1
|
Han XX, Zhang HY, Kong JW, Liu YX, Zhang KR, Ren WY. Inflammatory index is a promising biomarker for maintenance hemodialysis patients with cardiovascular disease. Eur J Med Res 2024; 29:544. [PMID: 39533444 PMCID: PMC11559046 DOI: 10.1186/s40001-024-02117-7] [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: 07/12/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Maintenance hemodialysis (MHD) patients are at a higher risk of cardiovascular disease (CVD), a common complication and leading cause of death. Persistent micro-inflammation is a unique feature of MHD. Given the established role of inflammation in the pathogenesis of atherosclerosis, this study aims to explore whether novel inflammatory markers (inflammation index) can serve as independent risk factors for CVD in MHD patients. METHODS A cross-sectional survey was conducted on patients from three dialysis centers, categorized into a CVD and non-CVD group based on medical history, laboratory tests, and physical examination. Fasting blood samples were collected from all participants for indicator testing. RESULTS The analysis of 209 patients revealed that 104 had concurrent CVD. Patients in the CVD group were significantly older and exhibited higher anxiety and depression scores. Forward stepwise multivariate logistic regression results identified the inflammation index neutrophil-to-lymphocyte ratio (NLR) (OR = 1.27, 95% CI 1.082-1.491, P < 0.05) and systemic immune-inflammation index (SII) (OR = 1.001, 95% CI 1.0001-1.002, P < 0.05) as independent risk factors for CVD in MHD patients. Receiver operating characteristic (ROC) curve analysis demonstrated that SII, platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR), and NLR all possess predictive diagnostic values for CVD events in this patient population. CONCLUSIONS Hemodialysis centers can utilize simple and cost-effective inflammatory markers to proactively identify patients at risk of CVD. Future research into how inflammation contributes to CVD in MHD is required.
Collapse
Affiliation(s)
- Xi-Xi Han
- Nephrology Department, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, 100039, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui-Ying Zhang
- Nephrology Department, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, 100039, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing-Wen Kong
- Nephrology Department, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, 100039, China
| | - Yu-Xin Liu
- Nephrology Department, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, 100039, China
| | - Ke-Ren Zhang
- Nephrology Department, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, 100039, China
| | - Wen-Ying Ren
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
2
|
Mosalmanzadeh N, Pence BD. Oxidized Low-Density Lipoprotein and Its Role in Immunometabolism. Int J Mol Sci 2024; 25:11386. [PMID: 39518939 PMCID: PMC11545486 DOI: 10.3390/ijms252111386] [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: 08/19/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Modified cholesterols such as oxidized low-density lipoprotein (OxLDL) contribute to atherosclerosis and other disorders through the promotion of foam cell formation and inflammation. In recent years, it has become evident that immune cell responses to inflammatory molecules such as OxLDLs depend on cellular metabolic functions. This review examines the known effects of OxLDL on immunometabolism and immune cell responses in atherosclerosis and several other diseases. We additionally provide context on the relationship between OxLDL and aging/senescence and identify gaps in the literature and our current understanding in these areas.
Collapse
Affiliation(s)
| | - Brandt D. Pence
- College of Health Sciences and Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN 38111, USA
| |
Collapse
|
3
|
Siew WS, Tang YQ, Goh BH, Yap WH. The senescent marker p16INK4a enhances macrophage foam cells formation. Mol Biol Rep 2024; 51:1021. [PMID: 39331194 DOI: 10.1007/s11033-024-09946-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND The senescence marker p16INK4a, which constitutes part of the genome 9p21.3 cardiovascular disease (CVD) risk allele, is believed to play a role in foam cells formation. This study aims to unravel the role of p16INK4a in mediating macrophage foam cells formation, cellular senescence, and autophagy lysosomal functions. METHODS The mammalian expression plasmid pCMV-p16INK4a was used to induce p16INK4a overexpression in THP-1 macrophages. Next, wild-type and p16INK4a-overexpressed macrophages were incubated with oxidized LDL to induce foam cells formation. Lipids accumulation was evaluated using Oil-red-O staining and cholesterol efflux assay, as well as expression of scavenger receptors CD36 and LOX-1. Cellular senescence in macrophage foam cells were determined through analysis of senescence-associated β-galactosidase activity and other SASP factors expression. Meanwhile, autophagy induction was assessed through detection of autophagosome formation and LC3B/p62 markers expression. RESULTS The findings showed that p16INK4a enhanced foam cells formation with increased scavenger receptors CD36 and LOX-1 expression and reduced cholesterol efflux in THP-1 macrophages. Besides, β-galactosidase activity was enhanced, and SASP factors such as IL-1α, TNF-α, and MMP9 were up-regulated. In addition, p16INK4a is also shown to induce autophagy, as well as increasing autophagy markers LC3B and p62 expression. CONCLUSIONS This study provides insights on p16INK4a in mediating macrophages foam cells formation, cellular senescence, and foam cells formation.
Collapse
Affiliation(s)
- Wei Sheng Siew
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, 47500, Malaysia
| | - Yin Quan Tang
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, 47500, Malaysia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Selangor, Malaysia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, 47500, Malaysia.
| |
Collapse
|
4
|
Jing M, Xi H, Wang Y, Zhu H, Sun Q, Zhang Y, Ren W, Xu Z, Deng L, Zhang B, Han T, Zhou J. Association between Pericoronary Fat Attenuation Index Values and Plaque Composition Volume Fraction Measured by Coronary Computed Tomography Angiography. Acad Radiol 2024; 31:3579-3589. [PMID: 38852002 DOI: 10.1016/j.acra.2024.05.001] [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: 02/17/2024] [Revised: 04/07/2024] [Accepted: 05/03/2024] [Indexed: 06/10/2024]
Abstract
RATIONALE AND OBJECTIVES The pericoronary fat attenuation index (FAI) values around plaques may reveal the relationship between periplaque vascular inflammation and different plaque component volume fractions. We aimed to evaluate the potential associations between periplaque FAI values and plaque component volume fractions. MATERIALS AND METHODS 496 patients (1078 lesions) with coronary artery disease, who underwent computed tomography angiography (CCTA) between September 2022 and August 2023, were analyzed retrospectively. Each lesion was characterized and the plaque component volume fractions and periplaque FAI values were measured. Multiple linear regression, weighted quantile sum (WQS) regression, and quantile g-computation (Qgcomp) were used to explore the relationship between plaque component volume fractions and the risk of elevated periplaque FAI values. RESULTS After adjusting for clinical characteristics, multiple linear regression identified that lipid components volume fraction (β = 0.162, P < 0.001) were independent risk factors for elevated periplaque FAI values whereas calcified components volume fraction (β = -0.066, P = 0.025) were independent protective factors. The WQS regression models indicated an increase in the overall confounding effect of the adjusted lipid indices and plaque composition volume fraction on the risk of elevated periplaque FAI values (P = 0.004). Qgcomp analysis indicated lipid component volume fraction and calcified component volume fraction was positively and negatively correlated with elevated plaque FAI values, respectively (all P < 0.05). CONCLUSIONS Periplaque FAI values quantified by CCTA were strongly correlated with lipid and calcification component volume fractions.
Collapse
Affiliation(s)
- Mengyuan Jing
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Huaze Xi
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Yuanyuan Wang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Hao Zhu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Qiu Sun
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Yuting Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Wei Ren
- GE Healthcare, Computed Tomography Research Center, Beijing, PR China
| | - Zheng Xu
- Shukun Technology Co., Beijing, China
| | - Liangna Deng
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Tao Han
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China.
| |
Collapse
|
5
|
Chen S, Tan Y, Tian L. Immunophenotypes in psychosis: is it a premature inflamm-aging disorder? Mol Psychiatry 2024; 29:2834-2848. [PMID: 38532012 PMCID: PMC11420084 DOI: 10.1038/s41380-024-02539-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
Immunopsychiatric field has rapidly accumulated evidence demonstrating the involvement of both innate and adaptive immune components in psychotic disorders such as schizophrenia. Nevertheless, researchers are facing dilemmas of discrepant findings of immunophenotypes both outside and inside the brains of psychotic patients, as discovered by recent meta-analyses. These discrepancies make interpretations and interrogations on their roles in psychosis remain vague and even controversial, regarding whether certain immune cells are more activated or less so, and whether they are causal or consequential, or beneficial or harmful for psychosis. Addressing these issues for psychosis is not at all trivial, as immune cells either outside or inside the brain are an enormously heterogeneous and plastic cell population, falling into a vast range of lineages and subgroups, and functioning differently and malleably in context-dependent manners. This review aims to overview the currently known immunophenotypes of patients with psychosis, and provocatively suggest the premature immune "burnout" or inflamm-aging initiated since organ development as a potential primary mechanism behind these immunophenotypes and the pathogenesis of psychotic disorders.
Collapse
Affiliation(s)
- Song Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, PR China
| | - Yunlong Tan
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, PR China
| | - Li Tian
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
Wai KW, Low LE, Goh BH, Yap WH. Nrf2 Connects Cellular Autophagy and Vascular Senescence in Atherosclerosis: A Mini-Review. J Lipid Atheroscler 2024; 13:292-305. [PMID: 39355399 PMCID: PMC11439754 DOI: 10.12997/jla.2024.13.3.292] [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: 12/09/2024] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 10/03/2024] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcriptional factor that maintains intracellular redox equilibrium, modulates the expression of antioxidant genes, scavenger receptors, and cholesterol efflux transporters, all of which contribute significantly to foam cell development and plaque formation. Nrf2 has recently emerged as a key regulator that connects autophagy and vascular senescence in atherosclerosis. Autophagy, a cellular mechanism involved in the breakdown and recycling of damaged proteins and organelles, and cellular senescence, a state of irreversible growth arrest, are both processes implicated in the pathogenesis of atherosclerosis. The intricate interplay of these processes has received increasing attention, shedding light on their cumulative role in driving the development of atherosclerosis. Recent studies have revealed that Nrf2 plays a critical role in mediating autophagy and senescence in atherosclerosis progression. Nrf2 activation promotes autophagy, which increases lipid clearance and prevents the development of foam cells. Meanwhile, the activation of Nrf2 also inhibits cellular senescence by regulating the expression of senescence markers to preserve cellular homeostasis and function and delay the progression of atherosclerosis. This review provides an overview of the molecular mechanisms through which Nrf2 connects cellular autophagy and vascular senescence in atherosclerosis. Understanding these mechanisms can provide insights into potential therapeutic strategies targeting Nrf2 to modulate cellular autophagy and vascular senescence, thereby preventing the progression of atherosclerosis.
Collapse
Affiliation(s)
- Kai Wen Wai
- School of Biosciences, Taylor’s University, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Liang Ee Low
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, Selangor, Malaysia
- Monash-Industry Plant Oils Research Laboratory (MIPO), Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, Selangor, Malaysia
- Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, Selangor, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, Bandar Sunway, Subang Jaya, Selangor Darul Ehsan, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wei Hsum Yap
- School of Biosciences, Taylor’s University, Subang Jaya, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
7
|
Meng Y, Zhang J, Liu Y, Zhu Y, Lv H, Xia F, Guo Q, Shi Q, Qiu C, Wang J. The biomedical application of inorganic metal nanoparticles in aging and aging-associated diseases. J Adv Res 2024:S2090-1232(24)00213-3. [PMID: 38821357 DOI: 10.1016/j.jare.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
Aging and aging-associated diseases (AAD), including neurodegenerative disease, cancer, cardiovascular diseases, and diabetes, are inevitable process. With the gradual improvement of life style, life expectancy is gradually extended. However, the extended lifespan has not reduced the incidence of disease, and most elderly people are in ill-health state in their later years. Hence, understanding aging and AAD are significant for reducing the burden of the elderly. Inorganic metal nanoparticles (IMNPs) predominantly include gold, silver, iron, zinc, titanium, thallium, platinum, cerium, copper NPs, which has been widely used to prevent and treat aging and AAD due to their superior properties (essential metal ions for human body, easily synthesis and modification, magnetism). Therefore, a systematic review of common morphological alternations of senescent cells, altered genes and signal pathways in aging and AAD, and biomedical applications of IMNPs in aging and AAD is crucial for the further research and development of IMNPs in aging and AAD. This review focus on the existing research on cellular senescence, aging and AAD, as well as the applications of IMNPs in aging and AAD in the past decade. This review aims to provide cutting-edge knowledge involved with aging and AAD, the application of IMNPs in aging and AAD to promote the biomedical application of IMNPs in aging and AAD.
Collapse
Affiliation(s)
- Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haining Lv
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qianli Shi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jigang Wang
- Department of Urology, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China.
| |
Collapse
|
8
|
Wang B, Zhou J, An N. Investigating molecular markers linked to acute myocardial infarction and cuproptosis: bioinformatics analysis and validation in the AMI mice model. PeerJ 2024; 12:e17280. [PMID: 38827298 PMCID: PMC11143973 DOI: 10.7717/peerj.17280] [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: 10/24/2023] [Accepted: 04/01/2024] [Indexed: 06/04/2024] Open
Abstract
Cuproptosis-related key genes play a significant role in the pathological processes of acute myocardial infarction (AMI). However, a complete understanding of the molecular mechanisms behind this participation remains elusive. This study was designed to identify genes and immune cells critical to AMI pathogenesis. Based on the GSE48060 dataset (31 AMI patients and 21 healthy persons, GPL570-55999), we identified genes associated with dysregulated cuproptosis and the activation of immune responses between normal subjects and patients with a first myocardial attack. Two molecular clusters associated with cuproptosis were defined in patients with AMI. Immune infiltration analysis showed that there was significant immunity heterogeneity among different clusters. Multiple immune responses were closely associated with Cluster2-specific differentially expressed genes (DEGs). The generalized linear model machine model presented the best discriminative performance with relatively lower residual and root mean square error, and a higher area under the curve (AUC = 0.870). A final two-gene-based generalized linear model was constructed, exhibiting satisfactory performance in two external validation datasets (AUC = 0.719, GSE66360 and AUC = 0.856, GSE123342). Column graph, calibration curve, and decision curve analyses also proved the accuracy of AMI prediction. We also constructed a mouse C57BL/6 model of AMI (3 h, 48 h, and 1 week) and used qRT-PCR and immunofluorescence to detect the expression changes of CBLB and ZNF302. In this study, we present a systematic analysis of the complex relationship between cuproptosis and a first AMI attack, and provide new insights into the diagnosis and treatment of AMI.
Collapse
Affiliation(s)
- Bingyu Wang
- Ningbo Medical Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Jianqing Zhou
- Ningbo Medical Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Ning An
- Ningbo Medical Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
9
|
Cheng X, Shihabudeen Haider Ali MS, Baki VB, Moran M, Su H, Sun X. Multifaceted roles of Meg3 in cellular senescence and atherosclerosis. Atherosclerosis 2024; 392:117506. [PMID: 38518516 PMCID: PMC11088985 DOI: 10.1016/j.atherosclerosis.2024.117506] [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: 05/25/2023] [Revised: 02/11/2024] [Accepted: 03/05/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND AND AIMS Long noncoding RNAs are involved in the pathogenesis of atherosclerosis. As long noncoding RNAs maternally expressed gene 3 (Meg3) prevents cellular senescence of hepatic vascular endothelium and obesity-induced insulin resistance, we decided to examine its role in cellular senescence and atherosclerosis. METHODS AND RESULTS By analyzing our data and human and mouse data from the Gene Expression Omnibus database, we found that Meg3 expression was reduced in humans and mice with cardiovascular disease, indicating its potential role in atherosclerosis. In Ldlr-/- mice fed a Western diet for 12 weeks, Meg3 silencing by chemically modified antisense oligonucleotides attenuated the formation of atherosclerotic lesions by 34.9% and 20.1% in male and female mice, respectively, revealed by en-face Oil Red O staining, which did not correlate with changes in plasma lipid profiles. Real-time quantitative PCR analysis of cellular senescence markers p21 and p16 revealed that Meg3 deficiency aggravates hepatic cellular senescence but not cellular senescence at aortic roots. Human Meg3 transgenic mice were generated to examine the role of Meg3 gain-of-function in the development of atherosclerosis induced by PCSK9 overexpression. Meg3 overexpression promotes atherosclerotic lesion formation by 29.2% in Meg3 knock-in mice independent of its effects on lipid profiles. Meg3 overexpression inhibits hepatic cellular senescence, while it promotes aortic cellular senescence likely by impairing mitochondrial function and delaying cell cycle progression. CONCLUSIONS Our data demonstrate that Meg3 promotes the formation of atherosclerotic lesions independent of its effects on plasma lipid profiles. In addition, Meg3 regulates cellular senescence in a tissue-specific manner during atherosclerosis. Thus, we demonstrated that Meg3 has multifaceted roles in cellular senescence and atherosclerosis.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE, 68588, USA
| | | | - Vijaya Bhaskar Baki
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE, 68588, USA
| | - Matthew Moran
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE, 68588, USA
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE, 68588, USA; Nebraska Center for the Prevention of Obesity Diseases Through Dietary Molecules, University of Nebraska - Lincoln, USA.
| |
Collapse
|
10
|
Singh J, Bisht P, Srivastav S, Kumar Y, Sharma V, Kumar A, Akhtar MS, Khan MF, Aldosari SA, Yadav S, Yadav NK, Mukherjee M, Sharma AK. Amelioration of endothelial integrity by 3,5,4'-trihydroxy-trans-stilbene against high-fat-diet-induced obesity and -associated vasculopathy and myocardial infarction in rats, targeting TLR4/MyD88/NF-κB/iNOS signaling cascade. Biochem Biophys Res Commun 2024; 705:149756. [PMID: 38460440 DOI: 10.1016/j.bbrc.2024.149756] [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: 12/11/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Exacerbated expression of TLR4 protein (foremost pattern recognition receptor) during obesity could trigger NF-κB/iNOS signaling through linker protein (MyD88), predisposed to an indispensable inflammatory response. The induction of this detrimental cascade leads to myocardial and vascular abnormalities. Molecular docking was studied for protein-ligand interaction between these potential targets and resveratrol. The pre-treatment of resveratrol (20 mg/kg/p.o/per day for ten weeks) was given to investigate the therapeutic effect against HFD-induced obesity and associated vascular endothelial dysfunction (VED) and myocardial infarction (MI) in Wistar rats. In addition to accessing the levels of serum biomarkers for VED and MI, oxidative stress, inflammatory cytokines, and histopathology of these tissues were investigated. Lipopolysaccharide (for receptor activation) and protein expression analysis were introduced to explore the mechanistic involvement of TLR4/MyD88/NF-κB/iNOS signaling. Assessment of in-silico analysis showed significant interaction between protein and ligand. The involvement of this proposed signaling (TLR4/MyD88/NF-κB/iNOS) was further endorsed by the impact of lipopolysaccharide and protein expression analysis in obese and treated rats. Moreover, resveratrol pre-treated rats showed significantly lowered cardio and vascular damage measured by the distinct down expression of the TLR4/MyD88/NF-κB/iNOS pathway by resveratrol treatment endorses its ameliorative effect against VED and MI.
Collapse
Affiliation(s)
- Jitender Singh
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, 122413, India
| | - Priyanka Bisht
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, 122413, India
| | - Srishti Srivastav
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, 122413, India
| | - Yash Kumar
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, 122413, India
| | - Vikash Sharma
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, 122413, India
| | - Ashish Kumar
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, 122413, India
| | - Md Sayeed Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Alfara, Abha, 62223, Saudi Arabia
| | - Mohd Faiyaz Khan
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 11942, Saudi Arabia
| | - Saad A Aldosari
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 11942, Saudi Arabia
| | - Snehlata Yadav
- Department of Pharmaceutical Sciences, Indra Gandhi University, Meerpur, Rewari, 122502, Haryana, India
| | - Nirmala K Yadav
- Department of Pharmaceutical Sciences, Indra Gandhi University, Meerpur, Rewari, 122502, Haryana, India
| | - Monalisa Mukherjee
- Molecular Sciences and Engineering Laboratory, Amity Institute of Click Chemistry Research and Studies, Amity University, Noida, Uttar Pradesh, 201303, India
| | - Arun K Sharma
- Department of Cardiovascular Pharmacology, Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, 122413, India.
| |
Collapse
|
11
|
Zietz A, Gorey S, Kelly PJ, Katan M, McCabe JJ. Targeting inflammation to reduce recurrent stroke. Int J Stroke 2024; 19:379-387. [PMID: 37800305 DOI: 10.1177/17474930231207777] [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: 10/07/2023]
Abstract
BACKGROUND Approximately one in four stroke patients suffer from recurrent vascular events, underlying the necessity to improve secondary stroke prevention strategies. Immune mechanisms are causally associated with coronary atherosclerosis. However, stroke is a heterogeneous disease and the relative contribution of inflammation across stroke mechanisms is not well understood. The optimal design of future randomized control trials (RCTs) of anti-inflammatory therapies to prevent recurrence after stroke must be informed by a clear understanding of the prognostic role of inflammation according to stroke subtype and individual patient factors. AIM In this narrative review, we discuss (1) inflammatory pathways in the etiology of ischemic stroke subtypes; (2) the evidence on inflammatory markers and vascular recurrence after stroke; and (3) review RCT evidence of anti-inflammatory agents for vascular prevention. SUMMARY OF REVIEW Experimental work, genetic epidemiological data, and plaque-imaging studies all implicate inflammation in atherosclerotic stroke. However, emerging evidence also suggests that inflammatory mechanisms are also important in other stroke mechanisms. Advanced neuroimaging techniques support the role of neuroinflammation in blood-brain barrier dysfunction in cerebral small vessel disease (cSVD). Systemic inflammatory processes also promote atrial cardiopathy, incident and recurrent atrial fibrillation (AF). Although several inflammatory markers have been associated with recurrence after stroke, interleukin-6 (IL-6) and high-sensitivity C-reactive protein (hsCRP) are presently the most promising markers to identify patients at increased vascular risk. Several RCTs have shown that anti-inflammatory therapies reduce vascular risk, including stroke, in coronary artery disease (CAD). Some, but not all of these trials, selected patients on the basis of elevated hsCRP. Although unproven after stroke, targeting inflammation to reduce recurrence is a compelling strategy and several RCTs are ongoing. CONCLUSION Evidence points toward the importance of inflammation across multiple stroke etiologies and potential benefit of anti-inflammatory targets in secondary stroke prevention. Taking the heterogeneous stroke etiologies into account, the use of serum biomarkers could be useful to identify patients with residual inflammatory risk and perform biomarker-led patient selection for future RCTs.
Collapse
Affiliation(s)
- Annaelle Zietz
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
- Neurology and Neurorehabilitation, University Department of Geriatric Medicine Felix Platter, University of Basel, Basel, Switzerland
| | - Sarah Gorey
- Health Research Board (HRB) Stroke Clinical Trials Network Ireland (SCTNI), Dublin, Ireland
- School of Medicine, University College Dublin (UCD), Dublin, Ireland
- Department of Geriatric Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Peter J Kelly
- Health Research Board (HRB) Stroke Clinical Trials Network Ireland (SCTNI), Dublin, Ireland
- School of Medicine, University College Dublin (UCD), Dublin, Ireland
- Department of Neurology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Mira Katan
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - John J McCabe
- Health Research Board (HRB) Stroke Clinical Trials Network Ireland (SCTNI), Dublin, Ireland
- School of Medicine, University College Dublin (UCD), Dublin, Ireland
- Department of Geriatric Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
12
|
Ali I, Zhang H, Zaidi SAA, Zhou G. Understanding the intricacies of cellular senescence in atherosclerosis: Mechanisms and therapeutic implications. Ageing Res Rev 2024; 96:102273. [PMID: 38492810 DOI: 10.1016/j.arr.2024.102273] [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: 11/14/2023] [Revised: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is currently the largest cause of mortality and disability globally, surpassing communicable diseases, and atherosclerosis is the main contributor to this epidemic. Aging is intimately linked to atherosclerosis development and progression, however, the mechanism of aging in atherosclerosis is not well known. To emphasize the significant research on the involvement of senescent cells in atherosclerosis, we begin by outlining compelling evidence that indicates various types of senescent cells and SASP factors linked to atherosclerotic phenotypes. We subsequently provide a comprehensive summary of the existing knowledge, shedding light on the intricate mechanisms through which cellular senescence contributes to the pathogenesis of atherosclerosis. Further, we cover that senescence can be identified by both structural changes and several senescence-associated biomarkers. Finally, we discuss that preventing accelerated cellular senescence represents an important therapeutic potential, as permanent changes may occur in advanced atherosclerosis. Together, the review summarizes the relationship between cellular senescence and atherosclerosis, and inspects the molecular knowledge, and potential clinical significance of senescent cells in developing senescent-based therapy, thus providing crucial insights into their biology and potential therapeutic exploration.
Collapse
Affiliation(s)
- Ilyas Ali
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Hongliang Zhang
- Shenzhen University General Hospital, Shenzhen University, Shenzhen 518060, PR China
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China.
| |
Collapse
|
13
|
Fraile-Martinez O, De Leon-Oliva D, Boaru DL, De Castro-Martinez P, Garcia-Montero C, Barrena-Blázquez S, García-García J, García-Honduvilla N, Alvarez-Mon M, Lopez-Gonzalez L, Diaz-Pedrero R, Guijarro LG, Ortega MA. Connecting epigenetics and inflammation in vascular senescence: state of the art, biomarkers and senotherapeutics. Front Genet 2024; 15:1345459. [PMID: 38469117 PMCID: PMC10925776 DOI: 10.3389/fgene.2024.1345459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Abstract
Vascular diseases pose major health challenges, and understanding their underlying molecular mechanisms is essential to advance therapeutic interventions. Cellular senescence, a hallmark of aging, is a cellular state characterized by cell-cycle arrest, a senescence-associated secretory phenotype macromolecular damage, and metabolic dysregulation. Vascular senescence has been demonstrated to play a key role in different vascular diseases, such as atherosclerosis, peripheral arterial disease, hypertension, stroke, diabetes, chronic venous disease, and venous ulcers. Even though cellular senescence was first described in 1961, significant gaps persist in comprehending the epigenetic mechanisms driving vascular senescence and its subsequent inflammatory response. Through a comprehensive analysis, we aim to elucidate these knowledge gaps by exploring the network of epigenetic alterations that contribute to vascular senescence. In addition, we describe the consequent inflammatory cascades triggered by these epigenetic modifications. Finally, we explore translational applications involving biomarkers of vascular senescence and the emerging field of senotherapy targeting this biological process.
Collapse
Affiliation(s)
- Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Joaquin García-García
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, Alcala deHenares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, Alcala deHenares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala deHenares, Spain
| |
Collapse
|
14
|
Molnár AÁ, Pásztor DT, Tarcza Z, Merkely B. Cells in Atherosclerosis: Focus on Cellular Senescence from Basic Science to Clinical Practice. Int J Mol Sci 2023; 24:17129. [PMID: 38138958 PMCID: PMC10743093 DOI: 10.3390/ijms242417129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Aging is a major risk factor of atherosclerosis through different complex pathways including replicative cellular senescence and age-related clonal hematopoiesis. In addition to aging, extracellular stress factors, such as mechanical and oxidative stress, can induce cellular senescence, defined as premature cellular senescence. Senescent cells can accumulate within atherosclerotic plaques over time and contribute to plaque instability. This review summarizes the role of cellular senescence in the complex pathophysiology of atherosclerosis and highlights the most important senotherapeutics tested in cardiovascular studies targeting senescence. Continued bench-to-bedside research in cellular senescence might allow the future implementation of new effective anti-atherosclerotic preventive and treatment strategies in clinical practice.
Collapse
Affiliation(s)
- Andrea Ágnes Molnár
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary; (D.T.P.); (Z.T.); (B.M.)
| | | | | | | |
Collapse
|
15
|
Huang Q, Zhou H, Yu S. Long non‑coding RNA PEG13 regulates endothelial cell senescence through the microRNA‑195/IRS1 axis. Exp Ther Med 2023; 26:584. [PMID: 38023368 PMCID: PMC10665998 DOI: 10.3892/etm.2023.12283] [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: 05/04/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction and plaque formation. The present study aimed to elucidate the pathological role of the long non-coding RNA (lncRNA) paternally expressed 13 (PEG13) in the onset and progression of atherosclerosis. Specifically, its effects on human umbilical vein endothelial cell (HUVEC) proliferation, angiogenesis, senescence and senescence-associated secretory phenotype (SASP)-related factors were investigated using cell proliferation, cellular angiogenesis, β-galactosidase staining, reverse transcription-quantitative PCR and enzyme-linked immunosorbent assays. The results showed that oxidized low-density lipoprotein (ox-LDL) inhibited lncRNA PEG13 expression and HUVEC viability in a dose-dependent manner and PEG13 overexpression partially reversed these effects. Additionally, PEG13 overexpression ameliorated the ox-LDL-induced impairment of angiogenesis, cellular senescence and SASP. Furthermore, lncRNA PEG13 directly targeted microRNA (miR/miRNA)-195-5p, suppressing the ox-LDL-induced upregulation of the miRNA. The gene coding for insulin receptor substrate 1 (IRS1), an activator of the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, was confirmed as a direct target of miR-195. PEG13 overexpression attenuated the ox-LDL-induced inhibition of IRS1 expression and PI3K/AKT signaling and its protective effects on HUVEC viability, angiogenesis and senescence were partially reversed by small interfering RNAs targeting IRS1. The present study demonstrated that lncRNA PEG13 attenuates ox-LDL-induced senescence in HUVECs by modulating the miR-195/IRS1/PI3K/AKT signaling pathway, suggesting a potential therapeutic target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Qin Huang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| | - Haiwen Zhou
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| | - Songping Yu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
16
|
Martyshkina YS, Tereshchenko VP, Bogdanova DA, Rybtsov SA. Reliable Hallmarks and Biomarkers of Senescent Lymphocytes. Int J Mol Sci 2023; 24:15653. [PMID: 37958640 PMCID: PMC10647376 DOI: 10.3390/ijms242115653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
The phenomenon of accumulation of senescent adaptive immunity cells in the elderly is attracting attention due to the increasing risk of global epidemics and aging of the global population. Elderly people are predisposed to various infectious and age-related diseases and are at higher risk of vaccination failure. The accumulation of senescent cells increases age-related background inflammation, "Inflammaging", causing lymphocyte exhaustion and cardiovascular, neurodegenerative, autoimmune and cancer diseases. Here, we present a comprehensive contemporary review of the mechanisms and phenotype of senescence in the adaptive immune system. Although modern research has not yet identified specific markers of aging lymphocytes, several sets of markers facilitate the separation of the aging population based on normal memory and exhausted cells for further genetic and functional analysis. The reasons for the higher predisposition of CD8+ T-lymphocytes to senescence compared to the CD4+ population are also discussed. We point out approaches for senescent-lymphocyte-targeting markers using small molecules (senolytics), antibodies and immunization against senescent cells. The suppression of immune senescence is the most relevant area of research aimed at developing anti-aging and anti-cancer therapy for prolonging the lifespan of the global population.
Collapse
Affiliation(s)
- Yuliya S. Martyshkina
- Division of Immunobiology and Biomedicine, Center for Genetics and Life Sciences, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia; (Y.S.M.)
| | - Valeriy P. Tereshchenko
- Resource Center for Cell Technology and Immunology, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia
| | - Daria A. Bogdanova
- Division of Immunobiology and Biomedicine, Center for Genetics and Life Sciences, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia; (Y.S.M.)
| | - Stanislav A. Rybtsov
- Resource Center for Cell Technology and Immunology, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia
| |
Collapse
|
17
|
Kim ME, Lee JS. Immune Diseases Associated with Aging: Molecular Mechanisms and Treatment Strategies. Int J Mol Sci 2023; 24:15584. [PMID: 37958564 PMCID: PMC10647753 DOI: 10.3390/ijms242115584] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Aging is associated with a decline in immune function, thereby causing an increased susceptibility to various diseases. Herein, we review immune diseases associated with aging, focusing on tumors, atherosclerosis, and immunodeficiency disorders. The molecular mechanisms underlying these conditions are discussed, highlighting telomere shortening, tissue inflammation, and altered signaling pathways, e.g., the mammalian target of the rapamycin (mTOR) pathway, as key contributors to immune dysfunction. The role of the senescence-associated secretory phenotype in driving chronic tissue inflammation and disruption has been examined. Our review underscores the significance of targeting tissue inflammation and immunomodulation for treating immune disorders. In addition, anti-inflammatory medications, including corticosteroids and nonsteroidal anti-inflammatory drugs, and novel approaches, e.g., probiotics and polyphenols, are discussed. Immunotherapy, particularly immune checkpoint inhibitor therapy and adoptive T-cell therapy, has been explored for its potential to enhance immune responses in older populations. A comprehensive analysis of immune disorders associated with aging and underlying molecular mechanisms provides insights into potential treatment strategies to alleviate the burden of these conditions in the aging population. The interplay among immune dysfunction, chronic tissue inflammation, and innovative therapeutic approaches highlights the importance of elucidating these complex processes to develop effective interventions to improve the quality of life in older adults.
Collapse
Affiliation(s)
| | - Jun Sik Lee
- Department of Biological Science, Immunology Research Lab & BK21-Four Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea;
| |
Collapse
|
18
|
Bu LL, Yuan HH, Xie LL, Guo MH, Liao DF, Zheng XL. New Dawn for Atherosclerosis: Vascular Endothelial Cell Senescence and Death. Int J Mol Sci 2023; 24:15160. [PMID: 37894840 PMCID: PMC10606899 DOI: 10.3390/ijms242015160] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/01/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Endothelial cells (ECs) form the inner linings of blood vessels, and are directly exposed to endogenous hazard signals and metabolites in the circulatory system. The senescence and death of ECs are not only adverse outcomes, but also causal contributors to endothelial dysfunction, an early risk marker of atherosclerosis. The pathophysiological process of EC senescence involves both structural and functional changes and has been linked to various factors, including oxidative stress, dysregulated cell cycle, hyperuricemia, vascular inflammation, and aberrant metabolite sensing and signaling. Multiple forms of EC death have been documented in atherosclerosis, including autophagic cell death, apoptosis, pyroptosis, NETosis, necroptosis, and ferroptosis. Despite this, the molecular mechanisms underlying EC senescence or death in atherogenesis are not fully understood. To provide a comprehensive update on the subject, this review examines the historic and latest findings on the molecular mechanisms and functional alterations associated with EC senescence and death in different stages of atherosclerosis.
Collapse
Affiliation(s)
- Lan-Lan Bu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.-L.B.); (D.-F.L.)
| | - Huan-Huan Yuan
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
| | - Ling-Li Xie
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Min-Hua Guo
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
| | - Duan-Fang Liao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.-L.B.); (D.-F.L.)
| | - Xi-Long Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
19
|
Fu L, Yu B, Zhang Y, Cao S, Lv B, Tian Y, Chen H, Yang S, Hu Y, Hua J, Ren P, Li J, Gong S. Integrative analysis of single-cell and bulk RNA sequencing unveils the senescence landscape in ischemic stroke. Aging (Albany NY) 2023; 15:5497-5513. [PMID: 37382646 PMCID: PMC10333064 DOI: 10.18632/aging.204804] [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: 04/06/2023] [Accepted: 05/27/2023] [Indexed: 06/30/2023]
Abstract
Ischemic stroke (IS) is a fatal neurological disease that occurs when the blood flow to the brain is disrupted, leading to brain tissue damage and functional impairment. Cellular senescence, a vital characteristic of aging, is associated with a poor prognosis for IS. This study explores the potential role of cellular senescence in the pathological process following IS by analyzing transcriptome data from multiple datasets (GSE163654, GSE16561, GSE119121, and GSE174574). By using bioinformatics methods, we identified hub-senescence-related genes such as ANGPTL4, CCL3, CCL7, CXCL16, and TNF and verified them using quantitative reverse transcription polymerase chain reaction. Further analysis of single-cell RNA sequencing data suggests that MG4 microglial is highly correlated with cellular senescence in MCAO, and might play a crucial role in the pathological process after IS. Additionally, we identified retinoic acid as a potential drug for improving the prognosis of IS. This comprehensive investigation of cellular senescence in various brain tissues and peripheral blood cell types provides valuable insights into the underlying mechanisms of the pathology of IS and identifies potential therapeutic targets for improving patient outcomes.
Collapse
Affiliation(s)
- Longhui Fu
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Beibei Yu
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Yongfeng Zhang
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Shuai Cao
- Department of Orthopedics, Civil Aviation General Hospital, Chaoyang, Beijing, China
| | - Boqiang Lv
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Yunze Tian
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
- Department of Thoracic Surgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Huangtao Chen
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Shijie Yang
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Yutian Hu
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Jinghua Hua
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Pengyu Ren
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Jianzhong Li
- Xi’an Jiaotong University, Xi’an, China
- Department of Thoracic Surgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Shouping Gong
- Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
- Xi’an Medical University, Xi’an, China
| |
Collapse
|
20
|
Mao Q, Deng M, Zhao J, Zhou D, Chen M, Liu Q, Xu S, Zhao X. Low serum Klotho reflects senile inflammation in middle-aged and elderly patients with coronary atherosclerosis. Cytokine 2023; 167:156213. [PMID: 37121091 DOI: 10.1016/j.cyto.2023.156213] [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: 02/04/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Anti-aging protein Klotho has been reported to be associated with atherosclerosis, which was considered as a chronic inflammatory disease. However, the relationship between Klotho and senile inflammation remained unclear. The present study aims to ascertain the correlation of Klotho with inflammation in middle-aged and elderly coronary atherosclerotic disease (CAD). METHODS A total of 302 patients with CAD were included in this study. Coronary atherosclerosis was confirmed and quantified for all patients by coronary angiography. Serum Klotho was detected by enzyme linked immunosorbent assay. Serum concentrations of IL-6 and IL-8 were quantified by chemiluminescence assay. T-lymphocyte subsets were measured using flow cytometry. RESULTS Multivariate linear regression analysis showed that serum Klotho was an independent predictor for circulating monocytes (standard β = -0.321, P < 0.001) and CD4+/CD8+ ratio (standard β = -0.522, P < 0.001). After adjustment, serum Klotho was still independently associated with IL-6 (standard β = -0.395, P < 0.001) and IL-8 (standard β = -0.296, P < 0.001). Moreover, circulating monocytes, CD4+ and CD8+ lymphocytes were correlated with increased serum concentrations of IL-6 and IL-8, independent of CRP (P < 0.05). In receiver operating characteristic curve analysis, CD4+/CD8+ ratio (AUC = 0.863, P < 0.001), IL-6 (AUC = 0.893, P < 0.001) and IL-8 (AUC = 0.884, P < 0.001) presented the excellent predictive performance for significant CAD. CONCLUSIONS Decreased concentrations in serum Klotho reflect senile inflammation, which is related to the severity of CAD in middle-aged and elderly patients.
Collapse
Affiliation(s)
- Qi Mao
- Department of Cardiology, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Mengyang Deng
- Department of Cardiology, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Jianhua Zhao
- Department of Cardiology, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Denglu Zhou
- Department of Cardiology, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Man Chen
- Department of Clinical Laboratory, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Qingsong Liu
- Department of Cardiology, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Shangcheng Xu
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China.
| | - Xiaohui Zhao
- Department of Cardiology, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| |
Collapse
|
21
|
Akram A, Jamshed A, Anwaar M, Rasheed HMF, Haider SI, Aslam N, Jabeen Q. Evaluation of Caralluma edulis for its Potential Against Obesity, Atherosclerosis and Hypertension. Dose Response 2023; 21:15593258231152112. [PMID: 36655145 PMCID: PMC9841870 DOI: 10.1177/15593258231152112] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Obesity, a chronic metabolic condition, is an increase in fat mass and blood lipid levels mainly causing atherosclerosis and hypertension, which further lead to cardiovascular complications. The objective of the study was to investigate the crude extract of Caralluma edulis (CE.Cr) for its potential against high-fat diet (HFD)-induced obesity and its related complications. Hyperlipidemia was induced in Wistar albino rats with HFD (1% cholesterol + 0.5% cholic acid) for 28 days. Treatment groups were administered with different doses of CE.Cr (100, 300 and 500 mg/Kg, p.o.) and the standard group received atorvastatin. At the end of study, sera were analyzed for biochemical markers and the aorta was dissected for microscopic examination. Antioxidant potential was evaluated and high-performance liquid chromatography (HPLC) analysis was performed. The hypotensive potential of CE.Cr was evaluated through an invasive technique. HPLC analysis of CE.Cr showed the presence of chlorogenic acid, caffeic acid, apigenin and naringenin. Histological examination of the aorta section showed anti-atherosclerotic effects which were also evident from decrease in serum total cholesterol, triglycerides and low-density lipoproteins levels. CE.Cr decreased mean arterial blood pressure and evoked significant hypotensive effects. The crude extract of C. edulis showed anti-obesity, antihypertensive, anti-atherosclerotic and antioxidant potential.
Collapse
Affiliation(s)
- Adnan Akram
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University, Bahawalpur, Pakistan,Department of Primary & Secondary Healthcare, Government of Punjab, Pakistan
| | - Ayesha Jamshed
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University, Bahawalpur, Pakistan,Ayesha Jamshed, Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| | - Mariya Anwaar
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University, Bahawalpur, Pakistan
| | - Hafiz Muhammad Farhan Rasheed
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University, Bahawalpur, Pakistan,Department of Primary & Secondary Healthcare, Government of Punjab, Pakistan
| | - Syed Ihtisham Haider
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University, Bahawalpur, Pakistan
| | - Naveed Aslam
- Department of Primary & Secondary Healthcare, Government of Punjab, Pakistan
| | - Qaiser Jabeen
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University, Bahawalpur, Pakistan,Qaiser Jabeen, Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| |
Collapse
|