1
|
Zhang YZ, Huo DY, Liu Z, Li XD, Wang Z, Li W. Review on ginseng and its potential active substance G-Rg2 against age-related diseases: Traditional efficacy and mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118781. [PMID: 39260708 DOI: 10.1016/j.jep.2024.118781] [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: 04/09/2024] [Revised: 08/04/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE According to the Shen Nong Herbal Classic, Ginseng (Panax ginseng C.A. Meyer) is documented to possess life-prolonging effects and is extensively utilized in traditional Chinese medicine for the treatment of various ailments such as qi deficiency, temper deficiency, insomnia, and forgetfulness. Ginseng is commonly employed for replenishing qi and nourishing blood, fortifying the body and augmenting immunity; it has demonstrated efficacy in alleviating fatigue, enhancing memory, and retarding aging. Furthermore, it exhibits a notable ameliorative impact on age-related conditions including cardiovascular diseases and neurodegenerative disorders. One of its active constituents - ginsenoside Rg2 (G-Rg2) - exhibits potential therapeutic efficacy in addressing these ailments. AIM OF THE REVIEW The aim of this review is to explore the traditional efficacy of ginseng in anti-aging diseases and the modern pharmacological mechanism of its potential active substance G-Rg2, in order to provide strong theoretical support for further elucidating the mechanism of its anti-aging effect. METHODS This review provides a comprehensive analysis of the traditional efficacy of ginseng and the potential mechanisms underlying the anti-age-related disease properties of G-Rg2, based on an extensive literature review up to March 12, 2024, from PubMed, Web of Science, Scopus, Cochrane, and Google Scholar databases. Potential anti-aging mechanisms of G-Rg2 were predicted using network pharmacology and molecular docking analysis techniques. RESULTS In traditional Chinese medicine theory, ginseng has been shown to improve aging-related diseases with a variety of effects, including tonifying qi, strengthening the spleen and stomach, nourishing yin, regulating yin and yang, as well as calming the mind. Its potential active ingredient G-Rg2 has demonstrated significant therapeutic potential in age-related diseases, especially central nervous system and cardiovascular diseases. G-Rg2 exhibited a variety of pharmacological activities, including anti-apoptotic, anti-inflammatory and antioxidant effects. Meanwhile, the network pharmacological analyses and molecular docking results were consistent with the existing literature review, further validating the potential efficacy of G-Rg2 as an anti-aging agent. CONCLUSION The review firstly explores the ameliorative effects of ginseng on a wide range of age-related diseases based on TCM theories. Secondly, the article focuses on the remarkable significance and value demonstrated by G-Rg2 in age-related cardiovascular and neurodegenerative diseases. Consequently, G-Rg2 has broad prospects for development in intervening in aging and treating age-related health problems.
Collapse
Affiliation(s)
- Yu-Zhuo Zhang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - De-Yang Huo
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Zhi Liu
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Xin-Dian Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; College of Life Sciences, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
2
|
Yang MM, Boin F, Wolters PJ. Molecular underpinnings of aging contributing to systemic sclerosis pathogenesis. Curr Opin Rheumatol 2025; 37:86-92. [PMID: 39600291 DOI: 10.1097/bor.0000000000001061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
PURPOSE OF REVIEW Systemic sclerosis (SSc) is a systemic autoimmune disease characterized by diffuse organ fibrosis and vasculopathy. Aberrant aging has been increasingly implicated in fibrotic diseases of the lung and other organs. The aim of this review is to summarize the established mechanisms of aging and how they may contribute to the pathogenesis of SSc. RECENT FINDINGS Shortened telomeres are present in SSc patients with interstitial lung disease (SSc-ILD) and associate with disease severity and mortality. Although the cause of telomere length shortening is unknown, immune mechanisms may be at play. Senescent cells accumulate in affected organs of SSc patients and contribute to a pathologic cellular phenotype that can be profibrotic and inflammatory. In addition to identifying patients with a more severe phenotype, biomarkers of aging may help identify patients who have worse outcomes with immunosuppression. SUMMARY Aging mechanisms, including telomere dysfunction and cellular senescence, likely contribute to the progressive fibrosis, vasculopathy, and immune dysfunction of SSc. Further work is needed to understand whether aberrant aging is an initiator or perpetuator of disease, and whether this is cell or organ specific. A better understanding of the role aging mechanisms play in SSc will contribute to our understanding of the underlying pathobiology and may also influence management of patients exhibiting the aging phenotype.
Collapse
Affiliation(s)
- Monica M Yang
- Division of Rheumatology, Department of Medicine, University of California, San Francisco
| | - Francesco Boin
- Division of Rheumatology, Kao Autoimmunity Institute, Cedar Sinai Medical Center, Los Angeles
| | - Paul J Wolters
- Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
3
|
Li M, Liu Z, Cao X, Xiao W, Wang S, Zhao C, Zhao Y, Xie Y. [Gly14]-Humanin ameliorates high glucose-induced endothelial senescence via SIRT6. Sci Rep 2024; 14:30924. [PMID: 39730568 DOI: 10.1038/s41598-024-81878-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
High glucose (HG) induced endothelial senescence is related to endothelial dysfunction and cardiovascular complications in diabetic patients. Humanin, a member of mitochondrial derived peptides (MDPs), is thought to contribute to aging-related cardiovascular protection. The goal of the study is to explore the pathogenesis of HG-induced endothelial senescence and potential anti-senescent effects of Humanin. Human umbilical vein endothelial cells (HUVECs) were exposed to glucose to induce senescence, determined by β-galactosidase staining and the expressions of p21, p53, and p16. A clinically relevant dose of HG (15 mM, HG) induced endothelial senescence after 72 h incubation without elevated apoptosis. HG-induced senescence was attributed to the induction of reactive oxygen species (ROS) caused by SIRT6 downregulation, as ROS inhibitor N-acetyl cysteine blocked HG-induced senescence, while inactivation of SIRT6 increased ROS levels and promoted senescence. Strikingly. pretreatment with [Gly14]-Humanin (HNG) antagonized the downregulation of SIRT6 in response to HG and alleviated ROS production and cell senescence. HG-induced reduction of SIRT6 results in ROS overproduction and endothelial senescence. Humanin protects against HG-induced endothelial senescence via SIRT6. This study provides new directions for biological products related to Humanin to be a potential candidate for the prevention of vascular aging in diabetes.
Collapse
Affiliation(s)
- Muqin Li
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Department of Endocrinology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, JiangSu, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215004, China
| | - Zhihua Liu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xueqin Cao
- Department of Endocrinology, The Fourth Affiliated Hospital of Soochow University, Chongwen Road No. 9, Suzhou, 215000, Jiangsu, China
| | - Wenjin Xiao
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Shurong Wang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Chengyuan Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Department of endocrinology, Taizhou school of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Ying Zhao
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Soochow Medical College of Soochow University, Suzhou, 215123, China.
| | - Ying Xie
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
4
|
Summer S, Borrell-Pages M, Bruno RM, Climie RE, Dipla K, Dogan A, Eruslanova K, Fraenkel E, Mattace-Raso F, Pugh CJA, Rochfort KD, Ross M, Roth L, Schmidt-Trucksäss A, Schwarz D, Shadiow J, Sohrabi Y, Sonnenberg J, Tura-Ceide O, Guvenc Tuna B, Julve J, Dogan S. Centenarians-the way to healthy vascular ageing and longevity: a review from VascAgeNet. GeroScience 2024:10.1007/s11357-024-01467-8. [PMID: 39725804 DOI: 10.1007/s11357-024-01467-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
The prevalence of centenarians, people who lived 100 years and longer, is steadily growing in the last decades. This exceptional longevity is based on multifaceted processes influenced by a combination of intrinsic and extrinsic factors such as sex, (epi-)genetic factors, gut microbiota, cellular metabolism, exposure to oxidative stress, immune status, cardiovascular risk factors, environmental factors, and lifestyle behavior. Epidemiologically, the incidence rate of cardiovascular diseases is reduced in healthy centenarians along with late onset of age-related diseases compared with the general aged population. Understanding the mechanisms that affect vascular ageing in centenarians and the underlying factors could offer valuable insights for developing strategies to improve overall healthy life span in the elderly. This review discusses these key factors influencing vascular ageing and how their modulation could foster healthy longevity.
Collapse
Affiliation(s)
- Sabrina Summer
- Department for Biomedical Research, University for Continuing Education Krems, Krems, Austria
| | - Maria Borrell-Pages
- Molecular Pathology and Therapeutic of Ischemic and Atherothrombotic Diseases, Institute de Recerca Sant Pau (IR-Sant Pau), Barcelona Spain. CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa-Maria Bruno
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center-PARCC, Paris, France
- Clinical Pharmacology Unit, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Rachel E Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Konstantina Dipla
- Department of Sports Sciences at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aysenur Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Kseniia Eruslanova
- Russian Gerontology Research and Clinical Centre, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Emil Fraenkel
- Department of Internal Medicine, University of Košice, Košice, Slovakia
| | | | | | - Keith D Rochfort
- School of Nursing, Psychotherapy and Community Health, Dublin City University, Dublin, Ireland
| | - Mark Ross
- Institute of Life and Earth Sciences, Heriot-Watt University, Edinburgh, UK
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Arno Schmidt-Trucksäss
- Department of Sport, Exercise and Health, Division of Sport and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Dennis Schwarz
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, University of Münster, Münster, Germany
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yahya Sohrabi
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, University of Münster, Münster, Germany
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Jannik Sonnenberg
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, University of Münster, Münster, Germany
| | | | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Josep Julve
- Endocrinology, Diabetes and Nutrition Group, Institut de Recerca SANT PAU, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Türkiye.
| |
Collapse
|
5
|
Song Y, Wang L, Wang H, Ma H, Xu J, Liu J, Qian L. Decoding aging in the heart via single cell dual omics of non-cardiomyocytes. iScience 2024; 27:111469. [PMID: 39735437 PMCID: PMC11681900 DOI: 10.1016/j.isci.2024.111469] [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: 06/11/2024] [Revised: 09/30/2024] [Accepted: 11/21/2024] [Indexed: 12/31/2024] Open
Abstract
To understand heart aging at the single-cell level, we employed single-cell dual omics (scRNA-seq and scATAC-seq) in profiling non-myocytes (non-CMs) from young, middle-aged, and elderly mice. Non-CMs, vital in heart development, physiology, and pathology, are understudied compared to cardiomyocytes. Our analysis revealed aging response heterogeneity and its dynamics over time. Immune cells, notably macrophages and neutrophils, showed significant aging alterations, while endothelial cells displayed moderate changes. We identified distinct aging signatures within the cell type, including differential gene expression, transcription factor activity, and motif variation. Sub-cluster analysis revealed intra-cell type heterogeneity, characterized by diverse aging patterns. The senescence-associated secretory phenotype emerged as a key aging-related phenotype. Moreover, aging significantly influenced cell-cell communication, especially impacting a fibroblast sub-cluster with high expression of ERBB4. This study elucidates the complex cellular and molecular landscape of cardiac aging and offers guidance for potential therapeutic avenues to treat aging-related heart diseases.
Collapse
Affiliation(s)
- Yiran Song
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Ma
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jun Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Fang Z, Wang L, Wang Y, Ma Y, Fang Y, Zhang W, Cao R, Zhang Y, Li H, Chen S, Tian L, Shen X, Cao F. Protective effects and bioinformatic analysis of narciclasine on vascular aging via cross-talk between inflammation and metabolism through inhibiting skeletal muscle-specific ceramide synthase 1. Mech Ageing Dev 2024; 223:112021. [PMID: 39706373 DOI: 10.1016/j.mad.2024.112021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE The senescence of smooth muscle is one of the independent risk factors in atherosclerosis progression in which the vascular inflammation plays an important role on vascular dysfunction. This study is designed to explore the novel vascular aging biomarkers and screen the potential molecular interventional targets through bioinformatic analysis. RESULTS Transcriptional analysis was conducted based on the GSE16487 open access database, which included 15 human vascular tissue samples from two groups: young group (≤ 60 years old, n = 8) and aged group (≥ 75 years old, n = 7). There were 275 differential expression genes (119 upregulated and 156 downregulated genes) with minimum 1.5-fold change between two groups. 9 genes were mainly participated in inflammation-related signaling pathways, in which narciclasine was validated as the most effective candidate for modulation the ceramide synthesis. In vitro and animal study demonstrated that narciclasine reversed vascular aging by inhibiting skeletal muscle-specific ceramide synthase 1 (CerS1), reducing the ceramide level derived from CerS1, and improving fat deposition and circulating glycolipid metabolism. CONCLUSION Narciclasine attenuates vascular aging and modulates the cross-talk between inflammation and metabolism via inhibiting skeletal muscle-specific ceramide synthase 1.
Collapse
Affiliation(s)
- Zhiyi Fang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 30071, China; Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Linghuan Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 30071, China; Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yabin Wang
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yan Ma
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yan Fang
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Weiwei Zhang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 30071, China; Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Ruihua Cao
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yingjie Zhang
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Hui Li
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Sijia Chen
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Lei Tian
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Xiaoying Shen
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Feng Cao
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 30071, China; Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China.
| |
Collapse
|
7
|
Auger C, Muzammel H, Diouf I, Schini-Kerth VB. Potential of Anthocyanin-rich Products to Prevent and Improve Endothelial Function and Senescence: Focus on Anthocyanins. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27590-27618. [PMID: 39629614 DOI: 10.1021/acs.jafc.4c04727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Endothelial dysfunction is a pivotal early event in the development of major cardiovascular diseases including hypertension, atherosclerosis, diabetes, and aging. The alteration of the endothelial function is often triggered by an imbalance between the endothelial formation of vasoprotective factors, including nitric oxide (NO) and endothelium-dependent hyperpolarization (EDH), and vasocontracting factors, such as arachidonic acid-derived mediators generated by cyclooxygenases, and an increased level of oxidative stress. Recently, endothelial senescence was reported to be an early trigger of endothelial dysfunction. Preclinical studies indicate that polyphenol-rich food, including anthocyanin-rich products, can activate pathways promoting an increased formation of vasoprotective factors and can prevent the induction of endothelial dysfunction in endothelial cells and isolated blood vessels. Similarly, intake of anthocyanin-rich products has been associated with the prevention and/or the improvement of an endothelial dysfunction in several experimental models of cardiovascular diseases, including physiological aging. Moreover, clinical data indicate that polyphenol-rich and anthocyanin-rich products can improve endothelial function and vascular health in humans with cardiovascular diseases. The present review will discuss both experimental and clinical evidence indicating that several polyphenol-rich foods and natural products, and especially anthocyanin-rich products, can promote endothelial and vascular health, as well as the underlying mechanisms.
Collapse
Affiliation(s)
- Cyril Auger
- University of Strasbourg, INSERM, Regenerative Nanomedicine UMR 1260, 67000 Strasbourg, France
| | - Hira Muzammel
- University of Strasbourg, Translational Cardiovascular Medicine UR 3074, 67000 Strasbourg, France
| | - Ibrahima Diouf
- University of Strasbourg, Translational Cardiovascular Medicine UR 3074, 67000 Strasbourg, France
| | - Valérie B Schini-Kerth
- University of Strasbourg, Translational Cardiovascular Medicine UR 3074, 67000 Strasbourg, France
| |
Collapse
|
8
|
Fesharaki NJ, Taylor A, Mosby K, Li R, Kim JH, Ress D. Global Impact of Aging on the Hemodynamic Response Function in the Gray Matter of Human Cerebral Cortex. Hum Brain Mapp 2024; 45:e70100. [PMID: 39692126 DOI: 10.1002/hbm.70100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/20/2024] [Accepted: 12/01/2024] [Indexed: 12/19/2024] Open
Abstract
In functional magnetic resonance imaging, the hemodynamic response function (HRF) is a stereotypical response to local changes in cerebral hemodynamics and oxygen metabolism due to briefly (< 4 s) evoked neural activity. Accordingly, the HRF is often used as an impulse response with the assumption of linearity in data analysis. In cognitive aging studies, it has been very common to interpret differences in brain activation as age-related changes in neural activity. Contrary to this assumption, however, evidence has accrued that normal aging may also significantly affect the vasculature, thereby affecting cerebral hemodynamics and metabolism, confounding interpretation of fMRI cognitive aging studies. In this study, use was made of a multisensory task to evoke the HRF in ~87% of cerebral cortex in cognitively intact adults with ages ranging from 22 to 75 years. This widespread activation enabled us to investigate age trends in the spatial distributions of HRF characteristics within the majority of cortical gray matter, which we termed as global age trends. The task evoked both positive and negative HRFs, which were characterized using model-free parameters in native-space coordinates. We found significant global age trends in the distributions of HRF parameters in terms of both amplitudes (e.g., peak amplitude and contrast-to-noise ratio) and temporal dynamics (e.g., full-width-at-half-maximum). Our findings offer insight into how age-dependent changes affect neurovascular coupling and show promise for use of HRF parameters as non-invasive indicators for age-related pathology.
Collapse
Affiliation(s)
- Nooshin J Fesharaki
- Department of Neurosurgery, University of Texas Health Science Center, Houston, Texas, USA
- Department of Neuroscience, High Resolution Brain Imaging Lab, Baylor College of Medicine, Houston, Texas, USA
| | - Amanda Taylor
- Department of Neuroscience, High Resolution Brain Imaging Lab, Baylor College of Medicine, Houston, Texas, USA
| | - Keisjon Mosby
- Department of Neuroscience, High Resolution Brain Imaging Lab, Baylor College of Medicine, Houston, Texas, USA
| | - Ruosha Li
- Department of Neurosurgery, University of Texas Health Science Center, Houston, Texas, USA
| | - Jung Hwan Kim
- Department of Neurosurgery, University of Texas Health Science Center, Houston, Texas, USA
| | - David Ress
- Department of Neuroscience, High Resolution Brain Imaging Lab, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
9
|
Song Y, Yang K, Su Y, Song K, Ding N. Nomogram for Predicting in-Hospital Severe Complications in Patients with Acute Myocardial Infarction Admitted in Emergency Department. Risk Manag Healthc Policy 2024; 17:3171-3186. [PMID: 39697902 PMCID: PMC11653858 DOI: 10.2147/rmhp.s485088] [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: 07/01/2024] [Accepted: 10/16/2024] [Indexed: 12/20/2024] Open
Abstract
Background There is lack of predictive models for the risk of severe complications during hospitalization in patients with acute myocardial infarction (AMI). In this study, we aimed to create a nomogram to forecast the likelihood of in-hospital severe complications in AMI. Methods From August 2020 to January 2023, 1024 patients with AMI including the modeling group (n=717) and the validation group (n=307) admitted in Changsha Central Hospital's emergency department. Conduct logistic regression analysis, both univariate and multivariate, on the pertinent patient data from the modeling cohort at admission, identify independent risk factors, create a nomogram to forecast the likelihood of severe complications in patients with AMI, and assess the accuracy of the graph's predictions in the validation cohort. Results Age, heart rate, mean arterial pressure, diabetes, hypertension, triglycerides and white blood cells were seven independent risk factors for serious complications in AMI patients. Based on these seven variables, the nomogram model was constructed. The nomogram has high predictive accuracy (AUC=0.793 for the modeling group and AUC=0.732 for the validation group). The calibration curve demonstrates strong consistency between the anticipated and observed values of the nomogram in the modeling and validation cohorts. Moreover, the DCA curve results show that the model has a wide threshold range (0.01-0.73) and has good practicality in clinical practice. Conclusion This study developed and validated an intuitive nomogram to assist clinicians in evaluating the probability of severe complications in AMI patients using readily available clinical data and laboratory parameters.
Collapse
Affiliation(s)
- Yaqin Song
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| | - Kongzhi Yang
- Department of Emergency Medicine, Clinical Research Center for Emergency and Critical Care in Hunan Province, Hunan Provincial Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People’s Republic of China
| | - Yingjie Su
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| | - Kun Song
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| | - Ning Ding
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
10
|
Velmurugan GV, Vekaria HJ, Patel SP, Sullivan PG, Hubbard WB. Astrocytic mitochondrial transfer to brain endothelial cells and pericytes in vivo increases with aging. J Cereb Blood Flow Metab 2024:271678X241306054. [PMID: 39668588 PMCID: PMC11638933 DOI: 10.1177/0271678x241306054] [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: 09/06/2024] [Revised: 11/01/2024] [Accepted: 11/22/2024] [Indexed: 12/14/2024]
Abstract
Intercellular mitochondrial transfer (IMT) is an intriguing biological phenomenon where mitochondria are transferred between different cells and notably, cell types. IMT is physiological, occurring in normal conditions, but also is utilized to deliver healthy mitochondria to cells in distress. Transferred mitochondria can be integrated to improve cellular metabolism, and mitochondrial function. Research on the mitochondrial transfer axis between astrocytes and brain capillaries in vivo is limited by the cellular heterogeneity of the neurovascular unit. To this end, we developed an inducible mouse model that expresses mitochondrial Dendra2 only in astrocytes and then isolated brain capillaries to remove all intact astrocytes. This method allows the visualization of in vivo astrocyte- endothelial cell (EC) and astrocyte-pericyte IMT. We demonstrate evidence of astrocyte-EC and astrocyte-pericyte mitochondrial transfer within brain capillaries. We also show that healthy aging enhances mitochondrial transfer from astrocytes to brain capillaries, revealing a potential link between brain aging and cellular mitochondrial dynamics. Finally, we observe that astrocyte-derived extracellular vesicles transfer mitochondria to brain microvascular endothelial cells, showing the potential route of in vivo IMT. These results represent a breakthrough in our understanding of IMT in the brain and a new target in brain aging and neurovascular metabolism.
Collapse
Affiliation(s)
- Gopal V Velmurugan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Hemendra J Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY, USA
| | - Samir P Patel
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY, USA
| | - W Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
11
|
Cai J, Liu Y, Fan H. Review on pathogenesis and treatment of Alzheimer's disease. Dev Dyn 2024. [PMID: 39651698 DOI: 10.1002/dvdy.762] [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: 08/17/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/11/2024] Open
Abstract
The rising incidence of Alzheimer's disease (AD) and the associated economic impacts has prompted a global focus in the field. In recent years, there has been a growing understanding of the pathogenic mechanisms of AD, including the aggregation of β-amyloid, hyperphosphorylated tau, and neuroinflammation. These processes collectively lead to neurodegeneration and cognitive decline, which ultimately results in the loss of autonomy in patients. Currently, there are three main types of AD treatments: clinical tools, pharmacological treatment, and material interventions. This review provides a comprehensive analysis of the underlying etiology and pathogenesis of AD, as well as an overview of the current prevalence of AD treatments. We believe this article can help deepen our understanding of the AD mechanism, and facilitate the clinical translation of scientific research or therapies, to address this global problem of AD.
Collapse
Affiliation(s)
- Jinxia Cai
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Yanqing Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| |
Collapse
|
12
|
Prokopidis K, Morgan PT, Veronese N, Morwani-Mangnani J, Triantafyllidis KK, Kechagias KS, Roberts J, Hurst C, Stevenson E, Vlachopoulos D, Witard OC. The effects of whey protein supplementation on indices of cardiometabolic health: A systematic review and meta-analysis of randomized controlled trials. Clin Nutr 2024; 44:109-121. [PMID: 39647241 DOI: 10.1016/j.clnu.2024.12.003] [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: 10/01/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
INTRODUCTION The increasing prevalence of cardiometabolic diseases highlights the urgent need for practical interventions to mitigate their associated public health burden. Whey protein supplementation has emerged as a potential intervention for improving markers of cardiometabolic health. The aim of this systematic review and meta-analysis was to examine the effect of whey protein ingestion on cardiometabolic profile in adults. METHODS A systematic search was conducted in PubMed, Web of Science, Scopus, and Cochrane Library from inception until June 2024. Eligible RCTs compared the effect of whey protein supplementation compared to placebo or a carbohydrate-based control on markers of cardiometabolic health. Using the random effects inverse-variance model, we estimated the mean difference (MD) in blood pressure, high- and low-density lipoproteins (HDL-cholesterol, LDL-cholesterol), total cholesterol, triglycerides, and homeostatic model assessment for insulin resistance (HOMA-IR) index. RESULTS This meta-analysis included 21 RCTs. Whey protein supplementation had no effect on HDL-cholesterol concentration but did elicit a reduction in LDL-cholesterol in individuals aged <50 years (P < 0.01) and when combined with exercise (MD: -5.38, 95 % confidence interval (95 % CI): -8.87 to -1.88, I2 = 0 %, P < 0.01). Total cholesterol was reduced with interventions that combined whey protein supplementation and exercise (MD: -8.58, -14.32 to -2.83, I2 = 55 %, P < 0.01), irrespective of age, protein dose, and body mass index ≥25 kg/m2 (MD: -6.71, 95 % CI: -11.60 to -1.83, I2 = 74 %, P < 0.01). Whey protein supplementation of ≥12 weeks was associated with reduced triglyceride levels (MD: -6.61, 95 % CI: -11.06 to -2.17, I2 = 70 %, P < 0.01). There was no clinically relevant effect of whey protein supplementation on blood pressure and HOMA-IR, however, changes pertinent to HDL-cholesterol, total cholesterol, and triglyceride reduction were primarily displayed in healthy adults. CONCLUSIONS Whey protein supplementation may be an effective intervention for reducing LDL and total cholesterol levels, particularly in healthy, overweight/obese adults aged <50 years, with the greatest benefits observed when combined with exercise. Healthy adults also showed a benefit regarding triglyceride levels.
Collapse
Affiliation(s)
- Konstantinos Prokopidis
- Department of Musculoskeletal and Ageing Science, University of Liverpool, Liverpool, UK; Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool Heart and Chest Hospital, Liverpool, UK.
| | - Paul T Morgan
- Department of Sport and Exercise Sciences, Institute of Sport, 99 Oxford Road, Manchester Metropolitan University, Manchester, UK
| | - Nicola Veronese
- Geriatric Unit, Department of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy
| | - Jordi Morwani-Mangnani
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Konstantinos S Kechagias
- Department of Obstetrics and Gynaecology, The Hillingdon Hospitals NHS Foundation Trust, Uxbridge, UK
| | - Justin Roberts
- Cambridge Centre for Sport and Exercise Sciences, School of Psychology and Sport Science, Anglia Ruskin University, Cambridge, UK
| | - Christopher Hurst
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Cumbria Northumberland Tyne and Wear NHS Foundation Trust and Faculty of Medical Sciences Newcastle University, Newcastle upon Tyne, UK
| | - Emma Stevenson
- Human Nutrition and Exercise Research Centre, School of Biomedical, Nutritional and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Dimitris Vlachopoulos
- Children's Health and Exercise Research Centre, Department of Public Health and Sport Science, University of Exeter, Exeter, UK
| | - Oliver C Witard
- Centre for Human and Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| |
Collapse
|
13
|
Behzadi P, St Hilaire C. Metabolites and metabolism in vascular calcification: links between adenosine signaling and the methionine cycle. Am J Physiol Heart Circ Physiol 2024; 327:H1361-H1375. [PMID: 39453431 PMCID: PMC11588312 DOI: 10.1152/ajpheart.00267.2024] [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: 04/29/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
The global population of individuals with cardiovascular disease is expanding, and a key risk factor for major adverse cardiovascular events is vascular calcification. The pathogenesis of cardiovascular calcification is complex and multifaceted, with external cues driving epigenetic, transcriptional, and metabolic changes that promote vascular calcification. This review provides an overview of some of the lesser understood molecular processes involved in vascular calcification and discusses the links between calcification pathogenesis and aspects of adenosine signaling and the methionine pathway; the latter of which salvages the essential amino acid methionine, but also provides the substrate critical for methylation, a modification that regulates the function and activity of DNA and proteins. We explore the complex and dynamic nature of osteogenic reprogramming underlying intimal atherosclerotic calcification and medial arterial calcification (MAC). Atherosclerotic calcification is more widely studied; however, emerging studies now show that MAC is a significant pathology independent from atherosclerosis. Furthermore, we emphasize metabolite and metabolic-modulating factors that influence vascular calcification pathogenesis. Although the contributions of these mechanisms are more well-define in relation to atherosclerotic intimal calcification, understanding these pathways may provide crucial mechanistic insights into MAC and inform future therapeutic approaches. Herein, we highlight the significance of adenosine and methyltransferase pathways as key regulators of vascular calcification pathogenesis.
Collapse
Affiliation(s)
- Parya Behzadi
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Cynthia St Hilaire
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
14
|
Peng X, Hu Y, Xu J, Chen L, Ren W, Cai W. Inverse association between serum klotho levels and C-reactive protein levels in the US population: a cross-sectional study. BMC Cardiovasc Disord 2024; 24:687. [PMID: 39614159 DOI: 10.1186/s12872-024-04375-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: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND The inverse relationship between serum Klotho levels and systemic inflammation, particularly C-reactive protein (CRP), has been suggested in limited studies. However, the association within a large and diverse population remains underexplored. METHODS We conducted a cross-sectional study using data from the National Health and Nutrition Examination Survey (NHANES) to investigate the association between serum Klotho levels and CRP among a nationally representative sample of the US population. Multiple linear regression analyses were performed to assess this relationship while adjusting for relevant covariates. Stratified analysis with interaction, restricted cubic splines (RCS) were employed to support the research objectives. RESULTS A total of 5901 participants had a mean age of 57.9 ± 11.0 years, with 49.4% of them being male and 50.6% of them being female. A negative association between serum Klotho and CRP was revealed in the fully adjusted model (β -0.26; 95% CI -0.41∼-0.11). When serum Klotho was taken as quartiles with Q1 as reference, the adjusted β that were lowest in Q4 were - 0.1 (95% CI -0.16∼-0.04, p-value = 0.002) in model 4, respectively. These statistics were robust in stratified analyses. CONCLUSION While our study demonstrates an inverse association between serum Klotho levels and CRP, suggesting a potential cardioprotective role of Klotho, it is important to note that our cross-sectional design does not permit the establishment of causality. Therefore, we cannot definitively conclude that increasing Klotho levels will directly reduce cardiovascular risk. Our findings do, however, highlight the need for further research to explore the potential of Klotho as a therapeutic target for cardiovascular health.
Collapse
Affiliation(s)
- Xuelan Peng
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yingjie Hu
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jiarong Xu
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ling Chen
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wei Ren
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wenzhi Cai
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China.
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
15
|
Deng S, Lao M, Zheng H, Hao J. Forkhead box P1 transcriptionally activates IGF-1 to lighten ox-LDL-induced endothelial cellular senescence by inactivating NLRP3 inflammasome. Biogerontology 2024; 26:15. [PMID: 39585426 DOI: 10.1007/s10522-024-10151-5] [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: 05/07/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
Endothelial cell (EC) senescence is a major contributor in atherosclerosis (AS) development. Herein, the role of forkhead box P transcription factor 1 (FOXP1) and insulin-like growth factor (IGF)-1 in regulating EC senescence during AS progression was investigated. The mRNA and protein expressions were assessed using qRT-PCR and western blot. IL-1β and IL-18 secretion levels were analyzed by ELISA. Cell viability and pyroptosis were determined by MTT assay and flow cytometry, respectively. SA β-Gal staining was used to measure cell senescence. Tube formation assay was adopted to detect the angiogenesis ability. Dual-luciferase reporter and ChIP assays were used to investigate the relationship between FOXP1 and IGF‑1. ox-LDL stimulation significantly reduced FOXP1 and IGF-1 expression levels in human aortic endothelial cells (HAECs). FOXP1 or IGF-1 overexpression both mitigated ox-LDL-induced cellular senescence and NLRP3 activation in HAECs. It was subsequently revealed that FOXB1 transcriptionally activated IGF-1 expression in HAECs by binding to IGF-1 promoter. Rescue experiments demonstrated that IGF-1 silencing abolished the inhibitory impact of FOXP1 overexpression on ox-LDL-induced cellular senescence and NLRP3 activation in HAECs. FOXP1 transcriptionally activated IGF-1 to lighten ox-LDL-induced endothelial cellular senescence by inactivating NLRP3 inflammasome.
Collapse
Affiliation(s)
- Siqi Deng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Meili Lao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Huihui Zheng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Jingwen Hao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China.
| |
Collapse
|
16
|
Wojtacha JJ, Morawin B, Wawrzyniak-Gramacka E, Tylutka A, de Freitas AKE, Zembron-Lacny A. Endothelial Dysfunction with Aging: Does Sex Matter? Int J Mol Sci 2024; 25:12203. [PMID: 39596269 PMCID: PMC11594464 DOI: 10.3390/ijms252212203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress and inflammation accompany endothelial dysfunction that results from the excessive or uncontrolled production of reactive oxygen and nitrogen species (RONS) in older adults. This study was designed to assess the usefulness of serum oxi-inflammatory component combinations in vascular disease prediction and prevention with regard to sex. Women (n = 145) and men (n = 50) aged 72.2 ± 7.8 years participated in this project. The females demonstrated the elevated production of hydrogen peroxide (H2O2) and nitric oxide (NO) responsible for intravascular low-density lipoprotein oxidation. NO generation was enhanced in the women, but its bioavailability was reduced, which was expressed by a high 3-nitrotyrosine (3-NitroT) concentration. The relation of NO/3-NitroT (rs = 0.811, p < 0.001) in the women and NO/3-NitroT (rs = -0.611, p < 0.001) in the men showed that sex determines endothelial dysfunction. RONS generation in the women simultaneously promoted endothelial regeneration, as demonstrated by a ~1.5-fold increase in circulating progenitor cells. Inflammation-specific variables, such as the neutrophil-to-lymphocyte ratio, the systemic immune inflammation index, and the neutrophil-to-high-density lipoprotein (HDL) ratio, were reduced in the women and showed their diagnostic utility for clinical prognosis in vascular dysfunction, especially the C-reactive-protein-to-HDL ratio (AUC = 0.980, specificity 94.7%, sensitivity 93.3%, OR = 252, 95% CI 65-967, p < 0.001). This study is the first to have revealed sex-specific changes in the oxi-inflammatory response, which can generate the risk of cardiovascular events at an older age.
Collapse
Affiliation(s)
- Jakub Jozue Wojtacha
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Barbara Morawin
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Edyta Wawrzyniak-Gramacka
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Anna Tylutka
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Ana Karyn Ehrenfried de Freitas
- School of Health Science, Positivo University, 5300 Professor Pedro Viriato Parigot de Souza Street, Campo Comprido, Curitiba 81280-330, PR, Brazil;
- Department of Cardiology, Hospital da Cruz Vermelha Brasileira Filial do Paraná, Av. Vicente Machado, 1280, R. Cap. Souza Franco, 50-Batel, Curitiba 80420-011, PR, Brazil
| | - Agnieszka Zembron-Lacny
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| |
Collapse
|
17
|
Kim Y, Mattos MK, Esquivel JH, Davis EM, Logan J. Sleep and blood pressure variability: A systematic literature review. Heart Lung 2024; 68:323-336. [PMID: 39217647 DOI: 10.1016/j.hrtlng.2024.08.016] [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: 04/20/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Blood pressure variability (BPV) is a prognostic marker of cardiovascular disease (CVD). Sleep is recognized as a significant risk factor for CVD; however, little is known about the relationship between sleep characteristics and BPV. OBJECTIVE In this systematic review, we aimed to (1) describe methods used to measure BPV and sleep and (2) describe the current evidence in the literature on the association between sleep and BPV. METHODS A systematic search was conducted using the search terms "sleep" AND ("blood pressure variability" OR "ambulatory blood pressure monitor") in CINAHL, PubMed, Web of Science, and PsycINFO databases. RESULTS Twenty-two studies were included in this systematic review. Sleep was measured using various methods, including polysomnography, actigraphy, sleep diaries, and questionnaires, while BPV was measured over various time intervals using different monitoring devices such as a beat-to-beat blood pressure (BP) monitoring device, a 24-h ambulatory BP monitor, or an automatic upper arm BP monitor. The studies demonstrated mixed results on the associations between sleep parameters (sleep quality, architecture, and duration) and increased BPV. CONCLUSIONS Although the mechanisms that explain the relationship between sleep and BPV are still unclear, accumulating evidence suggests potential associations between increased BPV with poor sleep quality and longer sleep duration. Given the recent development of sleep and BP monitoring technologies, further research is warranted to assess sleep and BPV under free-living conditions. Such studies will advance our understanding of complex interactions between sleep and CVD risk.
Collapse
Affiliation(s)
- Yeonsu Kim
- University of Virginia School of Nursing, 225 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA.
| | - Meghan Kathleen Mattos
- University of Virginia School of Nursing, 5012 McLeod Hall, 202 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA
| | - Jill Howie Esquivel
- University of California San Francisco School of Nursing, 2 Koret Way, San Francisco, CA, 94143, USA
| | - Eric M Davis
- Department of Medicine, University of Virginia, 1222 Jefferson Park Ave, Charlottesville, VA, 22903, USA
| | - Jeongok Logan
- University of Virginia School of Nursing, 4011 McLeod Hall, 202 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA
| |
Collapse
|
18
|
Liu WN, Huang HL, Lan Y, Li L, Yang ZL, Jiang L. Hyperhomocysteine promotes cataract development through mTOR-mediated inhibition of autophagy and connexins expression. Int Immunopharmacol 2024; 140:112827. [PMID: 39116497 DOI: 10.1016/j.intimp.2024.112827] [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: 03/13/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
AIM Hyperhomocysteine has been recognized as an independent risk factor of multiple diseases, including several eye diseases. In this study, we aim to investigate whether increased homocysteine (Hcy) is related to cataracts, and to explore whether dysregulation of mTOR-mediated autophagy and connexin expression are underlying mechanisms. METHOD We first developed a method of liquid chromatography tandem mass spectrometry to accurately measure serum concentrations of Hcy in 287 cataract patients and 334 healthy controls. Next, we treated human lens epithelial (HLC-B3) cells with Hcy at different concentrations and durations, and then analyzed expression of autophagy-related markers and connexins, as well as phosphorylated mTOR (p-mTOR) in these cells by Western blotting. Formation of autophagic vacuoles and intracellular Ca2+ in the Hcy-treated cells were observed by fluorescence microscopy. Further, we performed a rescue experiment in the Hcy-treated HLC-B3 cells by pre-incubation with rapamycin, an mTOR inhibitor. RESULTS The serum levels of Hcy in patients with cataracts were significantly increased compared to those in healthy controls. In cultured HLC-B3 cells, expression of autophagy related markers (LC3B and Beclin1) and connexins (Cx43 and Cx50) was inhibited by Hcy treatment in a dose- and duration-dependent manner. Accumulation of Ca2+ in the Hcy-treated lens epithelial cells was observed as a consequence of reduced connexin expression. Meanwhile, expression of p-mTOR increased, representing up-regulation of the mTOR pathway. Importantly, inhibition of autophagy and connexin expression due to hyperhomocysteine was rescued via mTOR suppression by pretreatment with rapamycin in HLC-B3 cells. CONCLUSION Our results demonstrate that hyperhomocysteine might promote cataract development through two mTOR-mediated pathways in the lens epithelial cells: 1) dysregulation of autophagy and 2) accumulation of intracellular calcium via decreased connexin expression.
Collapse
Affiliation(s)
- Wen-Na Liu
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong-Lang Huang
- Department of Clinical Laboratory, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei 442008 China
| | - Yu Lan
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Li
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zheng-Lin Yang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China
| | - Li Jiang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences, Chengdu, Sichuan 610072, China.
| |
Collapse
|
19
|
Wang M, Mo D, Zhou C, Guo M, Zhang W, Chen R, Xu J, Zhang N, Yu H. Association between Ward's triangle bone mineral density levels and abdominal aortic calcification: Data from the national health and nutrition examination survey 2013-2014. Clin Nutr ESPEN 2024; 64:344-357. [PMID: 39461592 DOI: 10.1016/j.clnesp.2024.10.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/05/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND & AIMS Despite extensive research into the cardiovascular implications of abdominal aortic calcification (AAC), there is a scarcity of robust studies exploring its association with Ward's triangle bone mineral density (BMD). This study aimed to evaluate this relationship in a nationally representative sample and compare the predictive value with femoral neck BMD and total femur BMD. METHODS We conducted a cross-sectional analysis of 2013-2014 National Health and Nutrition Examination Survey (NHANES) data, utilizing a complex, stratified, multistage, cluster sampling design. BMD measurements at Ward's triangle, femoral neck, and total femur were assessed using DXA scans. AAC severity was defined by a Kauppila score of ≥5. Predictors of AAC-24 scores were identified through correlation and linear regression models. Stratified regression and restricted cubic splines were applied to explore subgroup and dose-response relationships. RESULTS Of the 2965 participants representing 116, 562, 500 individuals in the U.S., 11 % had severe AAC. Ward's triangle BMD showed a significant negative association with AAC-24 scores (β = -1.90, 95 % CI: -2.80 to -1.00, P < 0.002) and a reduced risk of severe AAC (OR = 0.85, 95 % CI: 0.76 to 0.95, P = 0.010). Non-linear associations were observed between Ward's triangle BMD and AAC outcomes. Ward's triangle BMD outperformed femoral neck and total femur BMD in predicting AAC. CONCLUSIONS Higher Ward's triangle BMD is linked to lower odds of severe AAC, highlighting its potential for improved early detection of AAC over femoral neck and total femur BMD. Healthcare providers should consider the implications of reduced Ward's triangle BMD for systemic atherosclerosis and recommend early AAC screening for enhanced cardiovascular risk management.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China; Institute of Cardiovascular Disease, Qingdao University, Qingdao 266001, Shandong, China.
| | - Degang Mo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Chi Zhou
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Mengqi Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Wenqiang Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Rui Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Jiachao Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Ning Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China.
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China; Institute of Cardiovascular Disease, Qingdao University, Qingdao 266001, Shandong, China.
| |
Collapse
|
20
|
Cheng CK, Ye L, Zuo Y, Wang Y, Wang L, Li F, Chen S, Huang Y. Aged Gut Microbiome Induces Metabolic Impairment and Hallmarks of Vascular and Intestinal Aging in Young Mice. Antioxidants (Basel) 2024; 13:1250. [PMID: 39456503 PMCID: PMC11505429 DOI: 10.3390/antiox13101250] [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: 09/20/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Aging, an independent risk factor for cardiometabolic diseases, refers to a progressive deterioration in physiological function, characterized by 12 established hallmarks. Vascular aging is driven by endothelial dysfunction, telomere dysfunction, oxidative stress, and vascular inflammation. This study investigated whether aged gut microbiome promotes vascular aging and metabolic impairment. Fecal microbiome transfer (FMT) was conducted from aged (>75 weeks old) to young C57BL/6 mice (8 weeks old) for 6 weeks. Wire myography was used to evaluate endothelial function in aortas and mesenteric arteries. ROS levels were measured by dihydroethidium (DHE) staining and lucigenin-enhanced chemiluminescence. Vascular and intestinal telomere function, in terms of relative telomere length, telomerase reverse transcriptase expression and telomerase activity, were measured. Systemic inflammation, endotoxemia and intestinal integrity of mice were assessed. Gut microbiome profiles were studied by 16S rRNA sequencing. Some middle-aged mice (40-42 weeks old) were subjected to chronic metformin treatment and exercise training for 4 weeks to evaluate their anti-aging benefits. Six-week FMT impaired glucose homeostasis and caused vascular dysfunction in aortas and mesenteric arteries in young mice. FMT triggered vascular inflammation and oxidative stress, along with declined telomerase activity and shorter telomere length in aortas. Additionally, FMT impaired intestinal integrity, and triggered AMPK inactivation and telomere dysfunction in intestines, potentially attributed to the altered gut microbial profiles. Metformin treatment and moderate exercise improved integrity, AMPK activation and telomere function in mouse intestines. Our data highlight aged microbiome as a mechanism that accelerates intestinal and vascular aging, suggesting the gut-vascular connection as a potential intervention target against cardiovascular aging and complications.
Collapse
Affiliation(s)
- Chak-Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China;
| | - Lianwei Ye
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China; (L.Y.); (F.L.)
| | - Yuanyuan Zuo
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Yaling Wang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, China; (Y.W.); (S.C.)
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China;
| | - Fuyong Li
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China; (L.Y.); (F.L.)
| | - Sheng Chen
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, China; (Y.W.); (S.C.)
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China;
| |
Collapse
|
21
|
Ahmed B, Rahman AA, Lee S, Malhotra R. The Implications of Aging on Vascular Health. Int J Mol Sci 2024; 25:11188. [PMID: 39456971 PMCID: PMC11508873 DOI: 10.3390/ijms252011188] [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: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Vascular aging encompasses structural and functional changes in the vasculature, significantly contributing to cardiovascular diseases, which are the leading cause of death globally. The incidence and prevalence of these diseases increase with age, with most morbidity and mortality attributed to myocardial infarction and stroke. Diagnosing and intervening in vascular aging while understanding the mechanisms behind age-induced vascular phenotypic and pathophysiological alterations offers the potential for delaying and preventing cardiovascular mortality in an aging population. This review delves into various aspects of vascular aging by examining age-related changes in arterial health at the cellular level, including endothelial dysfunction, cellular senescence, and vascular smooth muscle cell transdifferentiation, as well as at the structural level, including arterial stiffness and changes in wall thickness and diameter. We also explore aging-related changes in perivascular adipose tissue deposition, arterial collateralization, and calcification, providing insights into the physiological and pathological implications. Overall, aging induces phenotypic changes that augment the vascular system's susceptibility to disease, even in the absence of traditional risk factors, such as hypertension, diabetes, obesity, and smoking. Overall, age-related modifications in cellular phenotype and molecular homeostasis increase the vulnerability of the arterial vasculature to structural and functional alterations, thereby accelerating cardiovascular risk. Increasing our understanding of these modifications is crucial for success in delaying or preventing cardiovascular diseases. Non-invasive techniques, such as measuring carotid intima-media thickness, pulse wave velocity, and flow-mediated dilation, as well as detecting vascular calcifications, can be used for the early detection of vascular aging. Targeting specific pathological mechanisms, such as cellular senescence and enhancing angiogenesis, holds promise for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Ahmed A. Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sujin Lee
- Division of Vascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
22
|
Mury P, Cagnone G, Dagher O, Wünnemann F, Voghel G, Beaudoin M, Lambert M, Miquel G, Noly PE, Perrault LP, Carrier M, Thorin-Trescases N, Joyal JS, Lettre G, Thorin E. Senescence and Inflamm-Aging Are Associated With Endothelial Dysfunction in Men But Not Women With Atherosclerosis. JACC Basic Transl Sci 2024; 9:1163-1177. [PMID: 39534645 PMCID: PMC11551873 DOI: 10.1016/j.jacbts.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 11/16/2024]
Abstract
Coronary artery disease (CAD) is more prevalent in men than in women, with endothelial dysfunction, prodromal to CAD, developing a decade earlier in middle-aged men. We investigated the molecular basis of this dimorphism ex vivo in arterial segments discarded during surgery of CAD patients. The results reveal a lower endothelial relaxant sensitivity in men, and a senescence-associated inflammaging transcriptomic signature in endothelial cells. In women, cellular metabolism and endothelial maintenance pathways are conserved. This suggests that senolytic therapies to reduce risk of cardiovascular events in women with CAD may not be as effective as in men.
Collapse
Affiliation(s)
- Pauline Mury
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Gael Cagnone
- University Hospital Sainte Justine Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Olina Dagher
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Calgary, Alberta, Canada
| | - Florian Wünnemann
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Guillaume Voghel
- Department of Family Medicine and Emergency Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Melissa Beaudoin
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Mélanie Lambert
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Géraldine Miquel
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Pierre-Emmanuel Noly
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Louis P. Perrault
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Michel Carrier
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | | | - Jean-Sébastien Joyal
- University Hospital Sainte Justine Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Department of Ophthalmology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Eric Thorin
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
23
|
Chen Q, Hu R, Qiu H, Li S, Xiang P, Lu Y, Wang X, Wang T, Zhou L, Zhang W, Wen E, Ma L, Yu C. REDD1 knockdown ameliorates endothelial cell senescence through repressing TXNIP-mediated oxidative stress. Mech Ageing Dev 2024; 221:111962. [PMID: 39004152 DOI: 10.1016/j.mad.2024.111962] [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: 05/11/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024]
Abstract
Endothelial cell senescence characterized by reactive oxygen species (ROS) accumulation and chronic inflammation is widely recognized as a key contributor to atherosclerosis (AS). Regulated in development and DNA damage response 1 (REDD1), a conserved stress-response protein that regulates ROS production, is involved in the pathogenesis of various age-related diseases. However, the role of REDD1 in endothelial cell senescence is still unclear. Here, we screened REDD1 as a differentially expressed senescence-related gene in the AS progression using bioinformatics methods, and validated the upregulation of REDD1 expression in AS plaques, senescent endothelial cells, and aging aorta by constructing AS mice, D-galactose (DG)-induced senescent endothelial cells and DG-induced accelerated aging mice, respectively. siRNA against REDD1 could improve DG-induced premature senescence of endothelial cells and inhibit ROS accumulation, similar to antioxidant N-Acetylcysteine (NAC) treatment. Meanwhile, NAC reduced the upregulation of REDD1 induced by DG, supporting the positive feedback loop between REDD1 and ROS contributes to endothelial cell senescence. Mechanistically, the regulatory effect of REDD1 on ROS might be related to the TXNIP-REDD1 interaction in DG-induced endothelial cell senescence. Collectively, experiments above provide evidence that REDD1 participates in endothelial cell senescence through repressing TXNIP-mediated oxidative stress, which may be involved in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Qingqiu Chen
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Rong Hu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Hongmei Qiu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Shan Li
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Peng Xiang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Yining Lu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Xianmin Wang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Tongchuan Wang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Lan Zhou
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - Wanping Zhang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China
| | - E Wen
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Limei Ma
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China.
| | - Chao Yu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
24
|
Sabouri M, Zheng X, Irwin BJ, Machin DR. Effects of excess sodium consumption on arterial function in C57BL/6 mice. Am J Physiol Heart Circ Physiol 2024; 327:H896-H907. [PMID: 39150393 PMCID: PMC11482244 DOI: 10.1152/ajpheart.00242.2024] [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: 04/17/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Excess sodium consumption contributes to arterial dysfunction in humans. The C57BL/6 strain of mice has been used to identify mechanisms by which arterial dysfunction occurs after excess sodium consumption. However, there are concerns that C57BL/6 mice have strain-specific resistance to high-sodium (HS) diet-induced hypertension. To address this concern, we performed a meta-analysis to determine if excess sodium consumption in C57BL/6 mice induces arterial dysfunction. Databases were searched for HS versus standard diet studies that measured arterial function [i.e., systolic blood pressure (BP), endothelium-dependent dilation (EDD), and central arterial stiffness] in C57BL/6 mice. A total of 39 studies were included, demonstrating that the HS condition resulted in higher systolic BP than control mice with a mean difference of 9.8 mmHg (95% confidence interval [CI] = [5.6, 14], P < 0.001). Subgroup analysis indicated that the systolic BP was higher in HS compared with the control condition when measured during night compared with daytime with telemetry (P < 0.001). We also identified that the difference in systolic BP between HS and control was ∼2.5-fold higher when administered through drinking water than through food (P < 0.001). A total of 12 studies were included, demonstrating that the HS condition resulted in lower EDD than control with a weighted mean difference of -12.0% (95% CI = [-20.0, -4.1], P = 0.003). It should be noted that there was considerable variability across studies with more than half of the studies showing no effect of the HS condition on systolic BP or EDD. In summary, excess sodium consumption elevates systolic BP and impairs EDD in C57BL/6 mice.NEW & NOTEWORTHY C57BL/6 mice are perceived as resistant to high-sodium diet-induced arterial dysfunction. This meta-analysis demonstrates that excess sodium consumption elevates blood pressure and impairs endothelium-dependent dilation in C57BL/6 mice. Nighttime measurements show more pronounced blood pressure elevation. In addition, sodium administration via drinking water, compared with food, induces a greater blood pressure elevation. These findings may be influenced by outlier studies, as the majority of studies showed no adverse effect of excess sodium consumption on arterial function.
Collapse
Affiliation(s)
- Mostafa Sabouri
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, United States
| | - Xiangyu Zheng
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, United States
| | - Bryan J Irwin
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States
| | - Daniel R Machin
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
25
|
Du R, Liu J, Tang X, Chen Z, Guan L, Gao W, Huang W. Correlation of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio with serum α-klotho levels in US middle-aged and older individuals: Results from NHANES 2007-2016. Prev Med Rep 2024; 46:102877. [PMID: 39309699 PMCID: PMC11415581 DOI: 10.1016/j.pmedr.2024.102877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Background The α-klotho (αKl) is widely accepted as an anti-aging and anti-inflammatory protein. However, it is rarely reported on the function and mechanism of αKl in the overall population (including healthy people and those with history of chronic disease). The neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) are established as predictors of systemic inflammation. This study aims to investigate the relationship between NLR, PLR, and αKl levels in the overall population. Methods Data from 10,124 adults aged 40 years old and above, collected from NHANES 2007-2016, were analyzed. Associations between NLR, PLR, and αKl levels were assessed by weighted multivariate linear regression analyses, adjusting for potential confounders. Subgroup analysis was conducted by gender, age, diabetes, cardiovascular disease, and chronic kidney disease. Results Weighted linear regression models showed that a significant negative correlation was observed between both NLR and PLR with αKl levels. Subgroup analysis revealed that the negative correlation between NLR and serum αKl levels was not significant in individuals aged 40-59 years and males, while this relationship remained stable across most other subgroups. The negative correlation between PLR and serum αKl levels was consistent across most subgroups but not significant in individuals with cardiovascular disease. Conclusion Our study revealed a significant negative relationship between inflammatory markers (NLR and PLR) and serum αKl levels, suggesting systemic inflammation may be linked to reduced αKl expression. Subgroup analyses showed that the relationship varies across different demographic and health-related factors. We provided insight into the significance of managing inflammation and preserving αKl levels.
Collapse
Affiliation(s)
- Rui Du
- Department of Ultrasound, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, Hubei, China
| | - Jie Liu
- Department of Ultrasound, International Mongolian Hospital of Inner Mongolia, No. 83, University East Street, Hohhot 010020, Inner Mongolia, China
| | - Xiaoyan Tang
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, Hubei, China
| | - Zili Chen
- Department of Ultrasound, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, Hubei, China
| | - Lei Guan
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, Hubei, China
| | - WenHong Gao
- Department of Ultrasound, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, Hubei, China
| | - Wei Huang
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, Hubei, China
| |
Collapse
|
26
|
Shabanian K, Shabanian T, Karsai G, Pontiggia L, Paneni F, Ruschitzka F, Beer JH, Saeedi Saravi SS. AQP1 differentially orchestrates endothelial cell senescence. Redox Biol 2024; 76:103317. [PMID: 39180980 PMCID: PMC11388013 DOI: 10.1016/j.redox.2024.103317] [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: 03/18/2024] [Revised: 07/05/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Accumulation of senescent endothelial cells (ECs) with age is a pivotal driver of cardiovascular diseases in aging. However, little is known about the mechanisms and signaling pathways that regulate EC senescence. In this report, we delineate a previously unrecognized role of aquaporin 1 (AQP1) in orchestrating extracellular hydrogen peroxide (H2O2)-induced cellular senescence in aortic ECs. Our findings underscore AQP1's differential impact on senescence hallmarks, including cell-cycle arrest, senescence-associated secretory phenotype (SASP), and DNA damage responses, intricately regulating angiogenesis. In proliferating ECs, AQP1 is crucial for maintaining angiogenic capacity, whereas disruption of AQP1 induces morphological and mitochondrial alterations, culminating in senescence and impaired angiogenesis. Conversely, Aqp1 knockdown or selective blockade of AQP1 in senescent ECs rescues the excess H2O2-induced cellular senescence phenotype and metabolic dysfunction, thereby ameliorating intrinsic angiogenic incompetence. Mechanistically, AQP1 facilitates H2O2 transmembrane transport, exacerbating oxidant-sensitive kinases CaMKII-AMPK. This process suppresses HDAC4 translocation, consequently de-repressing Mef2A-eNOS signaling in proliferating ECs. However, in senescent ECs, AQP1 overexpression is linked to preserved HDAC4-Mef2A complex and downregulation of eNOS signaling. Together, our studies identify AQP1 as a novel epigenetic regulator of HDAC4-Mef2A-dependent EC senescence and angiogenic potential, highlighting its potential as a therapeutic target for antagonizing age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Khatereh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Taraneh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zurich, 8952, Schlieren, Switzerland
| | - Luca Pontiggia
- Tissue Biology Research Unit, University Children's Hospital Zurich, 8952, Schlieren, Switzerland; Children's Research Center, University Children's Hospital Zurich, 8032, Zurich, Switzerland; Faculty of Medicine, University of Zurich, 8032, Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404, Baden, Switzerland.
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
27
|
Jeong JH, Heo M, Park S, Lee SH, Park O, Kim T, Yeo HJ, Jang JH, Cho WH, Yoo JW. Association between Age-Adjusted Endothelial Activation and Stress Index and Intensive Care Unit Mortality in Patients with Severe COVID-19. Tuberc Respir Dis (Seoul) 2024; 87:524-531. [PMID: 39044336 PMCID: PMC11468444 DOI: 10.4046/trd.2024.0081] [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: 05/31/2024] [Revised: 06/27/2024] [Accepted: 07/21/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Endothelial activation and stress index (EASIX) reflects endothelial dysfunction or damage. Because endothelial dysfunction is one of the key mechanisms, a few studies have shown the clinical usefulness of original and age-adjusted EASIX (age-EASIX) in patients with coronavirus disease 2019 (COVID-19). We aimed to evaluate the clinical utility of age-EASIX in predicting intensive care unit (ICU) mortality in critically ill patients with COVID-19 in South Korea. METHODS Secondary analysis was performed using clinical data retrospectively collected from 22 nationwide hospitals in South Korea between January 1, 2020, and August 31, 2021. Patients were at least 19 years old and admitted to the ICU for severe COVID-19, demanding at least high-flow nasal cannula oxygen therapy. EASIX [lactate dehydrogenase (U/L)×creatinine (mg/dL)/platelet count (109 cells/L)] and age-EASIX (EASIX×age) were calculated and log2-transformed. RESULTS The mean age of 908 critically ill patients with COVID-19 was 67.4 years with 59.7% male sex. The mean log2 age-EASIX was 7.38±1.45. Non-survivors (n=222, 24.4%) in the ICU had a significantly higher log2 age-EASIX than of survivors (8.2±1.52 vs. 7.1±1.32, p<0.001). log2 age-EASIX was significantly associated with ICU mortality (odds ratio, 1.541; 95% confidence interval, 1.322 to 1.796; p<0.001) and had a better area under the receiver operating characteristic curve than of the sequential organ failure assessment (SOFA) score in predicting ICU mortality (0.730 vs. 0.660, p=0.001). CONCLUSION Age-EASIX is significantly associated with ICU mortality and has better discriminatory ability than the SOFA score in predicting ICU mortality.
Collapse
Affiliation(s)
- Jong Hwan Jeong
- Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Manbong Heo
- Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Sunghoon Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Su Hwan Lee
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Onyu Park
- BioMedical Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Taehwa Kim
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Internal Medicine, Transplant Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Hye Ju Yeo
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Internal Medicine, Transplant Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jin Ho Jang
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Internal Medicine, Transplant Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Woo Hyun Cho
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Internal Medicine, Transplant Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jung-Wan Yoo
- Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - on behalf of the Korean Intensive Care Study Group
- Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
- Division of Pulmonary, Allergy and Critical Care Medicine, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- BioMedical Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Internal Medicine, Transplant Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| |
Collapse
|
28
|
Schini-Kerth VB, Diouf I, Muzammel H, Said A, Auger C. Natural Products to Promote Vascular Health. Handb Exp Pharmacol 2024. [PMID: 39317849 DOI: 10.1007/164_2024_721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Maintaining good vascular health is a major component in healthy ageing as it reduces the risk of cardiovascular diseases. Endothelial dysfunction, in particular, is a key mechanism in the development of major cardiovascular diseases including hypertension, atherosclerosis and diabetes. Recently, endothelial senescence has emerged as a pivotal early event in age-related endothelial dysfunction. Endothelial function is characterized by an imbalance between the endothelial formation of vasoprotective mechanisms, including the formation of nitric oxide (NO) and endothelium-dependent hyperpolarization responses, and an increased level of oxidative stress involving several pro-oxidant enzymes such as NADPH oxidases and, often also, the appearance of cyclooxygenase-derived vasoconstrictors. Pre-clinical studies have indicated that natural products, in particular several polyphenol-rich foods, can trigger activating pathways in endothelial cells promoting an increased formation of NO and endothelium-dependent hyperpolarization. In addition, some can even exert beneficial effects on endothelial senescence. Moreover, some of these products have been associated with the prevention and/or improvement of established endothelial dysfunction in several experimental models of cardiovascular diseases and in humans with cardiovascular diseases. Therefore, intake of certain natural products, such as dietary and plant-derived polyphenol-rich products, appears to be an attractive approach for a healthy vascular system in ageing.
Collapse
Affiliation(s)
- Valérie B Schini-Kerth
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France.
| | - Ibrahima Diouf
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Hira Muzammel
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Amissi Said
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Cyril Auger
- Regenerative Nanomedicine, INSERM UMR 1260, CRBS, University of Strasbourg, Strasbourg, France
| |
Collapse
|
29
|
Izemrane D, Benziane A, Makrelouf M, Hamdis N, Rabia SH, Boudjellaba S, Baz A, Benaziza D. Living donors kidney transplantation and oxidative stress: Nitric oxide as a predictive marker of graft function. PLoS One 2024; 19:e0307824. [PMID: 39312562 PMCID: PMC11419388 DOI: 10.1371/journal.pone.0307824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 07/10/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Glomerular filtration rate is the best indicator of renal function and a predictor of graft and patient survival after kidney transplantation. METHODS In a single-centre prospective analysis, we assessed the predictive performances of 4 oxidative stress biomarkers in estimating graft function at 6 months and 1 year after kidney transplantation from living donors. Blood samples were achieved on days (D-1, D1, D2, D3, D6 and D8), months (M1, M3 and M6) and after one year (1Y). For donors, a blood sample was collected on D-1. Malondialdehyde (MDA), nitric oxide (NO), glutathione s-transferase (GST), myeloperoxydase (MPO), and creatinine (Cr) were measured by spectrophotometric essays. The estimated glomerular filtration rate by the modification of diet in renal disease equation (MDRD-eGFR) was used to assess renal function in 32 consecutive donor-recipient pairs. Pearson's and Spearman's correlations have been applied to filter out variables and covariables that can be used to build predictive models of graft function at six months and one year. The predictive performances of NO and MPO were tested by multivariable stepwise linear regression to estimate glomerular filtration rate at six months. RESULTS Three models with the highest coefficients of determination stand out, combining the two variables nitric oxide at day 6 and an MDRD-eGFR variable at day 6 or MDRD-eGFR at day 21 or MDRD-eGFR at 3 months, associated for the first two models or not for the third model with donor age as a covariable (P = 0.000, r2 = 0.599, r2adj = 0.549; P = 0.000, r2 = 0.548, r2adj = 0.497; P = 0.000, r2 = 0.553, r2adj = 0.517 respectively). CONCLUSION Quantification of nitric oxide at day six could be useful in predicting graft function at six months in association with donor age and the estimated glomerular filtration rate in recipient at day 6, day 21 and 3 months after transplantation.
Collapse
Affiliation(s)
- Djamila Izemrane
- Laboratory of Biology and Animal Physiology, Higher Normal School, Kouba, Algiers, Algeria
- National Higher Veterinary School, Issad Abbes, Oued Smar, Algiers, Algeria
| | - Ali Benziane
- Department of Nephrology-Hemodialysis and Transplantation, Lamine Debaghine University Hospital, Bab El Oued, Algiers, Algeria
| | - Mohamed Makrelouf
- Central Biology Laboratory, Lamine Debaghine University Hospital, Bab El Oued, Algiers, Algeria
| | - Nacim Hamdis
- Laboratory of Food Technology Research, Faculty of Engineering Sciences-University M’Hamed Bougara, City Frantz Fanon, Boumerdes, Algeria
| | - Samia Hadj Rabia
- Laboratory of Biology and Animal Physiology, Higher Normal School, Kouba, Algiers, Algeria
- Department of Nuclear Applications, Nuclear Research Center, Sebala, Algiers, Algeria
| | - Sofiane Boudjellaba
- National Higher Veterinary School, Issad Abbes, Oued Smar, Algiers, Algeria
- Laboratory of Research Management of Local Animal Resources (GRAL), National Higher Veterinary School, Issad Abbes, Oued Smar, Algiers, Algeria
| | - Ahsene Baz
- Laboratory of Biology and Animal Physiology, Higher Normal School, Kouba, Algiers, Algeria
| | - Djamila Benaziza
- Laboratory of Biology and Animal Physiology, Higher Normal School, Kouba, Algiers, Algeria
| |
Collapse
|
30
|
Wang H, Zeng P, Zhu PH, Wang ZF, Cai YJ, Deng CY, Yang H, Mai LP, Zhang MZ, Kuang SJ, Rao F, Xu JS. Downregulation of stromal interaction molecule-1 is implicated in the age-associated vasoconstriction dysfunction of aorta, intrarenal, and coronary arteries. Eur J Pharmacol 2024; 979:176832. [PMID: 39038639 DOI: 10.1016/j.ejphar.2024.176832] [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: 03/13/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
The contractile function of vascular smooth muscle cells (VSMCs) typically undergoes significant changes with advancing age, leading to severe vascular aging-related diseases. The precise role and mechanism of stromal interaction molecule-1 (STIM1) in age-mediated Ca2+ signaling and vasocontraction remain unclear. The connection between STIM1 and age-related vascular dysfunction was investigated using a multi-myograph system, immunohistochemical analysis, protein blotting, and SA-β-gal staining. Results showed that vasoconstrictor responses in the thoracic aorta, intrarenal artery, and coronary artery decreased with age. STIM1 knockdown in the intrarenal and coronary arteries reduced vascular tone in young mice, while no change was observed in the thoracic aorta. A significant reduction in vascular tone occurred in the STIM1 knockout group with nifedipine. In the thoracic aorta, vasoconstriction significantly decreased with age following the use of nifedipine and thapsigargin and almost disappeared after STIM1 knockdown. The proportion of senescent VSMCs increased significantly in aged mice and further increased in sm-STIM1 KO aged mice. Moreover, the expression of senescence markers p21, p16, and IL-6 significantly increased with age, with p21 expression further increased in the STIM1 knockdown aged group, but not p16 or IL-6. These findings indicate that different arteries exhibit distinct organ-specific features and that STIM1 downregulation may contribute to age-related vasoconstrictive dysfunction through activation of the p21 pathway.
Collapse
Affiliation(s)
- Hao Wang
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Peng Zeng
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Peng-Hao Zhu
- The First Clinical College, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Zi-Fan Wang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yong-Jiang Cai
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Chun-Yu Deng
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hui Yang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Li-Ping Mai
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Meng-Zhen Zhang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Su-Juan Kuang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Fang Rao
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Jin-Song Xu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
31
|
de Lima EP, Tanaka M, Lamas CB, Quesada K, Detregiachi CRP, Araújo AC, Guiguer EL, Catharin VMCS, de Castro MVM, Junior EB, Bechara MD, Ferraz BFR, Catharin VCS, Laurindo LF, Barbalho SM. Vascular Impairment, Muscle Atrophy, and Cognitive Decline: Critical Age-Related Conditions. Biomedicines 2024; 12:2096. [PMID: 39335609 PMCID: PMC11428869 DOI: 10.3390/biomedicines12092096] [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: 08/05/2024] [Revised: 08/22/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The triad of vascular impairment, muscle atrophy, and cognitive decline represents critical age-related conditions that significantly impact health. Vascular impairment disrupts blood flow, precipitating the muscle mass reduction seen in sarcopenia and the decline in neuronal function characteristic of neurodegeneration. Our limited understanding of the intricate relationships within this triad hinders accurate diagnosis and effective treatment strategies. This review analyzes the interrelated mechanisms that contribute to these conditions, with a specific focus on oxidative stress, chronic inflammation, and impaired nutrient delivery. The aim is to understand the common pathways involved and to suggest comprehensive therapeutic approaches. Vascular dysfunctions hinder the circulation of blood and the transportation of nutrients, resulting in sarcopenia characterized by muscle atrophy and weakness. Vascular dysfunction and sarcopenia have a negative impact on physical function and quality of life. Neurodegenerative diseases exhibit comparable pathophysiological mechanisms that affect cognitive and motor functions. Preventive and therapeutic approaches encompass lifestyle adjustments, addressing oxidative stress, inflammation, and integrated therapies that focus on improving vascular and muscular well-being. Better understanding of these links can refine therapeutic strategies and yield better patient outcomes. This study emphasizes the complex interplay between vascular dysfunction, muscle degeneration, and cognitive decline, highlighting the necessity for multidisciplinary treatment approaches. Advances in this domain promise improved diagnostic accuracy, more effective therapeutic options, and enhanced preventive measures, all contributing to a higher quality of life for the elderly population.
Collapse
Affiliation(s)
- Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos Krt. 113, H-6725 Szeged, Hungary
| | - Caroline Barbalho Lamas
- Department of Gerontology, Universidade Federal de São Carlos, UFSCar, São Carlos 13565-905, SP, Brazil
| | - Karina Quesada
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
| | - Claudia Rucco P. Detregiachi
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Virgínia Maria Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Marcela Vialogo Marques de Castro
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Department of Odontology, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Edgar Baldi Junior
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | | | | | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17525-902, SP, Brazil
- Department of Administration, Associate Degree in Hospital Management, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Research Coordination, UNIMAR Charity Hospital (HBU), University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| |
Collapse
|
32
|
Zhao Q, Pedroza A, Sharma D, Gu W, Dalal A, Weldy C, Jackson W, Li DY, Ryan Y, Nguyen T, Shad R, Palmisano BT, Monteiro JP, Worssam M, Berezwitz A, Iyer M, Shi H, Kundu R, Limbu L, Kim JB, Kundaje A, Fischbein M, Wirka R, Quertermous T, Cheng P. A cell and transcriptome atlas of the human arterial vasculature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612293. [PMID: 39314359 PMCID: PMC11419041 DOI: 10.1101/2024.09.10.612293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Vascular beds show different propensities for different vascular pathologies, yet mechanisms explaining these fundamental differences remain unknown. We sought to build a transcriptomic, cellular, and spatial atlas of human arterial cells across multiple different arterial segments to understand this phenomenon. We found significant cell type-specific segmental heterogeneity. Determinants of arterial identity are predominantly encoded in fibroblasts and smooth muscle cells, and their differentially expressed genes are particularly enriched for vascular disease-associated loci and genes. Adventitial fibroblast-specific heterogeneity in gene expression coincides with numerous vascular disease risk genes, suggesting a previously unrecognized role for this cell type in disease risk. Adult arterial cells from different segments cluster not by anatomical proximity but by embryonic origin, with differentially regulated genes heavily influenced by developmental master regulators. Non-coding transcriptomes across arterial cells contain extensive variation in lnc-RNAs expressed in cell type- and segment-specific patterns, rivaling heterogeneity in protein coding transcriptomes, and show enrichment for non-coding genetic signals for vascular diseases.
Collapse
|
33
|
Di Carlo E, Sorrentino C. The multifaceted role of the stroma in the healthy prostate and prostate cancer. J Transl Med 2024; 22:825. [PMID: 39238004 PMCID: PMC11378418 DOI: 10.1186/s12967-024-05564-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
Prostate cancer (PC) is an age-related disease and represents, after lung cancer, the second cause of cancer death in males worldwide. Mortality is due to the metastatic disease, which mainly involves the bones, lungs, and liver. In the last 20 years, the incidence of metastatic PC has increased in Western Countries, and a further increase is expected in the near future, due to the population ageing. Current treatment options, including state of the art cancer immunotherapy, need to be more effective to achieve long-term disease control. The most significant anatomical barrier to overcome to improve the effectiveness of current and newly designed drug strategies consists of the prostatic stroma, in particular the fibroblasts and the extracellular matrix, which are the most abundant components of both the normal and tumor prostatic microenvironment. By weaving a complex communication network with the glandular epithelium, the immune cells, the microbiota, the endothelium, and the nerves, in the healthy prostatic microenvironment, the fibroblasts and the extracellular matrix support organ development and homeostasis. However, during inflammation, ageing and prostate tumorigenesis, they undergo dramatic phenotypic and genotypic changes, which impact on tumor growth and progression and on the development of therapy resistance. Here, we focus on the characteristics and functions of the prostate associated fibroblasts and of the extracellular matrix in health and cancer. We emphasize their roles in shaping tumor behavior and the feasibility of manipulating and/or targeting these stromal components to overcome the limitations of current treatments and to improve precision medicine's chances of success.
Collapse
Affiliation(s)
- Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti- Pescara, Via dei Vestini, Chieti, 66100, Italy.
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, Chieti, 66100, Italy.
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti- Pescara, Via dei Vestini, Chieti, 66100, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, Chieti, 66100, Italy
| |
Collapse
|
34
|
Chen Z, Li X, Sun X, Xiao S, Chen T, Ren L, Liu N. STING1-accelerated vascular smooth muscle cell senescence-associated vascular calcification in diabetes is ameliorated by oleoylethanolamide via improved mitochondrial DNA oxidative damage. Free Radic Biol Med 2024; 222:437-455. [PMID: 38889865 DOI: 10.1016/j.freeradbiomed.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/20/2024]
Abstract
Vascular calcification is a prevalent hallmark of cardiovascular risk in elderly and diabetic individuals. Senescent vascular smooth muscle cells (VSMCs) participate in calcification; however, the associated underlying mechanisms remain unknown. Aberrant activation of the cytosolic DNA sensing adaptor stimulator of interferon gene 1 (STING1) caused by cytosolic DNA, particularly that leaked from damaged mitochondria, is a catalyst for aging-related diseases. Although oleoylethanolamide (OEA) is an endogenous bioactive lipid mediator with lipid overload-associated vasoprotective effects, its benefit in diabetic vascular calcification remains uncharacterized. This study focused on the role of STING1 in mitochondrial dysfunction-mediated calcification and premature VMSC senescence in diabetes and the effects of OEA on these pathological processes. In diabetic in vivo rat/mouse aorta calcification models and an in vitro VSMC calcification model induced by Nε-carboxymethyl-lysine (CML), senescence levels, STING1 signaling activation, and mitochondrial damage markers were significantly augmented; however, these alterations were markedly alleviated by OEA, partially in a nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent manner, and similar anti-calcification and senescence effects were observed in STING1-knockout mice and STING1-knockdown VSMCs. Mechanistically, mitochondrial DNA (mtDNA) damage was aggravated by CML in a reactive oxygen species-dependent manner, followed by mtDNA leakage into the cytosol, contributing to VSMC senescence-associated calcification via STING1 pathway activation. OEA treatment significantly attenuated the aforementioned cytotoxic effects of CML by enhancing cellular antioxidant capacity through the maintenance of Nrf2 translocation to the nucleus. Collectively, targeting STING1, a newly defined VSMC senescence regulator, contributes to anti-vascular calcification effects.
Collapse
MESH Headings
- Animals
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Mice
- Cellular Senescence/drug effects
- Rats
- DNA, Mitochondrial/genetics
- DNA, Mitochondrial/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Vascular Calcification/drug therapy
- Vascular Calcification/genetics
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/genetics
- Oleic Acids/pharmacology
- Oxidative Stress/drug effects
- Male
- Endocannabinoids/metabolism
- Endocannabinoids/pharmacology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- DNA Damage/drug effects
- Mitochondria/metabolism
- Mitochondria/drug effects
- Mitochondria/pathology
- Signal Transduction/drug effects
- Humans
- Mice, Knockout
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Zhengdong Chen
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Xiaoxue Li
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Xuejiao Sun
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, 87 Dingjiaqiao, Nanjing, 210009, PR China; Nanjing First Hospital, Nanjing Medical University, Nanjing, 210000, PR China
| | - Shengjue Xiao
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Tian Chen
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Liqun Ren
- Department of Geriatrics, Zhongda Hospital, Southeast University School of Medicine, 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Naifeng Liu
- Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, 87 Dingjiaqiao, Nanjing, 210009, PR China.
| |
Collapse
|
35
|
Valeriani E, Bartimoccia S, Pignatelli P, Pastori D. Aging and Antithrombotic Treatment. Antioxid Redox Signal 2024; 41:542-556. [PMID: 37742116 DOI: 10.1089/ars.2023.0373] [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: 09/25/2023]
Abstract
Significance: Several aging-related pathophysiological mechanisms have been described to contribute to increased thrombotic risk in the elderly, including oxidative stress, endothelial dysfunction, and platelet and coagulation cascade activation. Antithrombotic treatment in the elderly should be individualized. Recent Advances: Recent studies have clarified some pathophysiological mechanisms of enhanced oxidative stress and thrombotic alterations in older adults. In the last decade, randomized trials have evaluated different antithrombotic strategies to reduce the risk of cardiovascular events in these patients. Critical Issues: The proportion of elderly patients included in clinical trials is generally low, thus not reflecting the daily clinical practice. There is no consensus on the most appropriate antithrombotic treatment in the elderly, also considering that bleeding risk management may be challenging in this high-risk subgroup of patients. Routine antiplatelet treatment is not a valid strategy for the primary prevention of cardiovascular events given the associated high risk of bleeding. In elderly patients with acute coronary syndrome, low-dose prasugrel or clopidogrel, shorter dual antiplatelet therapy, and no pretreatment before stent placement should be considered. Advanced age should not be the only reason for the underuse of oral anticoagulation in patients with atrial fibrillation, with direct oral anticoagulants preferred over warfarin for stroke prevention. Instead, a case-by-case clinical evaluation is warranted based on patient's bleeding risk also. Future Directions: There is a need for a structured tailored approach to manage thrombotic risk in elderly patients. The choice of the most appropriate antithrombotic treatment should balance efficacy and safety to reduce the risk of bleeding.
Collapse
Affiliation(s)
- Emanuele Valeriani
- Department of General Surgery, Surgical Specialties and Organ Transplantation "Paride Stefanini", Sapienza University of Rome, Rome, Italy
- Infectious Disease Department, Umberto I Hospital of Rome, Rome, Italy
| | - Simona Bartimoccia
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Daniele Pastori
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
36
|
Dzuko Kamga J, Floch R, Kerleguer K, Bourhis D, Le Pennec R, Hennebicq S, Salaün PY, Abgral R. Case-control study of the characteristics and risk factors of hot clot artefacts on 18F-FDG PET/CT. Cancer Imaging 2024; 24:114. [PMID: 39192363 DOI: 10.1186/s40644-024-00760-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
INTRODUCTION The pulmonary Hot Clot artifact (HCa) on 18F-FDG PET/CT is a poorly understood phenomenon, corresponding to the presence of a focal tracer uptake without anatomical lesion on combined CTscan. The hypothesis proposed in the literature is of microembolic origin. Our objectives were to determine the incidence of HCa, to analyze its characteristics and to identify associated factors. METHODS All 18F-FDG PET/CT retrieved reports containing the keywords (artifact/vascular adhesion/no morphological abnormality) during the period June 2021-2023 at Brest University Hospital were reviewed for HCa. Each case was associated with 2 control patients (same daily work-list). The anatomical and metabolic characteristics of HCa were analyzed. Factors related to FDG preparation/administration, patient and vascular history were investigated. Case-control differences between variables were tested using Chi-2 test and OR (qualitative) or Student's t-test (quantitative). RESULTS Of the 22,671 18F-FDG PET/CT performed over 2 years, 211 patients (0.94%) showed HCa. The focus was single in 97.6%, peripheral in 75.3%, and located independently in the right or left lung (51.1% vs. 48.9%). Mean ± SD values for SUVmax, SUVmean, MTV and TLG were 11.3 ± 16.5, 5.1 ± 5.0, 0.3 ± 0.3 ml and 1.5 ± 2.1 g respectively. The presence of vascular adhesion (p < 0.001), patient age (p = 0.002) and proximal venous access (p = 0.001) were statistically associated with the presence of HCa. CONCLUSION HCa is a real but rare phenomenon (incidence around 1%), mostly unique, intense, small in volume (< 1 ml), and associated with the presence of vascular FDG uptake, confirming the hypothesis of a microembolic origin due to probable vein wall trauma at the injection site.
Collapse
Affiliation(s)
- Jacques Dzuko Kamga
- Nuclear Medicine Department, CHRU Brest, Boulevard Tanguy Prigent, Brest, France.
| | - Romain Floch
- Nuclear Medicine Department, CHRU Brest, Boulevard Tanguy Prigent, Brest, France
| | - Kevin Kerleguer
- Nuclear Medicine Department, CHRU Brest, Boulevard Tanguy Prigent, Brest, France
| | - David Bourhis
- Nuclear Medicine Department, CHRU Brest, Boulevard Tanguy Prigent, Brest, France
- UMR Inserm GETBO 1304, University of Western Brittany, Brest, France
| | - Romain Le Pennec
- Nuclear Medicine Department, CHRU Brest, Boulevard Tanguy Prigent, Brest, France
- UMR Inserm GETBO 1304, University of Western Brittany, Brest, France
| | - Simon Hennebicq
- Nuclear Medicine Department, CHRU Brest, Boulevard Tanguy Prigent, Brest, France
| | - Pierre-Yves Salaün
- Nuclear Medicine Department, CHRU Brest, Boulevard Tanguy Prigent, Brest, France
- UMR Inserm GETBO 1304, University of Western Brittany, Brest, France
| | - Ronan Abgral
- Nuclear Medicine Department, CHRU Brest, Boulevard Tanguy Prigent, Brest, France.
- UMR Inserm GETBO 1304, University of Western Brittany, Brest, France.
| |
Collapse
|
37
|
Zhang J, Xia X, He S. Deciphering the causal association and underlying transcriptional mechanisms between telomere length and abdominal aortic aneurysm. Front Immunol 2024; 15:1438838. [PMID: 39234237 PMCID: PMC11371612 DOI: 10.3389/fimmu.2024.1438838] [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: 05/26/2024] [Accepted: 08/01/2024] [Indexed: 09/06/2024] Open
Abstract
Background The purpose of this study is to investigate the causal effect and potential mechanisms between telomere length and abdominal aortic aneurysm (AAA). Methods Summary statistics of telomere length and AAA were derived from IEU open genome-wide association studies and FinnGen R9, respectively. Bi-directional Mendelian randomization (MR) analysis was conducted to reveal the causal relationship between AAA and telomere length. Three transcriptome datasets were retrieved from the Gene Expression Omnibus database and telomere related genes was down-loaded from TelNet. The overlapping genes of AAA related differentially expressed genes (DEGs), module genes, and telomere related genes were used for further investigation. Telomere related diagnostic biomarkers of AAA were selected with machine learning algorisms and validated in datasets and murine AAA model. The correlation between biomarkers and immune infiltration landscape was established. Results Telomere length was found to have a suggestive negative associations with AAA [IVW, OR 95%CI = 0.558 (0.317-0.701), P < 0.0001], while AAA showed no suggestive effect on telomere length [IVW, OR 95%CI = 0.997 (0.990-1.004), P = 0.4061]. A total of 40 genes was considered as telomere related DEGs of AAA. PLCH2, PRKCQ, and SMG1 were selected as biomarkers after multiple algorithms and validation. Immune infiltration analysis and single cell mRNA analysis revealed that PLCH2 and PRKCQ were mainly expressed on T cells, while SMG1 predominantly expressed on T cells, B cells, and monocytes. Murine AAA model experiments further validated the elevated expression of biomarkers. Conclusion We found a suggestive effect of telomere length on AAA and revealed the potential biomarkers and immune mechanism of telomere length on AAA. This may shed new light for diagnosis and therapeutics on AAA.
Collapse
Affiliation(s)
- Jiyu Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujie He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
38
|
Ariko TA, Aimagambetova B, Gardener H, Gutierrez J, Elkind MSV, Wright CB, Zhao W, Rundek T. Estimated Pulse-Wave Velocity and Magnetic Resonance Imaging Markers of Cerebral Small-Vessel Disease in the NOMAS. J Am Heart Assoc 2024; 13:e035691. [PMID: 39023069 DOI: 10.1161/jaha.124.035691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Pulse-wave velocity is a measure of arterial stiffness and a risk factor for cardiovascular disease. Recently, an estimated pulse-wave velocity (ePWV) was introduced that was predictive of increased risk of cardiovascular disease. Our objective was to determine whether ePWV was associated with cerebral small-vessel disease on magnetic resonance imaging. METHODS AND RESULTS We included 1257 participants from the NOMAS (Northern Manhattan Study). The ePWV values were calculated using a nonlinear function of age and mean arterial blood pressure. The association between ePWV and white matter hyperintensity volume was assessed. Modification by race and ethnicity was evaluated. Associations between ePWV and other cerebral small-vessel disease markers, covert brain infarcts, cerebral microbleeds, and enlarged perivascular spaces, were explored as secondary outcomes. Mean±SD age of the cohort was 64±8 years; 61% were women; 18% self-identified as non-Hispanic Black, 67% as Hispanic, and 15% as non-Hispanic White individuals. Mean±SD ePWV was 11±2 m/s in the total NOMAS population and was similar across race and ethnic groups. The ePWV was significantly associated with white matter hyperintensity volume (β=0.23 [95% CI, 0.20-0.26]) after adjustment. Race and ethnicity modified the association between ePWV and white matter hyperintensity volume, with stronger associations in Hispanic and non-Hispanic Black individuals. Significant associations were found between ePWV and covert brain infarcts, cerebral microbleeds, and perivascular spaces after adjustment. CONCLUSIONS The ePWV function may provide a vascular mechanism for deleterious cerebrovascular outcomes in individuals with cerebral small-vessel disease and is particularly apparent in the racial and ethnic minorities represented in the NOMAS cohort.
Collapse
Affiliation(s)
- Taylor A Ariko
- Evelyn F. McKnight Brain Institute, University of Miami Miami FL
- Department of Biomedical Engineering University of Miami Miami FL
| | - Botagoz Aimagambetova
- Evelyn F. McKnight Brain Institute, University of Miami Miami FL
- Department of Neurology University of Miami Miller School of Medicine Miami FL
| | - Hannah Gardener
- Evelyn F. McKnight Brain Institute, University of Miami Miami FL
- Department of Neurology University of Miami Miller School of Medicine Miami FL
| | - Jose Gutierrez
- Department of Neurology, Vagelos College of Physicians and Surgeons Columbia University New York NY
| | - Mitchell S V Elkind
- Department of Neurology, Vagelos College of Physicians and Surgeons Columbia University New York NY
- American Heart Association Dallas TX
| | - Clinton B Wright
- National Institute of Neurological Disorders and Stroke Bethesda MD
| | - Weizhao Zhao
- Department of Biomedical Engineering University of Miami Miami FL
- Department of Neurology University of Miami Miller School of Medicine Miami FL
| | - Tatjana Rundek
- Evelyn F. McKnight Brain Institute, University of Miami Miami FL
- Department of Biomedical Engineering University of Miami Miami FL
- Department of Neurology University of Miami Miller School of Medicine Miami FL
| |
Collapse
|
39
|
Ma Y, Ma Z, Zhang Y, Luo C, Huang P, Tong J, Ding H, Liu H. Apigenin and baicalein ameliorate thoracic aortic structural deterioration and cognitive deficit via inhibiting AGEs/RAGE/NF-κB pathway in D-galactose-induced aging rats. Eur J Pharmacol 2024; 976:176660. [PMID: 38795756 DOI: 10.1016/j.ejphar.2024.176660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/04/2024] [Accepted: 05/16/2024] [Indexed: 05/28/2024]
Abstract
Apigenin and baicalein are structurally related flavonoids that have been reported to have multiple pharmacological activities. The aim of this study was to investigate the protective effects and potential mechanisms of apigenin and baicalein in D-galactose-induced aging rats. First, apigenin and baicalein showed remarkable antioxidant activity and anti-glycation activity in vitro. Secondly, the protective effects of apigenin and baicalein on aging rats were investigated. We found that apigenin and baicalein supplementation significantly ameliorated aging-related changes such as declines in the spatial learning and memory and histopathological damage of the hippocampus and thoracic aorta. In addition, our data showed that apigenin and baicalein alleviated oxidative stress as illustrated by decreasing MDA level, increasing SOD activity and GSH level. Further data showed that they significantly reduced the accumulation of advanced glycation end products (AGEs), inhibited the expression of RAGE, down-regulated phosphorylated nuclear factor (p-NF-κB (p65)). Our results suggested that the protective effects of apigenin and baicalein on aging rats were at least partially related to the inhibition of AGEs/RAGE/NF-κB pathway and the improvement of oxidative damage. Overall, apigenin and baicalein showed almost equal anti-aging efficacy. Our results provided an experimental basis for the application of apigenin and baicalein to delay the aging process.
Collapse
Affiliation(s)
- Yufang Ma
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China
| | - Zhenming Ma
- College of Software Engineering, Chengdu University of Information Technology, Chengdu, Sichuan, 610200, China
| | - Yiyuan Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China
| | - Chunyun Luo
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China
| | - Puxin Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China
| | - Jing Tong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China.
| | - Hong Ding
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China.
| | - Honghui Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China.
| |
Collapse
|
40
|
Pośpiech E, Bar A, Pisarek-Pacek A, Karaś A, Branicki W, Chlopicki S. Epigenetic clock in the aorta and age-related endothelial dysfunction in mice. GeroScience 2024; 46:3993-4002. [PMID: 38381284 PMCID: PMC11226569 DOI: 10.1007/s11357-024-01086-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/20/2024] [Indexed: 02/22/2024] Open
Abstract
While epigenetic age (EA) of mouse blood can be determined using DNA methylation analysis at three CpG sites in the Prima1, Hsf4 and Kcns1 genes it is not known whether this approach is useful for predicting vascular biological age. In this study we validated the 3-CpG estimator for age prediction in mouse blood, developed a new predictive model for EA in mouse aorta, and assessed whether epigenetic age acceleration (EAA) measured with blood and aorta samples correlates with age-dependent endothelial dysfunction. Endothelial function was characterized in vivo by MRI in 8-96-week-old C57BL/6 mice. Arterial stiffness was measured by USG-doppler. EA-related changes within 41 CpG sites in Prima1, Kcns1 and Hsf4 loci, were analyzed in the aorta and blood using bisulfite amplicon high-throughput sequencing. Progressive age-dependent endothelial dysfunction and changes in arterial stiffness were observed in 36-96-week-old C57BL/6 mice. Methylation levels of the investigated loci correlated with chronological age in blood and the aorta. The new model for EA estimation in aorta included three cytosines located in the Kcns1 and Hsf4, explained R2 = 87.8% of the variation in age, and predicted age with an mean absolute error of 9.6 weeks in the independent test set. EAA in the aorta was associated with endothelial dysfunction in the abdominal aorta and femoral artery what was consistent with the EAA direction estimated in blood samples. The rate of vascular biological ageing in mice, reflected by the age-dependent systemic endothelial dysfunction, could be estimated using DNA methylation measurements at three loci in aorta and blood samples.
Collapse
Affiliation(s)
- Ewelina Pośpiech
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Al. Powstancow Wielkopolskich 72, 70-204, Szczecin, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Aleksandra Pisarek-Pacek
- Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387, Krakow, Poland
| | - Agnieszka Karaś
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Wojciech Branicki
- Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland.
- Institute of Forensic Research, Westerplatte 9, 31-033, Kraków, Poland.
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland.
- Faculty of Medicine, Chair of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Krakow, Poland.
| |
Collapse
|
41
|
Luo Y, Zhang Z, Zheng W, Zeng Z, Fan L, Zhao Y, Huang Y, Cao S, Yu S, Shen L. Molecular Mechanisms of Plant Extracts in Protecting Aging Blood Vessels. Nutrients 2024; 16:2357. [PMID: 39064801 PMCID: PMC11279783 DOI: 10.3390/nu16142357] [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: 06/13/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Plant Extracts (PE) are natural substances extracted from plants, rich in various bioactive components. Exploring the molecular mechanisms and interactions involved in the vascular protective effects of PE is beneficial for the development of further strategies to protect aging blood vessels. For this review, the content was obtained from scientific databases such as PubMed, China National Knowledge Infrastructure (CNKI), and Google Scholar up to July 2024, using the search terms "Plant extracts", "oxidative stress", "vascular aging", "endothelial dysfunction", "ROS", and "inflammation". This review highlighted the effects of PE in protecting aging blood vessels. Through pathways such as scavenging reactive oxygen species, activating antioxidant signaling pathways, enhancing respiratory chain complex activity, inhibiting mitochondrial-reactive oxygen species generation, improving nitric oxide bioavailability, downregulating the secretion of inflammatory factors, and activating sirtuins 1 and Nrf2 signaling pathways, it can improve vascular structural and functional changes caused by age-related oxidative stress, mitochondrial dysfunction, and inflammation due to aging, thereby reducing the incidence of age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yuxin Luo
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Zeru Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Weijian Zheng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Zhi Zeng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Lei Fan
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yuquan Zhao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Yixin Huang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Suizhong Cao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Shumin Yu
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Liuhong Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| |
Collapse
|
42
|
Machi JF, Altilio I, Qi Y, Morales AA, Silvestre DH, Hernandez DR, Da Costa-Santos N, Santana AG, Neghabi M, Nategh P, Castro TL, Werneck-de-Castro JP, Ranji M, Evangelista FS, Vazquez-Padron RI, Bernal-Mizrachi E, Rodrigues CO. Endothelial c-Myc knockout disrupts metabolic homeostasis and triggers the development of obesity. Front Cell Dev Biol 2024; 12:1407097. [PMID: 39100099 PMCID: PMC11294153 DOI: 10.3389/fcell.2024.1407097] [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: 03/27/2024] [Accepted: 06/10/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction: Obesity is a major risk factor associated with multiple pathological conditions including diabetes and cardiovascular disease. Endothelial dysfunction is an early predictor of obesity. However, little is known regarding how early endothelial changes trigger obesity. In the present work we report a novel endothelial-mediated mechanism essential for regulation of metabolic homeostasis, driven by c-Myc. Methods: We used conditional knockout (EC-Myc KO) and overexpression (EC-Myc OE) mouse models to investigate the endothelial-specific role of c-Myc in metabolic homeostasis during aging and high-fat diet exposure. Body weight and metabolic parameters were collected over time and tissue samples collected at endpoint for biochemical, pathology and RNA-sequencing analysis. Animals exposed to high-fat diet were also evaluated for cardiac dysfunction. Results: In the present study we demonstrate that EC-Myc KO triggers endothelial dysfunction, which precedes progressive increase in body weight during aging, under normal dietary conditions. At endpoint, EC-Myc KO animals showed significant increase in white adipose tissue mass relative to control littermates, which was associated with sex-specific changes in whole body metabolism and increase in systemic leptin. Overexpression of endothelial c-Myc attenuated diet-induced obesity and visceral fat accumulation and prevented the development of glucose intolerance and cardiac dysfunction. Transcriptome analysis of skeletal muscle suggests that the protective effects promoted by endothelial c-Myc overexpression are associated with the expression of genes known to increase weight loss, energy expenditure and glucose tolerance. Conclusion: Our results show a novel important role for endothelial c-Myc in regulating metabolic homeostasis and suggests its potential targeting in preventing obesity and associated complications such as diabetes type-2 and cardiovascular dysfunction.
Collapse
Affiliation(s)
- Jacqueline F. Machi
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Isabella Altilio
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Yue Qi
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Alejo A. Morales
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diego H. Silvestre
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diana R. Hernandez
- DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Nicolas Da Costa-Santos
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Aline G. Santana
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Mehrnoosh Neghabi
- Department of Electrical Engineering and Computer Science, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Parisa Nategh
- Department of Electrical Engineering and Computer Science, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Thiago L. Castro
- School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - João P. Werneck-de-Castro
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Mahsa Ranji
- Department of Electrical Engineering and Computer Science, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, United States
| | | | - Roberto I. Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Claudia O. Rodrigues
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
43
|
Yu F, Zong B, Ji L, Sun P, Jia D, Wang R. Free Fatty Acids and Free Fatty Acid Receptors: Role in Regulating Arterial Function. Int J Mol Sci 2024; 25:7853. [PMID: 39063095 PMCID: PMC11277118 DOI: 10.3390/ijms25147853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The metabolic network's primary sources of free fatty acids (FFAs) are long- and medium-chain fatty acids of triglyceride origin and short-chain fatty acids produced by intestinal microorganisms through dietary fibre fermentation. Recent studies have demonstrated that FFAs not only serve as an energy source for the body's metabolism but also participate in regulating arterial function. Excess FFAs have been shown to lead to endothelial dysfunction, vascular hypertrophy, and vessel wall stiffness, which are important triggers of arterial hypertension and atherosclerosis. Nevertheless, free fatty acid receptors (FFARs) are involved in the regulation of arterial functions, including the proliferation, differentiation, migration, apoptosis, inflammation, and angiogenesis of vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs). They actively regulate hypertension, endothelial dysfunction, and atherosclerosis. The objective of this review is to examine the roles and heterogeneity of FFAs and FFARs in the regulation of arterial function, with a view to identifying the points of intersection between their actions and providing new insights into the prevention and treatment of diseases associated with arterial dysfunction, as well as the development of targeted drugs.
Collapse
Affiliation(s)
- Fengzhi Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (B.Z.); (P.S.)
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Lili Ji
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (B.Z.); (P.S.)
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| |
Collapse
|
44
|
Liu W, Wang S, Zhang X, Ke Z, Wen X, Zhao J, Zhuang X, Liao L. Enhanced Cardiomyocyte NLRP3 Inflammasome-Mediated Pyroptosis Promotes d-Galactose-Induced Cardiac Aging. J Am Heart Assoc 2024; 13:e032904. [PMID: 38979831 PMCID: PMC11292767 DOI: 10.1161/jaha.123.032904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 06/03/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Cardiac aging represents an independent risk factor for aging-associated cardiovascular diseases. Although evidence suggests an association between NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome formation and numerous cardiovascular diseases, its role in cardiac aging remains largely unclear. METHODS AND RESULTS The longevity of mice with wild-type and NLRP3 knockout (NLRP3-/-) genotypes was assessed, with or without d-galactose treatment. Cardiac function was evaluated using echocardiography, and cardiac histopathology was examined through hematoxylin and eosin and Masson's trichrome staining. Senescence-associated β-galactosidase (SA-β-gal) staining was employed to detect cardiac aging. Western blotting was used to assess aging-related proteins (p53, p21) and pyroptosis-related proteins. Additionally, dihydroethidium staining, lactate dehydrogenase release, and interleukin-1β ELISA assays were performed, along with measurements of total superoxide dismutase and malondialdehyde levels. In vitro, H9c2 cells were exposed to d-galactose for 24 hours in the absence or presence of N-acetyl-l-cysteine (reactive oxygen species inhibitor), BAY-117082 (nuclear factor κ-light-chain enhancer of activated B cells inhibitor), MCC950 (NLRP3 inhibitor), and VX-765 (Caspase-1 inhibitor). Immunofluorescence staining was employed to detect p53, gasdermin D, and apoptosis-associated speck-like protein proteins. Intracellular reactive oxygen species levels were assessed using fluorescence microscopy and flow cytometry. Senescence-associated β-galactosidase staining and Western blotting were also employed in vitro for the same purpose. The results showed that NLRP3 upregulation was implicated in aging and cardiovascular diseases. Inhibition of NLRP3 extended life span, mitigated the aging phenotype, improved cardiac function and blood pressure, ameliorated lipid metabolism abnormalities, inhibited pyroptosis in cardiomyocytes, and ultimately alleviated cardiac aging. In vitro, the inhibition of reactive oxygen species, nuclear factor κ-light-chain enhancer of activated B cells, NLRP3, or caspase-1 attenuated NLRP3 inflammasome-mediated pyroptosis. CONCLUSIONS The reactive oxygen species/nuclear factor κ-light-chain enhancer of activated B cells/NLRP3 signaling pathway loop contributes to d-galactose-treated cardiomyocyte senescence and cardiac aging.
Collapse
Affiliation(s)
- Wen‐bin Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Sui‐sui Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
- Department of Nuclear MedicineThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangdongChina
| | - Xu Zhang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Ze‐zhi Ke
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Xiu‐yun Wen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Jie Zhao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Xiao‐dong Zhuang
- Cardiology DepartmentThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangdongPeople’s Republic of China
| | - Li‐zhen Liao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| |
Collapse
|
45
|
Yuan M, Zhang Y, Hu R, Yuan L, Ma J, Xu Y. Linear-inverse associations of serum Klotho protein with prevalence of frailty among adults in the United States. Am J Transl Res 2024; 16:3090-3098. [PMID: 39114692 PMCID: PMC11301489 DOI: 10.62347/ozhf3072] [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: 04/04/2024] [Accepted: 05/17/2024] [Indexed: 08/10/2024]
Abstract
AIMS To investigate the potential linear relationship between serum concentrations of klotho and frailty. METHODS A retrospective analysis was conducted on the data of 9,597 middle-aged and older adults (aged 40-79 years) from the five cycles of the National Health and Nutrition Examination Survey (NHANES). Frailty was assessed using the Frailty Index, calculated as a percentage of accumulated deficits across 53 health items. Restricted cubic spline curves, subgroup analyses and logistic regression models were employed to evaluate the specific linear trend connection between circulating klotho protein concentration and frailty. RESULTS When taking Klotho into account as a continuous component in Models 1 and 2, there was a substantial association between the increasing Klotho level and the reduced risk of frailty. Model 3 revealed a strong negative correlation between the Klotho and Frailty, suggesting that high levels of Klotho protein decreases the frailty prevalence [Odd ratio (OR): 0.25; 95% confidence interval (CI): 0.15-0.43]. Furthermore, according to the quartile analyses, after fully adjusting for the covariates, it was observed that, comparing to the lowest quartile of Klotho, the highest quartile of Klotho demonstrated lowest risk of frailty (OR 0.69; 95% CI 0.58-0.81, Ptrend < 0.001). The restricted cubic spline curves showed a linear relationship and an inverse association between frailty and the Klotho levels (Plinearity < 0.001; Pnon-linearity = 0.736). CONCLUSION Klotho is inversely and linearly associated with physical frailty in the general population (aged 40-79 years), specifically in the population with an age < 65 and body mass index (BMI) ≥ 25 kg/m2. More necessary prospective studies should be done to further investigate the mechanisms underlying frailty and aging and to elucidate individual frailty causes.
Collapse
Affiliation(s)
- Menghan Yuan
- Department of Cardiology, Medical School of Chinese PLABeijing 100853, China
- Senior Department of Cardiology, The Sixth Medical Center of PLA General HospitalBeijing 100048, China
| | - Yingyue Zhang
- Department of Cardiology, Medical School of Chinese PLABeijing 100853, China
- Senior Department of Cardiology, The Sixth Medical Center of PLA General HospitalBeijing 100048, China
| | - Rong Hu
- Department of Cardiology, Medical School of Chinese PLABeijing 100853, China
- Senior Department of Cardiology, The Sixth Medical Center of PLA General HospitalBeijing 100048, China
| | - Lin Yuan
- Senior Department of Cardiology, The Sixth Medical Center of PLA General HospitalBeijing 100048, China
| | - Jing Ma
- Department of Cardiology, The First Medical Center of Chinese PLA General HospitalBeijing 100853, China
| | - Yong Xu
- Department of Cardiology, Medical School of Chinese PLABeijing 100853, China
- Senior Department of Cardiology, The Sixth Medical Center of PLA General HospitalBeijing 100048, China
| |
Collapse
|
46
|
Kim JH, Thiruvengadam R. Hypertension in an ageing population: Diagnosis, mechanisms, collateral health risks, treatments, and clinical challenges. Ageing Res Rev 2024; 98:102344. [PMID: 38768716 DOI: 10.1016/j.arr.2024.102344] [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: 07/20/2023] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Ageing population is considerably increasing worldwide, which is considered to reflect an improved quality of life. However, longevity in the human lifespan has increased the burden of late-life illnesses including cancer, neurodegeneration, and cardiovascular dysfunction. Of these, hypertension is the most common condition with huge health risks, with an increased prevalence among the elderly. In this review, we outline the current guidelines for defining hypertension and examine the detailed mechanisms underlying the relationship between hypertension and ageing-related outcomes, including sodium sensitivity, arterial stiffness, endothelial dysfunction, isolated systolic hypertension, white coat effect, and orthostatic hypertension. As hypertension-related collateral health risk increases among the elderly, the available management strategies are necessary to overcome the clinical treatment challenges faced among elderly population. To improve longevity and reduce adverse health effects, potential approaches producing crucial information into new era of medicine should be considered in the prevention and treatment of hypertension among elderly population. This review provides an overview of mechanisms underlying hypertension and its related collateral health risk in elderly population, along with multiple approaches and management strategies to improve the clinical challenges among elderly population.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, the Republic of Korea.
| | - Rekha Thiruvengadam
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, the Republic of Korea
| |
Collapse
|
47
|
Darvish S, Mahoney SA, Venkatasubramanian R, Rossman MJ, Clayton ZS, Murray KO. Socioeconomic status as a potential mediator of arterial aging in marginalized ethnic and racial groups: current understandings and future directions. J Appl Physiol (1985) 2024; 137:194-222. [PMID: 38813611 PMCID: PMC11389897 DOI: 10.1152/japplphysiol.00188.2024] [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: 03/13/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 05/31/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the United States. However, disparities in CVD-related morbidity and mortality exist as marginalized racial and ethnic groups are generally at higher risk for CVDs (Black Americans, Indigenous People, South and Southeast Asians, Native Hawaiians, and Pacific Islanders) and/or development of traditional CVD risk factors (groups above plus Hispanics/Latinos) relative to non-Hispanic Whites (NHW). In this comprehensive review, we outline emerging evidence suggesting these groups experience accelerated arterial dysfunction, including vascular endothelial dysfunction and large elastic artery stiffening, a nontraditional CVD risk factor that may predict risk of CVDs in these groups with advancing age. Adverse exposures to social determinants of health (SDOH), specifically lower socioeconomic status (SES), are exacerbated in most of these groups (except South Asians-higher SES) and may be a potential mediator of accelerated arterial aging. SES negatively influences the ability of marginalized racial and ethnic groups to meet aerobic exercise guidelines, the first-line strategy to improve arterial function, due to increased barriers, such as time and financial constraints, lack of motivation, facility access, and health education, to performing conventional aerobic exercise. Thus, identifying alternative interventions to conventional aerobic exercise that 1) overcome these common barriers and 2) target the biological mechanisms of aging to improve arterial function may be an effective, alternative method to aerobic exercise to ameliorate accelerated arterial aging and reduce CVD risk. Importantly, dedicated efforts are needed to assess these strategies in randomized-controlled clinical trials in these marginalized racial and ethnic groups.
Collapse
Affiliation(s)
- Sanna Darvish
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | | | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Kevin O Murray
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
48
|
Buscemi C, Randazzo C, Barile AM, Caruso R, Colombrita P, Lombardo M, Verde PL, Sottile N, Barbagallo M, Buscemi S. Endothelial function in healthy centenarians living in the Madonie's district (Italy). Exp Gerontol 2024; 192:112457. [PMID: 38728795 DOI: 10.1016/j.exger.2024.112457] [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: 04/01/2024] [Revised: 04/27/2024] [Accepted: 05/08/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Endothelial function declines with age and plays a critical role in cardiovascular health. Therefore, investigating endothelial function in successful aging models, such as centenarians, is of interest. Flow-mediated dilation (FMD) of the brachial artery is the gold standard for measuring endothelial function in vivo in humans. Therefore, we investigated, for the first time, the FMD of the brachial artery in a group of healthy centenarians. METHODS Selected as part of the ABCD project (nutrition, cardiovascular wellness, and diabetes) centenarians (aged ≥100 years) living in the municipalities of Madonie (Palermo, Italy) were compared with a younger (aged <65 years) sex-matched control group from the ABCD general cohort. FMD of the brachial artery was measured in all participants using a real-time computed video analysis system for B-mode ultrasound images. Body composition (bioimpedance), carotid intima-media thickness (IMT), and ankle-brachial index (ABI) were also measured. RESULTS Eleven participants (males 36.4 %; age: 101 ± 1 years) out of 28 healthy centenarians successfully cooperated with the FMD test procedures, which require remaining with the upper limb immobile for approximately 10 min. This subgroup was compared with a control group of 76 healthy and younger individuals (males 36.8 %; aged: 41 ± 14 years; P < 0.001). Centenarians exhibited better endothelial function than the control group (FMD: 12.1 ± 4.3 vs 8.6 ± 5.3 %; P < 0.05). The carotid IMT was higher in the centenarian group than in the control group (0.89 ± 0.09 vs 0.56 ± 0.18 mm; P < 0.001), whereas the ABI was comparable between the two groups. CONCLUSIONS This small group of centenarians demonstrated an unusually favorable endothelial function, which may contribute to their unique aging profile. Further research is needed to determine whether FMD is a valid prognostic marker for successful aging.
Collapse
Affiliation(s)
- Carola Buscemi
- Unit of Internal Medicine, "V. Cervello Hospital", Palermo, Italy; Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Cristiana Randazzo
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy; Unit of Clinical Nutrition, Obesity and Metabolic Diseases, University Hospital Policlinico "P. Giaccone", Palermo, Italy
| | - Anna Maria Barile
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy; Unit of Clinical Nutrition, Obesity and Metabolic Diseases, University Hospital Policlinico "P. Giaccone", Palermo, Italy.
| | - Roberta Caruso
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Piero Colombrita
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Martina Lombardo
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Piergiorgio Lo Verde
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Natalia Sottile
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Mario Barbagallo
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy; Geriatric Unit, University Hospital Policlinico "P. Giaccone", Palermo, Italy
| | - Silvio Buscemi
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy; Unit of Clinical Nutrition, Obesity and Metabolic Diseases, University Hospital Policlinico "P. Giaccone", Palermo, Italy.
| |
Collapse
|
49
|
Anwar S, Raut R, Alhumaydhi FA. A comprehensive investigation on alleviating oxidative stress and inflammation in hyperglycaemic conditions through in vitro experiments and computational analysis. Saudi J Biol Sci 2024; 31:104003. [PMID: 38766504 PMCID: PMC11097074 DOI: 10.1016/j.sjbs.2024.104003] [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: 02/22/2024] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/22/2024] Open
Abstract
Protein glycation, hyper-inflammatory reactions, and oxidative stress play a crucial role in the pathophysiology of numerous diseases. The current work evaluated the protective ability of ethyl alcohol extract of leaves from holy basil (Ocimum sanctum Linn) against inflammation, oxidative stress, glycation and advanced glycation endproducts formation. Various in vitro assays assessed prementioned properties of holy basil. In addition, molecular docking was conducted. The highest hydrogen peroxide reduction activity (72.7 %) and maximum percentage of DPPH scavenging (71.3 %) depicted its vigorous antioxidant abilities. Furthermore, it showed the most excellent protection against proteinase activity (67.247 %), prevention of denaturation of egg albumin (65.29 %), and BSA (bovine serum albumin) (68.87 %) with 600 µg/ml. Percent aggregation index (57.528 %), browning intensity (56.61 %), and amyloid structure (57.0 %) were all reduced significantly using 600 μg/ml of extract. Additionally, the antimicrobial potential was also confirmed. According to a molecular docking study, active leaf extract ingredients were found to bind with superoxide dismutase, catalase, and carbonic anhydrase. As a conclusion, O. sanctum has a variety of health-promoting properties that may reduce the severity of many diseases in diabetic patients. However, in order to ascertain the mechanisms of action of the components of its leaves in disease prevention, more thorough research based on pharmacological aspects is needed.
Collapse
Affiliation(s)
- Shehwaz Anwar
- Department of Medical Laboratory Technology, Mohan Institute of Nursing and Paramedical Sciences, Bareilly 243302, Uttar Pradesh, India
| | - Ravindra Raut
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
50
|
Mohammed KAK, Madeddu P, Avolio E. MEK inhibitors: a promising targeted therapy for cardiovascular disease. Front Cardiovasc Med 2024; 11:1404253. [PMID: 39011492 PMCID: PMC11247000 DOI: 10.3389/fcvm.2024.1404253] [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: 03/20/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Cardiovascular disease (CVD) represents the leading cause of mortality and disability all over the world. Identifying new targeted therapeutic approaches has become a priority of biomedical research to improve patient outcomes and quality of life. The RAS-RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase) pathway is gaining growing interest as a potential signaling cascade implicated in the pathogenesis of CVD. This pathway is pivotal in regulating cellular processes like proliferation, growth, migration, differentiation, and survival, which are vital in maintaining cardiovascular homeostasis. In addition, ERK signaling is involved in controlling angiogenesis, vascular tone, myocardial contractility, and oxidative stress. Dysregulation of this signaling cascade has been linked to cell dysfunction and vascular and cardiac pathological remodeling, which contribute to the onset and progression of CVD. Recent and ongoing research has provided insights into potential therapeutic interventions targeting the RAS-RAF-MEK-ERK pathway to improve cardiovascular pathologies. Preclinical studies have demonstrated the efficacy of targeted therapy with MEK inhibitors (MEKI) in attenuating ERK activation and mitigating CVD progression in animal models. In this article, we first describe how ERK signaling contributes to preserving cardiovascular health. We then summarize current knowledge of the roles played by ERK in the development and progression of cardiac and vascular disorders, including atherosclerosis, myocardial infarction, cardiac hypertrophy, heart failure, and aortic aneurysm. We finally report novel therapeutic strategies for these CVDs encompassing MEKI and discuss advantages, challenges, and future developments for MEKI therapeutics.
Collapse
Affiliation(s)
- Khaled A K Mohammed
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paolo Madeddu
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elisa Avolio
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|