1
|
Meyer-Lindemann U, Sager HB. Neuroimmune crosstalk : How mental stress fuels vascular inflammation. Herz 2024; 49:249-253. [PMID: 38954012 DOI: 10.1007/s00059-024-05254-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/04/2024]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Pathophysiologically, metabolic and inflammatory processes contribute substantially to the development and progression of cardiovascular diseases. Over the past decade, the role of disease-propagating inflammatory processes has been strengthened and reframed, leading to trials testing anti-inflammatory drugs for the treatment of atherosclerosis and its complications. Despite these achievements, further research in both pre-clinical and clinical studies is warranted to explore new targets, to better identify responders, and to refine therapy strategies to combat inflammation in human disease. Environmental disturbances, so-called lifestyle-associated cardiovascular risk factors, greatly alter the immune system in general and leukocytes in particular, thus affecting the progression of atherosclerosis. Epidemiological studies have shown that exposure to mental stress can be closely linked to the occurrence of cardiovascular disease. Here, we describe how acute and chronic mental stress alter the immune system via neuroimmune interactions, thereby modifying vascular inflammation. In addition, we identify gaps that still need to be addressed in the future.
Collapse
Affiliation(s)
- Ulrike Meyer-Lindemann
- Department of Cardiology, German Heart Centre Munich, Technical University Munich, Lazarettstr. 36, 80636, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Hendrik B Sager
- Department of Cardiology, German Heart Centre Munich, Technical University Munich, Lazarettstr. 36, 80636, Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
2
|
Shetty S, Duesman SJ, Patel S, Huynh P, Toh P, Shroff S, Das A, Chowhan D, Keller B, Alvarez J, Fisher-Foye R, Sebra R, Beaumont K, McAlpine CS, Rajbhandari P, Rajbhandari AK. Sex-specific role of high-fat diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. Biol Sex Differ 2024; 15:55. [PMID: 39010139 PMCID: PMC11247790 DOI: 10.1186/s13293-024-00628-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/11/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Scientific evidence highlights the influence of biological sex on the relationship between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress concurrently contribute to metabolic dysregulation in both males and females. Our study aimed to investigate the combined effects of high-fat diet (HFD) induced obesity and repeated stress on fear-related behaviors, metabolic, immune, and hypothalamic outcomes in male and female mice. METHODS To investigate this, we used a highly reliable rodent behavioral model that faithfully recapitulates key aspects of post-traumatic stress disorder (PTSD)-like fear. We subjected mice to footshock stressor followed by a weekly singular footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. At weeks 10 and 14 we conducted glucose tolerance and insulin sensitivity measurements. Additionally, we placed the mice in metabolic chambers to perform indirect calorimetric measurements. Finally, we collected brain and peripheral tissues for cellular analysis. RESULTS We observed that HFD-induced obesity disrupted fear memory extinction, increased glucose intolerance, and affected energy expenditure specifically in male mice. Conversely, female mice on HFD exhibited reduced respiratory exchange ratio (RER), and a significant defect in glucose tolerance only when subjected to repeated stress. Furthermore, the combination of repeated stress and HFD led to sex-specific alterations in proinflammatory markers and hematopoietic stem cells across various peripheral metabolic tissues. Single-nuclei RNA sequencing (snRNAseq) analysis of the ventromedial hypothalamus (VMH) revealed microglial activation in female mice on HFD, while male mice on HFD exhibited astrocytic activation under repeated stress. CONCLUSIONS Overall, our findings provide insights into complex interplay between repeated stress, high-fat diet regimen, and their cumulative effects on health, including their potential contribution to the development of PTSD-like stress and metabolic dysfunctions, emphasizing the need for further research to fully understand these interconnected pathways and their implications for health.
Collapse
Affiliation(s)
- Sanutha Shetty
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Samuel J Duesman
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sanil Patel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pacific Huynh
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pamela Toh
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sanjana Shroff
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anika Das
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Excellence in Youth Education, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Disha Chowhan
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Keller
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Johana Alvarez
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rachel Fisher-Foye
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Sebra
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin Beaumont
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Abha K Rajbhandari
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
3
|
Wu C, Yu Y, Zhao T, Xie H. Bidirectional causality between idiopathic sudden sensorineural hearing loss and depression: a Mendelian randomization study. Sci Rep 2024; 14:14932. [PMID: 38942925 PMCID: PMC11213911 DOI: 10.1038/s41598-024-65966-6] [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: 03/04/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024] Open
Abstract
Idiopathic Sudden Sensorineural Hearing Loss (ISSHL) is a sudden onset, unexplained sensorineural hearing loss. Depression is a common mental disorder and a leading cause of disability. Here, We used a two-sample Mendelian randomization approach using pooled statistics from genome-wide association studies of ISSHL (1491 cases, 196,592 controls) and depression (23,424 cases, 192,220 controls) in European populations. This study investigated the bidirectional relationship between single nucleotide polymorphisms associated with depression and ISSHL using inverse variance weighting.Additional sensitivity analyses, such as Mendelian randomization-Egger (MR-Egger), weighted median estimates, and leave-one-out analysis, were performed to assess the reliability of the findings. Significant causal association between genetic susceptibility to ISSHL and depression in a random-effects IVW approach (OR = 1.037, 95% CI = 1.004-1.072, P = 0.030). In contrast, genetic depression was not risk factors for ISSHL (OR = 1.134, 95% CI = 0.871-1.475, P = 0.350). After validation by different MR methods and the sensitivity analysis, all of the above results are consistent. The evidence we have gathered suggests a causal relationship between ISSHL and depression. The presence of the former induces or further exacerbates the latter, whereas a similar situation does not exist when the latter is an influencing factor.
Collapse
Affiliation(s)
- Chuanyu Wu
- Department of Otorhinolaryngology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Ya Yu
- Department of Otorhinolaryngology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Tongtong Zhao
- Department of Otorhinolaryngology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Hui Xie
- Department of Otorhinolaryngology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| |
Collapse
|
4
|
Jercălău CE, Andrei CL, Darabont RO, Guberna S, Staicu AM, Rusu CT, Ceban O, Sinescu CJ. Blood Cell Ratios Unveiled: Predictive Markers of Myocardial Infarction Prognosis. Healthcare (Basel) 2024; 12:824. [PMID: 38667586 PMCID: PMC11049867 DOI: 10.3390/healthcare12080824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Even if the management and treatment of patients with non-ST-elevation myocardial infarction (NSTEMI) have significantly evolved, it is still a burgeoning disease, an active volcano with very high rates of morbidity and mortality. Therefore, novel management and therapeutic strategies for this condition are urgently needed. Lately, theories related to the role of various blood cells in NSTEMI have emerged, with most of this research having so far been focused on correlating the ratios between various leukocyte types (neutrophil/lymphocyte ratio-NLR, neutrophil/monocyte ratio-NMR). But what about erythrocytes? Is there an interaction between these cells and leukocytes, and furthermore, can this relationship influence NSTEMI prognosis? Are they partners in crime? METHODS Through the present study, we sought, over a period of sixteen months, to evaluate the neutrophil/red blood cell ratio (NRR), monocyte/red blood cell ratio (MRR) and lymphocyte/red blood cell ratio (LRR), assessing their potential role as novel prognostic markers in patients with NSTEMI. RESULTS There was a statistically significant correlation between the NRR, LRR, MRR and the prognosis of NSTEMI patients. CONCLUSIONS These new predictive markers could represent the start of future innovative therapies that may influence crosstalk pathways and have greater benefits in terms of cardiac repair and the secondary prevention of NSTEMI.
Collapse
Affiliation(s)
- Cosmina Elena Jercălău
- Department of Cardiology, “Bagdasar Arseni” Emergency Hospital, University of Medicine and Pharmacy “Carol Davila”, 011241 Bucharest, Romania; (R.O.D.); (C.J.S.)
| | - Cătălina Liliana Andrei
- Department of Cardiology, “Bagdasar Arseni” Emergency Hospital, University of Medicine and Pharmacy “Carol Davila”, 011241 Bucharest, Romania; (R.O.D.); (C.J.S.)
| | - Roxana Oana Darabont
- Department of Cardiology, “Bagdasar Arseni” Emergency Hospital, University of Medicine and Pharmacy “Carol Davila”, 011241 Bucharest, Romania; (R.O.D.); (C.J.S.)
| | - Suzana Guberna
- Department of Cardiology, Emergency Hospital “Bagdasar-Arseni”, 050474 Bucharest, Romania; (S.G.); (A.M.S.)
| | - Arina Maria Staicu
- Department of Cardiology, Emergency Hospital “Bagdasar-Arseni”, 050474 Bucharest, Romania; (S.G.); (A.M.S.)
| | - Cătălin Teodor Rusu
- Department of Internal Medicine, “Coltea” Clinical Hospital, 030167 Bucharest, Romania;
| | - Octavian Ceban
- Economic Cybernetics and Informatics Department, The Bucharest University of Economic Studies, 010374 Bucharest, Romania;
| | - Crina Julieta Sinescu
- Department of Cardiology, “Bagdasar Arseni” Emergency Hospital, University of Medicine and Pharmacy “Carol Davila”, 011241 Bucharest, Romania; (R.O.D.); (C.J.S.)
| |
Collapse
|
5
|
Lechner K, Kia S, von Korn P, Dinges SM, Mueller S, Tjønna AE, Wisløff U, Van Craenenbroeck EM, Pieske B, Adams V, Pressler A, Landmesser U, Halle M, Kränkel N. Cardiometabolic and immune response to exercise training in patients with metabolic syndrome: retrospective analysis of two randomized clinical trials. Front Cardiovasc Med 2024; 11:1329633. [PMID: 38638882 PMCID: PMC11025358 DOI: 10.3389/fcvm.2024.1329633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
Background Metabolic syndrome (MetS) is defined by the presence of central obesity plus ≥two metabolic/cardiovascular risk factors (RF), with inflammation being a major disease-driving mechanism. Structured endurance exercise training (ET) may positively affect these traits, as well as cardiorespiratory fitness (V̇O2peak). Aims We explore individual ET-mediated improvements of MetS-associated RF in relation to improvements in V̇O2peak and inflammatory profile. Methods MetS patients from two randomized controlled trials, ExMET (n = 24) and OptimEx (n = 34), had performed 4- or 3-months supervised ET programs according to the respective trial protocol. V̇O2peak, MetS-defining RFs (both RCTs), broad blood leukocyte profile, cytokines and plasma proteins (ExMET only) were assessed at baseline and follow-up. Intra-individual changes in RFs were analysed for both trials separately using non-parametric approaches. Associations between changes in each RF over the exercise period (n-fold of baseline values) were correlated using a non-parametrical approach (Spearman). RF clustering was explored by uniform manifold approximation and projection (UMAP) and changes in RF depending on other RF or exercise parameters were explored by recursive partitioning. Results Four months of ET reduced circulating leukocyte counts (63.5% of baseline, P = 8.0e-6), especially effector subtypes. ET response of MetS-associated RFs differed depending on patients' individual RF constellation, but was not associated with individual change in V̇O2peak. Blood pressure lowering depended on cumulative exercise duration (ExMET: ≥102 min per week; OptimEx-MetS: ≥38 min per session) and baseline triglyceride levels (ExMET: <150 mg/dl; OptimEx-MetS: <174.8 mg/dl). Neuropilin-1 plasma levels were inversely associated with fasting plasma triglycerides (R: -0.4, P = 0.004) and changes of both parameters during the ET phase were inversely correlated (R: -0.7, P = 0.0001). Conclusions ET significantly lowered effector leukocyte blood counts. The improvement of MetS-associated cardiovascular RFs depended on individual basal RF profile and exercise duration but was not associated with exercise-mediated increase in V̇O2peak. Neuropilin-1 may be linked to exercise-mediated triglyceride lowering.
Collapse
Affiliation(s)
- Katharina Lechner
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Sylvia Kia
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
| | - Pia von Korn
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Sophia M. Dinges
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Stephan Mueller
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Arnt-Erik Tjønna
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisløff
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Emeline M. Van Craenenbroeck
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Volker Adams
- Department of Cardiology and Internal Medicine, Heart Center Dresden-University Hospital, TU Dresden, Dresden, Germany
| | - Axel Pressler
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Private Center for Sports and Exercise Cardiology, Munich, Germany
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
- Friede Springer—Centre of Cardiovascular Prevention at Charité, Charité University Medicine Berlin, Berlin, Germany
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nicolle Kränkel
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
- Friede Springer—Centre of Cardiovascular Prevention at Charité, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
6
|
Li C, Liu R, Xiong Z, Bao X, Liang S, Zeng H, Jin W, Gong Q, Liu L, Guo J. Ferroptosis: a potential target for the treatment of atherosclerosis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:331-344. [PMID: 38327187 PMCID: PMC10984869 DOI: 10.3724/abbs.2024016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Atherosclerosis (AS), the main contributor to acute cardiovascular events, such as myocardial infarction and ischemic stroke, is characterized by necrotic core formation and plaque instability induced by cell death. The mechanisms of cell death in AS have recently been identified and elucidated. Ferroptosis, a novel iron-dependent form of cell death, has been proven to participate in atherosclerotic progression by increasing endothelial reactive oxygen species (ROS) levels and lipid peroxidation. Furthermore, accumulated intracellular iron activates various signaling pathways or risk factors for AS, such as abnormal lipid metabolism, oxidative stress, and inflammation, which can eventually lead to the disordered function of macrophages, vascular smooth muscle cells, and vascular endothelial cells. However, the molecular pathways through which ferroptosis affects AS development and progression are not entirely understood. This review systematically summarizes the interactions between AS and ferroptosis and provides a feasible approach for inhibiting AS progression from the perspective of ferroptosis.
Collapse
Affiliation(s)
- Chengyi Li
- School of MedicineYangtze UniversityJingzhou434020China
| | - Ran Liu
- School of MedicineYangtze UniversityJingzhou434020China
| | - Zhenyu Xiong
- School of MedicineYangtze UniversityJingzhou434020China
| | - Xue Bao
- School of MedicineYangtze UniversityJingzhou434020China
| | - Sijia Liang
- Department of PharmacologyZhongshan School of MedicineSun Yat-Sen UniversityGuangzhou510120China
| | - Haotian Zeng
- Department of GastroenterologyShenzhen People’s HospitalThe Second Clinical Medical CollegeJinan UniversityShenzhen518000China
| | - Wei Jin
- Department of Second Ward of General PediatricsSuizhou Central HospitalHubei University of MedicineSuizhou441300China
| | - Quan Gong
- School of MedicineYangtze UniversityJingzhou434020China
| | - Lian Liu
- School of MedicineYangtze UniversityJingzhou434020China
| | - Jiawei Guo
- School of MedicineYangtze UniversityJingzhou434020China
| |
Collapse
|
7
|
Zhang J, Yu H, Pan R, Miao G, Wang Y, Li Z, Yu H, Lu L, Jin X. Airborne magnetite nanoparticles induced early vascular pathologies by disrupting lipid metabolism under high-fat dietary patterns. ENVIRONMENTAL TOXICOLOGY 2024; 39:1175-1186. [PMID: 37860912 DOI: 10.1002/tox.24005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/21/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023]
Abstract
Magnetite nanoparticles (MNPs) have been extensively detected in the atmospheric environment and implicated as a prominent threat to atherosclerosis, a chronic vascular inflammatory disease. Due to globalization and economic development, the dramatic shift in diet from traditional to high-fat dietary patterns aggravated atherosclerosis progression induced by environmental factors. However, limited knowledge is available regarding vascular risks and underlying mechanisms of airborne MNPs in high-risk populations with high-fat dietary habits. Herein, we demonstrated that MNPs exerted a proatherogenic effect under high-fat dietary patterns, leading to aortic wall thickening, elastic fiber disorganization, macrophage infiltration, and local inflammation. Based on the correlation analysis between MNPs and PM group, we identified that MNPs might be a key PM component in atherogenic toxicity. MNPs exposure disturbed the dynamic process of lipid metabolism, manifested as aortic lipid accumulation, dyslipidemia, and hepatic lipid metabolism disorder, which was modulated by the JAK-STAT pathway. Overall, these findings provide new insight into understanding the cardiovascular risks and mechanisms of MNPs among high-risk populations.
Collapse
Affiliation(s)
- Jingxu Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Hongyan Yu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Ruonan Pan
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Gan Miao
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Yu Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Ziyuan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Haiyi Yu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Lin Lu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Xiaoting Jin
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| |
Collapse
|
8
|
Luo X, Shi J, Wang R, Cao L, Gao Y, Wang J, Hong M, Sun X, Zhang Y. Near-Infrared Persistent Luminescence Nanoprobe for Early Detection of Atherosclerotic Plaque. ACS NANO 2024; 18:6500-6512. [PMID: 38348833 DOI: 10.1021/acsnano.3c12136] [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: 02/28/2024]
Abstract
Atherosclerosis (AS) is a crucial contributor to various cardiovascular diseases (CVDs), which seriously threaten human life and health. Early and accurate recognition of AS plaques is essential for the prevention and treatment of CVD. Herein, we introduce an AS-targeting nanoprobe based on near-infrared (NIR) persistent luminescence nanoparticles (PLNPs), developing a highly sensitive NIR persistent luminescence (PersL) AS plaque imaging technique and successfully realizing early AS plaque detection. The nanoprobe exhibits good monodispersity and regular spherical morphology and also owns exceptional NIR PersL performance upon repetitive irradiation by biological window light. The surface-conjugated antibody (anti-osteopontin) endowed nanoprobe excellent targeting ability to foam cells within plaques. After intravenously injected nanoprobe into AS model mice, the highly sensitive PersL imaging technique can accurately detect AS plaques prior to ultrasonography (US) and magnetic resonance imaging (MRI). Specifically, the NIR PersL imaging reveals AS plaques at the earliest within 2 weeks, with higher signal-to-background ratio (SBR) up to 5.72. Based on this technique, the nanoprobe has great potential for applications in the prevention and treatment of CVD, the study of AS pathogenesis, and the screening of anti-AS drugs.
Collapse
Affiliation(s)
- Xiaofang Luo
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
- School of Rare Earths, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Science, Ganzhou 341000, People's Republic of China
| | - Junpeng Shi
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
- School of Rare Earths, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Science, Ganzhou 341000, People's Republic of China
| | - Ruoping Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Longlong Cao
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Yan Gao
- School of Rare Earths, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Science, Ganzhou 341000, People's Republic of China
| | - Jinyuan Wang
- School of Rare Earths, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Science, Ganzhou 341000, People's Republic of China
| | - Maochun Hong
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Xia Sun
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350108, People's Republic of China
| | - Yun Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| |
Collapse
|
9
|
Yi W, Chen F, Yuan M, Wang C, Wang S, Wen J, Zou Q, Pu Y, Cai Z. High-fat diet induces cognitive impairment through repression of SIRT1/AMPK-mediated autophagy. Exp Neurol 2024; 371:114591. [PMID: 37898395 DOI: 10.1016/j.expneurol.2023.114591] [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: 03/22/2023] [Revised: 09/20/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
AIMS Recent evidence suggests an association between a high-fat diet (HFD) and cognitive decline. HFD may reduce synaptic plasticity and cause tau hyperphosphorylation, but the mechanisms involved remain unclear. The purpose of this study was to explore whether Sirtuin1 (SIRT1)/AMP-activated protein kinase (AMPK) pathway was involved in this pathogenic effect in the HFD exposed mice. METHODS C57BL/6 mice at 12 months of age were fed a standard (9% kcal fat) or high-fat (60% kcal fat) diet for 22 weeks, and Neuro-2a (N2a) cells were treated with normal culture medium or a palmitic acid (PA) medium (100uM) for 40 h. After that, cognitive function was tested by Morris water maze (MWM). The levels of proteins involved in SIRT1/AMPK pathway and autophagy were measured using western blotting and immunofluorescence. We also assessed the phosphorylation of tau protein and synapse. RESULTS The mice presented impaired learning and memory abilities. We further found decreased levels of synaptophysin (Syn) and brain-derived neurotrophic factor (BDNF), increased tau46 and phosphorylated tau protein, and damaged neurons in mice after HFD or in N2a cells treated with PA medium. Moreover, HFD can also reduce the expression of SIRT1, inhibit AMPK phosphorylation, and block autophagic flow in both mice and cells. After treating the cells with the SIRT1 agonist SRT1720, SIRT1/AMPK pathway and autophagy-related proteins were partially reversed and the number of PA-induced positive cells was alleviated in senescence-associated β-galactosidase (SA-β-gal) staining. CONCLUSIONS HFD may inhibit the expression of SIRT1/AMPK pathway and disrupt autophagy flux, and result in tau hyperphosphorylation and synaptic dysfunction during aging, which ultimately lead to cognitive decline.
Collapse
Affiliation(s)
- Wenmin Yi
- The fifth Clinical College of Chongqing Medical University, Chongqing 402160, China; Department of Neurology, Chongqing General Hospital, Chongqing 400013, China; Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China; Chongqing Medical University, Chongqing 400016, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing 400799, China
| | - Fei Chen
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China; Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China; Chongqing Medical University, Chongqing 400016, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing 400799, China
| | - Minghao Yuan
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China; Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China; Chongqing Medical University, Chongqing 400016, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing 400799, China
| | - Chuanling Wang
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China; Chongqing Medical University, Chongqing 400016, China
| | - Shengyuan Wang
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China; Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China; Chongqing Medical University, Chongqing 400016, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing 400799, China
| | - Jie Wen
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China; Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Qian Zou
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China; Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Yinshuang Pu
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China; Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Zhiyou Cai
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China; Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China; Chongqing Medical University, Chongqing 400016, China; Chongqing School, University of Chinese Academy of Sciences, Chongqing 400799, China.
| |
Collapse
|
10
|
Shetty S, Duesman SJ, Patel S, Huyhn P, Shroff S, Das A, Chowhan D, Sebra R, Beaumont K, McAlpine CS, Rajbhandari P, Rajbhandari AK. Sexually dimorphic role of diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567534. [PMID: 38014350 PMCID: PMC10680837 DOI: 10.1101/2023.11.17.567534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Scientific evidence underscores the influence of biological sex on the interplay between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress jointly contribute to metabolic dysregulation in both males and females. To address this gap, our study aimed to investigate the combined effects of a high-fat diet (HFD) and repeated footshock stress on fear-related behaviors and metabolic outcomes in male and female mice. Using a robust rodent model that recapitulates key aspects of post-traumatic stress disorder (PTSD), we subjected mice to footshock stressor followed by weekly reminder footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. Our findings revealed that HFD impaired fear memory extinction in male mice that received initial stressor but not in female mice. Blood glucose levels were influenced by both diet and sex, with HFD-fed female mice displaying elevated levels that returned to baseline in the absence of stress, a pattern not observed in male mice. Male mice on HFD exhibited higher energy expenditure, while HFD-fed female mice showed a decreased respiratory exchange ratio (RER). Sex-specific alterations in pro-inflammatory markers and abundance of hematopoietic stem cells were observed in chronically stressed mice on an HFD in different peripheral tissues, indicating the manifestation of distinct comorbid disorders. Single-nuclei RNA sequencing of the ventromedial hypothalamus from stressed mice on an HFD provided insights into sex-specific glial cell activation and cell-type-specific transcriptomic changes. In conclusion, our study offers a comprehensive understanding of the intricate interactions between stress, diet, sex, and various physiological and behavioral outcomes, shedding light on a potential brain region coordinating these interactions.
Collapse
Affiliation(s)
- Sanutha Shetty
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Samuel J. Duesman
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Sanil Patel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Pacific Huyhn
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Sanjana Shroff
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anika Das
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Center for Excellence in Youth Education, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Disha Chowhan
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin Beaumont
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S. McAlpine
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Prashant Rajbhandari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Senior authors
| | - Abha K. Rajbhandari
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Senior authors
| |
Collapse
|
11
|
Dai N, Tang X, Weng X, Cai H, Zhuang J, Yang G, Zhou F, Wu P, Liu B, Duan S, Yu Y, Guo W, Ju Z, Zhang L, Wang Z, Wang Y, Lu B, Shi H, Qian J, Ge J. Stress-Related Neural Activity Associates With Coronary Plaque Vulnerability and Subsequent Cardiovascular Events. JACC Cardiovasc Imaging 2023; 16:1404-1415. [PMID: 37269269 DOI: 10.1016/j.jcmg.2023.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND Stress-related neural activity (SNA) assessed by amygdalar activity can predict cardiovascular events. However, its mechanistic linkage with plaque vulnerability is not fully elucidated. OBJECTIVES The authors aimed to investigate the association of SNA with coronary plaque morphologic and inflammatory features as well as their ability in predicting major adverse cardiovascular events (MACE). METHODS A total of 299 patients with coronary artery disease (CAD) and without cancer underwent 18F-fluorodexoyglucose positron emission tomography/computed tomography (PET/CT) and available coronary computed tomographic angiography (CCTA) between January 1, 2013, and December 31, 2020. SNA and bone-marrow activity (BMA) were assessed with validated methods. Coronary inflammation (fat attenuation index [FAI]) and high-risk plaque (HRP) characteristics were assessed by CCTA. Relations between these features were analyzed. Relations between SNA and MACE were assessed with Cox models, log-rank tests, and mediation (path) analyses. RESULTS SNA was significant correlated with BMA (r = 0.39; P < 0.001) and FAI (r = 0.49; P < 0.001). Patients with heightened SNA are more likely to have HRP (40.7% vs 23.5%; P = 0.002) and increase risk of MACE (17.2% vs 5.1%, adjusted HR 3.22; 95% CI: 1.31-7.93; P = 0.011). Mediation analysis suggested that higher SNA associates with MACE via a serial mechanism involving BMA, FAI, and HRP. CONCLUSIONS SNA is significantly correlated with FAI and HRP in patients with CAD. Furthermore, such neural activity was associated with MACE, which was mediated in part by leukopoietic activity in the bone marrow, coronary inflammation, and plaque vulnerability.
Collapse
Affiliation(s)
- Neng Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xianglin Tang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Haidong Cai
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Shanghai, China
| | - Jianhui Zhuang
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai, China
| | - Guangjie Yang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Fan Zhou
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Ping Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Bao Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China; The Nuclear Medicine and Molecular Imaging Clinical Translation Institute of Soochow University, Changzhou, Jiangsu Province, China
| | | | - Yongfu Yu
- School of Public Health and The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Weifeng Guo
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China
| | - Zhiguo Ju
- College of Medical Imaging, Shanghai University of Medicine and Health Science, Shanghai, China
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Zhenguang Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China; The Nuclear Medicine and Molecular Imaging Clinical Translation Institute of Soochow University, Changzhou, Jiangsu Province, China
| | - Bin Lu
- Department of Radiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Lab and National Center for Cardiovascular Diseases, Beijing, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China.
| |
Collapse
|
12
|
Baumer Y, Singh K, Baez AS, Gutierrez-Huerta CA, Chen L, Igboko M, Turner BS, Yeboah JA, Reger RN, Ortiz-Whittingham LR, Bleck CK, Mitchell VM, Collins BS, Pirooznia M, Dagur PK, Allan DS, Muallem-Schwartz D, Childs RW, Powell-Wiley TM. Social Determinants modulate NK cell activity via obesity, LDL, and DUSP1 signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.556825. [PMID: 37745366 PMCID: PMC10515802 DOI: 10.1101/2023.09.12.556825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Adverse social determinants of health (aSDoH) are associated with obesity and related comorbidities like diabetes, cardiovascular disease, and cancer. Obesity is also associated with natural killer cell (NK) dysregulation, suggesting a potential mechanistic link. Therefore, we measured NK phenotypes and function in a cohort of African-American (AA) women from resource-limited neighborhoods. Obesity was associated with reduced NK cytotoxicity and a shift towards a regulatory phenotype. In vitro, LDL promoted NK dysfunction, implicating hyperlipidemia as a mediator of obesity-related immune dysregulation. Dual specific phosphatase 1 (DUSP1) was induced by LDL and was upregulated in NK cells from subjects with obesity, implicating DUSP1 in obesity-mediated NK dysfunction. In vitro, DUSP1 repressed LAMP1/CD107a, depleting NK cells of functional lysosomes to prevent degranulation and cytokine secretion. Together, these data provide novel mechanistic links between aSDoH, obesity, and immune dysregulation that could be leveraged to improve outcomes in marginalized populations.
Collapse
Affiliation(s)
- Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Komudi Singh
- Bioinformatics and Computational Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrew S. Baez
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christian A. Gutierrez-Huerta
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Long Chen
- Section of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Muna Igboko
- Section of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Briana S. Turner
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Josette A. Yeboah
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Robert N. Reger
- Section of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lola R. Ortiz-Whittingham
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher K.E. Bleck
- Electron Microscopy Core Facility, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Valerie M. Mitchell
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Billy S. Collins
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mehdi Pirooznia
- Bioinformatics and Computational Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pradeep K. Dagur
- Flow Cytometry Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - David S.J. Allan
- Section of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Richard W. Childs
- Section of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiffany M. Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Intramural Research Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
13
|
Abstract
The cardiovascular system is hardwired to the brain via multilayered afferent and efferent polysynaptic axonal connections. Two major anatomically and functionally distinct though closely interacting subcircuits within the cardiovascular system have recently been defined: The artery-brain circuit and the heart-brain circuit. However, how the nervous system impacts cardiovascular disease progression remains poorly understood. Here, we review recent findings on the anatomy, structures, and inner workings of the lesser-known artery-brain circuit and the better-established heart-brain circuit. We explore the evidence that signals from arteries or the heart form a systemic and finely tuned cardiovascular brain circuit: afferent inputs originating in the arterial tree or the heart are conveyed to distinct sensory neurons in the brain. There, primary integration centers act as hubs that receive and integrate artery-brain circuit-derived and heart-brain circuit-derived signals and process them together with axonal connections and humoral cues from distant brain regions. To conclude the cardiovascular brain circuit, integration centers transmit the constantly modified signals to efferent neurons which transfer them back to the cardiovascular system. Importantly, primary integration centers are wired to and receive information from secondary brain centers that control a wide variety of brain traits encoded in engrams including immune memory, stress-regulating hormone release, pain, reward, emotions, and even motivated types of behavior. Finally, we explore the important possibility that brain effector neurons in the cardiovascular brain circuit network connect efferent signals to other peripheral organs including the immune system, the gut, the liver, and adipose tissue. The enormous recent progress vis-à-vis the cardiovascular brain circuit allows us to propose a novel neurobiology-centered cardiovascular disease hypothesis that we term the neuroimmune cardiovascular circuit hypothesis.
Collapse
Affiliation(s)
- Sarajo K Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), Munich, Germany (S.K.M., C.Y., C.W., A.J.R.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (S.K.M., C.W., A.J.R.H.)
| | - Changjun Yin
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), Munich, Germany (S.K.M., C.Y., C.W., A.J.R.H.)
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (C.Y.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), Munich, Germany (S.K.M., C.Y., C.W., A.J.R.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (S.K.M., C.W., A.J.R.H.)
| | - Cristina Godinho-Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal (C.G.-S., H.V.-F.)
| | | | - Qian J Xu
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT (Q.J.X., R.B.C.)
| | - Rui B Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT (Q.J.X., R.B.C.)
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), Munich, Germany (S.K.M., C.Y., C.W., A.J.R.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (S.K.M., C.W., A.J.R.H.)
| |
Collapse
|
14
|
Sanford LD, Adkins AM, Boden AF, Gotthold JD, Harris RD, Shuboni-Mulligan D, Wellman LL, Britten RA. Sleep and Core Body Temperature Alterations Induced by Space Radiation in Rats. Life (Basel) 2023; 13:life13041002. [PMID: 37109531 PMCID: PMC10144689 DOI: 10.3390/life13041002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Sleep problems in astronauts can arise from mission demands and stress and can impact both their health and ability to accomplish mission objectives. In addition to mission-related physical and psychological stressors, the long durations of the proposed Mars missions will expose astronauts to space radiation (SR), which has a significant impact on the brain and may also alter sleep and physiological functions. Therefore, in this study, we assessed sleep, EEG spectra, activity, and core body temperature (CBT) in rats exposed to SR and compared them to age-matched nonirradiated rats. Male outbred Wistar rats (8-9 months old at the time of the study) received SR (15 cGy GCRsim, n = 15) or served as age- and time-matched controls (CTRL, n = 15) without irradiation. At least 90 days after SR and 3 weeks prior to recording, all rats were implanted with telemetry transmitters for recording EEG, activity, and CBT. Sleep, EEG spectra (delta, 0.5-4 Hz; theta, 4-8 Hz; alpha, 8-12 Hz; sigma, 12-16 Hz; beta, 16-24 Hz), activity, and CBT were examined during light and dark periods and during waking and sleeping states. When compared to the CTRLs, SR produced significant reductions in the amounts of dark period total sleep time, total nonrapid eye movement sleep (NREM), and total rapid eye movement sleep (REM), with significant decreases in light and dark period NREM deltas and dark period REM thetas as well as increases in alpha and sigma in NREM and REM during either light or dark periods. The SR animals showed modest increases in some measures of activity. CBT was significantly reduced during waking and sleeping in the light period. These data demonstrate that SR alone can produce alterations to sleep and temperature control that could have consequences for astronauts and their ability to meet mission demands.
Collapse
Affiliation(s)
- Larry D Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Austin M Adkins
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Alea F Boden
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Justin D Gotthold
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ryan D Harris
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Dorela Shuboni-Mulligan
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Laurie L Wellman
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Richard A Britten
- Center for Integrative Neuroscience and Inflammatory Diseases, Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
15
|
Meyer-Lindemann U, Moggio A, Dutsch A, Kessler T, Sager HB. The Impact of Exercise on Immunity, Metabolism, and Atherosclerosis. Int J Mol Sci 2023; 24:3394. [PMID: 36834808 PMCID: PMC9967592 DOI: 10.3390/ijms24043394] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Physical exercise represents an effective preventive and therapeutic strategy beneficially modifying the course of multiple diseases. The protective mechanisms of exercise are manifold; primarily, they are elicited by alterations in metabolic and inflammatory pathways. Exercise intensity and duration strongly influence the provoked response. This narrative review aims to provide comprehensive up-to-date insights into the beneficial effects of physical exercise by illustrating the impact of moderate and vigorous exercise on innate and adaptive immunity. Specifically, we describe qualitative and quantitative changes in different leukocyte subsets while distinguishing between acute and chronic exercise effects. Further, we elaborate on how exercise modifies the progression of atherosclerosis, the leading cause of death worldwide, representing a prime example of a disease triggered by metabolic and inflammatory pathways. Here, we describe how exercise counteracts causal contributors and thereby improves outcomes. In addition, we identify gaps that still need to be addressed in the future.
Collapse
Affiliation(s)
- Ulrike Meyer-Lindemann
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Aldo Moggio
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
| | - Alexander Dutsch
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
16
|
Páscoa R, Teixeira A, Monteiro H, Prazeres F, Martins C. Association of lifestyle and sociodemographic factors on multimorbidity: a cross-sectional study in Portugal. BMC Public Health 2022; 22:2341. [DOI: 10.1186/s12889-022-14640-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 11/15/2022] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
Lifestyle factors are widely recognized as modifiers and major risk factors for non-communicable diseases. Previous studies on the prevalence of multimorbidity in Portugal predict an unfavourable reality. The aim of the present study was to analyse 1) the prevalence of multimorbidity in Portugal and 2) the association of individuals’ lifestyles and sociodemographic factors with multimorbidity.
Methods
A cross-sectional, population-wide study was conducted on a representative sample of the general population of Portuguese adults aged ≥ 20 years. Categorical variables were described by their respective absolute and relative frequencies (n (%)). All variables with a p-value < 20% were included in the multiple logistic regression model. The variables were removed one by one in descending order of p-value (p) until the model contained only significant variables. The results are presented using the odds ratio and 95% confidence intervals. P-values < 5% were considered significant.
Results
The prevalence of multimorbidity was 48.9% (n = 436), and the odds of multimorbidity increased 4% (p < 0.001) for each year of increase in age. Participants with reasonable general health status had higher odds of multimorbidity (Odds ratio (OR) = 3.04; p < 0.001), and those with poor or very poor general health status had even higher odds (OR = 9.14; p < 0.001). Compared to those who never smoked, participants who quit smoking ≥ 1 year presented an increase of 91% (p = 0.005) in the odds of multimorbidity. Individuals with no good-quality sleep, non-moderate screen time, or non-moderate stress level had higher odds of multimorbidity (OR = 1.98; OR = 1.88; OR = 2.22, respectively. p < 0.001).
Conclusions
This study presented a new approach to multimorbidity in Portugal. Population-based, multidimensional lifestyle interventions are needed. It seems necessary to optimize and adjust measures to prevent non-communicable diseases to improve health in Portugal. In the future, longitudinal studies will be an asset to reinforce and clarify these conclusions.
Collapse
|
17
|
Zhou J, Yang R, Sun Y, Luo F, Zhang J, Ma H, Guan M. HClO-triggered interventional probe enabled early detection and intervention of atherosclerosis. Analyst 2022; 148:163-174. [PMID: 36464987 DOI: 10.1039/d2an01374f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of early atherosclerosis (AS). Targeting foam cell formation can be a promising approach for the early detection and prevention of AS. However, only a few studies have actually examined foam cells in vivo, and most methods combined nanotechnology with angiography, which is complex and could cause further damage to the endothelium. Herein, based on methylene blue, a biosafe NIR dye approved by the FDA, an interventional probe (HMB-NA@Mp) triggered by hypochlorous acid (HClO) was designed for imaging foam cells easily, safely, and effectively in the early stage of AS. Here, encapsulation of the probe by foam cells targeted platelet membrane (Mp) increased probe targeting and reduced toxicity. Cell and animal experimental results showed that the probe could accumulate at the lesion site and significantly enhance fluorescence in the early AS model group. Remarkably, at the same time, it could also release the metabolite niacin, which played a role in inhibiting atherosclerosis. Thus, HMB-NA@Mp is expected to be a powerful means for the early detection and timely intervention of early AS in the absence of clinical symptoms.
Collapse
Affiliation(s)
- Jie Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Ruhe Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Yiwen Sun
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Fusui Luo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Jin Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Huili Ma
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Min Guan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
18
|
Comprehensive Analysis of Novel Genes and Pathways Associated with Osteogenic Differentiation of Adipose Stem Cells. DISEASE MARKERS 2022; 2022:4870981. [PMID: 36133435 PMCID: PMC9484926 DOI: 10.1155/2022/4870981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/20/2022] [Indexed: 11/18/2022]
Abstract
Background. Adipose-derived stem cells (ADSCs) are an important alternative source of mesenchymal stem cells (MSCs) and show great promise in tissue engineering and regenerative medicine applications. However, identifying the novel genes and pathways and finding the underlying mechanisms regulating ADSCs osteogenic differentiation remain urgent. Methods. We downloaded the gene expression profiles of GSE63754 and GSE37329 from the Gene Expression Omnibus (GEO) Database. We derived differentially expressed genes (DEGs) before and after ADSC osteogenic differentiation, followed by Gene Ontology (GO) functional and KEGG pathway analysis and protein-protein interaction (PPI) network analysis. 211 differentially expressed genes (142 upregulated genes and 69 downregulated genes) were aberrantly expressed. GO analysis revealed that these DEGs were associated with extracellular matrix organization, protein extracellular matrix, and semaphorin receptor binding. Conclusions. Our study provides novel genes and pathways that play important roles in regulating ADSC osteogenic differentiation, which may have potential therapeutic targets for clinic.
Collapse
|
19
|
Zolotareva A, Shchebetenko S, Belousova S, Danilova I, Tseilikman V, Lapshin M, Sarapultseva L, Makhniova S, Sarapultseva M, Komelkova M, Hu D, Luo S, Lisovskaya E, Sarapultsev A. Big Five Traits as Predictors of a Healthy Lifestyle during the COVID-19 Pandemic: Results of a Russian Cross-Sectional Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10716. [PMID: 36078431 PMCID: PMC9517801 DOI: 10.3390/ijerph191710716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 06/02/2023]
Abstract
The healthy lifestyle of people around the world has changed dramatically during the COVID-19 pandemic. The personality risk factors for these processes from around the world remain understudied. This study aimed to examine the associations of the Big Five traits with a healthy lifestyle during the COVID-19 pandemic. In a cross-sectional study, data from 1215 Russian university students were analyzed. Participants completed the Big Five Inventory-10 and Short Multidimensional Inventory Lifestyle Evaluation. The results showed that personality traits predicted many dimensions of a healthy lifestyle during the COVID-19 pandemic. Diet and nutrition were positively predicted by extraversion, agreeableness, and conscientiousness, and it was negatively predicted by neuroticism. Substance abuse was positively predicted by agreeableness and conscientiousness, and it was negatively predicted by extraversion. Physical activity was positively predicted by extraversion and conscientiousness, and it was negatively predicted by neuroticism. Stress management was positively predicted by extraversion and conscientiousness, and it was negatively predicted by neuroticism. Restorative sleep was positively predicted by extraversion and conscientiousness, and it was negatively predicted by neuroticism. Social support for healthy practices was positively predicted by extraversion, agreeableness, and conscientiousness. Environmental exposures were positively predicted by extraversion, and neuroticism was positively and negatively predicted by conscientiousness. Our findings may be useful for further exploration of personality risk factors for healthy practices in challenging life circumstances.
Collapse
Affiliation(s)
- Alena Zolotareva
- School of Psychology, HSE University, 101000 Moscow, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| | | | - Svetlana Belousova
- Department of Psychology, Chelyabinsk State University, 454001 Chelyabinsk, Russia
| | - Irina Danilova
- Institute of Natural Sciences and Mathematics, Ural Federal University Named after the First President of Russia B.N. Yeltsin, 620002 Ekaterinburg, Russia
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| | - Vadim Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
| | - Maxim Lapshin
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| | - Lilia Sarapultseva
- Department of Mathematics and Natural Sciences, Russian State Vocational Pedagogical University, 620012 Ekaterinburg, Russia
| | - Svetlana Makhniova
- Department of Mathematics and Natural Sciences, Russian State Vocational Pedagogical University, 620012 Ekaterinburg, Russia
| | - Maria Sarapultseva
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
- Department of Pediatric Dentistry, Medical Firm Vital EBB, 620144 Ekaterinburg, Russia
| | - Maria Komelkova
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ekaterina Lisovskaya
- Department of Psychology, Chelyabinsk State University, 454001 Chelyabinsk, Russia
| | - Alexey Sarapultsev
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| |
Collapse
|
20
|
Kraemer MB, Garbuio ALP, Kaneko LO, Gobatto CA, Manchado-Gobatto FB, dos Reis IGM, Messias LHD. Associations among sleep, hematologic profile, and aerobic and anerobic capacity of young swimmers: A complex network approach. Front Physiol 2022; 13:948422. [PMID: 36091363 PMCID: PMC9448919 DOI: 10.3389/fphys.2022.948422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Although the link between sleep and hematological parameters is well-described, it is unclear how this integration affects the swimmer’s performance. The parameters derived from the non-invasive critical velocity protocol have been extensively used to evaluate these athletes, especially the aerobic capacity (critical velocity—CV) and the anaerobic work capacity (AWC). Thus, this study applied the complex network model to verify the influence of sleep and hematological variables on the CV and AWC of young swimmers. Thirty-eight swimmers (male, n = 20; female, n = 18) completed five experimental evaluations. Initially, the athletes attended the laboratory facilities for venous blood collection, anthropometric measurements, and application of sleep questionnaires. Over the 4 subsequent days, athletes performed randomized maximal efforts on distances of 100, 200, 400, and 800-m. The aerobic and anerobic parameters were determined by linear function between distance vs. time, where CV relates to the slope of regression and AWC to y-intercept. Weighted but untargeted networks were generated based on significant (p < 0.05) correlations among variables regardless of the correlation coefficient. Betweenness and eigenvector metrics were used to highlight the more important nodes inside the complex network. Regardless of the centrality metric, basophils and red blood cells appeared as influential nodes in the networks with AWC or CV as targets. The role of other hematologic components was also revealed in these metrics, along with sleep total time. Overall, these results trigger new discussion on the influence of sleep and hematologic profile on the swimmer’s performance, and the relationships presented by this targeted complex network can be an important tool throughout the athlete’s development.
Collapse
Affiliation(s)
- Mauricio Beitia Kraemer
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| | - Ana Luíza Paula Garbuio
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| | - Luisa Oliveira Kaneko
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| | - Claudio Alexandre Gobatto
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Ivan Gustavo Masseli dos Reis
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| | - Leonardo Henrique Dalcheco Messias
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
- *Correspondence: Leonardo Henrique Dalcheco Messias,
| |
Collapse
|
21
|
Devesa A, Lobo-González M, Martínez-Milla J, Oliva B, García-Lunar I, Mastrangelo A, España S, Sanz J, Mendiguren JM, Bueno H, Fuster JJ, Andrés V, Fernández-Ortiz A, Sancho D, Fernández-Friera L, Sanchez-Gonzalez J, Rossello X, Ibanez B, Fuster V. Bone marrow activation in response to metabolic syndrome and early atherosclerosis. Eur Heart J 2022; 43:1809-1828. [PMID: 35567559 PMCID: PMC9113301 DOI: 10.1093/eurheartj/ehac102] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/06/2022] [Accepted: 02/18/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS Experimental studies suggest that increased bone marrow (BM) activity is involved in the association between cardiovascular risk factors and inflammation in atherosclerosis. However, human data to support this association are sparse. The purpose was to study the association between cardiovascular risk factors, BM activation, and subclinical atherosclerosis. METHODS AND RESULTS Whole body vascular 18F-fluorodeoxyglucose positron emission tomography/magnetic resonance imaging (18F-FDG PET/MRI) was performed in 745 apparently healthy individuals [median age 50.5 (46.8-53.6) years, 83.8% men] from the Progression of Early Subclinical Atherosclerosis (PESA) study. Bone marrow activation (defined as BM 18F-FDG uptake above the median maximal standardized uptake value) was assessed in the lumbar vertebrae (L3-L4). Systemic inflammation was indexed from circulating biomarkers. Early atherosclerosis was evaluated by arterial metabolic activity by 18F-FDG uptake in five vascular territories. Late atherosclerosis was evaluated by fully formed plaques on MRI. Subjects with BM activation were more frequently men (87.6 vs. 80.0%, P = 0.005) and more frequently had metabolic syndrome (MetS) (22.2 vs. 6.7%, P < 0.001). Bone marrow activation was significantly associated with all MetS components. Bone marrow activation was also associated with increased haematopoiesis-characterized by significantly elevated leucocyte (mainly neutrophil and monocytes) and erythrocyte counts-and with markers of systemic inflammation including high-sensitivity C-reactive protein, ferritin, fibrinogen, P-selectin, and vascular cell adhesion molecule-1. The associations between BM activation and MetS (and its components) and increased erythropoiesis were maintained in the subgroup of participants with no systemic inflammation. Bone marrow activation was significantly associated with high arterial metabolic activity (18F-FDG uptake). The co-occurrence of BM activation and arterial 18F-FDG uptake was associated with more advanced atherosclerosis (i.e. plaque presence and burden). CONCLUSION In apparently healthy individuals, BM 18F-FDG uptake is associated with MetS and its components, even in the absence of systemic inflammation, and with elevated counts of circulating leucocytes. Bone marrow activation is associated with early atherosclerosis, characterized by high arterial metabolic activity. Bone marrow activation appears to be an early phenomenon in atherosclerosis development.[Progression of Early Subclinical Atherosclerosis (PESA); NCT01410318].
Collapse
Affiliation(s)
- Ana Devesa
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manuel Lobo-González
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Juan Martínez-Milla
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Belén Oliva
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Inés García-Lunar
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Cardiology Department, Hospital Ramón y Cajal, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Annalaura Mastrangelo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Samuel España
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, IdISSC, Madrid, Spain
| | - Javier Sanz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Hector Bueno
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiology Department, Hospital Universitario 12 de Octubre, and i+12 Research Institute, Madrid, Spain
| | - Jose J Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Antonio Fernández-Ortiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Hospital Clínico San Carlos, Universidad Complutense, IdISSC, Madrid, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Leticia Fernández-Friera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Hospital Universitario HM Montepríncipe-CIEC, Madrid, Spain
| | | | - Xavier Rossello
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiology Department, Hospital Universitari Son Espases-IDISBA, Palma de Mallorca, Spain
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
22
|
Susser LI, Rayner KJ. Through the layers: how macrophages drive atherosclerosis across the vessel wall. J Clin Invest 2022; 132:157011. [PMID: 35499077 PMCID: PMC9057606 DOI: 10.1172/jci157011] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Leah I. Susser
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Katey J. Rayner
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
23
|
Sara JDS, Toya T, Ahmad A, Clark MM, Gilliam WP, Lerman LO, Lerman A. Mental Stress and Its Effects on Vascular Health. Mayo Clin Proc 2022; 97:951-990. [PMID: 35512885 PMCID: PMC9058928 DOI: 10.1016/j.mayocp.2022.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 01/13/2023]
Abstract
Coronary artery disease continues to be a major cause of morbidity and mortality despite significant advances in risk stratification and management. This has prompted the search for alternative nonconventional risk factors that may provide novel therapeutic targets. Psychosocial stress, or mental stress, has emerged as an important risk factor implicated in a higher incidence of cardiovascular events, and although our understanding of this far ranging and interesting phenomenon has developed greatly over recent times, there is still much to be learned regarding how to measure mental stress and how it may impact physical health. With the current coronavirus disease 2019 global pandemic and its incumbent lockdowns and social distancing, understanding the potentially harmful biological effects of stress related to life-changing events and social isolation has become even more important. In the current review our multidisciplinary team discusses stress from a psychosocial perspective and aims to define psychological stress as rigorously as possible; discuss the pathophysiologic mechanisms by which stress may mediate cardiovascular disease, with a particular focus to its effects on vascular health; outline existing methods and approaches to quantify stress by means of a vascular biomarker; outline the mechanisms whereby psychosocial stressors may have their pathologic effects ultimately transduced to the vasculature through the neuroendocrine immunologic axis; highlight areas for improvement to refine existing approaches in clinical research when studying the consequences of psychological stress on cardiovascular health; and discuss evidence-based therapies directed at reducing the deleterious effects of mental stress including those that target endothelial dysfunction. To this end we searched PubMed and Google Scholar to identify studies evaluating the relationship between mental or psychosocial stress and cardiovascular disease with a particular focus on vascular health. Search terms included "myocardial ischemia," "coronary artery disease," "mental stress," "psychological stress," "mental∗ stress∗," "psychologic∗ stress∗," and "cardiovascular disease∗." The search was limited to studies published in English in peer-reviewed journals between 1990 and the present day. To identify potential studies not captured by our database search strategy, we also searched studies listed in the bibliography of relevant publications and reviews.
Collapse
Key Words
- cad, coronary artery disease
- cbt, cognitive behavioral therapy
- cvd, cardiovascular disease
- fmd, flow-mediated dilatation
- il, interleukin
- mi, myocardial infarction
- ms, mental stress
- msimi, mental stress induced myocardial ischemia
- pat, peripheral arterial tonometry
- ped, peripheral endothelial dysfunction
- pet, positron emission tomography
- rh, reactive hyperemia
- ses, socioeconomic status
- tnf, tumor necrosis factor
- vsmc, vascular smooth muscle cells
Collapse
Affiliation(s)
| | - Takumi Toya
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Ali Ahmad
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Matthew M Clark
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Wesley P Gilliam
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Lliach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
24
|
Roy P, Orecchioni M, Ley K. How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat Rev Immunol 2022; 22:251-265. [PMID: 34389841 PMCID: PMC10111155 DOI: 10.1038/s41577-021-00584-1] [Citation(s) in RCA: 216] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is the root cause of many cardiovascular diseases. Extensive research in preclinical models and emerging evidence in humans have established the crucial roles of the innate and adaptive immune systems in driving atherosclerosis-associated chronic inflammation in arterial blood vessels. New techniques have highlighted the enormous heterogeneity of leukocyte subsets in the arterial wall that have pro-inflammatory or regulatory roles in atherogenesis. Understanding the homing and activation pathways of these immune cells, their disease-associated dynamics and their regulation by microbial and metabolic factors will be crucial for the development of clinical interventions for atherosclerosis, including potentially vaccination-based therapeutic strategies. Here, we review key molecular mechanisms of immune cell activation implicated in modulating atherogenesis and provide an update on the contributions of innate and adaptive immune cell subsets in atherosclerosis.
Collapse
Affiliation(s)
- Payel Roy
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
25
|
Hinterdobler J, Schunkert H, Kessler T, Sager HB. Impact of Acute and Chronic Psychosocial Stress on Vascular Inflammation. Antioxid Redox Signal 2021; 35:1531-1550. [PMID: 34293932 PMCID: PMC8713271 DOI: 10.1089/ars.2021.0153] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 01/01/2023]
Abstract
Significance: Atherosclerosis and its complications, such as acute coronary syndromes, are the leading causes of death worldwide. A wide range of inflammatory processes substantially contribute to the initiation and progression of cardiovascular disease (CVD). In addition, epidemiological studies strongly associate both chronic stress and acute psychosocial stress with the occurrence of CVDs. Recent Advances: Extensive research during recent decades has not only identified major pathways in cardiovascular inflammation but also revealed a link between psychosocial factors and the immune system in the context of atherosclerosis. Both chronic and acute psychosocial stress drive systemic inflammation via neuroimmune interactions and promote atherosclerosis progression. Critical Issues: The associations human epidemiological studies found between psychosocial stress and cardiovascular inflammation have been substantiated by additional experimental studies in mice and humans. However, we do not yet fully understand the mechanisms through which psychosocial stress drives cardiovascular inflammation; consequently, specific treatment, although urgently needed, is lacking. Future Directions: Psychosocial factors are increasingly acknowledged as risk factors for CVD and are currently treated via behavioral interventions. Additional mechanistic insights might provide novel pharmacological treatment options to reduce stress-related morbidity and mortality. Antioxid. Redox Signal. 35, 1531-1550.
Collapse
Affiliation(s)
- Julia Hinterdobler
- Department of Cardiology, German Heart Centre Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Centre Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Centre Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
26
|
Wu J, Jin L, Tan JY, Chen XF, Wang QQ, Yuan GY, Chen TX. The effects of a biodegradable Mg-based alloy on the function of VSMCs via immunoregulation of macrophages through Mg-induced responses. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1292. [PMID: 34532429 PMCID: PMC8422083 DOI: 10.21037/atm-21-1375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
Background Restenosis is one of the worst side effects of percutaneous coronary intervention (PCI) due to neointima formation resulting from the excessive proliferation and migration of vascular smooth muscle cells (VSMCs) and continuous inflammation. Biodegradable Mg-based alloy is a promising candidate material because of its good mechanical properties and biocompatibility, and biodegradation of cardiovascular stents. Although studies have shown reduced neointima formation after Mg-based CVS implantation in vivo, these findings were inconsistent with in vitro studies, demonstrating magnesium-mediated promotion of the proliferation and migration of VSMCs. Given the vital role of activated macrophage-driven inflammation in neointima formation, along with the well-demonstrated crosstalk between macrophages and VSMCs, we investigated the interactions of a biodegradable Mg-Nd-Zn-Zr alloy (denoted JDBM), which is especially important for cardiovascular stents, with VSMCs via macrophages. Methods JDBM extracts and MgCl2 solutions were prepared to study their effect on macrophages. To study the effects of the JDBM extracts and MgCl2 solutions on the function of VSMCs via immunoregulation of macrophages, conditioned media (CM) obtained from macrophages was used to establish a VSMC-macrophage indirect coculture system. Results Our results showed that both JDBM extracts and MgCl2 solutions significantly attenuated the inflammatory response stimulated by lipopolysaccharide (LPS)-activated macrophages and converted macrophages into M2-type cells. In addition, JDBM extracts and MgCl2 solutions significantly decreased the expression of genes related to VSMC phenotypic switching, migration, and proliferation in macrophages. Furthermore, the proliferation, migration, and proinflammatory phenotypic switching of VSMCs were significantly inhibited when the cells were incubated with CMs from macrophages treated with LPS + extracts or LPS + MgCl2 solutions. Conclusions Taken together, our results suggested that the magnesium in the JDBM extract could affect the functions of VSMCs through macrophage-mediated immunoregulation, inhibiting smooth muscle hyperproliferation to suppress restenosis after implantation of a biodegradable Mg-based stent.
Collapse
Affiliation(s)
- Jing Wu
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Allergy/Immunology Innovation Team, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology/Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liang Jin
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology/Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jin-Yun Tan
- Department of Vascular Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Xia-Fang Chen
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Neonatology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | - Guang-Yin Yuan
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.,National Engineering Research Center of Light Alloys Net Forming and State Key Laboratory of Metal Matrix Composite, Shanghai Jiao Tong University, Shanghai, China
| | - Tong-Xin Chen
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Allergy/Immunology Innovation Team, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology/Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
27
|
Pérez-Olivares L, Soehnlein O. Contemporary Lifestyle and Neutrophil Extracellular Traps: An Emerging Link in Atherosclerosis Disease. Cells 2021; 10:1985. [PMID: 34440753 PMCID: PMC8394440 DOI: 10.3390/cells10081985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are networks of extracellular genetic material decorated with proteins of nuclear, granular and cytosolic origin that activated neutrophils expel under pathogenic inflammatory conditions. NETs are part of the host's innate immune defense system against invading pathogens. Interestingly, these extracellular structures can also be released in response to sterile inflammatory stimuli (e.g., shear stress, lipidic molecules, pro-thrombotic factors, aggregated platelets, or pro-inflammatory cytokines), as in atherosclerosis disease. Indeed, NETs have been identified in the intimal surface of diseased arteries under cardiovascular disease conditions, where they sustain inflammation via NET-mediated cell-adhesion mechanisms and promote cellular dysfunction and tissue damage via NET-associated cytotoxicity. This review will focus on (1) the active role of neutrophils and NETs as underestimated players of the inflammatory process during atherogenesis and lesion progression; (2) how these extracellular structures communicate with the main cell types present in the atherosclerotic lesion in the arterial wall; and (3) how these neutrophil effector functions interplay with lifestyle-derived risk factors such as an unbalanced diet, physical inactivity, smoking or lack of sleep quality, which represent major elements in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Laura Pérez-Olivares
- Center for Molecular Biology of Inflammation (ZMBE), Institute for Experimental Pathology (ExPat), Westfälische Wilhelms-Universität (WWU), 48149 Münster, Germany;
| | - Oliver Soehnlein
- Center for Molecular Biology of Inflammation (ZMBE), Institute for Experimental Pathology (ExPat), Westfälische Wilhelms-Universität (WWU), 48149 Münster, Germany;
- Department of Physiology and Pharmacology (FyFa), Karolinska Institute, 17165 Stockholm, Sweden
| |
Collapse
|
28
|
Bhattacharya R, Zekavat SM, Uddin MM, Pirruccello J, Niroula A, Gibson C, Griffin GK, Libby P, Ebert BL, Bick A, Natarajan P. Association of Diet Quality With Prevalence of Clonal Hematopoiesis and Adverse Cardiovascular Events. JAMA Cardiol 2021; 6:1069-1077. [PMID: 34106216 DOI: 10.1001/jamacardio.2021.1678] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Clonal hematopoiesis of indeterminate potential (CHIP), the expansion of somatic leukemogenic variations in hematopoietic stem cells, has been associated with atherosclerotic cardiovascular disease. Because the inherited risk of developing CHIP is low, lifestyle elements such as dietary factors may be associated with the development and outcomes of CHIP. Objective To examine whether there is an association between diet quality and the prevalence of CHIP. Design, Setting, and Participants This retrospective cohort study used data from participants in the UK Biobank, an ongoing population-based study in the United Kingdom that examines whole-exome sequencing data and survey-based information on health-associated behaviors. Individuals from the UK Biobank were recruited between 2006 and 2010 and followed up prospectively with linkage to health data records through May 2020. The present study included 44 111 participants in the UK Biobank who were age 40 to 70 years, had data available from whole-exome sequencing of blood DNA, and were free of coronary artery disease (CAD) or hematologic cancer at baseline. Exposures Diet quality was categorized as unhealthy if the intake of healthy elements (fruits and vegetables) was lower than the median of all survey responses, and the intake of unhealthy elements (red meat, processed food, and added salt) was higher than the median. Diets were classified as healthy if the intake of healthy elements was higher than the median, and the intake of unhealthy elements was lower than the median. The presence of CHIP was detected by data from whole-exome sequencing of blood DNA. Main Outcomes and Measures The primary outcome was CHIP prevalence. Multivariable logistic regression analysis was used to examine the association between diet quality and the presence of CHIP. Multivariable Cox proportional hazards models were used to assess the association of incident events (acute coronary syndromes, coronary revascularization, or death) in each diet quality category stratified by the presence of CHIP. Results Among 44 111 participants (mean [SD] age at time of blood sample collection, 56.3 [8.0] years; 24 507 women [55.6%]), 2271 individuals (5.1%) had an unhealthy diet, 38 552 individuals (87.4%) had an intermediate diet, and 3288 individuals (7.5%) had a healthy diet. A total of 2507 individuals (5.7%) had CHIP, and the prevalence of CHIP decreased as diet quality improved from unhealthy (162 of 2271 participants [7.1%]) to intermediate (2177 of 38 552 participants [5.7%]) to healthy (168 of 3288 participants [5.1%]; P = .003 for trend). Compared with individuals without CHIP who had an intermediate diet, the rates of incident cardiovascular events progressively decreased among those with CHIP who had an unhealthy diet (hazard ratio [HR], 1.52; 95% CI, 1.04-2.22) and those with CHIP who had a healthy diet (HR, 0.99; 95% CI, 0.62-1.58) over a median of 10.0 years (interquartile range, 9.6-10.4 years) of follow-up. Conclusions and Relevance This cohort study suggests that an unhealthy diet quality may be associated with a higher prevalence of CHIP and higher rates of adverse cardiovascular events and death independent of CHIP status.
Collapse
Affiliation(s)
- Romit Bhattacharya
- Cardiovascular Research Center, Massachusetts General Hospital, Boston.,Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Seyedeh Maryam Zekavat
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Cardiovascular Research Center, Massachusetts General Hospital, Boston.,Yale School of Medicine, New Haven, Connecticut
| | - Md Mesbah Uddin
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - James Pirruccello
- Cardiovascular Research Center, Massachusetts General Hospital, Boston.,Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Abhishek Niroula
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Christopher Gibson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gabriel K Griffin
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.,Epigenomics Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benjamin L Ebert
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alexander Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston.,Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Osborne MT, Abohashem S, Zureigat H, Abbasi TA, Tawakol A. Multimodality molecular imaging: Gaining insights into the mechanisms linking chronic stress to cardiovascular disease. J Nucl Cardiol 2021; 28:955-966. [PMID: 33205328 PMCID: PMC8126581 DOI: 10.1007/s12350-020-02424-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Positron emission tomography (PET) imaging can yield unique mechanistic insights into the pathophysiology of atherosclerosis. 18F-fluorodeoxyglucose (18F-FDG), a radiolabeled glucose analog, is retained by cells in proportion to their glycolytic activity. While 18F-FDG accumulates within several cell types in the arterial wall, its retention correlates with macrophage content, providing an index of arterial inflammation (ArtI) which predicts subsequent cardiovascular disease (CVD) events. Furthermore, 18F-FDG-PET imaging allows the simultaneous assessment of metabolic activity in several tissues (e.g., brain, bone marrow) and is performed in conjunction with cross-sectional imaging that enables multi-organ structural assessments. Accordingly, 18F-FDG-PET/computed tomography (CT) imaging facilitates evaluation of disease pathways that span multiple organ systems. Within this paradigm, 18F-FDG-PET/CT imaging has been implemented to study the mechanism linking chronic stress to CVD. To evaluate this, stress-associated neural activity can be quantified (as metabolic activity of the amygdala (AmygA)), while leukopoietic activity, ArtI, and coronary plaque burden are assessed concurrently. Such simultaneous quantification of tissue structures and activities enables the evaluation of multi-organ pathways with the aid of mediation analysis. Using this approach, multi-system 18F-FDG-PET/CT imaging studies have demonstrated that chronically heightened stress-associated neurobiological activity promotes leukopoietic activity and systemic inflammation. This in turn fuels more ArtI and greater non-calcified coronary plaque burden, which result in more CVD events. Subsequent studies have revealed that common stressors, such as chronic noise exposure and income disparities, drive the front end of this pathway to increase CVD risk. Hence, multi-tissue multimodality imaging serves as a powerful tool to uncover complex disease mechanisms.
Collapse
Affiliation(s)
- Michael T Osborne
- Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Yawkey 5E, Boston, MA, 02114-2750, USA
- Cardiovascular Imaging Research Center, Cardiology Division and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shady Abohashem
- Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Yawkey 5E, Boston, MA, 02114-2750, USA
- Cardiovascular Imaging Research Center, Cardiology Division and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hadil Zureigat
- Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Yawkey 5E, Boston, MA, 02114-2750, USA
- Cardiovascular Imaging Research Center, Cardiology Division and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Taimur A Abbasi
- Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Yawkey 5E, Boston, MA, 02114-2750, USA
- Cardiovascular Imaging Research Center, Cardiology Division and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ahmed Tawakol
- Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Yawkey 5E, Boston, MA, 02114-2750, USA.
- Cardiovascular Imaging Research Center, Cardiology Division and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Ludikhuize MC, Rodríguez Colman MJ. Metabolic Regulation of Stem Cells and Differentiation: A Forkhead Box O Transcription Factor Perspective. Antioxid Redox Signal 2021; 34:1004-1024. [PMID: 32847377 DOI: 10.1089/ars.2020.8126] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Stem cell activation and differentiation occur along changes in cellular metabolism. Metabolic transitions translate into changes in redox balance, cell signaling, and epigenetics, thereby regulating these processes. Metabolic transitions are key regulators of cell fate and exemplify the moonlighting nature of many metabolic enzymes and their associated metabolites. Recent Advances: Forkhead box O transcription factors (FOXOs) are bona fide regulators of cellular homeostasis. FOXOs are multitasking proteins able to regulate cell cycle, cellular metabolism, and redox state. Recent and ongoing research poses FOXOs as key factors in stem cell maintenance and differentiation in several tissues. Critical Issues: The multitasking nature of FOXOs and their tissue-specific expression patterns hinders to disclose a possible conserved mechanism of regulation of stem cell maintenance and differentiation. Moreover, cellular metabolism, cell signaling, and epigenetics establish complex regulatory interactions, which challenge the establishment of the causal/temporal nature of metabolic changes and stem cell activation and differentiation. Future Directions: The development of single-cell technologies and in vitro models able to reproduce the dynamics of stem cell differentiation are actively contributing to define the role of metabolism in this process. This knowledge is key to understanding and designing therapies for those pathologies where the balance between proliferation and differentiation is lost. Importantly, metabolic interventions could be applied to optimize stem cell cultures meant for therapeutical applications, such as transplantations, to treat autoimmune and degenerative disorders. Antioxid. Redox Signal. 34, 1004-1024.
Collapse
Affiliation(s)
- Marlies Corine Ludikhuize
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - María José Rodríguez Colman
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Zhu S, Xu K, Jiang Y, Zhu C, Suo C, Cui M, Wang Y, Yuan Z, Xue J, Wang J, Zhang T, Zhao G, Ye W, Huang T, Lu M, Tian W, Jin L, Chen X. The gut microbiome in subclinical atherosclerosis: a population-based multi-phenotype analysis. Rheumatology (Oxford) 2021; 61:258-269. [PMID: 33769467 DOI: 10.1093/rheumatology/keab309] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The altered microbiota, considered as quantitative traits, has also been identified to play pivotal roles in the host vascular physiology and might contribute to diseases. To understand the role of gut microbiota on vascular physiology in the sub-clinical elderly population and how lifestyles affect the composition of host gut microbiota to further impact the pathogenesis of vascular diseases. METHODS Performed a population-based fecal metagenomic study over 569 elderly asymptomatic sub-clinical individuals in rural China. An association network was built based on clinical measurements and detailed epidemiologic questionnaires, including blood chemistry, arterial stiffness, carotid ultrasonography, and metagenomic datasets. RESULTS Carotid arterial atherosclerosis indices, including intima-media thickness (IMT), were shown essentially in the network, and were significantly associated with living habits, socio-economic status, and diet. Using mediation analysis, we found that higher frequency of taking fresh fruits, fresh vegetables, and more exercise significantly reduces carotid arteries atherosclerosis in terms of IMT, PSV and EDV values the through the mediation of Alistepes, Oligella, and Prevotella. The gut microbes explained 16.5% of the mediation effect of lifestyles on the pathogenesis of carotid atherosclerosis. After adjusted, Faecalicatena (OR = 0.20∼0.30) was shown protective in the formation of carotid athersclerosis independently, while Libanicoccus (OR = 2.39∼2.43) were hazardous to carotid arterial IMTs. KEGG/KO analyses revealed a loss of anti-inflammation function in IMT subjects. CONCLUSIONS Our study provided a Chinese population-wide phenotype-metagenomic network, revealing association and mediation effect of gut microbiota on carotid artery atherosclerosis, hinting at a therapeutic and preventive potential of microbiota in vascular diseases.
Collapse
Affiliation(s)
- Sibo Zhu
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Kelin Xu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Department of Biostatistics, School of Public Health, Fudan University; Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Chengkai Zhu
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
| | - Chen Suo
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yingzhe Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ziyu Yuan
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Jiangli Xue
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Genming Zhao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Tingting Huang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ming Lu
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Weizhong Tian
- Department of Medical Imaging, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| |
Collapse
|
32
|
Páscoa R, Teixeira A, Gregório M, Carvalho R, Martins C. Patients' Perspectives about Lifestyle Behaviors and Health in the Context of Family Medicine: A Cross-Sectional Study in Portugal. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18062981. [PMID: 33799435 PMCID: PMC8001049 DOI: 10.3390/ijerph18062981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 11/17/2022]
Abstract
Lifestyle interventions are recognized as essential in the prevention and treatment of non-communicable diseases. Previous studies have shown that Portuguese patients tend to give more importance to diagnostic and laboratory tests than to lifestyle measures, and seem unaware that behavioral risks are the main modifiable risk factors. The study aimed to analyze patients’ perspectives about lifestyle behaviors and health in the context of family medicine in Portugal. A population-based cross-sectional study was carried out in Portugal (the mainland). A total of 900 Portuguese patients aged ≥20 years, representative of the population, were surveyed using face-to-face questionnaires. Participants were selected by the random route method. Descriptive statistics and non-parametric tests were performed to evaluate differences between the personal beliefs and the personal behavior self-assessment, as well as between the level of importance given to the family doctor to address health behaviors and the reported approach implemented by the family doctor, and its association with bio-demographic variables. The results indicate that the vast majority of this Portuguese cohort has informed beliefs regarding lifestyle behaviors, tends to overestimate their own behavior self-assessment, and strongly agrees that it is important that their family doctor asks/advises on these lifestyle behaviors, although the proportion of those who totally agree that their family doctor usually does this is significantly lower. Differences concerning bio-demographic variables were found. Future research directions should focus on the politics, economics, and policy aspects that may have an impact in this area. It will also be important to understand more broadly the relationships between lifestyle behaviors and clinical, physical, and sociodemographic variables.
Collapse
Affiliation(s)
- Rosália Páscoa
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal; (A.T.); (M.G.); (C.M.)
- Centre for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
- Correspondence: ; Tel.: +351-220-426-600
| | - Andreia Teixeira
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal; (A.T.); (M.G.); (C.M.)
- Centre for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
- Instituto Politécnico de Viana do Castelo (IPVC), 4900-347 Viana do Castelo, Portugal
| | - Micaela Gregório
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal; (A.T.); (M.G.); (C.M.)
| | - Rosa Carvalho
- #H4A Primary Healthcare Research Network, 4460-027 Porto, Portugal;
| | - Carlos Martins
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal; (A.T.); (M.G.); (C.M.)
- Centre for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
- #H4A Primary Healthcare Research Network, 4460-027 Porto, Portugal;
| |
Collapse
|
33
|
Haring B, Reiner AP, Liu J, Tobias DK, Whitsel E, Berger JS, Desai P, Wassertheil-Smoller S, LaMonte MJ, Hayden KM, Bick AG, Natarajan P, Weinstock JS, Nguyen PK, Stefanick M, Simon MS, Eaton CB, Kooperberg C, Manson JE. Healthy Lifestyle and Clonal Hematopoiesis of Indeterminate Potential: Results From the Women's Health Initiative. J Am Heart Assoc 2021; 10:e018789. [PMID: 33619969 PMCID: PMC8174283 DOI: 10.1161/jaha.120.018789] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background Presence of clonal hematopoiesis of indeterminate potential (CHIP) is associated with a higher risk of atherosclerotic cardiovascular disease, cancer, and mortality. The relationship between a healthy lifestyle and CHIP is unknown. Methods and Results This analysis included 8709 postmenopausal women (mean age, 66.5 years) enrolled in the WHI (Women's Health Initiative), free of cancer or cardiovascular disease, with deep‐coverage whole genome sequencing data available. Information on lifestyle factors (body mass index, smoking, physical activity, and diet quality) was obtained, and a healthy lifestyle score was created on the basis of healthy criteria met (0 point [least healthy] to 4 points [most healthy]). CHIP was derived on the basis of a prespecified list of leukemogenic driver mutations. The prevalence of CHIP was 8.6%. A higher healthy lifestyle score was not associated with CHIP (multivariable‐adjusted odds ratio [OR] [95% CI], 0.99 [0.80–1.23] and 1.13 [0.93–1.37]) for the upper (3 or 4 points) and middle category (2 points), respectively, versus referent (0 or 1 point). Across score components, a normal and overweight body mass index compared with obese was significantly associated with a lower odds for CHIP (OR, 0.71 [95% CI, 0.57–0.88] and 0.83 [95% CI, 0.68–1.01], respectively; P‐trend 0.0015). Having never smoked compared with being a current smoker tended to be associated with lower odds for CHIP. Conclusions A healthy lifestyle, based on a composite score, was not related to CHIP among postmenopausal women. However, across individual lifestyle factors, having a normal body mass index was strongly associated with a lower prevalence of CHIP. These findings support the idea that certain healthy lifestyle factors are associated with a lower frequency of CHIP.
Collapse
Affiliation(s)
- Bernhard Haring
- Department of Internal Medicine I University of Würzburg Bavaria Germany
| | - Alexander P Reiner
- Division of Public Health Sciences Department of Epidemiology Fred Hutchinson Cancer Research CenterUniversity of Washington Seattle WA
| | | | - Deirdre K Tobias
- Department of Nutrition Harvard T.H. Chan School of Public Health Boston MA.,Division of Preventive Medicine Department of Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Eric Whitsel
- Department of Epidemiology and Medicine University of North Carolina Chapel Hill NC
| | - Jeffrey S Berger
- Department of Medicine Center for the Prevention of Cardiovascular Disease New York University School of Medicine New York City NY
| | - Pinkal Desai
- Division of Hematology and Oncology Weill Cornell Medical College New York NY
| | | | - Michael J LaMonte
- Department of Epidemiology and Environmental Health School of Public Health and Health Professions University at Buffalo-SUNY Buffalo NY
| | - Kathleen M Hayden
- Division of Public Health Sciences Department of Social Sciences and Health Policy Wake Forest School of Medicine Winston-Salem NC
| | - Alexander G Bick
- Department of Medicine Program in Medical and Population Genetics Harvard Medical SchoolBroad Institute of Harvard and MIT Cambridge MA
| | - Pradeep Natarajan
- Department of Medicine Program in Medical and Population Genetics Harvard Medical SchoolBroad Institute of Harvard and MIT Cambridge MA
| | - Joshua S Weinstock
- Department of Biostatistics and Center for Statistical Genetics University of Michigan School of Public Health Ann Arbor MI
| | - Patricia K Nguyen
- Department of Medicine Stanford University Medical Center Palo Alto CA
| | - Marcia Stefanick
- Department of Medicine Stanford University Medical Center Palo Alto CA.,Departments of Obstetrics and Gynecology Stanford University Palo Alto CA
| | - Michael S Simon
- Department of Oncology Karmanos Cancer Institute at Wayne State University Detroit MI
| | - Charles B Eaton
- Department of Epidemiology Center for Primary Care and Prevention Brown University Providence RI
| | | | - JoAnn E Manson
- Division of Preventive Medicine Department of Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| |
Collapse
|
34
|
Hallmarks of aging and immunosenescence: Connecting the dots. Cytokine Growth Factor Rev 2021; 59:9-21. [PMID: 33551332 DOI: 10.1016/j.cytogfr.2021.01.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 01/24/2021] [Indexed: 12/11/2022]
Abstract
Aging is a natural physiological process that features various and variable challenges, associated with loss of homeostasis within the organism, often leading to negative consequences for health. Cellular senescence occurs when cells exhaust the capacity to renew themselves and their tissue environment as the cell cycle comes to a halt. This process is influenced by genetics, metabolism and extrinsic factors. Immunosenescence, the aging of the immune system, is a result of the aging process, but can also in turn act as a secondary inducer of senescence within other tissues. This review aims to summarize the current state of knowledge regarding hallmarks of aging in relation to immunosenescence, with a focus on aging-related imbalances in the medullary environment, as well as the components of the innate and adaptive immune responses. Aging within the immune system alters its functionality, and has consequences for the person's ability to fight infections, as well as for susceptibility to chronic diseases such as cancer and cardiovascular disease. The senescence-associated secretory phenotype is described, as well as the involvement of this phenomenon in the paracrine induction of senescence in otherwise healthy cells. Inflammaging is discussed in detail, along with the comorbidities associated with this process. A knowledge of these processes is required in order to consider possible targets for the application of senotherapeutic agents - interventions with the potential to modulate the senescence process, thus prolonging the healthy lifespan of the immune system and minimizing the secondary effects of immunosenescence.
Collapse
|
35
|
He M, Guo Z, Lu Z, Wei S, Wang Z. High milk consumption is associated with carotid atherosclerosis in middle and old-aged Chinese. INTERNATIONAL JOURNAL CARDIOLOGY HYPERTENSION 2021; 5:100031. [PMID: 33447760 PMCID: PMC7803059 DOI: 10.1016/j.ijchy.2020.100031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/06/2020] [Accepted: 05/08/2020] [Indexed: 10/25/2022]
Abstract
Background Carotid atherosclerosis (CA) can reflect atherosclerotic status of systemic vessels, and is associated with ischemic stroke and cognitive impairment. Milk consumption is growing in China, and data are limited on the association between milk and CA risk. Methods Participants aged ≥40 years in China National Stroke Prevention Project were recruited for carotid ultrasound examination, and those with carotid endarterectomy, carotid stenting stroke or coronary heart disease were excluded. Participants were defined as having CA when intima-media thickness (IMT) ≥1 mm or plaques were detected by ultrasonography. For milk consumption, though contents were not analyzed, people were divided into high consumption (≥200 ml/day and ≥5 day/week) and low consumption (occasional or never) groups. Multivariate logistics regression analyses were done in both full and propensity score-matched population to identify the association between milk consumption and CA. Results 84880 participants were included in the final analysis. After being adjusted by age, gender, living location, smoking habits, drinking habits, physical activity, obesity, hypertension, diabetes, and dyslipidemia, high milk consumption is associated with CA in both full population (odds ratio [OR] = 1.26, 95% confidence interval [CI] 1.19-1.33) and propensity score-matched population (OR = 1.25, 95%CI 1.17-1.34). High milk consumption is also a risk factor for IMT thickening (OR = 1.15, 95%CI 1.08-1.23), carotid plaque formation (OR = 1.17, 95%CI 1.09-1.25) and carotid stenosis over 50% (OR = 1.52, 95%CI 1.10-2.14) in the propensity score-matched population. Conclusions High milk consumption is related to CA in middle and old-aged Chinese population. More cautious advice on milk intake should be given in this population for prevention of atherosclerosis.
Collapse
Affiliation(s)
- Miao He
- Department of Epidemiology and Biostatistics, School of Public Health,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zaogeng Guo
- Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zuxun Lu
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Wei
- Department of Epidemiology and Biostatistics, School of Public Health,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihong Wang
- Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Innovation Platform of Translational Research for Cerebrovascular Diseases, Shenzhen, China
| |
Collapse
|
36
|
Kim KW, Ivanov S, Williams JW. Monocyte Recruitment, Specification, and Function in Atherosclerosis. Cells 2020; 10:E15. [PMID: 33374145 PMCID: PMC7823291 DOI: 10.3390/cells10010015] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022] Open
Abstract
Atherosclerotic lesions progress through the continued recruitment of circulating blood monocytes that differentiate into macrophages within plaque. Lesion-associated macrophages are the primary immune cells present in plaque, where they take up cholesterol and store lipids in the form of small droplets resulting in a unique morphology termed foam cell. Recent scientific advances have used single-cell gene expression profiling, live-cell imaging, and fate mapping approaches to describe macrophage and monocyte contributions to pro- or anti-inflammatory mechanisms, in addition to functions of motility and proliferation within lesions. Yet, many questions regarding tissue-specific regulation of monocyte-to-macrophage differentiation and the contribution of recruited monocytes at stages of atherosclerotic disease progression remain unknown. In this review, we highlight recent advances regarding the role of monocyte and macrophage dynamics in atherosclerotic disease and identify gaps in knowledge that we hope will allow for advancing therapeutic treatment or prevention strategies for cardiovascular disease.
Collapse
Affiliation(s)
- Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Stoyan Ivanov
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire C3M, Université Côte Azur, 06204 Nice, France;
| | - Jesse W. Williams
- Center for Immunology, Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
37
|
Fadini GP, Mehta A, Dhindsa DS, Bonora BM, Sreejit G, Nagareddy P, Quyyumi AA. Circulating stem cells and cardiovascular outcomes: from basic science to the clinic. Eur Heart J 2020; 41:4271-4282. [PMID: 31891403 PMCID: PMC7825095 DOI: 10.1093/eurheartj/ehz923] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/19/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
The cardiovascular and haematopoietic systems have fundamental inter-relationships during development, as well as in health and disease of the adult organism. Although haematopoietic stem cells (HSCs) emerge from a specialized haemogenic endothelium in the embryo, persistence of haemangioblasts in adulthood is debated. Rather, the vast majority of circulating stem cells (CSCs) is composed of bone marrow-derived HSCs and the downstream haematopoietic stem/progenitors (HSPCs). A fraction of these cells, known as endothelial progenitor cells (EPCs), has endothelial specification and vascular tropism. In general, the levels of HSCs, HSPCs, and EPCs are considered indicative of the endogenous regenerative capacity of the organism as a whole and, particularly, of the cardiovascular system. In the last two decades, the research on CSCs has focused on their physiologic role in tissue/organ homoeostasis, their potential application in cell therapies, and their use as clinical biomarkers. In this review, we provide background information on the biology of CSCs and discuss in detail the clinical implications of changing CSC levels in patients with cardiovascular risk factors or established cardiovascular disease. Of particular interest is the mounting evidence available in the literature on the close relationships between reduced levels of CSCs and adverse cardiovascular outcomes in different cohorts of patients. We also discuss potential mechanisms that explain this association. Beyond CSCs' ability to participate in cardiovascular repair, levels of CSCs need to be interpreted in the context of the broader connections between haematopoiesis and cardiovascular function, including the role of clonal haematopoiesis and inflammatory myelopoiesis.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Anurag Mehta
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| | - Devinder Singh Dhindsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| | | | - Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Prabhakara Nagareddy
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Arshed Ali Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| |
Collapse
|
38
|
Fernández-García V, González-Ramos S, Martín-Sanz P, Castrillo A, Boscá L. Contribution of Extramedullary Hematopoiesis to Atherosclerosis. The Spleen as a Neglected Hub of Inflammatory Cells. Front Immunol 2020; 11:586527. [PMID: 33193412 PMCID: PMC7649205 DOI: 10.3389/fimmu.2020.586527] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/06/2020] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) incidence is becoming higher. This fact is promoted by metabolic disorders such as obesity, and aging. Atherosclerosis is the underlying cause of most of these pathologies. It is a chronic inflammatory disease that begins with the progressive accumulation of lipids and fibrotic materials in the blood-vessel wall, which leads to massive leukocyte recruitment. Rupture of the fibrous cap of the atherogenic cusps is responsible for tissue ischemic events, among them myocardial infarction. Extramedullary hematopoiesis (EMH), or blood cell production outside the bone marrow (BM), occurs when the normal production of these cells is impaired (chronic hematological and genetic disorders, leukemia, etc.) or is altered by metabolic disorders, such as hypercholesterolemia, or after myocardial infarction. Recent studies indicate that the main EMH tissues (spleen, liver, adipose and lymph nodes) complement the hematopoietic function of the BM, producing circulating inflammatory cells that infiltrate into the atheroma. Indeed, the spleen, which is a secondary lymphopoietic organ with high metabolic activity, contains a reservoir of myeloid progenitors and monocytes, constituting an important source of inflammatory cells to the atherosclerotic lesion. Furthermore, the spleen also plays an important role in lipid homeostasis and immune-cell selection. Interestingly, clinical evidence from splenectomized subjects shows that they are more susceptible to developing pathologies, such as dyslipidemia and atherosclerosis due to the loss of immune selection. Although CVDs represent the leading cause of death worldwide, the mechanisms involving the spleen-atherosclerosis-heart axis cross-talk remain poorly characterized.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Unidad de Biomedicina, (Unidad Asociada al CSIC), Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Universidad de Las Palmas, Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Grupo de Investigación Medio Ambiente y Salud, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Unidad de Biomedicina, (Unidad Asociada al CSIC), Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Universidad de Las Palmas, Gran Canaria, Spain
| |
Collapse
|
39
|
Ghaemi Kerahrodi J, Michal M. The fear-defense system, emotions, and oxidative stress. Redox Biol 2020; 37:101588. [PMID: 32739155 PMCID: PMC7767737 DOI: 10.1016/j.redox.2020.101588] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/04/2020] [Accepted: 05/17/2020] [Indexed: 12/31/2022] Open
Abstract
Psychosocial stress has a profound impact on well-being and health. The response to stress is associated mainly with the amygdala, a crucial structure of the fear-defense system, essential for social cognition and emotion regulation. Recent neuroimaging-studies demonstrated how an increased metabolic activity of the amygdala enhances inflammation, and leads to cardiometabolic disease. The development of therapeutic strategies depends on our understanding of both which factors activate the fear-defense system and the subsequent molecular mechanisms that translate emotional stress into cell damage. Fear of emotions as an aftermath of attachment trauma is the most important trigger of the maladaptive activation of the fear-defense system. The central molecular pathways are enhanced myelopoiesis and upregulated proinflammatory gene expression, glucocorticoid and insulin resistance, and oxidative stress. Therapeutic strategies may benefit from holistic approaches. Psychotherapy can reduce the maladaptively increased activation of the fear-defense system. Biological interventions can buffer the detrimental effects of oxidative stress in the organism.
Collapse
Affiliation(s)
- Jasmin Ghaemi Kerahrodi
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Matthias Michal
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| |
Collapse
|
40
|
Osborne MT, Shin LM, Mehta NN, Pitman RK, Fayad ZA, Tawakol A. Disentangling the Links Between Psychosocial Stress and Cardiovascular Disease. Circ Cardiovasc Imaging 2020; 13:e010931. [PMID: 32791843 DOI: 10.1161/circimaging.120.010931] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Stress is a pervasive component of the human experience. While often considered an adversity to be ignored, chronic stress has important pathological consequences, including cardiovascular disease (CVD). Stress also increases the prevalence and severity of several CVD risk factors, including hypertension, diabetes mellitus, and obesity. Yet even after adjustment, stress' attributable CVD risk is similar to those risk factors, suggesting it is a particularly potent contributor. Nevertheless, there has been insufficient study of mechanisms linking stress to CVD or of methods to attenuate stress' pathological impact. This review covers the current concepts of how stress impacts CVD and emerging approaches to mitigate stress-attributable CVD risk.
Collapse
Affiliation(s)
- Michael T Osborne
- Cardiology Division (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Cardiovascular Imaging Research Center, Departments of Medicine and Imaging (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Lisa M Shin
- Department of Psychiatry (L.M.S., R.K.P.), Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Psychology, Tufts University, Boston, MA (L.M.S.)
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, MD (N.N.M.)
| | - Roger K Pitman
- Department of Psychiatry (L.M.S., R.K.P.), Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Zahi A Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (Z.A.F.)
| | - Ahmed Tawakol
- Cardiology Division (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Cardiovascular Imaging Research Center, Departments of Medicine and Imaging (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
41
|
Protective Activities of Dendrobium huoshanense C. Z. Tang et S. J. Cheng Polysaccharide against High-Cholesterol Diet-Induced Atherosclerosis in Zebrafish. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8365056. [PMID: 32724495 PMCID: PMC7366212 DOI: 10.1155/2020/8365056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/20/2020] [Indexed: 11/17/2022]
Abstract
Cardiovascular disease is the highest cause of death, and atherosclerosis (AS) is the primary pathogenesis of many cardiovascular diseases. In this study, we aim to investigate the possible pharmaceutical effects of Dendrobium huoshanense C. Z. Tang et S. J. Cheng polysaccharide (DHP) in AS. We fed zebrafish with high-cholesterol diet (HCD) to establish a zebrafish AS model and treated with DHP and observed plaque formation and neutrophil counts under a fluorescence microscope. Next, a parallel flow chamber was utilized to establish low shear stress- (LSS-) induced endothelial cell (EC) dysfunction model. We observed that DHP significantly improved HCD-induced lipid deposition, oxidative stress, and inflammatory response, mainly showing that DHP significantly increased superoxide dismutase (SOD) activity, decreased plaque formation, and decreased neutrophil recruitment and the levels of total cholesterol (TC), triglyceride (TG), malondialdehyde (MDA), and reactive oxygen species (ROS). Furthermore, DHP significantly improved LSS-induced oxidative stress and EC dysfunction. Our results indicated that DHP can exert treatment effects on AS, which may attribute to its hypolipidemic, antioxidant, anti-inflammatory activities and improving LSS-induced EC dysfunction. DHP has promising potential for further development as a functional natural medicine source targeted at AS prevention.
Collapse
|
42
|
Poznyak AV, Zhang D, Orekhova V, Grechko AV, Wetzker R, Orekhov AN. A brief overview of currently used atherosclerosis treatment approaches targeting lipid metabolism alterations. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2020; 10:62-71. [PMID: 32685264 PMCID: PMC7364272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/10/2020] [Indexed: 06/11/2023]
Abstract
Non-contagious diseases such as atherosclerosis, diabetes, cardiovascular disease, cancer, chronic respiratory diseases, and mental disorders hold responsibility for major health losses worldwide. Atherosclerosis was found to be the leading cause of deaths due to the major consequences, such as cardiovascular disease, stroke, ischemic heart disease, myocardial infarction, and others. The number of patients with atherosclerosis increases with every passing year. If treatment is not started on time, every second patient dies within 10 years. Moreover, the disease leads to persistent disability of patients, most of whom are of active working age. Atherosclerosis is a metabolic disorder characterized by hyperlipidemia and chronic inflammation. Although this disease annually kills a huge number of people, patients are now offered various therapeutic techniques, however, with different efficiencies. The scientific community is working to develop more effective means for treatment and precaution of the disease, regardless of the difficulties in understanding the causes of the health problem and the characteristics of its course. There are numerous strategies in the treatment and prevention of atherosclerosis, focusing on different aspects of the disease, such as inflammation, lipid metabolism alterations, or others, but none of them, unfortunately, is absolutely effective. In this review, we focused on the treatment approaches aimed at remedy the disruptions of lipid metabolism that are currently used in clinical practice.
Collapse
Affiliation(s)
- Anastasia V Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative CenterMoscow 121609, Russia
| | - Dongwei Zhang
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese MedicineBeijing 100029, China
| | - Varvara Orekhova
- Laboratory of Angiopathology, Institute of General Pathology and PathophysiologyMoscow 125315, Russia
| | - Andrey V Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology109240 Moscow, Russia
| | - Reinhard Wetzker
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum JenaAmKlinikum 1, D-07747 Jena, Germany
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and PathophysiologyMoscow 125315, Russia
- Institute of Human Morphology3 Tsyurupa Street, Moscow 117418, Russia
| |
Collapse
|
43
|
Perrino C, Ferdinandy P, Bøtker HE, Brundel BJJM, Collins P, Davidson SM, den Ruijter HM, Engel FB, Gerdts E, Girao H, Gyöngyösi M, Hausenloy DJ, Lecour S, Madonna R, Marber M, Murphy E, Pesce M, Regitz-Zagrosek V, Sluijter JPG, Steffens S, Gollmann-Tepeköylü C, Van Laake LW, Van Linthout S, Schulz R, Ytrehus K. Improving translational research in sex-specific effects of comorbidities and risk factors in ischaemic heart disease and cardioprotection: position paper and recommendations of the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2020; 117:367-385. [PMID: 32484892 DOI: 10.1093/cvr/cvaa155] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/29/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Ischaemic heart disease (IHD) is a complex disorder and a leading cause of death and morbidity in both men and women. Sex, however, affects several aspects of IHD, including pathophysiology, incidence, clinical presentation, diagnosis as well as treatment and outcome. Several diseases or risk factors frequently associated with IHD can modify cellular signalling cascades, thus affecting ischaemia/reperfusion injury as well as responses to cardioprotective interventions. Importantly, the prevalence and impact of risk factors and several comorbidities differ between males and females, and their effects on IHD development and prognosis might differ according to sex. The cellular and molecular mechanisms underlying these differences are still poorly understood, and their identification might have important translational implications in the prediction or prevention of risk of IHD in men and women. Despite this, most experimental studies on IHD are still undertaken in animal models in the absence of risk factors and comorbidities, and assessment of potential sex-specific differences are largely missing. This ESC WG Position Paper will discuss: (i) the importance of sex as a biological variable in cardiovascular research, (ii) major biological mechanisms underlying sex-related differences relevant to IHD risk factors and comorbidities, (iii) prospects and pitfalls of preclinical models to investigate these associations, and finally (iv) will provide recommendations to guide future research. Although gender differences also affect IHD risk in the clinical setting, they will not be discussed in detail here.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary.,Pharmahungary Group, Hajnoczy str. 6., H-6722 Szeged, Hungary
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 161, 8200 Aarhus, Denmark
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, Amsterdam, 1108 HV, the Netherlands
| | - Peter Collins
- Imperial College, Faculty of Medicine, National Heart & Lung Institute, South Kensington Campus, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Hester M den Ruijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), Schwabachanlage 12, 91054 Erlangen, Germany
| | - Eva Gerdts
- Department for Clinical Science, University of Bergen, PO Box 7804, 5020 Bergen, Norway
| | - Henrique Girao
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, and Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, 169609, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, 119228, Singapore.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, Chris Barnard Building, University of Cape Town, Private Bag X3 7935 Observatory, Cape Town, South Africa
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy.,Department of Internal Medicine, University of Texas Medical School in Houston, 6410 Fannin St #1014, Houston, TX 77030, USA
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Elizabeth Murphy
- Laboratory of Cardiac Physiology, Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD 20892, USA
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS Via Parea, 4, I-20138 Milan, Italy
| | - Vera Regitz-Zagrosek
- Berlin Institute of Gender in Medicine, Center for Cardiovascular Research, DZHK, partner site Berlin, Geschäftsstelle Potsdamer Str. 58, 10785 Berlin, Germany.,University of Zürich, Rämistrasse 71, 8006 Zürich, Germany
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands.,Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Can Gollmann-Tepeköylü
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstr.35, A - 6020 Innsbruck, Austria
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Ludwigstraße 23, 35390 Giessen, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9037 Tromsø, Norway
| |
Collapse
|
44
|
Carnevale D, Lembo G. Neuroimmune interactions in cardiovascular diseases. Cardiovasc Res 2020; 117:402-410. [PMID: 32462184 DOI: 10.1093/cvr/cvaa151] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/27/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Our body is continuously in contact with external stimuli that need a fine integration with the internal milieu in order to maintain the homoeostasis. Similarly, perturbations of the internal environment are responsible for the alterations of the physiological mechanisms regulating our main functions. The nervous system and the immune system represent the main interfaces between the internal and the external environment. In carrying out these functions, they share many similarities, being able to recognize, integrate, and organize responses to a wide variety of stimuli, with the final aim to re-establish the homoeostasis. The autonomic nervous system, which collectively refers to the ensemble of afferent and efferent neurons that wire the central nervous system with visceral effectors throughout the body, is the prototype system controlling the homoeostasis through reflex arches. On the other hand, immune cells continuously patrol our body against external enemies and internal perturbations, organizing acute responses and forming memory for future encounters. Interesting to notice, the integration of the two systems provides a further unique opportunity for fine tuning of our body's homoeostasis. In fact, the autonomic nervous system guides the development of lymphoid and myeloid organs, as well as the deployment of immune cells towards peripheral tissues where they can affect and control several physiological functions. In turn, every specific immune cell type can contribute to regulate neural circuits involved in cardiovascular function, metabolism, and inflammation. Here, we review current understanding of the cross-regulation between these systems in cardiovascular diseases.
Collapse
Affiliation(s)
- Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, 86077 Pozzilli IS, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, 86077 Pozzilli IS, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
45
|
Li Y, Sun D, Zheng Y, Cheng Y. Swimming exercise activates aortic autophagy and limits atherosclerosis in ApoE -/- mice. Obes Res Clin Pract 2020; 14:264-270. [PMID: 32444302 DOI: 10.1016/j.orcp.2020.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The aim of this study was to investigate the beneficial effect of swimming exercise on autophagy and atherosclerosis in mice aorta, so as to clarify the possible causal relationship between autophagy activation and atherosclerosis. METHODS The body weight was monitored regularly. Hematoxylin-eosin staining and Oil Red O staining was conducted to observe vascular morphology and plaque burden respectively. The levels of serum total cholesterol (TC), triglyceride (TG), soluble intercellular adhesion molecule-1 (sICAM-1), matrix metalloproteinase-9 (MMP-9) and interleukin-6 (IL-6) was examined via Enzyme-linked immu-nosorbent assays (ELISA). The mRNA expression level of autophagy markers, including LC3 and Beclin-1, was examined by real-time quantitative polymerase chain reaction (RT-PCR). The expressions of LC3-II/LC3-I and Beclin-1 are detected by Western blotting and immunohistochemistry. RESULTS Compared with the model group, long-term swimming exercise decreased the weight gain of ApoE-/- mice, improved the structural disorder of artery, reduced the load of atherosclerotic lesion, and attenuated the concentrations of serum TC, TG, sICAM-1, MMP-9, and IL-6. In addition, the expression of autophagy markers LC3 and Beclin-1 increased significantly at the mRNA and protein levels. CONCLUSION Long-term swimming exercise could activate the autophagy and reduce atherosclerotic lesion in the aorta of ApoE-/- mice. Autophagy activation may be one of the mechanisms by which atherosclerosis is improved through exercise.
Collapse
Affiliation(s)
- Yang Li
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, China
| | - Dakang Sun
- Clinical Medical Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Yuanyuan Zheng
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, China
| | - Yanli Cheng
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, China.
| |
Collapse
|
46
|
Abstract
Unhealthy diet, lack of exercise, psychosocial stress, and insufficient sleep are increasingly prevalent modifiable risk factors for cardiovascular disease. Accumulating evidence indicates that these risk factors may fuel chronic inflammatory processes that are active in atherosclerosis and lead to myocardial infarction and stroke. In concert with hyperlipidemia, maladaptive immune system activities can contribute to disease progression and increase the probability of adverse events. In this review, we discuss recent insight into how the above modifiable risk factors influence innate immunity. Specifically, we focus on pathways that raise systemic myeloid cell numbers and modulate immune cell phenotypes, reviewing hematopoiesis, leukocyte trafficking, and innate immune cell accumulation in cardiovascular organs. Often, relevant mechanisms that begin with lifestyle choices and lead to cardiovascular events span multiple organ systems, including the central nervous, endocrine, metabolic, hematopoietic, immune and, finally, the cardiovascular system. We argue that deciphering such pathways provides not only support for preventive interventions but also opportunities to develop biomimetic immunomodulatory therapeutics that mitigate cardiovascular inflammation.
Collapse
Affiliation(s)
- Maximilian J Schloss
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.)
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.).,Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.N.).,Department of Internal Medicine I, University Hospital Wuerzburg, Germany (M.N.)
| |
Collapse
|
47
|
Beneficial Effects of Melatonin on Apolipoprotein-E Knockout Mice by Morphological and 18F-FDG PET/CT Assessments. Int J Mol Sci 2020; 21:ijms21082920. [PMID: 32331251 PMCID: PMC7216051 DOI: 10.3390/ijms21082920] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis represents one of the main risk factors for the development of cardiovascular diseases. Their etiologies have been studied in recent years in order to better define therapeutic targets for intervention and to identify diagnostic methods. Two different subtypes of macrophages, M1 and M2, have been described in physiological conditions. They can also be found in the atherosclerotic process, where they both have opposite roles in disease progression. Perivascular brown adipose tissue is also involved in inflammation and endothelial damage. In this work, we provide insights into the protective role of melatonin in the atherosclerotic process by morphological and 18F-FDG-PET/CT analyses. In particular, we examined the effects of melatonin on pathways that are linked to atherosclerosis development. We showed that melatonin, by suppressing M1 activity, reduced inflammation and directed macrophage polarization toward the M2 macrophage subtype. Moreover, melatonin preserved the activity of perivascular brown adipose tissue. In addition, 18F-FDG uptake is very high in mice treated with melatonin, confirming that other factors may alter 18F-FDG distribution. In conclusion, we showed that melatonin affects inflammatory pathways that have been linked to atherosclerosis, assessed the relationships of the 18F-FDG PET/CT parameters with macrophage markers and the production of their cytokines, which that have been defined by morphological evaluations.
Collapse
|
48
|
Abstract
A central feature of atherosclerosis, the most prevalent chronic vascular disease and root cause of myocardial infarction and stroke, is leukocyte accumulation in the arterial wall. These crucial immune cells are produced in specialized niches in the bone marrow, where a complex cell network orchestrates their production and release. A growing body of clinical studies has documented a correlation between leukocyte numbers and cardiovascular disease risk. Understanding how leukocytes are produced and how they contribute to atherosclerosis and its complications is, therefore, critical to understanding and treating the disease. In this review, we focus on the key cells and products that regulate hematopoiesis under homeostatic conditions, during atherosclerosis and after myocardial infarction.
Collapse
Affiliation(s)
- Wolfram C Poller
- From the Center for Systems Biology (W.C.P., M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Matthias Nahrendorf
- From the Center for Systems Biology (W.C.P., M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston.,Department of Radiology (M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Filip K Swirski
- From the Center for Systems Biology (W.C.P., M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston.,Department of Radiology (M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
49
|
Monslow J, Todd L, Chojnowski JE, Govindaraju PK, Assoian RK, Puré E. Fibroblast Activation Protein Regulates Lesion Burden and the Fibroinflammatory Response in Apoe-Deficient Mice in a Sexually Dimorphic Manner. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1118-1136. [PMID: 32084369 DOI: 10.1016/j.ajpath.2020.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/19/2019] [Accepted: 01/02/2020] [Indexed: 01/10/2023]
Abstract
Fibroblast activation protein (FAP) has been established as an inducible and mesenchymal cell-specific mediator of disease progression in cancer and fibrosis. Atherosclerosis is a fibroinflammatory disease, and FAP was previously reported to be up-regulated in human atherosclerotic plaques compared with normal vessel. We investigated the spatial and temporal distribution of Fap-expressing cells in a murine model of atherosclerosis and used a genetic approach to determine if and how Fap affected disease progression. Fap was found to be expressed predominantly on vascular smooth muscle cells in lesions of athero-prone Apoe-/- mice. Global deletion of Fap (Fap-/-) in Apoe-/- mice accelerated atherosclerotic disease progression in both males and females, with the effect observed earlier in males. Sex-specific effects on lesion morphology were observed. Relative levels of extracellular matrix, fibrotic, and inflammatory cell content were comparable in lesions in male mice regardless of Fap status. In contrast, lesions in Fap-/- female mice were characterized by a more fibrotic composition due to a reduction in inflammation, specifically a reduction in Mox macrophages. Combined, these data suggest that Fap restrains the progression of atherosclerosis and may contribute to the sexually dimorphic susceptibility to atherosclerosis by regulating the balance between inflammation (an indicator of vulnerability to plaque rupture) and fibrosis (an indicator of plaque stability).
Collapse
Affiliation(s)
- James Monslow
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia Pennsylvania.
| | - Leslie Todd
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia Pennsylvania
| | - John E Chojnowski
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia Pennsylvania
| | - Priya K Govindaraju
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia Pennsylvania
| | - Richard K Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia Pennsylvania
| | - Ellen Puré
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia Pennsylvania.
| |
Collapse
|
50
|
Navarro E, Mallén A, Cruzado JM, Torras J, Hueso M. Unveiling ncRNA regulatory axes in atherosclerosis progression. Clin Transl Med 2020; 9:5. [PMID: 32009226 PMCID: PMC6995802 DOI: 10.1186/s40169-020-0256-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023] Open
Abstract
Completion of the human genome sequencing project highlighted the richness of the cellular RNA world, and opened the door to the discovery of a plethora of short and long non-coding RNAs (the dark transcriptome) with regulatory or structural potential, which shifted the balance of pathological gene alterations from coding to non-coding RNAs. Thus, disease risk assessment currently has to also evaluate the expression of new RNAs such as small micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), competing endogenous RNAs (ceRNAs), retrogressed elements, 3'UTRs of mRNAs, etc. We are interested in the pathogenic mechanisms of atherosclerosis (ATH) progression in patients suffering Chronic Kidney Disease, and in this review, we will focus in the role of the dark transcriptome (non-coding RNAs) in ATH progression. We will focus in miRNAs and in the formation of regulatory axes or networks with their mRNA targets and with the lncRNAs that function as miRNA sponges or competitive inhibitors of miRNA activity. In this sense, we will pay special attention to retrogressed genomic elements, such as processed pseudogenes and Alu repeated elements, that have been recently seen to also function as miRNA sponges, as well as to the use or miRNA derivatives in gene silencing, anti-ATH therapies. Along the review, we will discuss technical developments associated to research in lncRNAs, from sequencing technologies to databases, repositories and algorithms to predict miRNA targets, as well as new approaches to miRNA function, such as integrative or enrichment analysis and their potential to unveil RNA regulatory networks.
Collapse
Affiliation(s)
- Estanislao Navarro
- Independent Researcher, Barcelona, Spain. .,Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| | - Adrian Mallén
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Josep M Cruzado
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Joan Torras
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Miguel Hueso
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| |
Collapse
|