascular conditions. Most of the outcome indicators had low heterogeneity; sensitivity analyses were subsequently conducted on outcome measures demonstrating substantial heterogeneity, revealing that the findings remained consistent.
Collapse
Affiliation(s)
- Ruixue Jiang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| | - Ting Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| | - Kunlin Han
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| | - Peiqiang Peng
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| | - Gaoning Zhang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| | - Hanyu Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| | - Hang Liang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| | - Xuejiao Lv
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yanwei Du
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
34
|
Al-Kuraishy HM, Sulaiman GM, Mohammed HA, Dawood RA, Albuhadily AK, Al-Gareeb AI, Klionsky DJ, Abomughaid MM. Insight into the Mechanistic role of Colchicine in Atherosclerosis. Curr Atheroscler Rep 2025; 27:40. [PMID: 40111634 DOI: 10.1007/s11883-025-01291-1] [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] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE OF REVIEW Globally, the prevalence of atherosclerosis (AS) is rising. Currently, there is no specific drug for AS. Therefore, this review aims to discuss the protective mechanisms of colchicine against the development and progression of atherosclerosis (AS). RECENT FINDINGS Many studies highlighted that the anti-inflammatory drug colchicine reduces the severity of AS, although the underlying mechanism for the beneficial effect of colchicine was not fully clarified. AS is a chronic progressive vascular disorder characterized by the formation of atherosclerotic plaques. Endothelial dysfunction is an initial stage in the pathogenesis of AS that is induced by oxidized low-density lipoprotein (oxLDL). Engulfment of oxLDL by macrophages triggers the development of inflammation due to the release of pro-inflammatory cytokines and growth factors. Inflammatory and adhesion molecules are involved in the pathogenesis of AS. Infiltration and accumulation of leukocytes provoke erosion, rupture, and thrombosis of the atherosclerotic plaque. Therefore, targeting inflammation and leukocyte infiltration by anti-inflammatory agents may reduce AS progression and complications. The anti-inflammatory drug colchicine reduces the severity of AS, although the underlying mechanism for the beneficial effect of colchicine was not fully elucidated. IN CONCLUSION colchicine through inhibition of vascular inflammation, oxidative stress, platelet aggregation and the modulation of autophagy reduces the development and progression of AS.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ghassan M Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq.
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, 51452, Qassim, Saudi Arabia
| | - Retaj A Dawood
- Department of Biology, College of Science, Al-Mustaqbal University, Hilla, 51001, Iraq
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu, PO.Box13 Kufa, Najaf, Iraq
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Mosleh M Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 255, 67714, Bisha, Saudi Arabia
| |
Collapse
|
35
|
Radić M, Belančić A, Đogaš H, Vučković M, Sener YZ, Sener S, Fajkić A, Radić J. Cardiometabolic Risk in Psoriatic Arthritis: A Hidden Burden of Inflammation and Metabolic Dysregulation. Metabolites 2025; 15:206. [PMID: 40137170 PMCID: PMC11943837 DOI: 10.3390/metabo15030206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease that extends beyond musculoskeletal and dermatologic involvement to elevate cardiometabolic risk. Emerging evidence highlights the critical role of systemic inflammation in metabolic dysregulation, accelerating insulin resistance, dyslipidemia, and oxidative stress, all of which contribute to the increased burden of cardiovascular disease in PsA. This review explores the intricate interplay between inflammatory mediators-such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-17 (IL-17),-adipokine imbalances, and lipid metabolism abnormalities, all of which foster endothelial dysfunction and atherosclerosis. The dysregulation of adipokines, including leptin, adiponectin, and resistin, further perpetuates inflammatory cascades, exacerbating cardiovascular risk. Additionally, the metabolic alterations seen in PsA, particularly insulin resistance and lipid dysfunction, not only contribute to cardiovascular comorbidities but also impact disease severity and therapeutic response. Understanding these mechanistic links is imperative for refining risk stratification strategies and tailoring interventions. By integrating targeted immunomodulatory therapies with metabolic and cardiovascular risk management, a more comprehensive approach to PsA treatment can be achieved. Future research must focus on elucidating shared inflammatory and metabolic pathways, enabling the development of innovative therapeutic strategies to mitigate both systemic inflammation and cardiometabolic complications in PsA.
Collapse
Affiliation(s)
- Mislav Radić
- Department of Internal Medicine, Division of Rheumatology, Allergology and Clinical Immunology, Center of Excellence for Systemic Sclerosis in Croatia, University Hospital of Split, 21000 Split, Croatia;
- Internal Medicine Department, School of Medicine, University of Split, 21000 Split, Croatia
| | - Andrej Belančić
- Department of Basic and Clinical Pharmacology with Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Hana Đogaš
- Department of Neurology, University Hospital of Split, 21000 Split, Croatia;
| | - Marijana Vučković
- Department of Internal Medicine, Division of Nephrology and Dialysis, University Hospital of Split, 21000 Split, Croatia;
| | - Yusuf Ziya Sener
- Department of Pediatric Rheumatology, Sophia Children’s Hospital, Erasmus University Medical Center, 3000 CB Rotterdam, The Netherlands;
| | - Seher Sener
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, 3000 CB Rotterdam, The Netherlands;
| | - Almir Fajkić
- Department of Pathophysiology, Faculty of Medicine, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Josipa Radić
- Internal Medicine Department, School of Medicine, University of Split, 21000 Split, Croatia
- Department of Internal Medicine, Division of Nephrology and Dialysis, University Hospital of Split, 21000 Split, Croatia;
| |
Collapse
|
36
|
Fakir S, Sarker MMR, Sigdel M, Barabutis N. NEK2 inhibition alleviates lipopolysaccharide-induced endothelial injury. ASPET DISCOVERY 2025; 1:100002. [PMID: 40259950 PMCID: PMC12009629 DOI: 10.1016/j.aspetd.2025.100002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
The endothelial barrier regulates substance transfer across an extensive surface area, and vascular leakage may contribute to various inflammatory conditions, including acute respiratory distress syndrome and sepsis. NEK2 possesses a significant role in regulating cellular processes, and its overexpression has been linked to human disease. The present study investigates the effects of NEK2 inhibitor NCL 00017509 in endothelial barrier dysfunction and inflammation. Our results indicate that the aforementioned compound effectively suppresses lipopolysaccharide-induced activation of Cofilin and MLC2, which are crucial cytoskeletal components. NEK2 inhibition reduced endothelial paracellular permeability, reactive oxygen species generation, and phosphorylation of key inflammatory proteins (eg, ERK1/2, P38, STAT1, and STAT3) in cells exposed to lipopolysaccharide. Further investigation into the application of NEK2 inhibitors in preclinical models of direct and indirect lung injury will substantiate our findings.
Collapse
Affiliation(s)
- Saikat Fakir
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Md Matiur Rahman Sarker
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Madan Sigdel
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| |
Collapse
|
37
|
Ouyang J, Wu D, Gan Y, Tang Y, Wang H, Huang J. Unraveling the metabolic‒epigenetic nexus: a new frontier in cardiovascular disease treatment. Cell Death Dis 2025; 16:183. [PMID: 40102393 PMCID: PMC11920384 DOI: 10.1038/s41419-025-07525-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/16/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
Cardiovascular diseases are the leading causes of death worldwide. However, there are still shortcomings in the currently employed treatment methods for these diseases. Therefore, exploring the molecular mechanisms underlying cardiovascular diseases is an important avenue for developing new treatment strategies. Previous studies have confirmed that metabolic and epigenetic alterations are often involved in cardiovascular diseases across patients. Moreover, metabolic and epigenetic factors interact with each other and affect the progression of cardiovascular diseases in a coordinated manner. Lactylation is a novel posttranslational modification (PTM) that links metabolism with epigenetics and affects disease progression. Therefore, analyzing the crosstalk between cellular metabolic and epigenetic factors in cardiovascular diseases is expected to provide insights for the development of new treatment strategies. The purpose of this review is to describe the relationship between metabolic and epigenetic factors in heart development and cardiovascular diseases such as heart failure, myocardial infarction, and atherosclerosis, with a focus on acylation and methylation, and to propose potential therapeutic measures.
Collapse
Affiliation(s)
- Jun Ouyang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Deping Wu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yumei Gan
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yuming Tang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, China
| | - Hui Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, China.
| | - Jiangnan Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
38
|
Gu X, Du L, Lin R, Ding Z, Guo Z, Wei J, Li Y. How Advanced Is Nanomedicine for Atherosclerosis? Int J Nanomedicine 2025; 20:3445-3470. [PMID: 40125442 PMCID: PMC11928726 DOI: 10.2147/ijn.s508757] [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: 11/26/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in nanotechnology have opened new avenues for precision therapy, personalized medicine, and multifunctional theranostics in atherosclerosis (AS). This review provides a comprehensive overview of the role of nanoparticles (NPs) in precision medicine for AS, discussing their applications, challenges, and future prospects. The review first analyzes the current treatment landscape of AS and outlines potential biological targets for therapy. Various nanocarriers, including organic, inorganic, and hybrid systems, are evaluated for their therapeutic potential, with a focus on targeted drug delivery, anti-inflammatory therapy, vascular repair, plaque stabilization, and lipid clearance. Additionally, the review explores NP preparation methods, emphasizing strategies to enhance drug loading, stability, and controlled release. Finally, the translational challenges of NP-based therapies, including biocompatibility, large-scale production, regulatory hurdles, and clinical implementation, are critically analyzed. Future directions highlight the importance of interdisciplinary collaboration and technological innovation in advancing nanoparticle-based precision medicine for AS.
Collapse
Affiliation(s)
- Xiang Gu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lixin Du
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ruifang Lin
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zehui Ding
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhihua Guo
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiaming Wei
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
39
|
Li L, Lai L, Qiu D, Ding Y, Yu M, Zhang T, Wang Z, Wang S. P2Y 6 receptor: A promising therapeutic target for atherosclerosis. Eur J Pharmacol 2025; 998:177513. [PMID: 40097133 DOI: 10.1016/j.ejphar.2025.177513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Atherosclerosis is induced by lipid accumulation, inflammation, and endothelial dysfunction, and is the leading cause of death from cardiovascular disease worldwide. The P2Y6 receptor can be activated by the extracellular release of UDP. The evidence from the last decade has highlighted its critical therapeutic effect in atherosclerosis, yet with unclear mechanisms. This review introduced the P2Y6 receptor in atherosclerosis, and its mechanisms of atherosclerosis-promoting in macrophages, endothelial cells, and vascular smooth muscle cells. Finally, we discussed the development and potential of P2Y6 receptor antagonists in treating atherosclerosis.
Collapse
Affiliation(s)
- Lixia Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Liting Lai
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Dan Qiu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yang Ding
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Meiling Yu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Tingyu Zhang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zongbao Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Shuzhi Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
40
|
Parveen K, Hussain MA, Anwar S, Elagib HM, Kausar MA. Comprehensive review on diabetic foot ulcers and neuropathy: Treatment, prevention and management. World J Diabetes 2025; 16:100329. [PMID: 40093290 PMCID: PMC11885961 DOI: 10.4239/wjd.v16.i3.100329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/27/2024] [Accepted: 12/27/2024] [Indexed: 01/21/2025] Open
Abstract
Diabetic foot (DF) is a major public health concern. As evident from numerous previous studies, supervision of DF ulcer (DFU) is crucial, and a specific quality check-up is needed. Patients should be educated about glycaemic management, DFUs, foot lesions, proper care for injuries, diet, and surgery. Certain reasonably priced treatments, such as hyperbaric oxygen and vacuum-assisted closure therapy, are also available for DFUs, along with modern wound care products and techniques. Nonetheless, DF care (cleaning, applying antimicrobial cream when wounded, and foot reflexology), blood glucose monitoring to control diabetes, and monthly or quarterly examinations in individuals with diabetes are effective in managing DFUs. Between 50% and 80% of DF infections are preventable. Regardless of the intensity of the lesion, it needs to be treated carefully and checked daily during infection. Tissue regeneration can be aided by cleaning, dressing, and application of topical medicines. The choice of shoes is also important because it affects blood circulation and nerve impulses. In general, regular check-ups, monitoring of the patient's condition, measuring blood glucose levels, and providing frequent guidance regarding DFU care are crucial. Finally, this important clinical problem requires involvement of multiple professionals to properly manage it.
Collapse
Affiliation(s)
- Kehkashan Parveen
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Malik Asif Hussain
- Department of Pathology, College of Medicine, University of Ha’il, Ha'il 53962, Saudi Arabia
| | - Sadaf Anwar
- Department of Biochemistry, College of Medicine, University of Ha’il, Ha'il 53962, Saudi Arabia
| | | | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Ha’il, Ha'il 53962, Saudi Arabia
| |
Collapse
|
41
|
He L, Gong F, Wang J, Huang Y, Wang H. mpECs with high Piezo2 expression promote fracture healing by driving angiogenesis through the Notch signaling pathway. BMC Musculoskelet Disord 2025; 26:238. [PMID: 40069682 PMCID: PMC11895173 DOI: 10.1186/s12891-025-08476-4] [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: 09/10/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
Fractures will impair or disrupt angiogenesis, resulting in delayed union or non-union. Exploring angiogenic signaling molecules and related pathways can promote fracture healing. In this study, the roles of different endothelial cell (EC) subsets in fracture healing were observed using single-cell RNA sequencing (scRNA-seq). It was found that mpECs did affect the repair and regeneration of fracture sites, and could up-regulate genes related to the Notch signaling, angiogenesis, and cell cycle. In addition, in this study, Piezo2 expression was successfully knocked down by transfection of shRNA in human umbilical vein endothelial cells (HUVECs) for in vitro assays. The results suggested that the reduced expression of Piezo2 in HUVECs can suppress cell proliferation and cell cycle and further impair the activation of the Notch signaling pathway, inhibiting angiogenesis. Subsequently, HUVECs were intervened with the Notch pathway inhibitor DAPT and agonist Jagged1. It was found that inhibition of the Notch signaling pathway by Piezo2 knockdown was more significant in the presence of DAPT, whereas Jagged1 reversed the Piezo2 knockdown-caused changes in the downstream protein expression of the Notch pathway. With Jagged1, Piezo2 knockdown-induced decrease in HUVEC tube formation disappeared. Moreover, the tube formation was significantly enhanced, with a marked increase in tube length. Cell counting kit-8 (CCK-8) assay and flow cytometry demonstrated that Jagged1 can promote cell proliferation and trigger cell cycle entry. In conclusion, Piezo2 affects the phenotype of ECs by modulating the Notch signaling pathway and further promotes angiogenesis, thus accelerating fracture healing.
Collapse
Affiliation(s)
- Lingfeng He
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo No. 6 Hospital, Ningbo, China
| | - Fanyong Gong
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jiangyong Wang
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yi Huang
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Haifeng Wang
- Department of Neurosurgery, Ningbo Key Laboratory of Neurological Diseases and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, China.
- The First Affiliated Hospital of Ningbo University, Haishu District, Liuting RoadZhejiang Province, Ningbo, People's Republic of China.
| |
Collapse
|
42
|
Du J, Liu J, Wang X, Wang X, Ma Y, Zhang S, Li Z, Ma J, Liu J. The role of estrogen in the sex difference for the risk factors of heart failure with preserved ejection fraction. Biol Direct 2025; 20:28. [PMID: 40065410 PMCID: PMC11895175 DOI: 10.1186/s13062-025-00618-x] [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: 12/25/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major subtype of heart failure, primarily characterized by a normal or mildly reduced left ventricular ejection fraction along with left ventricular diastolic dysfunction. Recent studies have shown that the prevalence of HFpEF is higher in women than that in men, particularly in postmenopausal women. Concurrently, it has been observed that the incidence of risk factors contributing to HFpEF (such as obesity, hypertension, diabetes, and atrial fibrillation) also notably increases post-menopause, affecting the incidence of HFpEF. This review aimed to examine the relationship between estrogen and risk factors associated with HFpEF, clarifying the underlying mechanisms through which estrogen affects these risk factors from epidemiological and pathophysiological perspectives. This review also provides a comprehensive understanding of the association between estrogen and the risk factors for HFpEF, thus helping explore potential targets for HFpEF treatment.
Collapse
Affiliation(s)
- Jun Du
- Xi'an Medical University, Xi'an, People's Republic of China
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiaqi Liu
- Xi'an Medical University, Xi'an, People's Republic of China
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xiaoya Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yu Ma
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Sipan Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Zilin Li
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jipeng Ma
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China.
| | - Jincheng Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
43
|
Zhao Y, Lv R, He Y, Dong N, Wang X, Pu J, Yu Q. The miR-21-5p/DUSP8/MAPK signaling pathway mediates inflammation and apoptosis in vascular endothelial cells induced by intermittent hypoxia and contributes to the protective effects of N-acetylcysteine. Eur J Pharmacol 2025; 997:177462. [PMID: 40058751 DOI: 10.1016/j.ejphar.2025.177462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Obstructive sleep apnoea hypopnea syndrome (OSAHS) is a sleep disorder associated with significant cardiovascular complications, characterized by intermittent hypoxia (IH). IH causes endothelial dysfunction, an early event in cardiovascular disease. We investigated the role of dual-specificity phosphatase 8 (DUSP8), a key negative regulator of the mitogen-activated protein kinase (MAPK) signalling pathway, in IH-induced endothelial cell damage, and the therapeutic effects of N-acetylcysteine (NAC) by establishing IH models in human umbilical vein endothelial cells and C57BL/6 mice. DUSP8 and MAPK signalling pathway-related proteins were analysed by western blotting, and DUSP8 mRNA and miR-21-5p expression was assessed by RT-qPCR. Inflammatory cytokines were detected by an enzyme-linked immunosorbent assay, apoptosis-related proteins were analysed by western blotting, and apoptosis was assessed using flow cytometry. IH stimulation induced inflammation and apoptosis in endothelial cells, downregulated DUSP8 expression, and upregulated the phosphorylation of key molecules involved in the MAPK signalling pathway. However, DUSP8 overexpression alleviated IH-induced inflammation and apoptosis in endothelial cells and reduced the phosphorylation of key molecules in the MAPK signalling pathway. Bioinformatic analysis and dual-luciferase reporter assays confirmed that DUSP8 is a direct target of miR-21-5p. DUSP8 overexpression effectively reversed the damage caused by miR-21-5p upregulation under IH conditions. Furthermore, in cell and animal models of IH, NAC demonstrated protective effects against inflammation, apoptosis, and oxidative stress through a mechanism linked to the miR-21-5p/DUSP8/MAPK signalling pathway. Overall, this study elucidated the protective role of DUSP8 against IH-induced endothelial injury and confirmed the potential of NAC as a therapeutic agent for OSAHS-related diseases.
Collapse
Affiliation(s)
- Yan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Jiayuan Pu
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Qin Yu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China; Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
44
|
Zhang X, Sha Y, Wu Y, Guan H, Yang X, Wang W, Zhang W, Liu Y, Zhu L, Li Q. Targeting endothelial cells: A novel strategy for pulmonary fibrosis treatment. Eur J Pharmacol 2025; 997:177472. [PMID: 40054716 DOI: 10.1016/j.ejphar.2025.177472] [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: 09/30/2024] [Revised: 01/10/2025] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
Endothelial cells (ECs) are a monolayer of flat cells lining the inner surfaces of blood and lymphatic vessels. They play a key role in many physiological and pathological processes. Specifically, they maintain vascular permeability and structural stability and participate in immune responses, inflammation, coagulation, and other vital functions. ECs play a decisive role in various age-related diseases; however, their involvement in pulmonary fibrosis (PF) remains poorly understood. PF refers to a group of chronic interstitial lung diseases characterised by progressive scarring of the pulmonary parenchyma, primarily caused by aberrant tissue repair mechanisms. These changes lead to irreversible loss of lung function. Although the exact pathophysiological mechanism underlying PF has not yet been elucidated, recent studies have indicated that ECs may play a pivotal role in PF. This review outlines the involvement of pulmonary vascular ECs in PF, focusing on the regulation of vascular remodelling and endothelial barrier integrity and on the maintenance of angiogenesis through EC-specific markers, such as vascular endothelial growth factor. This review also explores processes such as endothelial-to-mesenchymal transition, immune cell interactions, anti-EC antibody reactions, metabolic dysregulation, and cellular senescence. By elucidating recent advancements in understanding the role of ECs in PF and examining drugs targeting ECs for the treatment of PF, this study provides novel insights into the pathological mechanisms of PF and the development of endothelium-based therapeutic agents.
Collapse
Affiliation(s)
- Xin Zhang
- Medical College, Anhui University of Science and Technology, Huainan, Anhui, 232001, China; Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yuxia Sha
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yu Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Haiyang Guan
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Xu Yang
- Medical College, Anhui University of Science and Technology, Huainan, Anhui, 232001, China
| | - Wenjin Wang
- Medical College, Anhui University of Science and Technology, Huainan, Anhui, 232001, China
| | - Wenlong Zhang
- Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yunyun Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Lili Zhu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Qing Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
45
|
Huang D, Wu H, Huang Y. Novel indicator for erectile dysfunction: the CALLY index, evidence from data of NHANES 2001-2004. Front Endocrinol (Lausanne) 2025; 16:1527506. [PMID: 40099259 PMCID: PMC11911170 DOI: 10.3389/fendo.2025.1527506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose This study explored the association between the C-reactive protein-albumin-lymphocyte (CALLY) index and erectile dysfunction (ED). Patients and methods Data from 2,128 participants in the 2001-2004 National Health and Nutrition Examination Survey (NHANES) were analyzed and classified into ED and non-ED groups.Additionally, a separate analysis of complete erectile dysfunction was conducted.A weighted multiple logistic regression model was used to assess the association between CALLY and ED, while smooth curve fitting was applied to explore their linear relationship.ROC analysis was conducted to compare the predictive accuracy (AUC) of CALLY, Systemic Inflammation Response Index (SIRI), Systemic Immune-Inflammation Index (SII), Aggregate Index of Systemic Inflammation (AISI), Neutrophil-to-Lymphocyte Ratio (NLR), Platelet-to-Lymphocyte Ratio (PLR), and the product of platelet count and neutrophil count (PPN) for ED. Results After adjustment, Ln-CALLY was negatively associated with ED (OR = 0.77, 95% CI: 0.69-0.85, p < 0.0001) and complete ED (OR = 0.88, 95% CI: 0.78-1.00, p = 0.0450).The highest Ln-CALLY tertile (Q3) was associated with a significantly lower risk of ED compared to Q1 (OR = 0.40, 95% CI: 0.30-0.55, p < 0.0001).A similar trend was observed for complete ED (OR = 0.57, 95% CI: 0.38-0.85, p = 0.006).Curve fitting revealed a negative correlation between CALLY and both types of ED.Subgroup analysis confirmed the consistent and independent association.CALLY exhibited superior predictive performance for ED (AUC = 0.6512) and complete ED (AUC = 0.6237) compared to other markers. Conclusion Higher CALLY levels were linked to a reduced ED risk and proved a superior predictor compared to other inflammatory markers.
Collapse
Affiliation(s)
- Dongli Huang
- Bishan Hospital of Chongqing Medical University (Bishan Hospital of Chongqing), Chongqing, China
| | - Hang Wu
- Bishan Hospital of Chongqing Medical University (Bishan Hospital of Chongqing), Chongqing, China
| | - Yanhua Huang
- Department of Infectious Diseases, Chongqing University Three Gorges Hospital, Chongqing, China
| |
Collapse
|
46
|
de Miranda JA, Cunha WR, Lovisi JCM, Moreira Lanna CM, Pinheiro LC, Lacchini R, Tanus-Santos JE, de Almeida Belo V. Oxidative stress and obesity are associated with endothelial dysfunction and subclinical atherosclerosis in adolescents. Clin Biochem 2025; 136:110889. [PMID: 39870127 DOI: 10.1016/j.clinbiochem.2025.110889] [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: 08/28/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 01/29/2025]
Abstract
OBJECTIVES This study explores the relationship between obesity, endothelial dysfunction, and the critical role of oxidative stress biomarkers in subclinical atherosclerosis. DESIGN & METHODS The study included 114 adolescents aged 12-17 years from Juiz de Fora, Brazil, divided into 40 individuals with obesity and 74 controls. Physical and biochemical assessments were conducted, including measurements of Brachial Flow-Mediated Dilation (BFMD), Carotid Intima-Media Thickness (IMT), and oxidative biomarkers such as nitrite, nitrate, and 8-isoprostane. Multiple regression analyses were used to evaluate associations between obesity, oxidative biomarkers, and endothelial function. RESULTS Adolescents with obesity exhibited significantly reduced BFMD at 60 s (5.44 ± 2.31 % vs. 7.82 ± 2.07 % in controls; p < 0.05) and 90 s (5.27 ± 2.64 % vs. 7.93 ± 2.12 % in controls; p < 0.05). IMT was significantly higher in the group with obesity for both the right carotid artery (0.054 ± 0.005 cm vs. 0.047 ± 0.004 cm in controls; p < 0.05) and the left carotid artery (0.053 ± 0.005 cm vs. 0.047 ± 0.004 cm in controls; p < 0.05). Additionally, 8-isoprostane levels were higher in adolescents with obesity (49.75 ± 22.62 pg/mL vs. 42.36 ± 17.35 pg/mL in controls; p < 0.05), indicating increased oxidative stress. Nitrite levels were significantly lower in adolescents with obesity (42.98 ± 10.62 nM vs. 49.94 ± 17.71 nM in controls; p < 0.05). Additionally, nitrate levels were inversely associated with IMT in both the right (p = 0.01) carotid arteries in the multiple linear regression analyses. CONCLUSIONS The study highlights the association between obesity and early vascular changes in adolescents, evidenced by reduced BFMD, increased IMT, and altered oxidative stress biomarkers.
Collapse
Affiliation(s)
| | - Warlley Rosa Cunha
- Center for Molecular Genetics and Genomics, Wayne State University Detroit MI USA
| | | | | | - Lucas Cézar Pinheiro
- Department of Pharmacology, Federal University of Santa Catarina Florianópolis SC Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Brazil
| | - José Eduardo Tanus-Santos
- Department of Pharmacology Ribeirao Preto Medical School, University of Sao Paulo Ribeirao Preto SP Brazil
| | | |
Collapse
|
47
|
Talebi S, Shirani M, Shokri-Mashhadi N, Sadeghi O, Karav S, Bagherniya M, Sahebkar A. The long-term and post-prandial effects of berry consumption on endothelial dysfunction in adults: a systematic review and meta-analysis of randomised controlled trials. Int J Food Sci Nutr 2025; 76:134-164. [PMID: 39828522 DOI: 10.1080/09637486.2025.2450666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/02/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
This systematic review and meta-analysis evaluated the long-term and post-prandial effects of berry consumption on endothelial dysfunction (ED) in adults. To identify relevant randomised controlled trials (RCTs), a systematic search was run and studies that examined the effects of berries consumption on ED markers were included. A total of 45 RCTs were included, focusing on markers such as flow-mediated dilation (FMD), pulse wave velocity (PWV), augmentation index (AIx), reactive hyperaemia index (RHI) and total peripheral resistance (TPR). The meta-analysis revealed that long-term berry consumption significantly increased FMD and decreased PWV. However, no significant effects were found for AIx, RHI or TPR. In post-prandial studies, berry consumption also improved FMD, but had no significant impact on PWV, AIx or RHI. Overall, berries were found to benefit endothelial function, particularly in improving FMD, though the effects on other cardiovascular markers were less consistent. Factors like trial design and berry type influenced outcomes.
Collapse
Affiliation(s)
- Shokoofeh Talebi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Shirani
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Shokri-Mashhadi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center Isfahan University of Medical Sciences, Isfahan, Iran
| | - Omid Sadeghi
- Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Mohammad Bagherniya
- Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
48
|
Ibrahim NNIN, Rasool AHG, Rahman RA, Azlan M, Aziz AA. Pulse wave analysis as a tool to assess endothelial function following lipid lowering intervention in hypercholesterolemia. Microvasc Res 2025; 158:104772. [PMID: 39586372 DOI: 10.1016/j.mvr.2024.104772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Pulse wave analysis (PWA) assesses endothelial dependent vasodilation (EDV) via the change in augmentation index (AIx) and has been used as a tool to assess endothelial function. However, its effectiveness in assessing the response to lipid lowering treatment has not been evaluated. The study aimed to describe and correlate the change in EDV following lipid lowering intervention in patients with hypercholesterolemia. METHODS 48 newly diagnosed patients with hypercholesterolemia underwent 6 months intervention with statin and/or therapeutic lifestyle changes (TLC) in clinical setting. Lipid profile measurement and endothelial function assessment using PWA were performed pre- and post-intervention. RESULTS Significant reductions in low density lipoprotein cholesterol (LDL-C), non-high density lipoprotein cholesterol (non-HDL-C) and total cholesterol (TC) with corresponding significant improvement in EDV (2.94 ± 3.69 % to 7.50 ± 3.79 %, p < 0.001) were observed following intervention. Sub-analyses revealed greater LDL-C reductions and EDV improvements in the statin group compared to TLC. There was a significant inverse correlation between the change in EDV and the change in LDL-C after intervention (r = -0.298, p = 0.040). CONCLUSION Endothelial function assessed by PWA showed a parallel change with lipid profile pattern following lipid lowering intervention. The simple and non-invasive method may provide a potential tool for evaluating endothelial function and treatment outcomes in patients with hypercholesterolemia.
Collapse
Affiliation(s)
- Nik Nor Izah Nik Ibrahim
- Department of Pharmacology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; Hospital Pakar USM, Jalan Raja Perempuan Zainab II, 16150 Kubang Kerian, Kelantan, Malaysia.
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; Hospital Pakar USM, Jalan Raja Perempuan Zainab II, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Razlina Abdul Rahman
- Department of Family Medicine, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; Hospital Pakar USM, Jalan Raja Perempuan Zainab II, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Maryam Azlan
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Aniza Abd Aziz
- Faculty of Medicine, Universiti Sultan Zainal Abidin, 20400 Kuala Terengganu, Terengganu, Malaysia
| |
Collapse
|
49
|
Wei X, He Y, Yu Y, Tang S, Liu R, Guo J, Jiang Q, Zhi X, Wang X, Meng D. The Multifaceted Roles of BACH1 in Disease: Implications for Biological Functions and Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412850. [PMID: 39887888 PMCID: PMC11905017 DOI: 10.1002/advs.202412850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/22/2024] [Indexed: 02/01/2025]
Abstract
BTB domain and CNC homolog 1 (BACH1) belongs to the family of basic leucine zipper proteins and is expressed in most mammalian tissues. It can regulate its own expression and play a role in transcriptionally activating or inhibiting downstream target genes. It has a crucial role in various biological processes, such as oxidative stress, cell cycle, heme homeostasis, and immune regulation. Recent research highlights BACH1's significant regulatory roles in a series of conditions, including stem cell pluripotency maintenance and differentiation, growth, senescence, and apoptosis. BACH1 is closely associated with cardiovascular diseases and contributes to angiogenesis, atherosclerosis, restenosis, pathological cardiac hypertrophy, myocardial infarction, and ischemia/reperfusion (I/R) injury. BACH1 promotes tumor cell proliferation and metastasis by altering tumor metabolism and the epithelial-mesenchymal transition phenotype. Moreover, BACH1 appears to show an adverse role in diseases such as neurodegenerative diseases, gastrointestinal disorders, leukemia, pulmonary fibrosis, and skin diseases. Inhibiting BACH1 may be beneficial for treating these diseases. This review summarizes the role of BACH1 and its regulatory mechanism in different cell types and diseases, proposing that precise targeted intervention of BACH1 may provide new strategies for human disease prevention and treatment.
Collapse
Affiliation(s)
- Xiangxiang Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Yunquan He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Yueyang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Sichong Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Ruiwen Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Jieyu Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Qingjun Jiang
- Department of Vascular & Endovascular Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| |
Collapse
|
50
|
Liu J, Chen Y, Pu H, Chen X, Yang W, Ouyang Z, Pang Q, Fan R. A new mechanism involved in cardiovascular senescence induced by environmentally relevant dose of 16 priority-controlled PAHs. ENVIRONMENT INTERNATIONAL 2025; 197:109326. [PMID: 39970779 DOI: 10.1016/j.envint.2025.109326] [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: 09/30/2024] [Revised: 01/06/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are closely related to the occurrence of cardiovascular diseases, nevertheless the toxicological mechanism remains ambiguous. To verify whether PAHs exposure leads to cardiovascular senescence, 8-week-old male sprague-dawley rats and primary human umbilical vein endothelial cells were exposed to different concentrations of 16 priority-controlled PAHs for 90 d and 48 h respectively. In in vitro study, PAHs exposure promoted aryl hydrocarbon receptor (AhR) activation, and then directly or indirectly inhibited SIRT6 expression leading to telomere dysfunction, which further caused DNA damage and subsequently promoted endothelial cells senescence. But the treatment of CH-223191 (an AhR inhibitor) rescued the aging phenotypes induced by PAHs, suggesting that AhR plays an important role in PAHs-induced endothelial cells senescence. In in vivo study, PAHs exposure raised AhR expression, affected SIRT6-related aging signaling pathway, and induced myocardial and vascular remodeling in rats. Molecular dynamics simulations demonstrated that, in addition to benzo[a]pyrene-7,8-diol-9,10-epoxide (the mediate metabolite of benzo[a]pyrene), typical parent PAHs (phenanthrene, benzo[a]pyrene) can directly bind to known DNA strand binding sites of SIRT6 through hydrophobic force, which was further validated by electrophoretic mobility shift assay. All above indicates for the first time that in addition to classical AhR dependent pathway, parent PAHs may affect DNA damage response and telomere maintenance function of SIRT6, which is a new mechanism of PAHs induced cardiovascular senescence.
Collapse
Affiliation(s)
- Jian Liu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Plant Protection Research Institute, Guangdong Academy of Agricultural Sciences, Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Guangzhou 510640, China
| | - Yuxin Chen
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Hao Pu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiaolin Chen
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Wucheng Yang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Zedong Ouyang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Qihua Pang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Ruifang Fan
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|