1
|
Graham EL, Weir TL, Gentile CL. Exploring the Impact of Intermittent Fasting on Vascular Function and the Immune System: A Narrative Review and Novel Perspective. Arterioscler Thromb Vasc Biol 2025; 45:654-668. [PMID: 40177772 PMCID: PMC12018117 DOI: 10.1161/atvbaha.125.322692] [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] [Indexed: 04/05/2025]
Abstract
Vascular function is a critical determinant of cardiovascular health and all-cause mortality. Recent studies have suggested that intermittent fasting, a popular dietary strategy, elicits beneficial effects on vascular function. These studies also suggest that fasting-mediated improvements in vascular function coincide with reductions in systemic inflammation. However, the mechanisms that connect fasting, the immune system, and vascular function remain largely underexplored. The current review summarizes the effects of different intermittent fasting modalities on vascular health, focusing on endothelial dysfunction and arterial stiffness, 2 critical indices of vascular function. Improvements in vascular function are associated with reduced inflammation and are mechanistically linked to decreased circulating immune cells and their accumulation within the vascular wall and perivascular tissue. Recent data show that fasting redistributes circulating and tissue-resident immune cells to the bone marrow, affecting their inflammatory actions. However, there is no direct evidence relating immune cell redistribution to cardiovascular health. By relating fasting-induced immune cell redistribution to reduced inflammation and improved vascular function, we propose an exciting avenue of further exploration is determining whether fasting-induced immune cell redistribution impacts cardiovascular health.
Collapse
Affiliation(s)
- Elliot L. Graham
- Integrative Cardiovascular Physiology Laboratory, Intestinal Health Laboratory, Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, United States
| | - Tiffany L. Weir
- Integrative Cardiovascular Physiology Laboratory, Intestinal Health Laboratory, Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, United States
| | - Christopher L. Gentile
- Integrative Cardiovascular Physiology Laboratory, Intestinal Health Laboratory, Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
2
|
Ghasempour Dabaghi G, Rabiee Rad M, Mortaheb M, Darouei B, Amani-Beni R, Mazaheri-Tehrani S, Izadan M, Touhidi A. The Neutrophil-to-Lymphocyte Ratio Predicts Cardiovascular Outcomes in Patients With Diabetes: A Systematic Review and Meta-Analysis. Cardiol Rev 2025; 33:202-211. [PMID: 39531515 DOI: 10.1097/crd.0000000000000820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The neutrophil-to-lymphocyte ratio (NLR) has been found as a potential biomarker for acute inflammation and the prognosis of different diseases. Here, we provided a meta-analysis of studies evaluating the association of NLR with cardiovascular outcomes among patients with diabetes. We searched PubMed, Scopus, and Web of Science databases from inception to April 06, 2024, to include papers based on eligible criteria. The outcomes of interest were all-cause mortality, cardiovascular mortality, major adverse cardiovascular events, myocardial infarction, and stroke. The pooled risk ratio (RR) and corresponding 95% confidence intervals (CI) were reported. Meta-analysis was performed using StataMP 14.0. A total of 15 studies involving 407,512 participants were included. Meta-analysis revealed that both categorical and continuous NLRs are linked to increased risk of all-cause mortality (RR = 1.68; 95% CI, 1.49-1.88; P < 0.001 and RR = 1.03; 95% CI, 1.03-1.03; P < 0.001, respectively) and cardiovascular mortality (RR = 2.04; 95% CI, 1.58-2.63; P < 0.001 and RR = 1.25; 95% CI, 1.19-1.32; P < 0.001, respectively) in patients with diabetes. However, NLR was not associated with the risk of major adverse cardiovascular events, myocardial infarction, and stroke in patients with diabetes. Subgroup analysis revealed sample size as the main source of the heterogeneity found between studies. The findings suggest NLR as a prognostic marker for mortality outcomes in patients with diabetes, providing clinicians with a noninvasive and readily available indicator for risk assessment and patient management.
Collapse
Affiliation(s)
- Ghazal Ghasempour Dabaghi
- From the Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrdad Rabiee Rad
- From the Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadreza Mortaheb
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Science, Isfahan, Iran
| | - Bahar Darouei
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Science, Isfahan, Iran
| | - Reza Amani-Beni
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Science, Isfahan, Iran
| | - Sadegh Mazaheri-Tehrani
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Science, Isfahan, Iran
| | - Mahshad Izadan
- School of Nursing and Midwifery, Isfahan University of Medical Science, Isfahan, Iran
| | - Ali Touhidi
- School of Nursing and Midwifery, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
3
|
Ajike RA, Afolabi OA, Alabi BA, Ajayi AF, Oyekunle OS, Lawal SK, Olojede SO, Nku-Ekpang OA, Hezekiah OS, Hammed OS. Sequential administration of febuxostat and vitamin E protects against testicular ischemia/reperfusion injury via inhibition of sperm DNA damage in Wistar rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04095-x. [PMID: 40261353 DOI: 10.1007/s00210-025-04095-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/22/2025] [Indexed: 04/24/2025]
Abstract
The pathway of testicular ischemia-reperfusion injury (TIRI) has been shown to involve reactive oxygen species (ROS) generation in the ischemic phase and later phase of reperfusion. This study was therefore designed to investigate the effect of blockage of ROS in the ischemic and reperfusion phases of TIRI. Thirty male Wistar rats were grouped into five groups (n = 6 rats each): sham, torsion + detorsion (TD), febuxostat (FEB)-administered (TFD) group, vitamin E (V)-administered (TDV) group, and FEB and vitamin E-administered (TFDV) group. Blood samples (for inflammatory and hormonal assay), testicular (for oxidative stress and histopathology), and epididymal (for sperm DNA damage and indices) tissues were collected after 3 days of detorsion. The TFD and TFDV groups showed a significant reduction in XO and MDA (p < 0.001; η2 > 0.7), as well as a concomitant increase in CAT, thiols, and SOD levels when compared with the TD group (p < 0.01, η2 > 0.5). The TFD group significantly reduced all inflammatory markers (p < 0.05; η2 = 0.75). The observed increase (p < 0.05; η2 = 0.92) in LH level, in response to a low level of testosterone in the TD group, was significantly raised in TFD and TFDV groups. The observed decrease (p < 0.001) in inhibin level in the TD group was raised (p < 0.05; η2 = 0.90) in the TDV group only. A significant increase (p < 0.001) in sperm DNA damage in the TD group was significantly reduced (p < 0.05; η2 = 0.88) in all the treatment groups while the reduced sperm viability (p < 0.01) in the TD group was increased (p < 0.05) in the TFDV group only. There was an improvement in the testicular cytoarchitecture in the TFD and TFDV groups. This study showed that sequential administration of febuxostat in the ischemic phase of TT and vitamin E in the later phase of reperfusion protects the testes against TIRI via inhibition of oxidative stress, inflammation, and sperm DNA damage.
Collapse
Affiliation(s)
- Richard Adedamola Ajike
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oladele Ayobami Afolabi
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - Babatunde Adebola Alabi
- Department of Pharmacology & Therapeutics, Bowen University, Iwo, Osun State, Nigeria
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Pharmacy, Kampala International University in Tanzania, Dar Es Salaam, United Republic of Tanzania
| | - Ayodeji Folorunsho Ajayi
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Olubunmi Simeon Oyekunle
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Sodiq Kolawole Lawal
- School of Nursing, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana
| | - Samuel Oluwaseun Olojede
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Okot-Asi Nku-Ekpang
- Department of Physiology, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Oluwaseun Samuel Hezekiah
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Opeyemi Sodiq Hammed
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
4
|
Grego A, Fernandes C, Fonseca I, Dias-Neto M, Costa R, Leite-Moreira A, Oliveira SM, Trindade F, Nogueira-Ferreira R. Endothelial dysfunction in cardiovascular diseases: mechanisms and in vitro models. Mol Cell Biochem 2025:10.1007/s11010-025-05289-w. [PMID: 40259179 DOI: 10.1007/s11010-025-05289-w] [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: 02/09/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Endothelial cells (ECs) are arranged side-by-side to create a semi-permeable monolayer, forming the inner lining of every blood vessel (micro and macrocirculation). Serving as the first barrier for circulating molecules and cells, ECs represent the main regulators of vascular homeostasis being able to respond to environmental changes, either physical or chemical signals, by producing several factors that regulate vascular tone and cellular adhesion. Healthy endothelium has anticoagulant properties that prevent the adhesion of leukocytes and platelets to the vessel walls, contributing to resistance to thrombus formation, and regulating inflammation, and vascular smooth muscle cell proliferation. Many risk factors of cardiovascular diseases (CVDs) promote the endothelial expression of chemokines, cytokines, and adhesion molecules. The resultant endothelial activation can lead to endothelial cell dysfunction (ECD). In vitro models of ECD allow the study of cellular and molecular mechanisms of disease and provide a research platform for screening potential therapeutic agents. Even though alternative models are available, such as animal models or ex vivo models, in vitro models offer higher experimental flexibility and reproducibility, making them a valuable tool for the understanding of pathophysiological mechanisms of several diseases, such as CVDs. Therefore, this review aims to synthesize the currently available in vitro models regarding ECD, emphasizing CVDs. This work will focus on 2D cell culture models (endothelial cell lines and primary ECs), 3D cell culture systems (scaffold-free and scaffold-based), and 3D cell culture models (such as organ-on-a-chip). We will dissect the role of external stimuli-chemical and mechanical-in triggering ECD.
Collapse
Affiliation(s)
- Ana Grego
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Cristiana Fernandes
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ivo Fonseca
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Raquel Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Sandra Marisa Oliveira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
5
|
Wang B, Li Y, Ouyang Q, Xu MT, Wang YY, Fu SJ, Liu WQ, Liu XT, Ling H, Zhang X, Xiu RJ, Liu MM. Strain- and sex-dependent variability in hepatic microcirculation and liver function in mice. World J Gastroenterol 2025; 31:101058. [DOI: 10.3748/wjg.v31.i15.101058] [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: 09/04/2024] [Revised: 02/02/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The integrity and functionality of the hepatic microcirculation are essential for maintaining liver health, which is influenced by sex and genetic background. Understanding these variations is crucial for addressing disparities in liver disease outcomes.
AIM To investigate the sexual dimorphism and genetic heterogeneity of liver microcirculatory function in mice.
METHODS We assessed hepatic microhemodynamics in BALB/c, C57BL/6J, and KM mouse strains using laser Doppler flowmetry and wavelet analysis. We analyzed the serum levels of alanine transaminase, glutamic acid aminotransferase, total bile acid, total protein, alkaline phosphatase, and glucose. Histological and immunohistochemical staining were employed to quantify microvascular density and the expression levels of cluster of differentiation (CD) 31, and estrogen receptor α, and β. Statistical analyses, including the Mantel test and Pearson correlation, were conducted to determine the relationships among hepatic function, microcirculation, and marcocirculation between different sexes and across genetic backgrounds.
RESULTS We identified sex-based disparities in hepatic microhemodynamics across all strains, with males exhibiting higher microvascular perfusion and erythrocyte concentration, but lower blood velocity. Strain-specific differences were evident, particularly in the endothelial oscillatory characteristics of the erythrocyte concentration. No sex-dependent differences in estrogen receptor expression were observed, while significant variations in CD31 expression and microvascular density were observed. The correlations highlighted relationships between hepatic microhemodynamics and liver function indicators.
CONCLUSION Our findings indicate the influence of genetic and sex differences on hepatic microcirculation and liver function, highlighting the necessity of incorporating both genetic background and sex into hepatic physiology studies and potential liver disease management strategies.
Collapse
Affiliation(s)
- Bing Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yuan Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Qin Ouyang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Science, Beijing 100102, China
| | - Meng-Ting Xu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Ying-Yu Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Sun-Jing Fu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Wei-Qi Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xue-Ting Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Hao Ling
- Department of Radiology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, Hunan Province, China
| | - Xu Zhang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing 100034, China
| | - Rui-Juan Xiu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Ming-Ming Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
6
|
Liu Z, Shi X, Ke T, Yan Z, Xiong L, Tang F. USP7 promotes endothelial activation to aggravate sepsis-induced acute lung injury through PDK1/AKT/NF-κB signaling pathway. Cell Death Discov 2025; 11:183. [PMID: 40246841 PMCID: PMC12006344 DOI: 10.1038/s41420-025-02481-1] [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: 12/05/2024] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Disruption of the endothelial cell barrier and the subsequent inflammatory response represent a central pathological feature of acute lung injury (ALI). Ubiquitination plays a pivotal role in regulating protein stability, intracellular transport, and enzyme activity, which is typically reversed by deubiquitinating enzymes. Nevertheless, the function of deubiquitinating enzymes in endothelial biology and in ALI remains largely uninvestigated. The present study demonstrates that the expression of USP7 is increased in instances of endothelial inflammation and ALI. The knockdown or inhibition of USP7 using specific inhibitors was observed to significantly reduce the TNF-α-induced inflammatory response of endothelial cells and their adhesion capacity to monocytes. Conversely, the overexpression of USP7 was observed to promote the inflammatory response and adhesion capacity of endothelial cells. The specific inhibitors of USP7 were found to be effective in mitigating acute lung injury induced by LPS. From a mechanistic perspective, our findings indicate that USP7 binds and deubiquitinates PDK1, thereby stabilizing PDK1 and promoting the activity of the inflammatory pathway in endothelial cells. In conclusion, our findings demonstrate the role of a novel USP7-PDK1 signaling axis in regulating TNF-α-induced vascular endothelial injury and reveal that USP7 is a deubiquitylating enzyme of PDK1. These observations suggest that targeting the USP7-PDK1 axis may offer a promising therapeutic strategy for the treatment of acute lung injury.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoyun Shi
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Tiantian Ke
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhisu Yan
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Lei Xiong
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fang Tang
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
7
|
Simats A, Sager HB, Liesz A. Heart-brain axis in health and disease: role of innate and adaptive immunity. Cardiovasc Res 2025; 120:2325-2335. [PMID: 39180327 DOI: 10.1093/cvr/cvae185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 08/26/2024] Open
Abstract
The importance of the brain-heart interaction has been increasingly recognized as a critical physiological axis that is altered in disease. In this review, we explore the intricate relationship between the central nervous system and cardiovascular health, focusing particularly on immunological mechanisms that influence the course of both neurological and cardiovascular diseases. While previous studies have established a key role of the autonomic nervous system (ANS) in linking brain and the heart, more recent studies have expanded our understanding of the multifaceted inter-organ interactions. As such, circulating mediators include immune cells of the adaptive and innate immune system and their secreted immunogenic factors have come into the focus as mediators along this bidirectional communication. Hence, in this review we briefly discuss the contribution of the ANS and then focus on innate and adaptive immune mechanisms along the heart-to-brain and brain-to-heart axes, illustrating how cardiovascular diseases affect cognitive functions and how brain pathologies lead to cardiac complications.
Collapse
Affiliation(s)
- Alba Simats
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), c/Rosselló 161, 08036 Barcelona, Spain
| | - Hendrik B Sager
- DZHK (German Center for Cardiovascular Research), Partner site Munich Heart Alliance, Munich, Germany
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University Medical Center Munich, Feodor-Lynen-Straße 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| |
Collapse
|
8
|
Jannesar K, Soraya H. MPO and its role in cancer, cardiovascular and neurological disorders: An update. Biochem Biophys Res Commun 2025; 755:151578. [PMID: 40043618 DOI: 10.1016/j.bbrc.2025.151578] [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: 10/25/2024] [Revised: 01/31/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
Myeloperoxidase (MPO) is an enzyme that contains a heme group, found mostly in neutrophils and in small amounts in monocytes and plays a major role in their anti-microbial activity. However, excessive levels of MPO have been linked to various disorders and identified as a major cause of tissue destruction. Inhibiting its activity can reduce the severity and extent of tissue damage. Over activity of MPO during chronic inflammation has been shown to be involved in tumorigenesis by inducing a hyper-mutagenic environment through oxidant interaction with DNA, causing DNA modification. Vascular endothelium is one of the most important targets of MPO and high levels have been associated with increased rates of cardiomyopathy, ischemic stroke, heart failure, myocardial infarction, and atrial fibrillation. Therefore, it may be considered a therapeutic target in the treatment of cardiovascular disorders. MPO also participates in the pathogenesis of neurodegenerative diseases. For example, an increase in MPO levels has been observed in the brain tissue of patients with Alzheimer's, Multiple sclerosis (MS), and Parkinson's diseases. In Alzheimer's disease, active MPO is mostly found in the location of beta amyloids and microglia. Therefore, targeting MPO may be a potential treatment and prevention strategy for neurological disorders. This review will discuss MPO's physiological and pathological role in cancer, cardiovascular, and neurological disorders.
Collapse
Affiliation(s)
- Kosar Jannesar
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
9
|
Meng Q, Song C, Ma J, Cui J, Zhan J, Zhao J, Zhang Y, Zhu Z, Du X. Quercetin Prevents Hyperuricemia Associated With Gouty Arthritis by Inactivating the NLRP3/NF-κB Signaling Pathway. Chem Biol Drug Des 2025; 105:e70103. [PMID: 40230265 DOI: 10.1111/cbdd.70103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/15/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Quercetin (QCT) shows great therapeutic potential for hyperuricemia (HUA) associated with gouty arthritis (GA). However, the underlying mechanism of QCT in inhibiting the progression of HUA and GA remains unclear. HUA mouse model was established by injection of oteracil potassium (OXO) combined with ethambutol (EMB). The GA mouse model was established by intraarticular injection of sodium urate (MSU). MSU-induced HK-2 cells as well as lipopolysaccharide (LPS) and MSU-induced THP-1/M0 macrophages were used as cell models. The ankle perimeter of each mouse was measured to evaluate ankle swelling. The study also detected serum levels of uric acid (UA), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β (IL-1β), and IL-6 and analyzed the pathological conditions of synovial tissues and renal tissues. QCT treatment inhibited ankle joint swelling, TNF-α, IL-1β, and IL-6 serum levels as well as UA production in HUA and GA mice. Treatment with QCT inhibited oxidative stress in the renal tissues of HUA and GA mice and MSU-induced HK-2 cells. QCT treatment inhibited the inflammatory response in LPS and MSU-induced THP-1/M0 macrophages. QCT treatment inactivated the NOD-like receptor thermal protein domain associated protein 3 (NLRP3)/nuclear factor kappa-B (NF-κB) pathway. QCT inactivated the NLRP3/NF-κB signaling pathway to prevent HUA associated with GA.
Collapse
Affiliation(s)
- Qingliang Meng
- Department of Rheumatism, Henan Povince Hospital of TCM, Zhengzhou, Henan, China
| | - Chenyang Song
- Academy of Orthopedics and Traumatology, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Junfu Ma
- Department of Rheumatism, Henan Povince Hospital of TCM, Zhengzhou, Henan, China
| | - Jiakang Cui
- Department of Rheumatism, Henan Povince Hospital of TCM, Zhengzhou, Henan, China
| | - Junping Zhan
- Department of Rheumatism, Henan Povince Hospital of TCM, Zhengzhou, Henan, China
| | - Jing Zhao
- Academy of Orthopedics and Traumatology, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yunan Zhang
- Academy of Orthopedics and Traumatology, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zelin Zhu
- Academy of Orthopedics and Traumatology, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xuzhao Du
- Department of Orthopaedic DiseasesI, Henan Povince Hospital of TCM, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Wacker BK, Bi L, Saenz-Pipaon G, Sanford N, Regan AZ, Lim NS, Liu L, Kim F, Dichek DA. Overexpression of ABCA1 in Carotid Endothelium of Hyperlipidemic Rabbits Modulates Vascular Inflammation. Hum Gene Ther 2025; 36:750-764. [PMID: 40111153 DOI: 10.1089/hum.2024.166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
Endothelial activation and dysfunction are key early steps in atherogenesis. Vascular gene therapy targeting endothelial inflammation and cholesterol accumulation could decrease atherosclerosis progression. ATP-binding cassette subfamily A member 1 (ABCA1) exhibits anti-inflammatory properties and promotes cholesterol efflux. A mouse model showed that systemic endothelial overexpression of ABCA1 decreased diet-induced atherosclerosis. To test if local ABCA1 endothelial overexpression protects against atherosclerosis, we used helper-dependent adenoviral vectors (HDAd) to express ABCA1 or a "Null" control in the carotid endothelium of hyperlipidemic rabbits. Both ABCA1 mRNA and endothelial protein were increased 3 days after vector infusion. After 24 weeks on a high-fat diet, laser-microdissected endothelium showed increased ABCA1 mRNA expression, but whole-vessel ABCA1 mRNA was decreased with HDAdABCA1. Endothelial ABCA1 protein could not be measured at 24 weeks, so its overexpression may be transient. CD68 expression was decreased (-23%, p < 0.001), but ITGAM (-15%, p = 0.3) was unchanged. Macrophage markers for both M1-like macrophages (IL1B: -44% [p = 0.02]; IL6: -40% [p = 0.02]; CCL2: -25% [p = 0.02]) and M2-like macrophages (ARG1: -27% [p = 0.03]; IL10: -23% [p = 0.09]; TGFB1: -13% [p < 0.001]) were also decreased. The inflammatory cytokines IL6 (-100%; p < 0.001) and TNF (p < 0.05) were significantly decreased in the laser-microdissected endothelium, but VCAM1 (+5%, p = 1.0) was unchanged and ICAM1 (+101%; p = 0.03) increased. Lesion size, intimal lipid, and intimal macrophage content were all unchanged (p > 0.5 for all), and vascular cholesterol measured by mass spectrometry (-11%; p = 0.9) also showed no difference. There was a small decrease in the intimal/medial ratio. scRNAseq revealed that vector transcripts were not restricted to endothelial cells after 24+ weeks but were detected in most cell types. The exception was modulated smooth muscle cells, which were found in substantial numbers in larger lesions. Overall, transient overexpression of ABCA1 in the vascular endothelium subtly alters the expression of inflammatory markers, providing only a modest atheroprotection.
Collapse
Affiliation(s)
- Bradley K Wacker
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Lianxiang Bi
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Goren Saenz-Pipaon
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Nicole Sanford
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Abigail Z Regan
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Natalie S Lim
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Li Liu
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Francis Kim
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - David A Dichek
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Montagnino J, Kaufman MW, Shetty M, Centeno C, Fredericson M. Optimizing orthobiologic therapies with exercise, diet, and supplements. PM R 2025; 17:452-462. [PMID: 39853939 DOI: 10.1002/pmrj.13320] [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: 01/30/2024] [Revised: 10/09/2024] [Accepted: 11/22/2024] [Indexed: 01/26/2025]
Abstract
Orthobiologic injections including platelet-rich plasma (PRP) and cell-based injections are becoming increasingly popular. Evidence suggests that these therapies can be effective in certain situations. The efficacy of these injections may be more dependent on the quality of the injectate, which given their autologous nature, may be dependent on lifestyle choices like exercise, diet, and supplements. The literature describing PRP injections shows that the number and activity of platelets can improve their efficacy. A multitude of lifestyle modifications can affect those factors. Exercise intensity appears to increase platelet count and increases adhesion as well as release of growth factors. Low inflammatory diets increase platelet counts and activity overall. Stress, some supplements, high cholesterol, or processed sugar diets can increase inflammation and potentially decrease platelet counts as well as quality of PRP injectate. Similarly, cell-based therapies can be affected by mesenchymal stromal cell (MSC) number and quality. Cell-based therapy is based upon limiting cellular senescence and increasing replication and differentiation. Exercise may limit senescence and improve replication and differentiation of these cell-based therapies, especially in older adult populations. There are a multitude of supplements that may potentiate these types of injections and patients should discuss the potential benefits and concerns when starting a supplement regimen. Certain foods as well as changes in oxygenation may limit cellular senescence and lower calorie intake may affect MSC viability and function as well. Overall, the current state of literature describes biologic plausibility of how exercise, diet and supplements might affect orthobiologic injection efficacy. Further translational research needs to be completed to describe the effect size and improve recommendations for clinical implementation.
Collapse
Affiliation(s)
- Jami Montagnino
- Department of Orthopaedic Surgery, Stanford University, Redwood City, California, USA
- Department of Orthopedics, Tulane University, Tulane Institute of Sports Medicine, New Orleans, Louisiana, USA
| | - Matthew W Kaufman
- Department of Orthopaedic Surgery, Stanford University, Redwood City, California, USA
- Stanford Research and Prevention Center, Stanford Lifestyle Medicine, Redwood City, California, USA
| | - Maya Shetty
- Department of Orthopaedic Surgery, Stanford University, Redwood City, California, USA
- Stanford Research and Prevention Center, Stanford Lifestyle Medicine, Redwood City, California, USA
| | | | - Michael Fredericson
- Department of Orthopaedic Surgery, Stanford University, Redwood City, California, USA
- Stanford Research and Prevention Center, Stanford Lifestyle Medicine, Redwood City, California, USA
| |
Collapse
|
12
|
Zhang S, Li S, Cui L, Xie S, Wang Y. Astragaloside IV Attenuates Angiotensin II-Induced Inflammatory Responses in Endothelial Cells: Involvement of Mitochondria. J Inflamm Res 2025; 18:3951-3967. [PMID: 40125084 PMCID: PMC11927501 DOI: 10.2147/jir.s504427] [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: 11/03/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Background Angiotensin II (Ang II)-triggered endothelial inflammation is a critical mechanism contributing to Ang II-related cardiovascular diseases. The inflammation is highly correlated with mitochondrial function. Although astragaloside IV (AS-IV), a primary bioactive ingredient extracted from the traditional Chinese medicine Astragalus membranaceus Bunge that can effectively treat numerous cardiovascular diseases, posses the actions of antiinflammation and antioxidation in vivo, limited data are made available on the impacts of AS-IV on mitochondrial function in endothelial inflammation triggered by Ang II. This study was performed to evaluate the in vitro actions of AS-IV on Ang II-triggered inflammatory responses in endothelial cells, and to further clarify the potential role of mitochondria in the actions. Methods Human umbilical vein endothelial cells (HUVECs) were preincubated with AS-IV and then exposed to Ang II for 12 h. Results The exposure of HUVECs to Ang II triggered cytokine and chemokine production, the upregulation of adhesive molecules, monocyte attachment, and nuclear factor-kappa B activation. Additionally, our results showed that the inflammatory responses triggered by Ang II were associated with the impairment of mitochondrial function, as evidenced by the reductions of mitochondrial membrane potential, ATP synthesis, and mitochondrial complexes I and III activities. Moreover, the concentrations of malondialdehyde, cellular reactive oxygen species, and mitochondrial superoxide enhanced after HUVECs challenged with Ang II, which were concurrent with the decreases in total superoxide dismutase (SOD) and its isoenzyme activities such as Mn-SOD. These Ang II-induced alterations were reversed by preincubation with AS-IV. Conclusion Our data indicate that AS-IV attenuates Ang II-triggered endothelial inflammation possibly via ameliorating mitochondrial function.
Collapse
Affiliation(s)
- Shiyu Zhang
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Shijie Li
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Lin Cui
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Shiyang Xie
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Youping Wang
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| |
Collapse
|
13
|
Grossini E, Bellan M, Venkatesan S, Ola Pour MM, Mennuni M, D’Amario D, Bruno S, Ferrante D, Capello D, Sainaghi PP, Pirisi M, Patti G. Characterization of Circulating Vesicles of Complicated and Uncomplicated Systemic Sclerosis Patients and Their Role in Vascular Dysfunction. Int J Mol Sci 2025; 26:2380. [PMID: 40141024 PMCID: PMC11942416 DOI: 10.3390/ijms26062380] [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: 12/11/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Extracellular vesicles (EVs) could be involved in the onset of systemic sclerosis (SSc) through the modulation of vascular function. Anyway, available data are contradictory, and further investigation would be necessary to clarify this aspect. Here, we characterized circulating EVs isolated from SSc patients and evaluated their effects on human vascular endothelial cells (HUVECs) and smooth muscle cells. In EVs from 13 complicated and 27 uncomplicated SSc patients and five healthy controls (HCs), we analyzed the size, concentration, and surface marker expression. In addition, EVs were used to stimulate HUVECs, and we evaluated cell viability, mitochondrial membrane potential, and nitric oxide (NO) and mitochondrial reactive oxygen species (MitoROS) release. In smooth muscle cells, the effects of EVs on calcium movement were examined. The results showed that the EVs of SSc patients expressed markers of T-lymphocyte/platelet/endothelial cell origin and were larger and more concentrated than those from HCs. In addition, the EVs of SSc patients reduced cell viability and mitochondrial membrane potential and increased NO and MitoROS release in HUVECs and intracellular calcium in smooth muscle cells. In conclusion, we found a specific pattern for EVs isolated from SSc patients, which could have a pathogenic role through direct actions on endothelial and smooth muscle cells.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Mattia Bellan
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy; (M.B.); (P.P.S.); (M.P.)
- CAAD, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy
| | - Sakthipriyan Venkatesan
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Mohammad Mostafa Ola Pour
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Marco Mennuni
- Cardiology Unit, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy; (M.M.); (D.D.); (G.P.)
| | - Domenico D’Amario
- Cardiology Unit, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy; (M.M.); (D.D.); (G.P.)
| | - Stefania Bruno
- Laboratory of Translational Research, Department of Medical Sciences, University of Torino, 10126 Torino, Italy;
| | - Daniela Ferrante
- Statistic Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy;
| | - Daniela Capello
- Laboratory of Clinical Biochemistry, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy;
- UPO Biobank, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Pier Paolo Sainaghi
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy; (M.B.); (P.P.S.); (M.P.)
- CAAD, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy
| | - Mario Pirisi
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy; (M.B.); (P.P.S.); (M.P.)
- CAAD, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy
| | - Giuseppe Patti
- Cardiology Unit, Department of Translational Medicine, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria Maggiore della Carità, 28100 Novara, Italy; (M.M.); (D.D.); (G.P.)
| |
Collapse
|
14
|
Kulpa J, Paduch J, Szczepanik M, Gorący-Rosik A, Rosik J, Tchórz M, Pawlik A, Gorący J. Catestatin in Cardiovascular Diseases. Int J Mol Sci 2025; 26:2417. [PMID: 40141061 PMCID: PMC11942146 DOI: 10.3390/ijms26062417] [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: 02/10/2025] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Cardiovascular diseases are one of the leading causes of mortality and morbidity worldwide. The pathogenesis of this group of disorders is highly complex and involves interactions between various cell types and substances, among others, catestatin (CTS). In recent years, numerous researchers have expanded our knowledge about CTS's role in development and its potential for the treatment of a variety of diseases. In this review, the authors discuss CTS's importance in the pathogenesis of arterial hypertension, coronary artery disease, and heart failure. Moreover, we present CTS's influence on heart and vessel function.
Collapse
Affiliation(s)
- Joanna Kulpa
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.K.); (J.P.); (M.S.); (M.T.); (A.P.)
| | - Jarosław Paduch
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.K.); (J.P.); (M.S.); (M.T.); (A.P.)
| | - Marcin Szczepanik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.K.); (J.P.); (M.S.); (M.T.); (A.P.)
| | - Anna Gorący-Rosik
- Independent Laboratory of Invasive Cardiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (A.G.-R.); (J.G.)
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.K.); (J.P.); (M.S.); (M.T.); (A.P.)
| | - Magdalena Tchórz
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.K.); (J.P.); (M.S.); (M.T.); (A.P.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.K.); (J.P.); (M.S.); (M.T.); (A.P.)
| | - Jarosław Gorący
- Independent Laboratory of Invasive Cardiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (A.G.-R.); (J.G.)
| |
Collapse
|
15
|
Meneghini V, Tebar WR, Santos IS, Janovsky CCPS, Varella AC, Lotufo PA, Sgarbi JA, Teixeira PFS, Benseñor IM. Thyroid peroxidase antibodies were associated with prevalent carotid intima-media thickness in middle-age and older adults: the ELSA-Brasil cohort. J Endocrinol Invest 2025; 48:597-606. [PMID: 39382627 DOI: 10.1007/s40618-024-02475-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE Understanding the relationship between antithyroperoxidase antibodies (TPOAb) and carotid intima-media thickness (cIMT) could provide insights into the mechanisms linking thyroid autoimmunity and cardiovascular disease. We aimed to explore the association of multiple categories of TPOAb with the increased cIMT at baseline and at follow-up in participants from the ELSA-Brasil Study. METHODS This prospective cohort study analyzed data from 9,264 participants (51.5 ± 8.9 years old, 55.9% women) without a history of cardiovascular disease. Fasting serum TPOAb levels were determined. Values of cIMT equal to or above one deviation standard of the sample's mean were classified as increased cIMT at baseline. The increased cIMT after the 8-year follow-up was calculated after excluding participants with increased cIMT at baseline. Multivariate analyses were done using binary logistic and Poisson regression models. RESULTS The increased cIMT was prevalent in 14.3% of the participants at baseline and its development occurred in 16.8% participants during the cohort. After adjustment for all confounder variables, TPOAb detectability (OR = 1.84, 95%CI = 1.21-2.79), and low detectable (OR = 1.81, 95%CI = 1.18-2.75), high detectable (OR = 2.01, 95%CI = 1.29-3.11) and positive (OR = 1.70, 95%CI = 1.07-2.70) TPOAb were positively associated with increased cIMT at baseline. The associations of low and high detectable TPOAb and increased cIMT at baseline were consistent when excluding those with thyroid dysfunction. There was no statistically significant association between TPOAb levels and increased cIMT at follow-up (p > 0.05), neither for all sample nor for euthyroid individuals. CONCLUSION Different levels of TPOAb, including its detectability, were associated with increased cIMT at baseline in the studied sample. We highlight that may be relevant to consider the levels of TPOAb detectability as possible marker of increased cardiovascular risk.
Collapse
Grants
- baseline grants: 01 06 0010.00 RS, 01 06 0212.00 BA, 01 06 0300.00 ES, 01 06 0278.00 MG, 01 06 0115.00 SP, and 01 06 0071.00 RJ; 8-year follow-up grants: 405551/2015-0 BA, 405544/2015-4 RJ, 405547/2015-3 SP, 405552/2015-7 MG, 405543/2015-8 ES, and 405545/2015-0 RS Ministério da Saúde
- 2011/12256-4 e 2019/23734-6 Fundação de Amparo à Pesquisa do Estado de São Paulo
- 2021/04416-3 Fundação de Amparo à Pesquisa do Estado de São Paulo
- 2021/06798-0 Fundação de Amparo à Pesquisa do Estado de São Paulo
Collapse
Affiliation(s)
- Vandrize Meneghini
- Center for Clinical and Epidemiological Research, Universidade de São Paulo, São Paulo, SP, Brazil
| | - William R Tebar
- Center for Clinical and Epidemiological Research, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Itamar S Santos
- Center for Clinical and Epidemiological Research, Universidade de São Paulo, São Paulo, SP, Brazil
- Department of Internal Medicine, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Carolina C P S Janovsky
- Center for Clinical and Epidemiological Research, Universidade de São Paulo, São Paulo, SP, Brazil
- Division of Endocrinology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Ana C Varella
- Center for Clinical and Epidemiological Research, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Paulo A Lotufo
- Center for Clinical and Epidemiological Research, Universidade de São Paulo, São Paulo, SP, Brazil
- Department of Internal Medicine, Universidade de São Paulo, São Paulo, SP, Brazil
| | - José A Sgarbi
- Division of Endocrinology and Metabolism, Department of Medicine, Faculdade de Medicina de Marilia, Marilia, SP, Brazil
| | - Patrícia F S Teixeira
- Faculty of Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Isabela M Benseñor
- Center for Clinical and Epidemiological Research, Universidade de São Paulo, São Paulo, SP, Brazil.
- Department of Internal Medicine, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
16
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. Harnessing Gasotransmitters to Combat Age-Related Oxidative Stress in Smooth Muscle and Endothelial Cells. Pharmaceuticals (Basel) 2025; 18:344. [PMID: 40143122 PMCID: PMC11946800 DOI: 10.3390/ph18030344] [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: 01/25/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Age-related oxidative stress is a critical factor in vascular dysfunction, contributing to hypertension and atherosclerosis. Smooth muscle cells and endothelial cells are particularly susceptible to oxidative damage, which exacerbates vascular aging through cellular senescence, chronic inflammation, and arterial stiffness. Gasotransmitters-hydrogen sulfide (H2S), nitric oxide (NO), and carbon monoxide (CO)-are emerging as promising therapeutic agents for counteracting these processes. This review synthesizes findings from recent studies focusing on the mechanisms by which H2S, NO, and CO influence vascular smooth muscle and endothelial cell function. Therapeutic strategies involving exogenous gasotransmitter delivery systems and combination therapies were analyzed. H2S enhances mitochondrial bioenergetics, scavenges ROS, and activates antioxidant pathways. NO improves endothelial function, promotes vasodilation, and inhibits platelet aggregation. CO exhibits cytoprotective and anti-inflammatory effects by modulating heme oxygenase activity and ROS production. In preclinical studies, gasotransmitter-releasing molecules (e.g., NaHS, SNAP, CORMs) and targeted delivery systems show significant promise. Synergistic effects with lifestyle modifications and antioxidant therapies further enhance their therapeutic potential. In conclusion, gasotransmitters hold significant promise as therapeutic agents to combat age-related oxidative stress in vascular cells. Their multifaceted mechanisms and innovative delivery approaches make them potential candidates for treating vascular dysfunction and promoting healthy vascular aging. Further research is needed to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
17
|
Wang J, Lin C, Li X, Wei J, Wang Y, Li R, Ming Y, Tang J, Liu J, Zhang M, Ji H. Polydopamine nanoparticles loaded with sodium ferulate for targeted therapy of myocardial infarction in endothelial cells. Int J Pharm 2025; 671:125187. [PMID: 39855278 DOI: 10.1016/j.ijpharm.2025.125187] [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: 11/03/2024] [Revised: 12/16/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025]
Abstract
Myocardial infarction (MI) is a leading cause of heart failure and death in cardiovascular diseases. Most drug trials currently fail due to inadequate local drug activity and side effects. In this study, we developed a novel polydopamine (PDA) nano delivery system that carries sodium ferulate (SF) and is modified with RGD peptides (SF/RGD-PDA NPs) for precise targeted delivery. SF is a clinical adjuvant for cardiovascular and cerebrovascular diseases but lacks targeting and has a short half-life. PDA, known for its excellent biocompatibility and surface modifiability, serves as an effective carrier. SF was loaded onto PDA through π-π stacking, while RGD was attached via a Michael addition reaction, resulting in stable SF/RGD-PDA NPs with an average particle size of 206.13 nm. In vitro and in vivo studies indicate that this targeted formulation has good safety. Targeting studies showed a 2.19-fold increase in nanoparticle accumulation in the heart and a 5.94-fold increase in cellular uptake efficiency. Pharmacodynamic studies revealed a 1.45-fold increase in endothelial cell proliferation, a 1.46-fold increase in angiogenesis rate, and a significant reduction in MI area. These findings suggest that SF/RGD-PDA NPs can improve endothelial cell function and reduce MI area through targeted delivery.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, the Second Affiliated Hospital, Harbin Medical University (Key Laboratory of Medications Research, College of Heilongjiang Province), Harbin 150086, China; Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Congcong Lin
- Department of Pharmacy, the Second Affiliated Hospital, Harbin Medical University (Key Laboratory of Medications Research, College of Heilongjiang Province), Harbin 150086, China; Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Xiaoyang Li
- Department of Pharmacy, the Second Affiliated Hospital, Harbin Medical University (Key Laboratory of Medications Research, College of Heilongjiang Province), Harbin 150086, China; Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Junyao Wei
- Department of Pharmacy, the Second Affiliated Hospital, Harbin Medical University (Key Laboratory of Medications Research, College of Heilongjiang Province), Harbin 150086, China; Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Yihua Wang
- Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Rongtao Li
- Department of Pharmacy, the Second Affiliated Hospital, Harbin Medical University (Key Laboratory of Medications Research, College of Heilongjiang Province), Harbin 150086, China; Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Yan Ming
- Department of Pharmacy, the Second Affiliated Hospital, Harbin Medical University (Key Laboratory of Medications Research, College of Heilongjiang Province), Harbin 150086, China; Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Jingling Tang
- Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Jiaxin Liu
- Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Manjie Zhang
- Department of Pharmacy, the Second Affiliated Hospital, Harbin Medical University (Key Laboratory of Medications Research, College of Heilongjiang Province), Harbin 150086, China; Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China.
| | - Hongyu Ji
- Department of Pharmacy, the Second Affiliated Hospital, Harbin Medical University (Key Laboratory of Medications Research, College of Heilongjiang Province), Harbin 150086, China; Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
18
|
Penna C, Pagliaro P. Endothelial Dysfunction: Redox Imbalance, NLRP3 Inflammasome, and Inflammatory Responses in Cardiovascular Diseases. Antioxidants (Basel) 2025; 14:256. [PMID: 40227195 PMCID: PMC11939635 DOI: 10.3390/antiox14030256] [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: 01/26/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 04/15/2025] Open
Abstract
Endothelial dysfunction (ED) is characterized by an imbalance between vasodilatory and vasoconstrictive factors, leading to impaired vascular tone, thrombosis, and inflammation. These processes are critical in the development of cardiovascular diseases (CVDs) such as atherosclerosis, hypertension and ischemia/reperfusion injury (IRI). Reduced nitric oxide (NO) production and increased oxidative stress are key contributors to ED. Aging further exacerbates ED through mitochondrial dysfunction and increased oxidative/nitrosative stress, heightening CVD risk. Antioxidant systems like superoxide-dismutase (SOD), glutathione-peroxidase (GPx), and thioredoxin/thioredoxin-reductase (Trx/TXNRD) pathways protect against oxidative stress. However, their reduced activity promotes ED, atherosclerosis, and vulnerability to IRI. Metabolic syndrome, comprising insulin resistance, obesity, and hypertension, is often accompanied by ED. Specifically, hyperglycemia worsens endothelial damage by promoting oxidative stress and inflammation. Obesity leads to chronic inflammation and changes in perivascular adipose tissue, while hypertension is associated with an increase in oxidative stress. The NLRP3 inflammasome plays a significant role in ED, being triggered by factors such as reactive oxygen and nitrogen species, ischemia, and high glucose, which contribute to inflammation, endothelial injury, and exacerbation of IRI. Treatments, such as N-acetyl-L-cysteine, SGLT2 or NLRP3 inhibitors, show promise in improving endothelial function. Yet the complexity of ED suggests that multi-targeted therapies addressing oxidative stress, inflammation, and metabolic disturbances are essential for managing CVDs associated with metabolic syndrome.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy;
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy;
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
19
|
Chen Z, Di X, Chen H, Song S, Chen R, Kou L, Chu M. MEF2C mitigates coronary artery lesions in Kawasaki disease by enhancing endothelial barrier function through KLF2 regulation. Int Immunopharmacol 2025; 148:114030. [PMID: 39826452 DOI: 10.1016/j.intimp.2025.114030] [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: 11/01/2024] [Revised: 12/16/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Coronary artery lesions constitute a significant complication of Kawasaki disease (KD) and represents one of the primary etiologies of acquired cardiovascular disease in pediatric populations. In the present study, we observed a downregulation of MEF2C expression in the whole blood of KD patients and in human coronary artery endothelial cells (HCAECs) during the pathophysiological progression of KD. Furthermore, transcriptomic data analysis, in conjunction with observations from HCAECs stimulated with KD serum, indicates that the downregulation of MEF2C in KD is correlated with increased inflammatory levels and the activation of inflammatory pathways. Overexpression of MEF2C has the potential to mitigate inflammation and apoptosis in HCAECs, whereas MEF2C knockdown exhibits contrary effects. Furthermore, MEF2C overexpression may alleviate inflammation and apoptosis in the coronary endothelium, attenuate abdominal aortic dilation, and prevent the decline of cardiac function in a CAWS-induced KD murine model. Mechanistically, MEF2C overexpression safeguards against KD-induced endothelial barrier disruption and the downregulation of endothelial junction proteins in coronary injury associated with KD. Additionally, through RNA sequencing, we identified that KLF2 might be involved in the MEF2C-mediated protection against coronary endothelial injury. Employing a gene interference methodology, we substantiated that MEF2C mitigates coronary artery injury in KD via KLF2-regulated endothelial barrier protection in HCAECs. These findings suggest that MEF2C could serve as a potential therapeutic target for the prevention and treatment of coronary lesions in KD.
Collapse
Affiliation(s)
- Zhiwei Chen
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinyu Di
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Heyan Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shengnan Song
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Maoping Chu
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
20
|
Jiang Y, Ayaz Z, Xiang L, Zhou L, Mamizadeh L, Wang Y, Dong X, Huang N, Leng Y, Akhavan B, Jing F. Sodium Carboxymethylcellulose/Polydopamine Biocellulose Coatings with Enhanced Wet Stability for Implantable Medical Devices. ACS APPLIED BIO MATERIALS 2025; 8:1050-1064. [PMID: 39899808 DOI: 10.1021/acsabm.4c01278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Sodium carboxymethylcellulose (CMC) is a biocompatible and biodegradable derivative of cellulose, making it a promising material for biomedical applications. However, its poor stability in aqueous environments has significantly limited its use in long-term biomedical devices. Here, we present for the first time a simple and controllable method to enhance the wet stability of CMC coatings by cross-linking of CMC and polydopamine (PDA) and self-polymerization of PDA for widespread applications in biomedical devices. A series of CMC/PDA coatings were fabricated on the initial PDA layers by using dip coating and subsequently heated at 200 °C. The performance of the CMC/PDA coatings and their chemical and structural stability in aqueous media have been systematically analyzed, and the mechanisms underpinning their robust performance have been revealed. FITR, X-ray photoelectron spectroscopy (XPS), and gel permeation chromatography (GPC) results showed that CMC/PDA coatings involved amidation and esterification reactions as well as self-polymerization of PDA. Degradation studies in phosphate-buffered saline (PBS) solution at 37 °C indicated degradation via ester and amide bond cleavage, with the stability of CMC/PDA coatings surpassing that of individual PDA and CMC coatings over a 30-day immersion period. The CMC/PDA coating with a CMC concentration of 15 mg/mL exhibited the highest adhesion strength in an aqueous environment, which was attributed to the high cross-linking of CMC and PDA, as well as the intrinsic stability of PDA. The CMC/PDA coatings demonstrated favorable viability, growth, and proliferation of endothelial cells. The stable and biocompatible biocellulose coatings can be easily applied from aqueous solutions onto almost any type of solid metal and ceramic material, providing a promising dimension for surface engineering of vascular scaffolds and tissue engineering constructs.
Collapse
Affiliation(s)
- Yehao Jiang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zainab Ayaz
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Long Xiang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lili Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Leila Mamizadeh
- School of Engineering, University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute (HMRI), Precision Medicine Program, New Lambton Heights, NSW 2305, Australia
- School of Biomedical Engineering, University of Sydney, Sydney, New South Wales 2006, Australia
- School of Physics, University of Sydney, Sydney, NSW 2006, Australia
| | - Yong Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xie Dong
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Nan Huang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yongxiang Leng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Behnam Akhavan
- School of Engineering, University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute (HMRI), Precision Medicine Program, New Lambton Heights, NSW 2305, Australia
- School of Biomedical Engineering, University of Sydney, Sydney, New South Wales 2006, Australia
- School of Physics, University of Sydney, Sydney, NSW 2006, Australia
- Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Fengjuan Jing
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
21
|
Huang C, Wei Z, Zheng N, Yan J, Zhang J, Ye X, Zhao W. The interaction between dysfunction of vasculature and tauopathy in Alzheimer's disease and related dementias. Alzheimers Dement 2025; 21:e14618. [PMID: 39998958 PMCID: PMC11854360 DOI: 10.1002/alz.14618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/01/2025] [Accepted: 01/12/2025] [Indexed: 02/27/2025]
Abstract
Tauopathy is one of the pathological features of Alzheimer's disease and related dementias (ADRD). At present, there have been many studies on the formation, deposition, and intercellular transmission of tau in neurons and immune cells. The vasculature is an important component of the central nervous system. This review discusses the interaction between vasculature and tau in detail from three aspects. (1) The vascular risk factors (VRFs) discussed in this review include diabetes mellitus (DM), abnormal blood pressure (BP), and hypercholesterolemia. (2) In ADRD pathology, the hyperphosphorylation and deposition of tau interact with disrupted vasculature, such as different cells (endothelial cells, smooth muscular cells, and pericytes), the blood-brain barrier (BBB), and the cerebral lymphatic system. (3) The functions of vasculature are regulated by various signaling transductions. Endothelial nitric oxide synthase/nitric oxide, calcium signaling, Rho/Rho-associated coiled-coil containing Kinase, and receptors for advanced glycation end products are discussed in this review. Our findings indicate that the prevention and treatment of vascular health may be a potential target for ADRD combination therapy. HIGHLIGHTS: Persistent VRFs increase early disruption of vascular mechanisms and are strongly associated with tau pathology in ADRD. Cell dysfunction in the vasculature causes BBB leakage and drainage incapacity of the cerebral lymphatic system, which interacts with tau pathology. Signaling molecules in the vasculature regulate vasodilation and contraction, angiogenesis, and CBF. Abnormal signaling transduction is related to tau hyperphosphorylation and deposition.
Collapse
Affiliation(s)
- Chuyao Huang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Zhenwen Wei
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Ningxiang Zheng
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Jingsi Yan
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Jiayu Zhang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Xinyi Ye
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Wei Zhao
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| |
Collapse
|
22
|
Midtlien JP, Ashraf O, Hui FK, Tsappidi S, Zhang YJ, Forster AA, Chang E, Wiater AH, Starke RM, Abdelsalam A, Rai AT, Tarabishy AR, Boo S, Vu P, Ehrig MR, Kittel C, Brown PA, Wolfe SQ, Fargen KM. One in six patients exhibit changes in reperfusion on 10-minute repeat cerebral angiography during mechanical thrombectomy for stroke. J Neurointerv Surg 2025:jnis-2024-022448. [PMID: 39613324 DOI: 10.1136/jnis-2024-022448] [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: 08/30/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Post-recanalization target vessel re-occlusion (TVR) following endovascular thrombectomy (EVT) is a known complication of the procedure, and it is associated with worse long-term functional outcomes. The incidence and factors that contribute to TVR are not well understood, particularly within the immediate timeframe following EVT. METHODS A prospective, multicenter study was performed across four comprehensive stroke centers on adult patients undergoing EVT for acute large vessel occlusion. Modified Thrombolysis in Cerebral Infarction (TICI) score was recorded at the end of the standard procedure, and another TICI score was recorded 10 min later to evaluate for TVR. RESULTS 167 patients underwent EVT for a large vessel occlusion, 93.4% of which were in the anterior circulation. Twenty-seven patients (16.2%) had a change in their TICI score 10 min after EVT, with 19 of these patients (70%) having a worsening in their score. Of the total sample, 13% had their post-procedure care altered by any intervention, and 8% underwent further endovascular interventions due to the change in reperfusion over the 10 min time period. Functional independence (modified Rankin Scale score 0-2) at 90 days was observed in 31% of the entire cohort and in 21% of patients with a worse TICI score at 10 min. CONCLUSIONS This is the first study to prospectively assess for TVR in the immediate timeframe following EVT. One in six patients had a change in their TICI score, and one in 11 patients had additional intervention. Accordingly, neurointerventionalists should consider integrating angiographic evaluation at 10 min following EVT.
Collapse
Affiliation(s)
- Jackson P Midtlien
- Neurological Surgery and Radiology, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Omar Ashraf
- Neurological Surgery and Radiology, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Ferdinand K Hui
- Neurointerventional Surgery, The Neuroscience Institute, The Queen's Medical Center, Honolulu, Hawaii, USA
| | - Sam Tsappidi
- Neurointerventional Surgery, The Neuroscience Institute, The Queen's Medical Center, Honolulu, Hawaii, USA
| | - Yi Jonathan Zhang
- Neurological Surgery, The Neuroscience Institute, The Queen's Medical Center, Honolulu, Hawaii, USA
| | - Aren A Forster
- Neurological Surgery and Radiology, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Emily Chang
- Neurological Surgery and Radiology, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Angelina H Wiater
- Neurological Surgery and Radiology, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Robert M Starke
- Neurological Surgery, Radiology, Neurosciences, and Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ahmed Abdelsalam
- Neurological Surgery, Radiology, Neurosciences, and Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ansaar T Rai
- Neuroradiology, West Virginia University Rockefeller Neuroscience Institute, Morgantown, West Virginia, USA
| | - Abdul R Tarabishy
- Neuroradiology, West Virginia University Rockefeller Neuroscience Institute, Morgantown, West Virginia, USA
| | - SoHyun Boo
- Neuroradiology, West Virginia University Rockefeller Neuroscience Institute, Morgantown, West Virginia, USA
| | - Phong Vu
- Neuroradiology, West Virginia University Rockefeller Neuroscience Institute, Morgantown, West Virginia, USA
| | - Molly R Ehrig
- Biostatistics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Carol Kittel
- Biostatistics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Patrick A Brown
- Neurological Surgery and Radiology, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Stacey Q Wolfe
- Neurological Surgery and Radiology, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Kyle M Fargen
- Neurological Surgery and Radiology, Wake Forest University, Winston-Salem, North Carolina, USA
| |
Collapse
|
23
|
Chen Y, Chen D, Peng Y, Wang M, Wang W, Shi F, Wang Y, Hua L. The effect of vitamin D supplementation on endothelial function: An umbrella review of interventional meta-analyses. Nutr Metab Cardiovasc Dis 2025:103871. [PMID: 39986938 DOI: 10.1016/j.numecd.2025.103871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 12/10/2024] [Accepted: 01/22/2025] [Indexed: 02/24/2025]
Abstract
AIMS There is no consensus in the existing literature regarding the effect of vitamin D supplementation on endothelial function. This umbrella review aimed to assess meta-analyses of randomized controlled trials (RCTs) conducted in this field. DATA SYNTHESIS We systematically searched English-language databases, including PubMed, Embase, Scopus, and Web of Science, up to January 2024. Flow-Mediated Dilation (FMD), Pulse Wave Velocity (PWV), and Augmentation Index (AIx) were the primary endpoints evaluated. A total of 16 meta-analyses were included in the review. The results indicated that vitamin D supplementation significantly improved FMD as assessed by Standardized Mean Difference (SMD) (SMD = 0.72, 95 % CI: 0.34, 1.11; p < 0.001; I2 = 79.4 %, p < 0.001) and Weighted Mean Difference (WMD) (WMD = 1.91; 95 % CI: 0.66, 3.16; p = 0.003; I2 = 94.5 %, p < 0.001). PWV also showed a modest but significant improvement (SMD = -0.06, 95 % CI: -0.12, -0.00; p = 0.03; I2 = 0.0 %, p = 0.66). However, vitamin D had no significant impact on AIx based on SMD (SMD = -0.03, 95 % CI: -0.13, 0.06; p = 0.48; I2 = 0.0 %, p = 0.49) or WMD (WMD = 0.02, 95 % CI: -2.22, 2.25; p = 0.98; I2 = 29.2 %, p = 0.23). CONCLUSION These findings suggest that vitamin D supplementation may be a beneficial intervention for improving endothelial function, particularly in populations with low FMD. The effects on PWV were modest, while AIx remained unaffected. REGISTRATION NUMBER PROSPERO, CRD42024451215.
Collapse
Affiliation(s)
- Yirui Chen
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Hangzhou, Zhejiang, 310014, PR China
| | - Dandan Chen
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Hangzhou, Zhejiang, 310014, PR China
| | - Ye Peng
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Hangzhou, Zhejiang, 310014, PR China
| | - Manling Wang
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Hangzhou, Zhejiang, 310014, PR China
| | - Wensong Wang
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Hangzhou, Zhejiang, 310014, PR China
| | - Fangfang Shi
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Hangzhou, Zhejiang, 310014, PR China
| | - Yanzhong Wang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, PR China.
| | - Liqun Hua
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Hangzhou, Zhejiang, 310014, PR China.
| |
Collapse
|
24
|
Kaltsas A, Giannakodimos I, Markou E, Adamos K, Stavropoulos M, Kratiras Z, Zachariou A, Dimitriadis F, Sofikitis N, Chrisofos M. The Role of Gut Microbiota Dysbiosis in Erectile Dysfunction: From Pathophysiology to Treatment Strategies. Microorganisms 2025; 13:250. [PMID: 40005617 PMCID: PMC11857656 DOI: 10.3390/microorganisms13020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Erectile dysfunction (ED) is a prevalent male sexual disorder characterized by the persistent inability to achieve or maintain an erection sufficient for satisfactory sexual performance. While its etiology is multifactorial, encompassing vascular, neurological, hormonal, and psychological components, emerging evidence suggests a significant role for gut microbiota dysbiosis in its development. The gut microbiota influences various metabolic, inflammatory, and neuropsychological processes critical to erectile function. Dysbiosis can lead to systemic inflammation, endothelial dysfunction, hormonal imbalances, and altered neurotransmitter production, all of which are key factors in ED pathogenesis. This narrative review synthesizes current research on the association between gut microbiota alterations and ED, highlighting specific bacterial taxa implicated in ED through mechanisms involving inflammation, metabolic disturbances, and hormonal regulation. This review explores potential mechanisms linking gut microbiota and ED, including pro-inflammatory cytokines, gut barrier integrity disruption, metabolic disorders, psychological factors via the gut-brain axis, and hormonal regulation. Furthermore, the gut microbiota offers promising avenues for developing non-invasive biomarkers and therapeutic interventions such as probiotics, prebiotics, dietary modifications, and fecal microbiota transplantation. Future research should focus on longitudinal studies, mechanistic explorations, and clinical trials to validate these findings and translate them into clinical practice. Understanding the interplay between the gut microbiota and erectile function could unveil novel diagnostic biomarkers and pave the way for innovative treatments targeting the microbiota, ultimately improving men's sexual and overall health.
Collapse
Affiliation(s)
- Aris Kaltsas
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Ilias Giannakodimos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Eleftheria Markou
- Department of Microbiology, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Konstantinos Adamos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Marios Stavropoulos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Zisis Kratiras
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Athanasios Zachariou
- Laboratory of Spermatology, Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Nikolaos Sofikitis
- Laboratory of Spermatology, Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Michael Chrisofos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| |
Collapse
|
25
|
Donadini MP, Calcaterra F, Romualdi E, Ciceri R, Cancellara A, Lodigiani C, Bacci M, Della Bella S, Ageno W, Mavilio D. The Link Between Venous and Arterial Thrombosis: Is There a Role for Endothelial Dysfunction? Cells 2025; 14:144. [PMID: 39851572 PMCID: PMC11763525 DOI: 10.3390/cells14020144] [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: 11/23/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Venous thromboembolism (VTE) and arterial thrombosis (AT) are distinct yet closely related pathological processes. While traditionally considered separate entities, accumulating evidence suggests that they share common risk factors, such as inflammation and endothelial dysfunction (ED). This review explores the parallels and differences between venous and arterial thrombosis, with particular attention to the role of unprovoked VTE and its potential links to atherosclerosis and systemic inflammation. A key focus is the role of ED, which is emerging as a critical factor in thrombogenesis across both the venous and arterial systems. We examine the current methods for clinically detecting ED, including the use of biomarkers and advanced imaging techniques. Additionally, we discuss novel research avenues, such as the potential of endothelial colony-forming cells and other innovative methodologies, to further unravel the complex mechanisms of thrombosis. Finally, we propose future clinical scenarios where targeting endothelial health could pave the way for more effective prevention and treatment strategies in thrombosis management.
Collapse
Affiliation(s)
- Marco Paolo Donadini
- Department of Medicine and Surgery, Research Center on Thromboembolic Diseases and Antithrombotic Therapies, University of Insubria, 21100 Varese, Italy;
- Centro Trombosi e TAO, Azienda Socio Sanitaria Territoriale dei Sette Laghi, 21100 Varese, Italy;
| | - Francesca Calcaterra
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Erica Romualdi
- Centro Trombosi e TAO, Azienda Socio Sanitaria Territoriale dei Sette Laghi, 21100 Varese, Italy;
| | - Roberta Ciceri
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Assunta Cancellara
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Corrado Lodigiani
- Center for Thrombosis and Hemorrhagic Diseases, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.L.); (M.B.)
| | - Monica Bacci
- Center for Thrombosis and Hemorrhagic Diseases, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.L.); (M.B.)
| | - Silvia Della Bella
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Walter Ageno
- Department of Medicine and Surgery, Research Center on Thromboembolic Diseases and Antithrombotic Therapies, University of Insubria, 21100 Varese, Italy;
- Department of Internal Medicine, Ospedale Regionale di Bellinzona e Valli, 6500 Bellinzona, Switzerland
| | - Domenico Mavilio
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| |
Collapse
|
26
|
Kumar V, S H, Huligowda LKD, Umesh M, Chakraborty P, Thazeem B, Singh AP. Environmental Pollutants as Emerging Concerns for Cardiac Diseases: A Review on Their Impacts on Cardiac Health. Biomedicines 2025; 13:241. [PMID: 39857824 PMCID: PMC11759859 DOI: 10.3390/biomedicines13010241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Comorbidities related to cardiovascular disease (CVD) and environmental pollution have emerged as serious concerns. The exposome concept underscores the cumulative impact of environmental factors, including climate change, air pollution, chemicals like PFAS, and heavy metals, on cardiovascular health. Chronic exposure to these pollutants contributes to inflammation, oxidative stress, and endothelial dysfunction, further exacerbating the global burden of CVDs. Specifically, carbon monoxide (CO), ozone, particulate matter (PM2.5), nitrogen dioxide (NO2), sulfur dioxide (SO2), heavy metals, pesticides, and micro- and nanoplastics have been implicated in cardiovascular morbidity and mortality through various mechanisms. PM2.5 exposure leads to inflammation and metabolic disruptions. Ozone and CO exposure induce oxidative stress and vascular dysfunction. NO2 exposure contributes to cardiac remodeling and acute cardiovascular events, and sulfur dioxide and heavy metals exacerbate oxidative stress and cellular damage. Pesticides and microplastics pose emerging risks linked to inflammation and cardiovascular tissue damage. Monitoring and risk assessment play a crucial role in identifying vulnerable populations and assessing pollutant impacts, considering factors like age, gender, socioeconomic status, and lifestyle disorders. This review explores the impact of cardiovascular disease, discussing risk-assessment methods, intervention strategies, and the challenges clinicians face in addressing pollutant-induced cardiovascular diseases. It calls for stronger regulatory policies, public health interventions, and green urban planning.
Collapse
Affiliation(s)
- Vinay Kumar
- Biomaterials & Tissue Engineering (BITE) Laboratory, Department of Community Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai 602105, Tamil Nadu, India; (V.K.)
| | - Hemavathy S
- Biomaterials & Tissue Engineering (BITE) Laboratory, Department of Community Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai 602105, Tamil Nadu, India; (V.K.)
| | | | - Mridul Umesh
- Department of Life Sciences, Christ University, Hosur Road, Bengaluru 560029, Karnataka, India
| | - Pritha Chakraborty
- Area of Molecular Medicine, Department of Allied Healthcare and Sciences, JAIN (Deemed to be University), Bangalore 560066, Karnataka, India
| | - Basheer Thazeem
- Waste Management Division, Integrated Rural Technology Centre (IRTC), Palakkad 678592, Kerala, India
| | - Anand Prakash Singh
- Frankel Cardiovascular Center, Department of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
Wang HS, Hsu BG, Wang JH, Yang CF. Increased serum galectin-3 level is associated with endothelial dysfunction and cardiovascular events in patients with hypertension. Heliyon 2025; 11:e41111. [PMID: 39758383 PMCID: PMC11699377 DOI: 10.1016/j.heliyon.2024.e41111] [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: 03/28/2024] [Revised: 09/15/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background Endothelial dysfunction can lead to various harmful cardiovascular complications. The importance of galectin-3 (Gal-3) has been proposed in some cardiac diseases related to chronic inflammation. However, its role in hypertension-induced endothelial dysfunction remains unclear. Methods We enrolled 120 patients with hypertension, assessed their baseline characteristics, and monitored their 7-year cardiovascular outcomes. We performed an enzyme-linked immunosorbent assay to measure serum Gal-3 levels. The vascular reactivity index (VRI) was examined by digital thermal monitoring. Patients with VRI <1.0, 1.0 to <2.0, and ≥2.0 were defined as having poor, intermediate, and good vascular reactivity, respectively. Results Among the recruited patients, 12 had poor vascular reactivity, 57 had intermediate vascular reactivity, and 51 had good vascular reactivity. Older age, higher total cholesterol levels, higher low-density lipoprotein cholesterol levels, lower estimated glomerular filtration rate, and higher Gal-3 levels were associated with poor endothelial dysfunction. Multivariate linear regression analysis showed that age and Gal-3 levels were correlated with VRI. During the 7-year follow-up period, patients with higher Gal-3 levels had more cardiovascular events. Conclusions Higher Gal-3 levels are associated with endothelial dysfunction and unfavorable cardiovascular outcomes in patients with hypertension, suggesting its potential role in the hypertension-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Hui-Sheng Wang
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ji-Hung Wang
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Cardiovascular Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chiu-Fen Yang
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Cardiovascular Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| |
Collapse
|
28
|
Rankin S, Fountain C, Gemmell AJ, Quinn D, Henderson A, McClure J, Small S, Venugopal B, McKay P, Slomka PJ, Colville D, Petrie MC, Meléndez GC, Lang NN. Arterial effects of anthracycline: structural and inflammatory assessments in non-human primates and lymphoma patients. Clin Sci (Lond) 2025; 139:29-41. [PMID: 39680089 DOI: 10.1042/cs20241529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/17/2024]
Abstract
Anthracyclines, such as doxorubicin, are important anti-cancer therapies but are associated with arterial injury. Histopathological insights have been limited to small animal models, and the role of inflammation in the arterial toxic effects of anthracycline is unclear in humans. Our aims were (1) to evaluate aortic media fibrosis and injury in non-human primates treated with anthracyclines; (2) to assess the effect of anthracycline on aortic inflammation in patients treated for lymphoma. African Green monkeys (AGMs) received doxorubicin (30-60 mg/m2/biweekly intravenously, cumulative dose: 240 mg/m2). Blinded histopathologic analyses of the ascending aorta were performed 15 weeks after the last doxorubicin dose and compared to five age- and gender-matched healthy, untreated AGMs. Analysis of the thoracic aorta of patients with diffuse large B-cell lymphoma (DLBCL), at baseline and after doxorubicin exposure, was performed using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) in this observational study by maximal tissue-to-background ratio (TBRmax). In AGMs, doxorubicin exposure was associated with greater aortic fibrosis (collagen deposition: doxorubicin 6.23 ± 0.88% vs. controls 4.67 ± 0.54%; P=0.01) and intracellular vacuolization (doxorubicin 66.3 ± 10.1 vs. controls 11.5 ± 4.2 vacuoles/field, P<0.0001) than untreated controls. In 101 patients with DLBCL, there was no change in aortic TBRmax after anthracycline exposure (TBRmax 1.46 ± 0.16 vs. 1.44 ± 0.14, respectively, P=0.14). Univariate analyses yielded similar results. In a large animal model, anthracycline exposure was associated with aortic fibrosis. In patients with lymphoma, anthracycline exposure was not associated with aortic inflammation. Further research is required to elucidate the mechanisms of anthracycline-related vascular harm.
Collapse
MESH Headings
- Animals
- Humans
- Female
- Doxorubicin/adverse effects
- Male
- Middle Aged
- Fibrosis/chemically induced
- Antibiotics, Antineoplastic/adverse effects
- Antibiotics, Antineoplastic/toxicity
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/diagnostic imaging
- Positron Emission Tomography Computed Tomography
- Aged
- Anthracyclines/adverse effects
- Inflammation/pathology
- Inflammation/chemically induced
- Adult
- Aorta, Thoracic/pathology
- Aorta, Thoracic/diagnostic imaging
- Aorta, Thoracic/drug effects
- Aorta/pathology
- Aorta/diagnostic imaging
- Aorta/drug effects
Collapse
Affiliation(s)
- Stephen Rankin
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Caitlin Fountain
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, U.S.A
| | - Alastair J Gemmell
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Daire Quinn
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Alasdair Henderson
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - John McClure
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Sandy Small
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Balaji Venugopal
- School of Medicine, Dentistry and Nursing, University of Glasgow
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Pamela McKay
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Piotr J Slomka
- Cedars-Sinai, Division of Artificial Intelligence in Medicine, Department of Medicine, Los Angeles, U.S.A
| | - David Colville
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Mark C Petrie
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Giselle C Meléndez
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, U.S.A
| | - Ninian N Lang
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| |
Collapse
|
29
|
Azam MNK, Biswas P, Khandker A, Tareq MMI, Tauhida SJ, Shishir TA, Bibi S, Alam MA, Zilani MNH, Albekairi NA, Alshammari A, Rahman MS, Hasan MN. Profiling of antioxidant properties and identification of potential analgesic inhibitory activities of Allophylus villosus and Mycetia sinensis employing in vivo, in vitro, and computational techniques. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118695. [PMID: 39142619 DOI: 10.1016/j.jep.2024.118695] [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: 05/20/2024] [Revised: 07/30/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional use of plants for medicinal purposes, called phytomedicine, has been known to provide relief from pain. In Bangladesh, the Chakma indigenous community has been using Allophylus villosus and Mycetia sinensis to treat various types of pain and inflammation. AIM OF THE STUDY The object of this research is to evaluate the effectiveness of these plants in relieving pain and their antioxidant properties using various approaches such as in vitro, in vivo, and computational techniques. Additionally, the investigation will also analyse the phytochemicals present in these plants. MATERIALS AND METHODS We conducted in vivo analgesic experiment on Swiss albino mice and in-silico inhibitory activities on COX-2 & 15-LOX-2 enzymes. Assessment of DPPH, Anti Radical Activities (ARA), FRAP, H2O2 Free Radical Scavenging, Reducing the power of both plants performed significant % inhibition with tolerable IC50. Qualitative screening of functional groups of phytochemicals was précised by FTIR and GC-MS analysis demonstrated phytochemical investigations. RESULTS The ethyl acetate (EtOAc) fractioned Mycetia sinensis extract as well as the ethanoic extract and all fractioned extracts of Allophylus villosus have reported a significant percentage (%) of writhing inhibition (p < 0.05) with the concentrated doses 250 mg as well as 500 mg among the Swiss albino mice for writhing observation of analgesic effect. In the silico observation, a molecular-docking investigation has performed according to GC-MS generated 43 phyto-compounds of both plants to screen their binding affinity by targeting COX-2 and 15-LOX-2 enzymes. Consequently, in order to assess and ascertain the effectiveness of the sorted phytocompounds, ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) investigation, DFT (Density-functional theory) by QM (Quantum mechanics), and MDS (Molecular dynamics simulation) were carried out. As the outcome, compounds like 5-(2,4-ditert-butylphenoxy)-5-oxopentanoic acid; 2,4-ditert-butylphenyl 5-hydroxypentanoate; 3,3-diphenyl-5-methyl-3H-pyrazole; 2-O-(6-methylheptan-2-yl) 1-O-octyl benzene-1,2-dicarboxylate and dioctan-3-yl benzene-1,2-dicarboxylate derived from the ethnic plant A. villosus and another ethnic plant M. sinensis extracts enchants magnificent analgesic inhibitions and performed more significant drug like activities with the targeted enzymes. CONCLUSIONS Phytocompounds from A. villosus & M. sinensis exhibited potential antagonist activity against human 15-lipoxygenase-2 and cyclooxygenase-2 proteins. The effective ester compounds from these plants performed more potential anti-nociceptive activity which could be used as a drug in future.
Collapse
Affiliation(s)
- Md Nur Kabidul Azam
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh; ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh
| | - Amia Khandker
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh; Biotechnology & Natural Medicine Division, TechB Nutrigenomics, Dhanmondi, Dhaka, 1209, Bangladesh
| | - Md Mohaimenul Islam Tareq
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Sadia Jannat Tauhida
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Tushar Ahmed Shishir
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka, 1212, Bangladesh
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad, 41000, Pakistan
| | - Md Asraful Alam
- School of Chemical Engineering, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Md Nazmul Hasan Zilani
- Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.
| | - Norah A Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Mohammad Shahedur Rahman
- Bioresources Technology & Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Md Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.
| |
Collapse
|
30
|
Murphy GF. White Depressed Areas and Tumor Infiltrating Lymphocytes: The Cancer Cure That Lies Within? J Cutan Pathol 2025. [PMID: 39777741 DOI: 10.1111/cup.14768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 01/11/2025]
Abstract
This brief overview is inspired by seminal contributions by the late Dr. Martin C. Mihm, Jr. who provided a basis for recognition and better understanding of interactions between lymphocytes (tumor-infiltrating lymphocytes [TILs]) that home to and permeate cancers. In primary melanomas, this phenomenon may produce what Dr. Mihm called white depressed areas, prescient clues to what would fuel future attempts at harnessing anticancer immunity. The critical and sequential TIL attributes of antigenic stimulation, homing, and effector-target cell apoptotic injury herein are briefly reviewed in light of more recent advances in the field of immuno-oncology. The intent is to emphasize how fundamental clinical and histopathological observations, as forged by Dr. Mihm and his associates, have led to critically important prognostic paradigms as well as to translational insights that now have become transformative in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- George F Murphy
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital/Mass General Brigham, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Tiwari E, Shrimankar D, Maindarkar M, Bhagawati M, Kaur J, Singh IM, Mantella L, Johri AM, Khanna NN, Singh R, Chaudhary S, Saba L, Al-Maini M, Anand V, Kitas G, Suri JS. Artificial intelligence-based cardiovascular/stroke risk stratification in women affected by autoimmune disorders: a narrative survey. Rheumatol Int 2025; 45:14. [PMID: 39745536 DOI: 10.1007/s00296-024-05756-5] [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: 09/26/2024] [Accepted: 11/20/2024] [Indexed: 01/25/2025]
Abstract
Women are disproportionately affected by chronic autoimmune diseases (AD) like systemic lupus erythematosus (SLE), scleroderma, rheumatoid arthritis (RA), and Sjögren's syndrome. Traditional evaluations often underestimate the associated cardiovascular disease (CVD) and stroke risk in women having AD. Vitamin D deficiency increases susceptibility to these conditions. CVD risk prediction in AD can benefit from surrogate biomarker for coronary artery disease (CAD), such as carotid ultrasound. Due to non-linearity in the CVD risk stratification, we use artificial intelligence-based system using AD biomarkers and carotid ultrasound. Investigate the relationship between AD and CVD/stroke markers including autoantibody-influenced plaque load. Second, to study the surrogate biomarkers for the CAD and gather radiomics-based features such as carotid intima-media thickness (cIMT), and plaque area (PA). Third and final, explore the automated CVD/stroke risk identification using advanced machine learning (ML) and deep learning (DL) paradigms. Analysed biomarker data from women with AD, including carotid ultrasonography imaging, clinical parameters, autoantibody profiles, and vitamin D levels. Proposed artificial intelligence (AI) models to predict CVD/stroke risk accurately in AD for women. There is a strong association between AD duration and elevated cIMT/PA, with increased CVD risk linked to higher rheumatoid factor (RF) and anti-citrullinated peptide antibodies (ACPAs) levels. AI models outperformed conventional methods by integrating imaging data and disorder-specific factors. Interdisciplinary collaboration is crucial for managing CVD/stroke in women with chronic autoimmune diseases. AI-based assisted risk stratification methods may improve treatment decision-making and cardiovascular outcomes.
Collapse
Affiliation(s)
- Ekta Tiwari
- Vishvswarya National Institute of Technology, Nagpur, India
| | | | - Mahesh Maindarkar
- School of Bioengineering and Sciences and Research, MIT Art Design and Technology University, Pune, 4123018, India
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, 95661, USA
| | - Mrinalini Bhagawati
- Department of Biomedical Engineering, North-Eastern Hill University, Shillong, India
| | - Jiah Kaur
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, 95661, USA
| | - Inder M Singh
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, 95661, USA
| | - Laura Mantella
- Allergy, Clinical Immunology and Rheumatology Institute, Toronto, ON, L4Z 4C4, Canada
| | - Amer M Johri
- Division of Cardiology, Department of Medicine, Queen's University, Kingston, Canada
| | - Narendra N Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi, 110001, India
| | - Rajesh Singh
- Department of Research and Innovation, UIT, Uttaranchal University, Dehradun, 248007, India
| | - Sumit Chaudhary
- Department of Research and Innovation, UIT, Uttaranchal University, Dehradun, 248007, India
| | - Luca Saba
- Department of Pathology, Azienda Ospedaliero Universitaria, 09124, Cagliari, Italy
| | - Mustafa Al-Maini
- Allergy, Clinical Immunology and Rheumatology Institute, Toronto, ON, L4Z 4C4, Canada
| | - Vinod Anand
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, 95661, USA
| | - George Kitas
- Academic Affairs, Dudley Group NHS Foundation Trust, Dudley, DY1 2HQ, UK
- Arthritis Research UK Epidemiology Unit, Manchester University, Mancheser, M13 9PL, UK
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, 95661, USA.
- Department of Computer Engineering, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248002, India.
- University Centre for Research & Development, Chandigarh University, Mohali, India.
- Symbiosis Institute of Technology, Nagpur Campus, Symbiosis International (Deemed University), Pune, India.
| |
Collapse
|
32
|
Arachchillage DJ, Mobayen G, Laffan M, Randi AM, Ahnström J, Pericleous C. A cell-based model to study mechanisms of endothelial-dependent thrombin generation in response to inflammation and its modulation by hydroxychloroquine. Res Pract Thromb Haemost 2025; 9:102665. [PMID: 39877523 PMCID: PMC11772944 DOI: 10.1016/j.rpth.2024.102665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/05/2024] [Indexed: 01/31/2025] Open
Abstract
Background Inflammation is a driver of thrombosis, but the phenomenon of thromboinflammation has been defined only recently, bringing together the multiple pathways involved. In vitro models can support the development of new therapeutics targeting the endothelium and also assess the existing immunomodulatory drugs, such as hydroxychloroquine, in modulating the inflammation-driven endothelial prothrombotic phenotype. Objectives To develop a model for thrombin generation (TG) on the surface of human endothelial cells (ECs) to assess pro/antithrombotic properties in response to inflammation. Furthermore, to elucidate the mechanisms of TG regulation and its modulation by immunomodulatory therapies. Methods Cytokine-induced (tumor necrosis factor [TNF]-α, interleukin [IL]-1β, and interferon-γ) effects on ECs isolated from umbilical veins or human aortic tissue were assessed using calibrated automated thrombography in platelet-poor plasma. The expression of key coagulant and inflammatory regulators was measured at the mRNA level. Tissue factor (TF) protein levels were further assessed by flow cytometry. Results Endothelial stimulation with TNF-α or IL-1β caused ECs to trigger TG without the addition of exogenous TF, with higher TG observed after 6 hours of stimulation than 24 hours. IL-1β induced higher peak thrombin (170 ± 5.9 nM vs 115 ± 4.9 nM), endogenous thrombin potential (1632 ± 35 nM ∗ min vs 1370 ± 23 nM ∗ min) presented as mean ± SD, and TF expression (∼2.8-fold higher) compared with TNF-α at 6 hours. Interferon-γ stimulation failed to induce TG and TF expression. The immunomodulatory drug, hydroxychloroquine, reduced cytokine-induced TG and downregulated TF expression. Conclusion We provide detailed optimization of a robust in vitro model to assess the induction of an inflammation-driven endothelial prothrombotic phenotype that is also sensitive to immunomodulatory therapies, providing a tool for investigating mechanisms of disease and new drugs.
Collapse
Affiliation(s)
- Deepa J. Arachchillage
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Golzar Mobayen
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Mike Laffan
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Anna M. Randi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Josefin Ahnström
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Charis Pericleous
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
33
|
Rashki M, Ghasemzadeh Rahbardar M, Boskabady MH. Nutritional Advantages of Walnut ( Juglans regia L.) for Cardiovascular Diseases: A Comprehensive Review. Food Sci Nutr 2025; 13:e4526. [PMID: 39803290 PMCID: PMC11717060 DOI: 10.1002/fsn3.4526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/07/2024] [Accepted: 09/24/2024] [Indexed: 01/16/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain one of the leading causes of morbidity and mortality worldwide. In recent years, the potential role of dietary interventions in preventing and managing CVDs has gained significant attention. Among these dietary components, walnuts (Juglans regia L.) have emerged as a promising candidate due to their unique nutrient profile and potential cardiovascular benefits. This review aims to provide a comprehensive analysis of the existing literature on the role of walnuts in cardiovascular health. Using databases from Scopus, Google Scholar, and PubMed, the most relevant in vitro, in vivo, and clinical trial research has been collected from the time of inception until 2024. Several studies have shown that walnut consumption has a positive effect on a variety of cardiovascular risk factors. Walnut bioactive ingredients, including omega-3 fatty acids, antioxidants, fiber, and polyphenols, have been demonstrated to improve lipid profiles, blood pressure, endothelial function, inflammation, oxidative stress, and thrombosis. These processes all contribute to the possible cardioprotective properties of walnuts. Epidemiological and clinical research indicates that daily walnut consumption can reduce the risk of CVDs like coronary heart disease and stroke. Walnuts may aid in managing CVDs through mechanisms such as enhancing lipid profiles, reducing inflammation, and improving overall cardiovascular function. This review highlights the potential role of walnuts as a dietary strategy for the prevention and management of CVDs. Further understanding of the mechanisms and long-term effects of walnut consumption is crucial for optimizing their therapeutic potential and integrating them into clinical practice. Future research should focus on elucidating specific dose-response relationships and exploring the synergistic effects of walnuts in combination with other dietary and lifestyle interventions.
Collapse
Affiliation(s)
- Mostafa Rashki
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| | | | - Mohammad Hossein Boskabady
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Physiology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
34
|
Kong D, Ryu JC, Shin N, Lee SE, Kim NG, Kim HY, Kim MJ, Choi J, Kim DH, Kang KS. In Vitro Modeling of Atherosclerosis Using iPSC-Derived Blood Vessel Organoids. Adv Healthc Mater 2025; 14:e2400919. [PMID: 39580678 DOI: 10.1002/adhm.202400919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 11/07/2024] [Indexed: 11/26/2024]
Abstract
As modeling of atherosclerosis requires recapitulating complex interactions with vasculature and immune cells, previous in vitro models have limitations due to their insufficient 3D vascular structures. However, induced pluripotent stem cell-derived blood vessel organoids (BVOs) are applicable for modeling vascular diseases, containing multiple cell types, including endothelial and vascular smooth muscle cells self-assembled into a blood vessel structure. Atherosclerotic BVOs with a microenvironment associated with atherogenesis, such as shear stress, low-density lipoprotein, pro-inflammatory cytokine, and monocyte co-culture are successfully developed. In atherosclerotic BVOs, representative atherosclerotic phenotypes, including endothelial dysfunction, inflammatory responses, formation of foam cells and fibrous plaque, and moreover, calcification of the plaques are observed. To verify the drug response in this model, it is treated with clinically used lovastatin and confirm phenotype attenuation. Furthermore, the therapeutic efficacy of nano-sized graphene oxides (NGOs) is evaluated on atherosclerosis. Due to their anti-inflammatory effects, NGOs effectively alleviate the pathologic lesions in atherosclerotic BVOs by promoting macrophage polarization toward M2. These results suggest that atherosclerotic BVOs are advanced in vitro models suitable for drug discovery and elucidation of therapeutic mechanisms. From the perspective of precision medicine, this platform using patient-derived BVOs can be further employed for personalized drug screening in the future.
Collapse
Affiliation(s)
- Dasom Kong
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae-Chul Ryu
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Bio and Nano Convergence, Biogo Co., LTD, Seoul, 08826, Republic of Korea
| | - Nari Shin
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung-Eun Lee
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nam Gyo Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee-Yeong Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min-Ji Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jungju Choi
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Da-Hyun Kim
- Department of Biotechnology, Sungshin Women's University, Seoul, 01133, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
35
|
Ordashev KA, Milojevic Samanovic A, Omarov IA, Zivkovic V, Nikolic Turnic T, Novakovic J, Stojanovic A, Nikolic M, Pindovic B, Sretenovic J, Fisenko VP, Bolevich S, Bolevich S, Jakovljevic V. Biochemical, pathohistological, radiographic and cardiological analysis reveals the possible association between apical periodontitis and cardiac function in diabetic rats. Arch Oral Biol 2025; 169:106089. [PMID: 39255649 DOI: 10.1016/j.archoralbio.2024.106089] [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: 05/01/2024] [Revised: 08/10/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVE To evaluate the possible effects of apical periodontitis (AP) on cardiac function, structure, and oxidative stress (OS) in rats with diabetes mellitus type 2 (T2DM). DESIGN Forty-eight (Wistar albino, male) rats were randomized into four groups: control healthy (CTRL), normoglycemic with AP (AP), T2DM, and T2DM with AP (T2DM+AP). T2DM was induced by streptozotocin and a high-fat diet. AP was induced by pulp exposure to the oral environment for 4 weeks and analyzed radiographically. In the blood samples insulin and glucose were established. In vivo, cardiac function was evaluated by echocardiography. Ex vivo cardiac function was assessed by the Langendorff technique. Heart tissue was analyzed pathophysiologically. OS was determined in cardiac tissue homogenate and coronary venous effluent, spectrophotometrically. RESULTS Impaired glycoregulation was observed in the T2DM+AP group compared to the T2DM, AP, and CTRL groups. The T2DM+AP group was associated with disturbed echocardiography and cardiodynamic parameters. The levels of superoxide anion radical, nitrite, and index of lipid peroxidation were significantly increased, while the superoxide dismutase and catalase were significantly decreased in the T2DM+AP group compared to T2DM, AP, and CTRL groups. The radiographic AP area was significantly larger in the T2DM+AP compared to the AP group. CONCLUSION AP was associated with increased glucose levels, impaired cardiac function, structure, and OS in diabetic rats. Diabetes was related to an increased radiographic AP area. The study may be a starting point for further research to clarify the effects of AP on cardiac function in various models of systemic diseases.
Collapse
Affiliation(s)
- Khasan Alievich Ordashev
- Department of Surgical Dentistry and Maxillofacial Surgery, Dagestan State Medical, University of the Ministry of Health, 47 Lyakhova St., 367000 Mahachkala, Dagestan Republic, Russian Federation
| | - Andjela Milojevic Samanovic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia.
| | - Israpil Alisultanovich Omarov
- Medical and Health Center of the Ministry of Foreign Affairs of Russia, 32/34 Smolenskaya-Sennaya St., 119200 Moscow, Russian Federation
| | - Vladimir Zivkovic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991 Moscow, Russian Federation
| | - Tamara Nikolic Turnic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; N.A. Semashko Public Health and Healthcare Department, F.F. Erisman Institute of Public Health, I.M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991 Moscow, Russian Federation
| | - Jovana Novakovic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia
| | - Aleksandra Stojanovic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia
| | - Marina Nikolic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia
| | - Bozidar Pindovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia
| | - Jasmina Sretenovic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia
| | - Vladimir Petrovich Fisenko
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991 Moscow, Russian Federation
| | - Sergey Bolevich
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991 Moscow, Russian Federation
| | - Stefani Bolevich
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991 Moscow, Russian Federation; Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991 Moscow, Russian Federation
| | - Vladimir Jakovljevic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica St., 34000 Kragujevac, Serbia; Department of Human Pathology, I.M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991 Moscow, Russian Federation
| |
Collapse
|
36
|
Brinkley L, Brock MA, Stinson G, Bilgili A, Jacobs JP, Bleiweis M, Peek GJ. The biological role and future therapeutic uses of nitric oxide in extracorporeal membrane oxygenation, a narrative review. Perfusion 2025; 40:83-91. [PMID: 38226651 DOI: 10.1177/02676591241228169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
BACKGROUND Nitric oxide (NO) is a gas naturally produced by the human body that plays an important physiological role. Specifically, it binds guanylyl cyclase to induce smooth muscle relaxation. NO's other protective functions have been well documented, particularly its protective endothelial functions, effects on decreasing pulmonary vascular resistance, antiplatelet, and anticoagulation properties. The use of nitric oxide donors as vasodilators has been known since 1876. Inhaled nitric oxide has been used as a pulmonary vasodilator and to improve ventilation perfusion matching since the 1990s. It is currently approved by the United States Food and Drug Administration for neonates with hypoxic respiratory failure, however, it is used off-label for acute respiratory distress syndrome, acute bronchiolitis, and COVID-19. PURPOSE In this article we review the currently understood biological action and therapeutic uses of NO through nitric oxide donors such as inhaled nitric oxide. We will then explore recent studies describing use of NO in cardiopulmonary bypass and extracorporeal membrane oxygenation and speculate on NO's future uses.
Collapse
|
37
|
Fan Z, Yuan X, Yuan Y. Circular RNAs in coronary heart disease: From molecular mechanism to promising clinical application (Review). Int J Mol Med 2025; 55:11. [PMID: 39513584 PMCID: PMC11573316 DOI: 10.3892/ijmm.2024.5452] [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: 08/06/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Coronary heart disease (CHD) remains a leading cause of morbidity and mortality worldwide, posing a substantial public health burden. Despite advancements in treatment, the complex etiology of CHD necessitates ongoing exploration of novel diagnostic markers and therapeutic targets. Circular RNAs (circRNAs), a distinct class of non‑coding RNAs with a covalently closed loop structure, have emerged as significant regulators in various diseases, including CHD. Their high stability, tissue‑specific expression and evolutionary conservation underscore their potential as biomarkers and therapeutic agents in CHD. This review discusses the current knowledge on circRNAs in the context of CHD and explores the molecular mechanisms by which circRNAs influence the pathophysiology of CHD, including cardiomyocyte death, endothelial injury, vascular dysfunction and inflammation. It also summarizes the emerging evidence highlighting the differential expression of circRNAs in patients with CHD and their potential utilities as non‑invasive diagnostic and prognostic biomarkers and therapeutic targets for this disease.
Collapse
Affiliation(s)
- Zengguang Fan
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330006, P.R. China
| | - Xingxing Yuan
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, Heilongjiang 150006, P.R. China
| | - Ye Yuan
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
38
|
Liu S, Wu J, Banerjee O, Xue B, Shi H, Ding Z. Big data analytics and scRNA-seq in human aortic aneurysms and dissections: role of endothelial MerTK. Theranostics 2025; 15:202-215. [PMID: 39744231 PMCID: PMC11667232 DOI: 10.7150/thno.103851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 10/30/2024] [Indexed: 01/25/2025] Open
Abstract
Rationale: Aortic aneurysms and dissections (AAD) cause more than 10,000 deaths in the United States each year. However, there are no medications that can effectively prevent the pathogenesis of AAD. MER proto-oncogene tyrosine kinase (MerTK) is a key receptor for efferocytosis, a process for the clearance of apoptotic cells. Here, we mainly focused on ascending aortic aneurysms and dissections (AAAD) and investigated the role of endothelial MerTK in AAAD progression. Methods: Single-cell RNA sequencing (scRNA-seq) analysis in human AAAD samples and RNA-seq big data analytics, combined with our unique MerTKflox/flox/Tie2Cre mouse model with MerTK deficiency in endothelial cells (ECs), were applied to define the role of endothelial MerTK in AAAD. Results: Through comparative analyses of scRNA-seq in human AAAD (communications of ECs with other cells) and comprehensive big data analytics including about 600,000 cross analyses, we found that the expression of endothelial MerTK is significantly inhibited in human AAAD, resulting in decreased ability of ECs to engulf antigen presenting cells, phagocytes, leukocytes, blood cells and myeloid cells. Our in vivo data showed a significantly higher incidence of AAAD in MerTK flox/flox/Tie2Cre mice compared to that of their littermate controls of MerTK flox/flox mice (100% vs. 11.1%). MerTK deficiency in ECs induces both endothelial dysfunction and SMC phenotypic alterations, subsequently promoting AAAD development. Conclusions: Our findings indicate that endothelial MerTK impairment and subsequent endothelial dysfunction and SMC phenotypic alterations represent novel mechanisms promoting AAAD.
Collapse
Affiliation(s)
| | | | | | | | | | - Zufeng Ding
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| |
Collapse
|
39
|
Zhu M, Mao X, Huang X, Gan M, Zhang K, Chen Y. Novel Serum Markers that Distinguish Behcet's Disease from Idiopathic Recurrent Aphthous Stomatitis. Immunol Invest 2025; 54:1-17. [PMID: 39356129 DOI: 10.1080/08820139.2024.2410743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
BACKGROUND Behcet's disease (BD) is a rare and recurrent autoinflammatory disorder characterized by systemic vasculitis, frequently manifested as recurrent aphthous stomatitis (RAS). We aim to identify specific serum proteins to discriminate between BD and idiopathicRAS. METHOD Peripheral blood was collected from 12 BD patients, 12 idiopathic RAS patients, and 21 healthy volunteers. The serum samples underwent Tandem Mass Tag-based mass spectrometry analysis. Differentially expressed proteins (DEPs) were identified for KEGG pathway enrichment, Gene Ontology (GO), and protein-protein interaction (PPI) analyses. ELISA was utilized to verify two BD-specific DEPs in another cohort consisting of 18 BD patients, 18 idiopathic RAS patients, and 18 controls. RESULTS Compared with RAS serum, BD serum showed 242 DEPs. 49 proteins were differentially expressed in BD but not RAS serum compared to healthy controls. KEGG pathway and GO analyses revealed that DEPs in BD and RAS have similar biological functions and cellular distributions, featuring a significant association with pathways regulating blood coagulation and immune response. When comparing DEPs between BD and RAS, several keratins emerged as markers that distinguish RAS from BD. We also identified multiple DEPs in BD but not RAS patients. PPI analysis uncovered that lipoprotein metabolism regulators serve as hub proteins, indicating their potentially essential roles in BD pathology. In addition, ELISA results confirmed the elevated LRG1 and SOD3 levels in BD, but not RAS patients, compared to healthy donors. CONCLUSION Our data uncovered novel serum proteins that distinguish BD from RAS, which may potentially be useful in BD diagnosis and treatment.
Collapse
Affiliation(s)
- Mengya Zhu
- Department of Rheumatology and Immunology, Ningbo No.2 hospital, Ningbo, China
| | - Xinliang Mao
- Emergency Department, Ningbo No.2 hospital, Ningbo, China
| | - Xianqian Huang
- Department of Rheumatology and Immunology, Ningbo No.2 hospital, Ningbo, China
| | - Minzhi Gan
- Department of Rheumatology and Immunology, Ningbo No.2 hospital, Ningbo, China
| | - Keyue Zhang
- Department of Rheumatology and Immunology, Ningbo No.2 hospital, Ningbo, China
| | - Yong Chen
- Department of Rheumatology and Immunology, Ningbo No.2 hospital, Ningbo, China
| |
Collapse
|
40
|
Zhang M, Sun Y, Zhang L, Xu Y, Liu Y, Li K. The application of mass defect percentage in the evaluation of acute coronary syndrome. Nucl Med Commun 2025; 46:1-6. [PMID: 39363632 PMCID: PMC11634191 DOI: 10.1097/mnm.0000000000001907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024]
Abstract
OBJECTIVES White blood cells, neutrophils, lymphocytes, and neutrophil-to-lymphocyte ratio (NLR) distribution patterns in patients with anatomic coronary disease have previously been associated with cardiac events such as myocardial infarct size, complications, and prognosis. However, it remains unknown whether myocardial perfusion mass defect percentage (MDP) obtained from gated myocardial perfusion imaging (G-MPI) correlates with these hematological parameters. Therefore, our research aimed to investigate the application of MDP in the evaluation of acute coronary syndrome (ACS). METHODS Thirty-six patients with ACS underwent single-photon emission computed tomography/computed tomography using retrospective electrocardiography gating during the resting state. The primary outcome was the percentage of left ventricular mass with abnormal myocardial perfusion (i.e. MDP) in G-MPI. Furthermore, the correlation between myocardial perfusion MDP and lymphocyte count, neutrophil count, white blood cell count, and NLR was calculated. In addition, we explored the relationship of myocardial perfusion MDP with other cardiac function parameters obtained from G-MPI, such as summed rest score, left ventricular ejection fraction, end-systolic volume, and end-diastolic volume. RESULTS Myocardial perfusion MDP significantly correlated with white blood cell count, neutrophil count, and NLR ( P < 0.01). Furthermore, these hematological parameters were significantly different between low and high MDP groups. Additionally, myocardial perfusion MDP negatively correlated with end-systolic volume ( r = -0.615) and left ventricular ejection fraction ( r = -0.657). CONCLUSION Myocardial perfusion MDP has a high correlation with inflammatory cell counts and cardiac function parameters obtained from G-MPI in ACS; this may be of help in the evaluation and treatment of these patients.
Collapse
Affiliation(s)
- Man Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, China
| | - Yinuo Sun
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, China
| | - Limeng Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, China
| | - Yu Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, China
| | - Yifan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, China
| | - Kun Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, China
| |
Collapse
|
41
|
Mascarenhas D, Mohammadi A, Higazy R, Ivanovska J, Gauda E, Jasani B. L-Citrulline in Neonates: From Bench to Bed Side. CHILDREN (BASEL, SWITZERLAND) 2024; 12:42. [PMID: 39857873 PMCID: PMC11763423 DOI: 10.3390/children12010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025]
Abstract
L-citrulline (L-CIT), a precursor to L-arginine (L-ARG), is a key contributor to the nitric oxide (NO) signaling pathway. Endothelial dysfunction, characterized by deficient nitric oxide synthesis, is implicated in the pathogenesis of various neonatal conditions such as necrotizing enterocolitis (NEC) and bronchopulmonary dysplasia (BPD) associated pulmonary hypertension (PH). This review summarizes the current evidence around the possible role of L-CIT supplementation in the treatment of these conditions. Detoxification of endogenously produced superoxide radicals is inadequate in preterm infants due to immature antioxidants that leads to the production of peroxynitrite, a reactive oxygen-free radical that is cytotoxic and causes damage to organelles and cellular membranes, further disrupting the coupling of endothelial NO synthase enzyme and the generation of high levels of reactive nitrogen and oxygen species. Animal studies in lipopolysaccharide-induced models of chorioamnionitis and hyperoxia- and inflammation-induced BPD-PH in rodent lung models revealed that L-CIT supplementation significantly mitigated structural changes in the pulmonary vasculature, preserved alveolar growth, and increased vascular endothelial growth factor gene expression, highlighting the anti-inflammatory and antioxidant effects of L-CIT supplementation. Similar benefits were noted in newborn piglet models of chronic hypoxia-induced PH and NEC. Pharmacokinetic studies in neonates have shown doses of 100-300 mg/kg/day to be safe and well tolerated. A few studies have shown the beneficial effects of L-CIT supplementation in pulmonary hypertension secondary to congenital heart disease, but evidence of efficacy in the neonatal population is lacking. While L-CIT shows promise in the treatment of various neonatal conditions, adequately powered studies to evaluate the safety and efficacy of L-CIT supplementation post-surgical NEC and BPD ± PH in the extremely preterm population are needed to translate this novel therapy to clinical practice.
Collapse
Affiliation(s)
- Dwayne Mascarenhas
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (D.M.); (E.G.)
| | - Atefeh Mohammadi
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 1E8, Canada; (A.M.); (J.I.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 3K3, Canada;
| | - Randa Higazy
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 3K3, Canada;
| | - Julijana Ivanovska
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 1E8, Canada; (A.M.); (J.I.)
| | - Estelle Gauda
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (D.M.); (E.G.)
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 1E8, Canada; (A.M.); (J.I.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 3K3, Canada;
| | - Bonny Jasani
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (D.M.); (E.G.)
| |
Collapse
|
42
|
Kiatsoonthon K, Phimthong N, Potikanond S, Wikan N, Nimlamool W. Panduratin A Inhibits TNF Alpha-Stimulated Endothelial Cell Activation Through Suppressing the NF-κB Pathway. Biomolecules 2024; 15:34. [PMID: 39858429 PMCID: PMC11762725 DOI: 10.3390/biom15010034] [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: 11/28/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Upon exposure to inflammatory stimuli including TNF-α, endothelial cells are activated leading to the adhesion of monocytes to their surface. These events are involved in the pathophysiology of atherosclerosis. Since TNF-α activates the NF-κB pathway, which contributes to atherosclerosis, targeting this signaling pathway may help prevent the risk of developing the disease. The current study elucidated the inhibitory effect of panduratin A (PA) on TNF-α-induced endothelial activation and monocyte adhesion. We discovered that PA reduced the level of pro-inflammatory cytokine IL-6 and chemokine MCP-1 in the media collected from endothelial cells stimulated with TNF-α. In addition, PA inhibited the expression of ICAM-1 and VCAM-1 on the surface of TNF-α-induced endothelial cells resulting in a decrease in the number of monocytes attached to endothelial cell surface. Mechanistically, PA prevented IκB degradation and specifically suppressed NF-κB phosphorylation and nuclear translocation in endothelial cells. However, PA had no inhibitory effect on the phosphorylation of AKT, ERK1/2, p38, and JNK. Taken together, PA blocked the production of cytokine and chemokine, adhesion molecules, and monocyte adhesion in response to TNF-α stimulation, in part, through NF-κB inhibition. Our study suggests that PA may possibly be effective in blocking the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Kriangkrai Kiatsoonthon
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Nitchakarn Phimthong
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
- PhD’s Degree Program in Pharmacology, Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| |
Collapse
|
43
|
Mahmood NMS, Mahmud AMR, Maulood IM. Melatonin attenuates responses to angiotensin II in isolated aortic rings of STZ-induced type 1-like DM rats. Endocr Res 2024:1-13. [PMID: 39719865 DOI: 10.1080/07435800.2024.2445264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/18/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND In patients with diabetes mellitus (DM), vascular endothelial dysfunction (VED) is the main reason for impaired life expectancy. Melatonin (MEL) demonstrates wide-ranging effects across various organs and exhibits pleiotropic characteristics. The current study aims to investigate the modulatory roles of MEL vascular response to angiotensin II (Ang II) and its receptors including angiotensin type 1 receptor (AT-1 R) and angiotensin type 2 receptor (AT-2 R) in isolated thoracic aorta of non-diabetes (non-DM) and diabetes (DM) rats. METHODS The thoracic aortae were isolated in order to investigate the influence of MEL on AT-1 R, using valsartan (VAL) and MT-2Rusing luzindole (LUZ) via dose-response curve (DRC) measurement of Ang II reactivity. In addition, AT-1 R was involved in this study, under PD123319 with ADInstrument organ bath (Panlab apparatus, Harvard University, USA). RESULTS The maximum response of Ang II was increased significantly in DM condition. In addition, AT-1 R was completely blocked under VAL, while AT-2 R was upregulated in the DM group. The combination of VAL and PD123319 led to abolishing the Ang II effect dramatically as well. Melatonin alone reduced Ang II in the DM group dramatically. This effect was also observed with MEL, PD1213319, and VAL combination, as well as, with MEL, LUZ, and PD1213319 combination. CONCLUSIONS Melatonin has been demonstrated to modulate both AT-1 R and AT-2 R and has influenced the reactivity of Ang II in the aortas of diabetic rats through highly complex mechanisms.
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
|
44
|
Skrobucha A, Pindlowski P, Krajewska N, Grabowski M, Jonik S. Anti-inflammatory effects of glucagon-like peptide-1 (GLP-1) in coronary artery disease: a comprehensive review. Front Cardiovasc Med 2024; 11:1446468. [PMID: 39741663 PMCID: PMC11685754 DOI: 10.3389/fcvm.2024.1446468] [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: 06/09/2024] [Accepted: 11/21/2024] [Indexed: 01/03/2025] Open
Abstract
Coronary artery disease (CAD)-cardiovascular condition occuring due to atherosclerotic plaque accumulation in the epicardial arteries-is responsible for disabilities of millions of people worldwide and remains the most common single cause of death. Inflammation is the primary pathological mechanism underlying CAD, since is involved in atherosclerotic plaque formation. Glucagon-like peptide-1 (GLP-1) is a peptide hormone which role extends beyond well-known carbohydrates metabolism. In in vitro studies GLP-1 receptor agonism is associated with regulation of several inflammatory pathways, including cytokine production, lypotoxicity and macrophages differentiation. In this review, we aimed to provide a comprehensive summary of the potential relationship between anti-inflammatory effects of GLP-1 and CAD. We have described a well-established association of anti-inflammatory properties of GLP-1 and atherosclerosis in animals. Pre-clinical studies showed that anti-atherogenic effect of GLP-1 is independent of modulation of plasma lipid levels and depends on anti-inflammatory response. Human studies in this area are limited by small sample size and often nonrandomized character. However, beneficial impact of GLP-1 on endothelial function and microcirculatory integrity in patients with CAD have been described. Understanding atherosclerosis as a chronic inflammatory disease offers new opportunities for the prevention and treatment of CAD. Therefore, we emphasize the need for larger randomized controlled trials focusing on cardiovascular morbidity and mortality to verify the cardioprotective properties of GLP-1R agonists in patients with CAD.
Collapse
Affiliation(s)
- Alicja Skrobucha
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | | | | | | | | |
Collapse
|
45
|
Begum M, Choubey M, Tirumalasetty MB, Arbee S, Sadik S, Mohib MM, Srivastava S, Minhaz N, Alam R, Mohiuddin MS. Exploring the Molecular Link Between Diabetes and Erectile Dysfunction Through Single-Cell Transcriptome Analysis. Genes (Basel) 2024; 15:1596. [PMID: 39766863 PMCID: PMC11675191 DOI: 10.3390/genes15121596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Erectile dysfunction (ED) is a pathophysiological condition in which the patients cannot achieve an erection during sexual activity, and it is often overlooked yet prevalent among diabetic men, globally affecting approximately 35-75% of diabetic individuals. The precise mechanisms through which diabetes contributes to ED remain elusive, but the existing literature suggests the potential involvement of nerve and vascular damage that affects the penile supply. In the present review, we reanalyze the existing human single-cell transcriptomic data from patients having diabetes mellitus-associated ED with normal erections. The analysis validates the expression of genes associated with antioxidative pathways, growth factors, adipokines, angiogenesis, vascular functions, penile erection, sexual function, and inflammation in diverse cell types from healthy individuals and those with ED. Our transcriptomic analysis reveals alterations in the expression of adiponectin receptors in the pathogenesis of ED compared to their counterparts in healthy subjects. This comprehensive review sheds light on the molecular underpinnings of ED in the context of diabetes, providing an in-depth understanding of the biological and cellular alterations involved and paving the way for possible targeted therapeutic discoveries in the field of diabetes-associated male infertility.
Collapse
Affiliation(s)
- Mahmuda Begum
- Department of Internal Medicine, HCA-St. David’s Medical Center, 919 E 32nd St, Austin, TX 78705, USA;
| | - Mayank Choubey
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA or (M.C.); (M.B.T.)
| | - Munichandra Babu Tirumalasetty
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA or (M.C.); (M.B.T.)
| | - Shahida Arbee
- Institute for Molecular Medicine, Aichi Medical University, 1-Yazako, Karimata, Aichi, Nagakute 480-1103, Japan;
| | - Sibly Sadik
- National Institute of Preventive and Social Medicine (NIPSOM), Mohakhali, Dhaka 1212, Bangladesh;
| | - Mohammad Mohabbulla Mohib
- Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Magdeburger Straße 6, 06112 Halle, Germany;
| | | | - Naofel Minhaz
- PGY1, Family Medicine, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada;
| | - Riffat Alam
- Alberta Hospital Edmonton, 17480 Fort Rd NW, Edmonton, AB T5Y 6A8, Canada;
| | - Mohammad Sarif Mohiuddin
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA or (M.C.); (M.B.T.)
| |
Collapse
|
46
|
Gwozdzinski L, Pieniazek A, Gwozdzinski K. The Roles of Oxidative Stress and Red Blood Cells in the Pathology of the Varicose Vein. Int J Mol Sci 2024; 25:13400. [PMID: 39769165 PMCID: PMC11678264 DOI: 10.3390/ijms252413400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
This review discusses sources of reactive oxygen species, enzymatic antioxidant systems, and low molecular weight antioxidants. We present the pathology of varicose veins (VVs), including factors such as hypoxia, inflammation, dysfunctional endothelial cells, risk factors in varicose veins, the role of RBCs in venous thrombus formation, the influence of reactive oxygen species (ROS) and RBCs on VV pathology, and the role of hemoglobin in the damage of particles and macromolecules in VVs. This review discusses the production of ROS, enzymatic and nonenzymatic antioxidants, the pathogenesis of varicose veins as a pathology based on hypoxia, inflammation, and oxidative stress, as well as the participation of red blood cells in the pathology of varicose veins.
Collapse
Affiliation(s)
- Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (A.P.); (K.G.)
| | - Krzysztof Gwozdzinski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (A.P.); (K.G.)
| |
Collapse
|
47
|
Kopaliani I, Elsaid B, Speier S, Deussen A. Immune and Metabolic Mechanisms of Endothelial Dysfunction. Int J Mol Sci 2024; 25:13337. [PMID: 39769104 PMCID: PMC11728141 DOI: 10.3390/ijms252413337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Endothelial dysfunction is a strong prognostic factor in predicting the development of cardiovascular diseases. Dysfunctional endothelium loses its homeostatic ability to regulate vascular tone and prevent overactivation of inflammation, leading to vascular dysfunction. These functions are critical for vascular homeostasis and arterial pressure control, the disruption of which may lead to hypertension. Hypertension itself can also cause endothelial dysfunction, as endothelial cells are susceptible to haemodynamic changes. Although it is unclear which of those factors appear first, they create a vicious circle further damaging multiple organs, including the heart and vessels. There are also sex-specific differences in homeostatic functions of the endothelium regarding vessel tone regulation, which may contribute to differences in arterial blood pressure between men and women. Even more importantly, there are sex-differences in the development of endothelial dysfunction and vessel remodelling. Hence, an understanding of the mechanisms of endothelial dysfunction and its contribution to pathological vascular remodelling during hypertension is of critical importance. This review addresses immunological and metabolic aspects in mechanisms of endothelial dysfunction and the resulting mechanisms in vascular remodelling with respect to arterial hypertension, including the potential role of sex-specific differences.
Collapse
Affiliation(s)
- Irakli Kopaliani
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| | - Basant Elsaid
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo 1181, Egypt
| | - Stephan Speier
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Andreas Deussen
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| |
Collapse
|
48
|
Garcia VP, Stockelman KA, Levy MV, Fandl HK, Goulding A, Hijmans JG, Ruzzene ST, Berry AR, Greiner JJ, DeSouza CA. Microvesicles Derived from Nitric Oxide Synthase-Inhibited Endothelial Cells Promote Cell Dysfunction. J Vasc Res 2024; 62:10-21. [PMID: 39657607 DOI: 10.1159/000542280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION The aims of this study were to determine (1) whether endothelial nitric oxide synthase (eNOS) inhibition stimulates endothelial microvesicles (EMVs) release and (2) the effect of EMVs derived from eNOS-inhibited cells on endothelial cell eNOS, inflammation, apoptosis, and tissue-type plasminogen activator (t-PA). METHODS Human umbilical vein endothelial cells (HUVECs) were treated with the eNOS inhibitor (NG-nitro-l-arginine methyl ester [L-NAME], 300 µM) for 24 h. EMVs from untreated and L-NAME-treated cells were isolated, quantified, and exposed to HUVECs for 24 h. RESULTS eNOS-inhibited cells released significantly higher EMVs than untreated cells (81 ± 13 vs. 41 ± 15 EMV/μL; p = 0.005). Expression of total eNOS (97.1 ± 16.4 vs. 157.5 ± 31.2 arbitrary units [AUs]; p = 0.01), p-eNOS (4.9 ± 1.2 vs. 9.1 ± 12.6 AUs; p = 0.02), and NO production (5.0 ± 0.8 vs. 7.0 ± 1.3 µmol/L; p = 0.04) were significantly lower in cells treated with EMVs from L-NAME-treated cells. L-NAME-derived EMVs induced significantly higher IL-6 (38.3 ± 10.3 vs. 21.0 ± 3.8 pg/mL; p = 0.01) and IL-8 (38.9 ± 7.0 vs. 27.2 ± 6.2 pg/mL; p = 0.04) production concurrent with higher expression of p-NF-κB p65 (Ser536) (9.7 ± 1.6 vs. 6.1 ± 1.2 AUs; p = 0.01). Expression of activated caspase-3 was higher (9.5 ± 1.1 vs. 6.4 ± 0.4 AUs) and t-PA lower (24.2 ± 4.3 vs. 36.2 ± 8.4 AUs; p = 0.04) in cells treated with L-NAME-derived EMVs. CONCLUSION eNOS inhibition induces an increase in EMV release and an EMV phenotype with adverse cellular effects.
Collapse
Affiliation(s)
- Vinicius P Garcia
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Kelly A Stockelman
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Ma'ayan V Levy
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Hannah K Fandl
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Anabel Goulding
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Jamie G Hijmans
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Samuel T Ruzzene
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Auburn R Berry
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Jared J Greiner
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Christopher A DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
49
|
Bell DA, Miller CM, Sullivan R. A continuous mode of action of nitric oxide in hard-to-heal wound healing. J Wound Care 2024; 33:912-925. [PMID: 39630551 DOI: 10.12968/jowc.2024.0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Nitric oxide (NO) is one of the most studied molecules in medical science. The role of NO as an endogenous regulator of inflammation, as an antibacterial agent and as an endogenous gasotransmitter is well established. Even so, despite a plethora of excellent wound healing data, hard-to-heal (chronic) wounds are of epidemic proportions, and still growing in number. However, yet to be established and sorely needed is the identification of a single, continuous NO mechanism of action (MoA), where phase-to-phase variance in the complex sequence of cellular and molecular wound healing may elucidate the potential for placing hard-to-heal wounds on positive healing trajectories. Hence, the objectives of this review were to: identify salient MoAs for NO in each phase of skin wound healing; and to select and validate a single MoA that is both ubiquitous and continuous in NO across acute and hard-to-heal wound sequences, and which potentiates the ability to supplementally motivate and guide the recovery of a hard-to-heal wound onto a positive healing trajectory. The search began by selecting a detailed, multipart wound healing model. Next, as guided by the literature, was the identification of salient NO functionalities for each model segment. These modes of action were then be used to identify and validate a single NO MoA that is continuous across the healing spectrum. Finally, by using the principle of 'super position' of two continuous functions, this acute healing NO MoA solution was compared to a similar solution set describing a hard-to-heal or chronic wound. As both solution sets are continuous in a NO function, the resultant 'overlay' then helped to identify and guide the use of a NO MoA capable of placing any hard-to-heal wound on a positive healing trajectory.
Collapse
|
50
|
Doro L, Peana AT, Migheli R, Capobianco G, Criscione M, Montella A, Campesi I. Effect of (R)-(-)-Linalool on endothelial damage: Sex differences. Biochem Biophys Rep 2024; 40:101846. [PMID: 39483177 PMCID: PMC11525626 DOI: 10.1016/j.bbrep.2024.101846] [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: 08/28/2024] [Revised: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
Oxidative stress and inflammation are responsible for endothelial damage displaying many sex differences. Lipopolysaccharide (LPS) is a pathogenic stimulus that can trigger inflammation, contributing to endothelial dysfunction. Given the scientific evidence on the effectiveness of herbal extracts in managing endothelial dysfunction, we considered the (R)-(-)-Linalool (LIN), an aromatic monoterpene alcohol, as a bioactive phytochemical compound that could prevent and improve endothelial injury. In this study, we evaluated the effect of the LIN on LPS-induced damage in female and male human umbilical vein endothelial cells (FHUVECs and MHUVECs), measuring cell viability, cytokines release (IL-6 and TNF-α), malondialdehyde (MDA), and nitrites. LPS significantly reduced viability both in MHUVECs and FHUVECs. Moreover, LPS increased the IL-6, TNF-α, and MDA level only in FHUVECs if compared to basal value; despite that, LPS reduced nitrites only in MHUVECs. LIN alone did not affect the parameters measured except for an increase in nitrites in FHUVECs. Nevertheless, LIN reduced damage and restored endothelium viability reduced by LPS without a clear sex difference. Under LPS, LIN inhibited IL-6 release and reduced MDA levels only in FHUVECs. The present data confirm the existence of sex differences in the behavior of HUVECs under LPS conditions. The administration of LIN seems to have a more evident effect on FHUVECs after damage induced by LPS. These LIN effects are important to conduct further well-designed studies on the sex-specific use of this compound on vascular endothelial injury.
Collapse
Affiliation(s)
- Laura Doro
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Alessandra T. Peana
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Rossana Migheli
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Giampiero Capobianco
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
- Gynecologic and Obstetric Clinic, AOU, Viale San Pietro 12, 07100, Sassari, Italy
| | - Massimo Criscione
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| |
Collapse
|