1
|
Cai S, Fu Y, Chen J, Tian M, Li X. Causal Relationship Between Branched-Chain Amino Acids and Hypertension: A Mendelian Randomization Study. J Am Heart Assoc 2024; 13:e032084. [PMID: 38420789 PMCID: PMC10944042 DOI: 10.1161/jaha.123.032084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/12/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND This study aimed to investigate the causal relationships between branched-chain amino acids (BCAAs) and the risks of hypertension via meta-analysis and Mendelian randomization analysis. METHODS AND RESULTS A meta-analysis of 32 845 subjects was conducted to evaluate the relationships between BCAAs and hypertension. In Mendelian randomization analysis, independent single-nucleotide polymorphisms associated with BCAAs at the genome-wide significance level were selected as the instrumental variables. Meanwhile, the summary-level data for essential hypertension and secondary hypertension end points were obtained from the FinnGen study. As suggested by the meta-analysis results, elevated BCAA levels were associated with a higher risk of hypertension (isoleucine: summary odds ratio, 1.26 [95% CI, 1.08-1.47]; leucine: summary odds ratio, 1.28 [95% CI, 1.07-1.52]; valine: summary odds ratio, 1.32 [95% CI, 1.12-1.57]). Moreover, the inverse variance-weighted method demonstrated that an elevated circulating isoleucine level might be the causal risk factor for essential hypertension but not secondary hypertension (essential hypertension: odds ratio, 1.22 [95% CI, 1.12-1.34]; secondary hypertension: odds ratio, 0.96 [95% CI, 0.54-1.68]). CONCLUSIONS The increased levels of 3 BCAAs positively correlated with an increased risk of hypertension. Particularly, elevated isoleucine level is a causal risk factor for essential hypertension. Increased levels of leucine and valine also tend to increase the risk of essential hypertension, but further verification is still warranted.
Collapse
Affiliation(s)
- Shiyuan Cai
- School of Public Health, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuanyuan Fu
- School of Public Health, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Chen
- School of Public Health, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mingjie Tian
- Department of Clinical Nutrition, Shanghai Deji HospitalAffiliated to Qingdao UniversityShanghaiChina
| | - Xue Li
- School of Public Health, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
2
|
Fraile-Martinez O, De Leon-Oliva D, Boaru DL, De Castro-Martinez P, Garcia-Montero C, Barrena-Blázquez S, García-García J, García-Honduvilla N, Alvarez-Mon M, Lopez-Gonzalez L, Diaz-Pedrero R, Guijarro LG, Ortega MA. Connecting epigenetics and inflammation in vascular senescence: state of the art, biomarkers and senotherapeutics. Front Genet 2024; 15:1345459. [PMID: 38469117 PMCID: PMC10925776 DOI: 10.3389/fgene.2024.1345459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Abstract
Vascular diseases pose major health challenges, and understanding their underlying molecular mechanisms is essential to advance therapeutic interventions. Cellular senescence, a hallmark of aging, is a cellular state characterized by cell-cycle arrest, a senescence-associated secretory phenotype macromolecular damage, and metabolic dysregulation. Vascular senescence has been demonstrated to play a key role in different vascular diseases, such as atherosclerosis, peripheral arterial disease, hypertension, stroke, diabetes, chronic venous disease, and venous ulcers. Even though cellular senescence was first described in 1961, significant gaps persist in comprehending the epigenetic mechanisms driving vascular senescence and its subsequent inflammatory response. Through a comprehensive analysis, we aim to elucidate these knowledge gaps by exploring the network of epigenetic alterations that contribute to vascular senescence. In addition, we describe the consequent inflammatory cascades triggered by these epigenetic modifications. Finally, we explore translational applications involving biomarkers of vascular senescence and the emerging field of senotherapy targeting this biological process.
Collapse
Affiliation(s)
- Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Joaquin García-García
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, Alcala deHenares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, Alcala deHenares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala deHenares, Spain
| |
Collapse
|
3
|
Reijnders E, van der Laarse A, Jukema JW, Cobbaert CM. High residual cardiovascular risk after lipid-lowering: prime time for Predictive, Preventive, Personalized, Participatory, and Psycho-cognitive medicine. Front Cardiovasc Med 2023; 10:1264319. [PMID: 37908502 PMCID: PMC10613690 DOI: 10.3389/fcvm.2023.1264319] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023] Open
Abstract
As time has come to translate trial results into individualized medical diagnosis and therapy, we analyzed how to minimize residual risk of cardiovascular disease (CVD) by reviewing papers on "residual cardiovascular disease risk". During this review process we found 989 papers that started off with residual CVD risk after initiating statin therapy, continued with papers on residual CVD risk after initiating therapy to increase high-density lipoprotein-cholesterol (HDL-C), followed by papers on residual CVD risk after initiating therapy to decrease triglyceride (TG) levels. Later on, papers dealing with elevated levels of lipoprotein remnants and lipoprotein(a) [Lp(a)] reported new risk factors of residual CVD risk. And as new risk factors are being discovered and new therapies are being tested, residual CVD risk will be reduced further. As we move from CVD risk reduction to improvement of patient management, a paradigm shift from a reductionistic approach towards a holistic approach is required. To that purpose, a personalized treatment dependent on the individual's CVD risk factors including lipid profile abnormalities should be configured, along the line of P5 medicine for each individual patient, i.e., with Predictive, Preventive, Personalized, Participatory, and Psycho-cognitive approaches.
Collapse
Affiliation(s)
- E. Reijnders
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - A. van der Laarse
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - J. W. Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
| | - C. M. Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
4
|
Gao S, Zhao J, Liu X, Liu L, Chen R. Metabolomics reveals serum metabolic signatures in H-type hypertension based on mass spectrometry multi-platform. Eur J Clin Invest 2023; 53:e14063. [PMID: 37458276 DOI: 10.1111/eci.14063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/14/2023] [Accepted: 06/03/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND H-type hypertension (HHT) is a disease combined with hyperhomocysteinaemia and hypertension (HT). This study aims to find specific metabolic changes and reveal the pathophysiological mechanism of HHT, which provide the theoretical basis for the early prevention and treatment of HHT. METHODS Serum samples from three groups including 53 HHT patients, 36 HT patients and 46 healthy controls (HC) were collected. The targeted and untargeted metabolomics analyses were performed to determine the metabolic changes. Based on multivariate statistical analysis, the serum potential metabolites were screened and different metabolic pathways were explored. RESULTS Our results demonstrated that there were 28 important potential metabolites for distinguishing HT from HHT patients. Metabolic pathway analysis showed that the different metabolic pathways between HHT and HC group were arginine biosynthesis, arginine and proline metabolism, and tyrosine metabolism. The changed metabolic pathway of HT and HC group included linoleic acid metabolism. The specific metabolic pathways of HT-HHT comparison group had phenylalanine metabolism; phenylalanine, tyrosine and tryptophan biosynthesis; glycine, serine and threonine metabolism. CONCLUSIONS Metabolomics analysis by mass spectrometry multi-platform revealed the differences of metabolic profiles between HHT and HT subjects. This work laid the groundwork for understanding the aetiology of HHT, and these findings may provide the useful information for explaining the HHT metabolic alterations and try to prevent HHT.
Collapse
Affiliation(s)
- Siqi Gao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Jinhui Zhao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Xiaowei Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Liyan Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Rui Chen
- Department of Orthopedics, Jiangnan University Medical Center, Wuxi, China
| |
Collapse
|
5
|
Sinclair SH, Miller E, Talekar KS, Schwartz SS. Diabetes mellitus associated neurovascular lesions in the retina and brain: A review. FRONTIERS IN OPHTHALMOLOGY 2022; 2:1012804. [PMID: 38983558 PMCID: PMC11182219 DOI: 10.3389/fopht.2022.1012804] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/14/2022] [Indexed: 07/11/2024]
Abstract
Diabetes mellitus (DM) is now recognized as a system-wide, autoimmune, inflammatory, microvascular disorder, which, in the retina and brain results in severe multifocal injury now recognized as a leading cause, world-wide, of progressive vision loss and dementia. To address this problem, resulting primarily from variations in glycemia in the prediabetic and overt diabetic states, it must be realized that, although some of the injury processes associated with diabetes may be system wide, there are varying responses, effector, and repair mechanisms that differ from organ to organ or within varying cell structures. Specifically, within the retina, and similarly within the brain cortex, lesions occur of the "neurovascular unit", comprised of focal microvascular occlusions, inflammatory endothelial and pericyte injury, with small vessel leakage resulting in injury to astrocytes, Müller cells, and microglia, all of which occur with progressive neuronal apoptosis. Such lesions are now recognized to occur before the first microaneurysms are visible to imaging by fundus cameras or before they result in detectable symptoms or signs recognizable to the patient or clinician. Treatments, therefore, which currently are not initiated within the retina until edema develops or there is progression of vascular lesions that define the current staging of retinopathy, and in the brain only after severe signs of cognitive failure. Treatments, therefore are applied relatively late with some reduction in progressive cellular injury but with resultant minimal vision or cognitive improvement. This review article will summarize the multiple inflammatory and remediation processes currently understood to occur in patients with diabetes as well as pre-diabetes and summarize as well the current limitations of methods for assessing the structural and functional alterations within the retina and brain. The goal is to attempt to define future screening, monitoring, and treatment directions that hopefully will prevent progressive injury as well as enable improved repair and attendant function.
Collapse
Affiliation(s)
- Stephen H Sinclair
- Pennsylvania College of Optometry, Salus University, Philadelphia, PA, United States
| | - Elan Miller
- Division of Vascular Neurology, Vickie & Jack Farber Institute for Institute for Neuroscience, Sidney Kimmel Medical College (SKMC) Thomas Jefferson University, Philadelphia, PA, United States
| | - Kiran S Talekar
- Department of Radiology, Section of Neuroradiology and ENT Radiology, Clinical Functional Magnetic Resonance Imaging and Diffusion Tensor Imaging at Thomas Jefferson University Hospital and The Jefferson Integrated Magnetic Resonance Imaging Center (JIMRIC) Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| | - Stanley S Schwartz
- Department of Endocrinology and Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Main Line Health System, Philadelphia, PA, United States
| |
Collapse
|
6
|
Shi H, Wu H, Winkler MA, Belin de Chantemèle EJ, Lee R, Kim HW, Weintraub NL. Perivascular adipose tissue in autoimmune rheumatic diseases. Pharmacol Res 2022; 182:106354. [PMID: 35842184 PMCID: PMC10184774 DOI: 10.1016/j.phrs.2022.106354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/27/2022] [Accepted: 07/11/2022] [Indexed: 01/14/2023]
Abstract
Perivascular adipose tissue (PVAT) resides at the outermost boundary of the vascular wall, surrounding most conduit blood vessels, except for the cerebral vessels, in humans. A growing body of evidence suggests that inflammation localized within PVAT may contribute to the pathogenesis of cardiovascular disease (CVD). Patients with autoimmune rheumatic diseases (ARDs), e.g., systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), psoriasis, etc., exhibit heightened systemic inflammation and are at increased risk for CVD. Data from clinical studies in patients with ARDs support a linkage between dysfunctional adipose tissue, and PVAT in particular, in disease pathogenesis. Here, we review the data linking PVAT to the pathogenesis of CVD in patients with ARDs, focusing on the role of novel PVAT imaging techniques in defining disease risk and responses to biological therapies.
Collapse
Affiliation(s)
- Hong Shi
- Division of Rheumatology, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Hanping Wu
- Department of Radiology and Imaging, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Michael A Winkler
- Department of Radiology and Imaging, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric J Belin de Chantemèle
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Richard Lee
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ha Won Kim
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Neal L Weintraub
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
7
|
Antonopoulos AS, Angelopoulos A, Tsioufis K, Antoniades C, Tousoulis D. Cardiovascular risk stratification by coronary computed tomography angiography imaging: current state-of-the-art. Eur J Prev Cardiol 2022; 29:608-624. [PMID: 33930129 DOI: 10.1093/eurjpc/zwab067] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022]
Abstract
Current cardiovascular risk stratification by use of clinical risk score systems or plasma biomarkers is good but less than satisfactory in identifying patients at residual risk for coronary events. Recent clinical evidence puts now further emphasis on the role of coronary anatomy assessment by coronary computed tomography angiography (CCTA) for the management of patients with stable ischaemic heart disease. Available computed tomography (CT) technology allows the quantification of plaque burden, identification of high-risk plaques, or the functional assessment of coronary lesions for ischaemia detection and revascularization for refractory angina symptoms. The current CT armamentum is also further enhanced by perivascular Fat Attenuation Index (FAI), a non-invasive metric of coronary inflammation, which allows for the first time the direct quantification of the residual vascular inflammatory burden. Machine learning and radiomic features' extraction and spectral CT for tissue characterization are also expected to maximize the diagnostic and prognostic yield of CCTA. The combination of anatomical, functional, and biological information on coronary circulation by CCTA offers a unique toolkit for the risk stratification of patients, and patient selection for targeted aggressive prevention strategies. We hereby provide a review of the current state-of-the art in the field and discuss how integrating the full capacities of CCTA into clinical care pathways opens new opportunities for the tailored management of coronary artery disease.
Collapse
Affiliation(s)
- Alexios S Antonopoulos
- 1st Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens, 114 Vas. Sofias Avenue, 11527, Athens, Greece
- RDM Division of Cardiovascular Medicine, Oxford Academic CT Programme, University of Oxford, John Radcliffe Hospital, Headley Way, OX3 9DU Oxford, UK
| | - Andreas Angelopoulos
- 1st Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens, 114 Vas. Sofias Avenue, 11527, Athens, Greece
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens, 114 Vas. Sofias Avenue, 11527, Athens, Greece
| | - Charalambos Antoniades
- RDM Division of Cardiovascular Medicine, Oxford Academic CT Programme, University of Oxford, John Radcliffe Hospital, Headley Way, OX3 9DU Oxford, UK
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens, 114 Vas. Sofias Avenue, 11527, Athens, Greece
| |
Collapse
|
8
|
Gopcevic KR, Gkaliagkousi E, Nemcsik J, Acet Ö, Bernal-Lopez MR, Bruno RM, Climie RE, Fountoulakis N, Fraenkel E, Lazaridis A, Navickas P, Rochfort KD, Šatrauskienė A, Zupkauskienė J, Terentes-Printzios D. Pathophysiology of Circulating Biomarkers and Relationship With Vascular Aging: A Review of the Literature From VascAgeNet Group on Circulating Biomarkers, European Cooperation in Science and Technology Action 18216. Front Physiol 2021; 12:789690. [PMID: 34970157 PMCID: PMC8712891 DOI: 10.3389/fphys.2021.789690] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Impairment of the arteries is a product of sustained exposure to various deleterious factors and progresses with time; a phenomenon inherent to vascular aging. Oxidative stress, inflammation, the accumulation of harmful agents in high cardiovascular risk conditions, changes to the extracellular matrix, and/or alterations of the epigenetic modification of molecules, are all vital pathophysiological processes proven to contribute to vascular aging, and also lead to changes in levels of associated circulating molecules. Many of these molecules are consequently recognized as markers of vascular impairment and accelerated vascular aging in clinical and research settings, however, for these molecules to be classified as biomarkers of vascular aging, further criteria must be met. In this paper, we conducted a scoping literature review identifying thirty of the most important, and eight less important, biomarkers of vascular aging. Herein, we overview a selection of the most important molecules connected with the above-mentioned pathological conditions and study their usefulness as circulating biomarkers of vascular aging.
Collapse
Affiliation(s)
- Kristina R. Gopcevic
- Laboratory for Analytics of Biomolecules, Department of Chemistry in Medicine, Faculty of Medicine, Belgrade, Serbia
| | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - János Nemcsik
- Department of Family Medicine, Semmelweis University, Budapest, Hungary
- Health Service of ZUGLO, Department of Family Medicine, Budapest, Hungary
| | - Ömür Acet
- Vocational School of Health Science, Pharmacy Services Program, Tarsus University, Tarsus, Turkey
| | - M. Rosa Bernal-Lopez
- Internal Medicine Department, Regional University Hospital of Malaga, Instituto de Investigacion Biomedica de Malaga, University of Malaga, CIBER Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Málaga, Spain
| | - Rosa M. Bruno
- Unversite de Paris, INSERM, U970, Paris Cardiovascular Research Center, Paris, France
| | - Rachel E. Climie
- Unversite de Paris, INSERM, U970, Paris Cardiovascular Research Center, Paris, France
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Sports Cardiology Lab, Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Nikolaos Fountoulakis
- Faculty of Life Sciences and Medicine, King’s College London - Waterloo Campus, London, United Kingdom
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital and Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Petras Navickas
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Keith D. Rochfort
- School of Nursing, Psychotherapy and Community Health, Dublin City University, Dublin, Ireland
| | - Agnė Šatrauskienė
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Centre of Cardiology and Angiology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Jūratė Zupkauskienė
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
9
|
Deng Y, Huang C, Su J, Pan CW, Ke C. Identification of biomarkers for essential hypertension based on metabolomics. Nutr Metab Cardiovasc Dis 2021; 31:382-395. [PMID: 33495028 DOI: 10.1016/j.numecd.2020.11.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/10/2020] [Accepted: 11/20/2020] [Indexed: 01/09/2023]
Abstract
AIM Essential hypertension (EH) is one of the most important public health problems worldwide. However, the pathogenesis of EH is unclear and early diagnostic methods are lacking. Metabolomics demonstrates great potential for biomarker discovery and the mechanistic exploration of metabolic diseases. DATA SYNTHESIS This review included human and animal metabolomics studies related to EH in the PubMed and Web of Science databases between February 1996 and May 2020. The study designs, EH standards, and reported metabolic biomarkers were systematically examined and compared. The pathway analysis was conducted through the online software MetaboAnalyst 4.0. Twenty-two human studies and fifteen animal studies were included in this systematic review. There were many frequently reported biomarkers with consistent trends (e.g., pyruvate, lactic acid, valine, and tryptophan) in human and animal studies, and thus had potential as biomarkers of EH. In addition, several shared metabolic pathways, including alanine, aspartate, and glutamate metabolism, aminoacyl-tRNA biosynthesis, and arginine biosynthesis, were identified in human and animal metabolomics studies. These biomarkers and pathways, closely related to insulin resistance, the inflammatory state, and impaired nitric oxide production, were demonstrated to contribute to EH development. CONCLUSIONS This study summarized valuable metabolic biomarkers and pathways that could offer opportunities for the early diagnosis or prediction of EH and the discovery of the metabolic mechanisms of EH.
Collapse
Affiliation(s)
- Yueting Deng
- Medical College of Soochow University, Suzhou, 215123, PR China
| | - Chen Huang
- Medical College of Soochow University, Suzhou, 215123, PR China
| | - Jingyue Su
- Medical College of Soochow University, Suzhou, 215123, PR China
| | - Chen-Wei Pan
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China.
| | - Chaofu Ke
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China.
| |
Collapse
|
10
|
Association of Major Histocompatibility Complex Class I Related Chain A/B Positive Microparticles with Acute Myocardial Infarction and Disease Severity. Diagnostics (Basel) 2020; 10:diagnostics10100766. [PMID: 33003303 PMCID: PMC7656305 DOI: 10.3390/diagnostics10100766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/23/2020] [Accepted: 09/26/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Various cell types undergo activation and stress during atherosclerosis resulting in the development of acute myocardial infarction (AMI) in coronary artery disease (CAD). Major histocompatibility complex class I related chain A and B (MICA/B) can be expressed on the surface of activated and stressed cells and released into blood circulation in several forms including microparticles (MICA/B+ MPs) from various cell types. We aimed to investigate the association of these MICA/B+ MPs with the presence of AMI. Fifty-one AMI and 46 age-matched control subjects were recruited. Methods: Levels of MICA/B+ MPs derived from various parent cells including endothelial cells, platelets, monocytes, neutrophils, and T lymphocytes were determined by flow cytometry. Results: The levels and proportion of MICA/B+ MPs from all types of cell origin were significantly increased in AMI patients compared to those of the controls. A multivariate regression model showed an independent association between MICA/B+ MPs and AMI (OR = 11.6; 95% CI = 2.8, 47.3). Interestingly, based on the disease severity, we found that the levels of MICA/B+ MPs were significantly elevated in the ST-segment elevation myocardial infarction (STEMI) compared to the non-STEMI (NSTEMI) patients. Moreover, an independent association of MICA/B+ MPs with the occurrence of STEMI was also demonstrated (OR = 4.1; 95% CI = 1.5, 16.7). Conclusions: These results suggest that MICA/B+ MPs are associated with AMI and disease severity. They may act as mediators contributing to the pathological process of AMI. Alternatively, they are the results of various cell activations contributing to AMI.
Collapse
|
11
|
Slíva J, Charalambous C, Bultas J, Karetová D. A new strategy for the treatment of atherothrombosis - inhibition of inflammation. Physiol Res 2020; 68:S17-S30. [PMID: 31755287 DOI: 10.33549/physiolres.934327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Improvement in the prognosis of patients at risk of atherothrombotic events is based on three pillars - slowing down the process of atherogenesis (i.e. the development of atherosclerotic plaque), stabilizing the current atherosclerotic plaque, and reducing the risk of thrombotic occlusion in cases with unstable atherosclerotic plaque. The current prophylaxis has so far taken into consideration the adjustment of several risk factors, including dyslipidemia, arterial hypertension, smoking, and diabetes through lifestyle changes or pharmacological therapies. An essential part of prophylaxis is the anti-thrombotic strategy, especially anti-platelet therapy. Recently, a new pathway has been developed, based on reducing the activity of the inflammatory process with NLRP3 inflammasome, specifically a monoclonal antibody against interleukin 1beta (canakinumab). The efficacy and safety of this treatment, in secondary prevention, were documented in the CANTOS study. Other therapeutic procedures, including suppression of the inflammatory component of atherogenesis, are at the stage of clinical assessment.
Collapse
Affiliation(s)
- J Slíva
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | |
Collapse
|
12
|
Analysis of monocitary subpopulations in relation to cardiovascular risk factors. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2019; 31:152-159. [PMID: 30967277 DOI: 10.1016/j.arteri.2019.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/17/2019] [Accepted: 02/06/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Monocytes play an important role in atherosclerotic progression having both pro and anti-inflammatory effects depending on different circulating monocyte subpopulations. The objective of this study is to characterize these subpopulations and their association with cardiovascular risk factors. METHODS Transversal study including 102 selected patients, mean age: 65 years-old (range 41-86), 69% males. A set of specific antibodies against classical monocytes (Mon1, CD14+CD16- CD300e+HLADR+), intermediate (Mon2, CD14+CD16+CD300e+HLADR+) and non-classical (Mon3, CD14-CD16+CD300e+HLADR+) was assayed. Three groups of patients were included: 17 asymptomatic with more than one cardiovascular risk factor (group 1), 56 subjects asymptomatic but with vascular pathology assessed by ultrasound or microalbuminuria (group 2) and 19 patients with a previous atherothrombotic event (group 3). The cardiovascular risk was determined by Framingham and REGICOR scores. RESULTS An association between study groups and the percentage of Mon1 and Mon2 was observed (ANOVA, p<.05), being independent of age and sex for Mon2. Likewise Mon1 and Mon2 subpopulations were associated with cardiovascular adverse events (β=0.86, p=.02 y β=0.1 p=.002, respectively), independently of age and sex in the case of Mon2. Moreover the percentage of Mon3 was associated with the presence of several cardiovascular risk factors (β=0.21, p=.04) in the univariate analysis. In addition, there was a correlation between the levels of Mon1 and Mon2 and leukocytes (r=0.7, p<.001 and r=0.26, p=.01, respectively). CONCLUSIONS The analysis of monocyte subpopulations may be clinically useful to stratify the inflammatory profile related to the different cardiovascular risk groups.
Collapse
|
13
|
Antoniades C, Kotanidis CP, Berman DS. State-of-the-art review article. Atherosclerosis affecting fat: What can we learn by imaging perivascular adipose tissue? J Cardiovasc Comput Tomogr 2019; 13:288-296. [PMID: 30952610 PMCID: PMC6928589 DOI: 10.1016/j.jcct.2019.03.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/04/2019] [Accepted: 03/25/2019] [Indexed: 01/05/2023]
Abstract
Perivascular adipose tissue (PVAT) surrounding the human coronary arteries, secretes a wide range of adipocytokines affecting the biology of the adjacent vascular wall in a paracrine way. However, we have recently found that PVAT also behaves as a sensor of signals coming from the vascular wall, to which it reacts by changing its morphology and secretory profile. Indeed, vascular inflammation, a key feature of vascular disease pathogenesis, leads to the release of inflammatory signals that disseminate into local fat, inducing local lipolysis and inhibiting adipogenesis. This ability of PVAT to sense inflammatory signals from the vascular wall, can be used as a "thermometer" of the vascular wall, allowing for non-invasive detection of coronary inflammation. Vascular inflammation induces a shift of PVAT's composition from lipid to aqueous phase, resulting into increased computed tomography (CT) attenuation around the inflamed artery, forming a gradient with increasing attenuation closer to the inflamed coronary artery wall. These spatial changes in PVAT's attenuation are easily detected around culprit lesions during acute coronary syndromes. A new biomarker designed to captured these spatial changes in PVAT's attenuation around the human coronary arteries, the Fat Attenuation Index (FAI), has additional predictive value in stable patients for cardiac mortality and non-fatal heart attacks, above the prediction provided by the current state of the art that includes risk factors, calcium score and presence of high risk plaque features. The use of perivascular FAI in clinical practice may change the way we interpret cardiovascular CT angiography, as it is applicable to any coronary CT angiogram, and it offers dynamic information about the inflammatory burden of the coronary arteries, providing potential guidance for preventive measures and invasive treatments.
Collapse
Affiliation(s)
- Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Christos P Kotanidis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel S Berman
- Department of Imaging and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
14
|
Affiliation(s)
- Heather Y Small
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, UK
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, UK.,Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, 31-008 Anny 12, Krakow, Poland
| |
Collapse
|
15
|
Batko B, Maga P, Urbanski K, Ryszawa-Mrozek N, Schramm-Luc A, Koziej M, Mikolajczyk T, McGinnigle E, Czesnikiewicz-Guzik M, Ceranowicz P, Guzik TJ. Microvascular dysfunction in ankylosing spondylitis is associated with disease activity and is improved by anti-TNF treatment. Sci Rep 2018; 8:13205. [PMID: 30181568 PMCID: PMC6123474 DOI: 10.1038/s41598-018-31550-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/20/2018] [Indexed: 12/26/2022] Open
Abstract
Ankylosing spondylitis (AS) is associated with high cardiovascular morbidity and mortality. Recent studies indicate that microvascular dysfunction may underlie cardiovascular risk in AS. We hypothesized, that microvascular morphology and dysfunction is linked to AS activity and is modifiable by TNF-α inhibitor (TNFi) treatment. Functional Laser Doppler Flowmetry with post-occlusive reactive hyperemia, and structural nailfold capillaroscopy were performed in 54 patients with AS and 28 matched controls. Active AS was diagnosed based on BASDAI ≥ 4 (n = 37). Effects of 3-month TNFi on microcirculation in active AS were studied. AS was associated with prolonged time to peak hyperemia compared to healthy controls. High disease activity was associated with increased time to peak hyperemia and decreased peak hyperemia when compared to patients with inactive AS. In capillaroscopy, AS was associated with morphological abnormalities indicating increased neoangiogenesis and pericapillary edema compared to controls. Microvascular function improved following 3 months of TNFi in reference to basal flow as well as post-occlusive parameters. TNFi reduced pericapillary edema, while other parameters of capillary morphology remained unchanged. Microvascular dysfunction and capillary neovascular formation are associated with disease activity of AS. Anti-TNF-α treatment may restore microcirculation function and capillary edema but does not modify microvascular structural parameters.
Collapse
Affiliation(s)
- Bogdan Batko
- Department of Rheumatology, J Dietl Hospital, Krakow, Poland
| | - Pawel Maga
- 0000 0001 2162 9631grid.5522.0Department of Angiology, II Chair of Internal Medicine, Jagiellonian University School of Medicine, Krakow, Poland
| | - Karol Urbanski
- 0000 0001 2162 9631grid.5522.0Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland
| | - Natalia Ryszawa-Mrozek
- 0000 0001 2162 9631grid.5522.0Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland
| | - Agata Schramm-Luc
- 0000 0001 2162 9631grid.5522.0Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland
| | - Mateusz Koziej
- 0000 0001 2162 9631grid.5522.0Department of Anatomy, Jagiellonian University School of Medicine, Krakow, Poland
| | - Tomasz Mikolajczyk
- 0000 0001 2193 314Xgrid.8756.cBHF Centre of Research Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Eilidh McGinnigle
- 0000 0001 2193 314Xgrid.8756.cBHF Centre of Research Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Marta Czesnikiewicz-Guzik
- 0000 0001 2162 9631grid.5522.0Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland ,0000 0001 2193 314Xgrid.8756.cInstitute of Infection Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Piotr Ceranowicz
- 0000 0001 2162 9631grid.5522.0Department of Physiology, Jagiellonian University School of Medicine, Krakow, Poland
| | - Tomasz J. Guzik
- 0000 0001 2162 9631grid.5522.0Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland ,0000 0001 2193 314Xgrid.8756.cBHF Centre of Research Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
16
|
Small HY, Migliarino S, Czesnikiewicz-Guzik M, Guzik TJ. Hypertension: Focus on autoimmunity and oxidative stress. Free Radic Biol Med 2018; 125:104-115. [PMID: 29857140 DOI: 10.1016/j.freeradbiomed.2018.05.085] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022]
Abstract
Understanding the causal role of the immune and inflammatory responses in hypertension has led to questions regarding the links between hypertension and autoimmunity. Immune pathology in primary hypertension mimics several autoimmune mechanisms observed in the pathogenesis of systemic lupus erythematosus, psoriasis, systemic sclerosis, rheumatoid arthritis and periodontitis. More importantly, the prevalence of hypertension in patients with these autoimmune diseases is significantly increased, when compared to control populations. Clinical and epidemiological evidence is reviewed along with possible mechanisms linking hypertension and autoimmunity. Inflammation and oxidative stress are linked in a self-perpetuating cycle that significantly contributes to the vascular dysfunction and renal damage associated with hypertension. T cell, B cell, macrophage and NK cell infiltration into these organs is essential for this pathology. Effector cytokines such as IFN-γ, TNF-α and IL-17 affect Na+/H+ exchangers in the kidney. In blood vessels, they lead to endothelial dysfunction and loss of nitric oxide bioavailability and cause vasoconstriction. Both renal and vascular effects are, in part, mediated through induction of reactive oxygen species-producing enzymes such as superoxide anion generating NADPH oxidases and dysfunction of anti-oxidant systems. These mechanisms have recently become important therapeutic targets of novel therapies focused on scavenging oxidative (isolevuglandin) modification of neo-antigenic peptides. Effects of classical immune targeted therapies focused on immunosuppression and anti-cytokine treatments are also reviewed.
Collapse
Affiliation(s)
- Heather Y Small
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Serena Migliarino
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Marta Czesnikiewicz-Guzik
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Department of Dental Prophylaxis and Experimental Dentistry, Dental School of Jagiellonian University, Krakow, Poland
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland.
| |
Collapse
|
17
|
Ma F, Li X, Wang Y, Liang N, Pan S, Yang G, Liao Y, Zhang C, Zhang Q, Lin Y. Effectiveness of traditional Chinese exercises on stroke risk factors in individuals with pre-hypertension or mild-to-moderate essential hypertension: A systematic review and meta-analysis. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2018. [DOI: 10.1016/j.jtcms.2018.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
18
|
Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role of infiltrating immune cells in dysfunctional adipose tissue. Cardiovasc Res 2018; 113:1009-1023. [PMID: 28838042 PMCID: PMC5852626 DOI: 10.1093/cvr/cvx108] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue (AT) dysfunction, characterized by loss of its homeostatic functions, is a hallmark of non-communicable diseases. It is characterized by chronic low-grade inflammation and is observed in obesity, metabolic disorders such as insulin resistance and diabetes. While classically it has been identified by increased cytokine or chemokine expression, such as increased MCP-1, RANTES, IL-6, interferon (IFN) gamma or TNFα, mechanistically, immune cell infiltration is a prominent feature of the dysfunctional AT. These immune cells include M1 and M2 macrophages, effector and memory T cells, IL-10 producing FoxP3+ T regulatory cells, natural killer and NKT cells and granulocytes. Immune composition varies, depending on the stage and the type of pathology. Infiltrating immune cells not only produce cytokines but also metalloproteinases, reactive oxygen species, and chemokines that participate in tissue remodelling, cell signalling, and regulation of immunity. The presence of inflammatory cells in AT affects adjacent tissues and organs. In blood vessels, perivascular AT inflammation leads to vascular remodelling, superoxide production, endothelial dysfunction with loss of nitric oxide (NO) bioavailability, contributing to vascular disease, atherosclerosis, and plaque instability. Dysfunctional AT also releases adipokines such as leptin, resistin, and visfatin that promote metabolic dysfunction, alter systemic homeostasis, sympathetic outflow, glucose handling, and insulin sensitivity. Anti-inflammatory and protective adiponectin is reduced. AT may also serve as an important reservoir and possible site of activation in autoimmune-mediated and inflammatory diseases. Thus, reciprocal regulation between immune cell infiltration and AT dysfunction is a promising future therapeutic target.
Collapse
Affiliation(s)
- Tomasz J Guzik
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Dominik S Skiba
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Rhian M Touyz
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - David G Harrison
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Department of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
19
|
Wierzbicki AS. Further options for treating lipids in people with diabetes: targeting LDL-cholesterol and beyond. Diabet Med 2018; 35:1173-1180. [PMID: 29766560 DOI: 10.1111/dme.13667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2018] [Indexed: 12/11/2022]
Abstract
Diabetes is associated with increased cardiovascular disease (CVD) risk. Previous studies with statins have established that a 1 mmol/l reduction in LDL-cholesterol reduces CVD events by 21% over 5 years in people with diabetes. More recently, trials in people with acute coronary syndromes showed that ezetimibe reduced CVD events by 6% at 5 years and achieved a LDL-cholesterol of 1.6 mmol/l with better results in people with Type 2 diabetes. Several novel lipid-lowering therapies have recently been developed. Most data have been accumulated with proprotein convertase subtilisin kexin-9 (PCSK-9) inhibitors, which reduce LDL-cholesterol by 50-55%. A large CVD outcome trial with evolocumab, in which 40% of participants had diabetes, achieved a LDL-cholesterol of 0.8 mmol/l and showed a consistent 20% relative risk reduction within 2 years, including in people with diabetes. Trials to increase HDL-cholesterol using cholesterol ester transfer protein (CETP) inhibitors have generally underwhelmed. Although anacetrapib reduced coronary ischaemic events by 7% in a population with chronic CVD, more expansive CVD endpoints were not improved. The complex nature of CETP inhibitor trial outcomes means that these compounds are not being developed further. Trials targeting inflammation-associated lipids have been generally unsuccessful but recent data on the interleukin-1B receptor antagonist canakinumab have shown a reduction in acute coronary intervention, validating this target although at the cost of increased infections. The ability to achieve low LDL-cholesterol with off-patent medications and the costs of novel therapies will confine the use of novel agents to subgroups of people at highest risk of CVD.
Collapse
Affiliation(s)
- A S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas' Hospitals, London, UK
| |
Collapse
|
20
|
Petrie JR, Guzik TJ, Touyz RM. Diabetes, Hypertension, and Cardiovascular Disease: Clinical Insights and Vascular Mechanisms. Can J Cardiol 2018; 34:575-584. [PMID: 29459239 PMCID: PMC5953551 DOI: 10.1016/j.cjca.2017.12.005] [Citation(s) in RCA: 846] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022] Open
Abstract
Hypertension and type 2 diabetes are common comorbidities. Hypertension is twice as frequent in patients with diabetes compared with those who do not have diabetes. Moreover, patients with hypertension often exhibit insulin resistance and are at greater risk of diabetes developing than are normotensive individuals. The major cause of morbidity and mortality in diabetes is cardiovascular disease, which is exacerbated by hypertension. Accordingly, diabetes and hypertension are closely interlinked because of similar risk factors, such as endothelial dysfunction, vascular inflammation, arterial remodelling, atherosclerosis, dyslipidemia, and obesity. There is also substantial overlap in the cardiovascular complications of diabetes and hypertension related primarily to microvascular and macrovascular disease. Common mechanisms, such as upregulation of the renin-angiotensin-aldosterone system, oxidative stress, inflammation, and activation of the immune system likely contribute to the close relationship between diabetes and hypertension. In this article we discuss diabetes and hypertension as comorbidities and discuss the pathophysiological features of vascular complications associated with these conditions. We also highlight some vascular mechanisms that predispose to both conditions, focusing on advanced glycation end products, oxidative stress, inflammation, the immune system, and microRNAs. Finally, we provide some insights into current therapies targeting diabetes and cardiovascular complications and introduce some new agents that may have vasoprotective therapeutic potential in diabetes.
Collapse
Affiliation(s)
- John R Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom.
| |
Collapse
|
21
|
Oikonomou EK. Clinical and translational science in cardiovascular research: highlights from the American Heart Association Scientific Sessions 2017: Changing view on the concept of cardiovascular risk and blood pressure targets. Cardiovasc Res 2018; 114:e6-e8. [PMID: 29202178 DOI: 10.1093/cvr/cvx233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Evangelos K Oikonomou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Level 6, West Wing, Jonh Radcliffe Hospital, Headley Way, OX3 9DU, Oxford, UK
| |
Collapse
|
22
|
Huang Z, Jiang H, Cui X, Liang G, Chen Y, Wang T, Sun Z, Qi L. Elevated serum levels of lipoprotein‑associated phospholipase A2 predict mortality rates in patients with sepsis. Mol Med Rep 2017; 17:1791-1798. [PMID: 29138849 DOI: 10.3892/mmr.2017.8034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 01/09/2017] [Indexed: 11/06/2022] Open
Abstract
Sepsis remains one of the leading contributors to mortality rates in the intensive care unit (ICU) and emergency intensive care unit (EICU). Therefore, any treatments against the agents which produce sepsis in a medical emergency, are welcome. Elevated serum levels of lipoprotein‑associated phospholipase A2 (Lp‑PLA2) have been reported in a small cohort of patients with inflammation. The present study evaluated serum levels of Lp‑PLA2 in patients with sepsis and investigated the role of Lp‑PLA2 in sepsis. The investigation involved the selection of 151 patients with sepsis admitted to the emergency department of the Affiliated Hospital of Nantong University (Nantong, China) and 30 healthy controls. All patients (39 with sepsis, 55 with severe sepsis and 57 with septic shock) were examined on admission to the EICU. A complete blood count was performed, and serum levels of Lp‑PLA2, C‑reactive protein, procalcitonin, and interleukin 6, sequential organ failure (SOFA) scores and Acute Physiology and Chronic Health Evaluation II (APACHE II) scores were determined on hospital admission. The EICU and overall mortality rates were evaluated at baseline. The present study also assessed various laboratory parameters, clinical data and inflammatory cytokines. The patient follow up duration was 90 days. The data suggested that the serum levels of Lp‑PLA2 on admission to the EICU in patients with sepsis were elevated, compared with those in healthy controls. The concentrations of Lp‑PLA2 were correlated with the severity of disease, and were significantly associated with experimental markers of inflammation and established prognostic scores. In the total cohort, persistently elevated levels of Lp‑PLA2 on admission for EICU treatment was a predictor of poor prognosis, and provided superior diagnostic use, compared with the prognostic scoring systems, including SOFA or APACHE II scores. Taken together, the results suggested that Lp‑PLA2, with respect to other markers of inflammation, may have a role as a prognostic marker in sepsis, and provide background evidence for further trials to evaluate the clinical and pathophysiologic roles of Lp‑PLA2 in sepsis. Persistently elevated serum concentrations of Lp‑PLA2 indicated an unfavorable outcome in patients with sepsis. In addition, the results indicated the potential role of Lp‑PLA2 as a prognostic biomarker in patients with sepsis during the early course of EICU treatment.
Collapse
Affiliation(s)
- Zhongwei Huang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Haiyan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaohui Cui
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Guiwen Liang
- Department of Geriatric Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yu Chen
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Ting Wang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhichao Sun
- Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
23
|
Paul P, Picard C, Sampol E, Lyonnet L, Di Cristofaro J, Paul-Delvaux L, Lano G, Nicolino-Brunet C, Ravis E, Collart F, Dignat-George F, Dussol B, Sabatier F, Mouly-Bandini A. Genetic and Functional Profiling of CD16-Dependent Natural Killer Activation Identifies Patients at Higher Risk of Cardiac Allograft Vasculopathy. Circulation 2017; 137:1049-1059. [PMID: 29097449 DOI: 10.1161/circulationaha.117.030435] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/19/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cardiac transplantation is an effective therapy for end-stage heart failure. Because cardiac allograft vasculopathy (CAV) is the major cause of late mortality after heart transplant (HT), there is a need to identify markers that reflect inflammatory or cytotoxic immune mechanisms contributing to its onset. Noninvasive and early stratification of patients at risk remains a challenge for adapting individualized therapy. The CD16 (Fc-gamma receptor 3A [FCGR3A]) receptor was recently identified as a major determinant of antibody-mediated natural killer (NK) cell activation in HT biopsies; however, little is known about the role of CD16 in promoting allograft vasculopathy. This study aimed to investigate whether markers that reflect CD16-dependent circulating NK cell activation may identify patients at higher risk of developing CAV after HT. METHODS Blood samples were collected from 103 patients undergoing routine coronarography angiography for CAV diagnosis (median 5 years since HT). Genomic and phenotypic analyses of FCGR3A/CD16 Fc-receptor profiles were compared in CAV-positive (n=52) and CAV-free patients (n=51). The levels of CD16 expression and rituximab-dependent cell cytotoxic activity of peripheral NK cells in HT recipients were evaluated using a noninvasive NK-cellular humoral activation test. RESULTS Enhanced levels of CD16 expression and antibody-dependent NK cell cytotoxic function of HT recipients were associated with the FCGR3A-VV genotype. The frequency of the FCGR3A-VV genotype was significantly higher in the CAV+ group (odds ratio, 3.9; P=0.0317) than in the CAV- group. The FCGR3A-VV genotype was identified as an independent marker correlated with the presence of CAV at the time of coronary angiography by using multivariate logistic regression models. The FCGR3A-VV genotype was also identified as a baseline-independent predictor of CAV risk (odds ratio, 4.7; P=0.023). CONCLUSIONS This study unravels a prominent role for the CD16-dependent NK cell activation pathway in the complex array of factors that favor the progression of transplant arteriosclerosis. It highlights the clinical potential of a noninvasive evaluation of FCGR3A/CD16 in the early stratification of CAV risk. The recognition of CD16 as a major checkpoint that controls immune surveillance may promote the design of individualized NK cell-targeted therapies to limit vascular damage in highly responsive sensitized patients. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT01569334.
Collapse
Affiliation(s)
- Pascale Paul
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.). .,INSERM, Aix-Marseille Université (AMU), VRCM, UMR-1076, France (P.P., F.D.G., F.S.)
| | - Christophe Picard
- Immunogenetic Laboratory, Établissement Français du Sang, Marseille, France (C.P.).,Aix Marseille Université, Etablissement Français du Sang, CNRS, UMR 7268 ADÉS, France (C.P., J.D.C.)
| | - Emmanuelle Sampol
- Assistance Publique-Hôpitaux Marseille (AP-HM), Pharmacokinetics Department, France (E.S.)
| | - Luc Lyonnet
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.)
| | - Julie Di Cristofaro
- Assistance Publique-Hôpitaux Marseille (AP-HM), Pharmacokinetics Department, France (E.S.)
| | - Louise Paul-Delvaux
- ENSAE-Paristech, Data Science & Statistics Department, Paris, France (L.P.-D.)
| | - Guillaume Lano
- Assistance Publique-Hôpitaux Marseille (AP-HM), Aix Marseille Univ, Center for Clinical Investigation, France (G.L., B.D.)
| | - Corinne Nicolino-Brunet
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.)
| | - Eleonore Ravis
- Assistance Publique Hopitaux de Marseille (AP-HM), Adult Cardiac Surgery Department, France (E.R., F.C., A.M.B.)
| | - Frederic Collart
- Assistance Publique Hopitaux de Marseille (AP-HM), Adult Cardiac Surgery Department, France (E.R., F.C., A.M.B.)
| | - Francoise Dignat-George
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.).,INSERM, Aix-Marseille Université (AMU), VRCM, UMR-1076, France (P.P., F.D.G., F.S.)
| | - Bertrand Dussol
- Assistance Publique-Hôpitaux Marseille (AP-HM), Aix Marseille Univ, Center for Clinical Investigation, France (G.L., B.D.)
| | - Florence Sabatier
- Assistance Publique-Hôpitaux Marseille (AP-HM), Vascular Biology and Cell Therapy Department, France (P.P., L.L., C.N.B., F.D.G., F.S.).,INSERM, Aix-Marseille Université (AMU), VRCM, UMR-1076, France (P.P., F.D.G., F.S.)
| | - Annick Mouly-Bandini
- Assistance Publique Hopitaux de Marseille (AP-HM), Adult Cardiac Surgery Department, France (E.R., F.C., A.M.B.)
| |
Collapse
|
24
|
Schnitzler JG, Bernelot Moens SJ, Tiessens F, Bakker GJ, Dallinga-Thie GM, Groen AK, Nieuwdorp M, Stroes ESG, Kroon J. Nile Red Quantifier: a novel and quantitative tool to study lipid accumulation in patient-derived circulating monocytes using confocal microscopy. J Lipid Res 2017; 58:2210-2219. [PMID: 28972117 PMCID: PMC5665660 DOI: 10.1194/jlr.d073197] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 09/07/2017] [Indexed: 01/19/2023] Open
Abstract
The inflammatory profile of circulating monocytes is an important biomarker for atherosclerotic plaque vulnerability. Recent research revealed that peripheral lipid uptake by monocytes alters their phenotype toward an inflammatory state and this coincides with an increased lipid droplet (LD) content. Determination of lipid content of circulating monocytes is, however, not very well established. Based on Nile Red (NR) neutral LD imaging, using confocal microscopy and computational analysis, we developed NR Quantifier (NRQ), a novel quantification method to assess LD content in monocytes. Circulating monocytes were isolated from blood and used for the NR staining procedure. In monocytes stained with NR, we clearly distinguished, based on 3D imaging, phospholipids and exclusively intracellular neutral lipids. Next, we developed and validated NRQ, a semi-automated quantification program that detects alterations in lipid accumulation. NRQ was able to detect LD alterations after ex vivo exposure of isolated monocytes to freshly isolated LDL in a time- and dose-dependent fashion. Finally, we validated NRQ in patients with familial hypercholesterolemia and obese subjects in pre- and postprandial state. In conclusion, NRQ is a suitable tool to detect even small differences in neutral LD content in circulating monocytes using NR staining.
Collapse
Affiliation(s)
- Johan G Schnitzler
- Departments of Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands
| | | | - Feiko Tiessens
- Departments of Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands
| | - Guido J Bakker
- Departments of Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands
| | - Geesje M Dallinga-Thie
- Departments of Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands.,Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Albert K Groen
- Departments of Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Laboratory of Metabolic Diseases, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Max Nieuwdorp
- Departments of Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands.,Wallenberg Laboratory, University of Gothenberg, Gothenberg, Sweden.,Department of Internal Medicine, Diabetes Center, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands.,Institute for Cardiovascular Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
| | - Erik S G Stroes
- Departments of Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Departments of Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Guzik TJ, Touyz RM. Oxidative Stress, Inflammation, and Vascular Aging in Hypertension. Hypertension 2017; 70:660-667. [PMID: 28784646 DOI: 10.1161/hypertensionaha.117.07802] [Citation(s) in RCA: 451] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Tomasz J Guzik
- From the British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, United Kingdom (T.J.G., R.M.T.); and Department of Internal and Agricultural Medicine, Translational Medicine Laboratory, Collegium Medicum Jagiellonian University, Krakow, Poland (T.J.G.).
| | - Rhian M Touyz
- From the British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, United Kingdom (T.J.G., R.M.T.); and Department of Internal and Agricultural Medicine, Translational Medicine Laboratory, Collegium Medicum Jagiellonian University, Krakow, Poland (T.J.G.)
| |
Collapse
|
26
|
Beierlein JM, McNamee LM, Walsh MJ, Kaitin KI, DiMasi JA, Ledley FD. Landscape of Innovation for Cardiovascular Pharmaceuticals: From Basic Science to New Molecular Entities. Clin Ther 2017; 39:1409-1425.e20. [PMID: 28652015 DOI: 10.1016/j.clinthera.2017.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/23/2017] [Accepted: 06/05/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE This study examines the complete timelines of translational science for new cardiovascular therapeutics from the initiation of basic research leading to identification of new drug targets through clinical development and US Food and Drug Administration (FDA) approval of new molecular entities (NMEs) based on this research. METHODS This work extends previous studies by examining the association between the growth of research on drug targets and approval of NMEs associated with these targets. Drawing on research on innovation in other technology sectors, where technological maturity is an important determinant in the success or failure of new product development, an analytical model was used to characterize the growth of research related to the known targets for all 168 approved cardiovascular therapeutics. FINDINGS Categorizing and mapping the technological maturity of cardiovascular therapeutics reveal that (1) there has been a distinct transition from phenotypic to targeted methods for drug discovery, (2) the durations of clinical and regulatory processes were significantly influenced by changes in FDA practice, and (3) the longest phase of the translational process was the time required for technology to advance from initiation of research to a statistically defined established point of technology maturation (mean, 30.8 years). IMPLICATIONS This work reveals a normative association between metrics of research maturation and approval of new cardiovascular therapeutics and suggests strategies for advancing translational science by accelerating basic and applied research and improving the synchrony between the maturation of this research and drug development initiatives.
Collapse
Affiliation(s)
- Jennifer M Beierlein
- Center for Integration of Science and Industry, Department of Natural & Applied Sciences, Bentley University, Waltham, Massachusetts
| | - Laura M McNamee
- Center for Integration of Science and Industry, Department of Natural & Applied Sciences, Bentley University, Waltham, Massachusetts
| | - Michael J Walsh
- Center for Integration of Science and Industry, Department of Natural & Applied Sciences, Bentley University, Waltham, Massachusetts
| | - Kenneth I Kaitin
- Tufts Center for the Study of Drug Development, Tufts University School of Medicine, Boston, Massachusetts
| | - Joseph A DiMasi
- Tufts Center for the Study of Drug Development, Tufts University School of Medicine, Boston, Massachusetts
| | - Fred D Ledley
- Center for Integration of Science and Industry, Department of Natural & Applied Sciences, Bentley University, Waltham, Massachusetts; Department of Management, Bentley University, Waltham, Massachusetts.
| |
Collapse
|
27
|
Affiliation(s)
- Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, UK.,Department of Internal and Agricultural Medicine, Jagiellonian University, Collegium Medicum, ul. Skarbowa 1, 31-101, Krakow, Poland
| |
Collapse
|
28
|
A comparison of heart rate variability, n-3 PUFA status and lipid mediator profile in age- and BMI-matched middle-aged vegans and omnivores. Br J Nutr 2017; 117:669-685. [PMID: 28366178 DOI: 10.1017/s0007114517000629] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Low heart rate variability (HRV) predicts sudden cardiac death. Long-chain (LC) n-3 PUFA (C20-C22) status is positively associated with HRV. This cross-sectional study investigated whether vegans aged 40-70 years (n 23), whose diets are naturally free from EPA (20 : 5n-3) and DHA (22 : 6n-3), have lower HRV compared with omnivores (n 24). Proportions of LC n-3 PUFA in erythrocyte membranes, plasma fatty acids and concentrations of plasma LC n-3 PUFA-derived lipid mediators were significantly lower in vegans. Day-time interbeat intervals (IBI), adjusted for physical activity, age, BMI and sex, were significantly shorter in vegans compared with omnivores (mean difference -67 ms; 95 % CI -130, -3·4, P50 % and high-frequency power) were similarly lower in vegans, with no differences during sleep. In conclusion, vegans have higher 24 h SDNN, but lower day-time HRV and shorter day-time IBI relative to comparable omnivores. Vegans may have reduced availability of precursor markers for pro-resolving lipid mediators; it remains to be determined whether there is a direct link with impaired cardiac function in populations with low-n-3 status.
Collapse
|
29
|
Cannatà A, Marcon G, Cimmino G, Camparini L, Ciucci G, Sinagra G, Loffredo FS. Role of circulating factors in cardiac aging. J Thorac Dis 2017; 9:S17-S29. [PMID: 28446965 PMCID: PMC5383555 DOI: 10.21037/jtd.2017.03.95] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 01/31/2017] [Indexed: 12/11/2022]
Abstract
Worldwide increase in life expectancy is a major contributor to the epidemic of chronic degenerative diseases. Aging, indeed, simultaneously affects multiple organ systems, and it has been hypothesized that systemic alterations in regulators of tissue physiology may regulate this process. Cardiac aging itself is a major risk factor for cardiovascular diseases and, because of the intimate relationship with the brain, may contribute to increase the risk of neurodegenerative disorders. Blood-borne factors may play a major role in this complex and still elusive process. A number of studies, mainly based on the revival of parabiosis, a surgical technique very popular during the 70s of the 20th century to study the effect of a shared circulation in two animals, have indeed shown the potential that humoral factors can control the aging process in different tissues. In this article we review the role of circulating factors in cardiovascular aging. A better understanding of these mechanisms may provide new insights in the aging process and provide novel therapeutic opportunities for chronic age-related disorders.
Collapse
Affiliation(s)
- Antonio Cannatà
- Molecular Cardiology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata and University of Trieste, Trieste, Italy
| | - Gabriella Marcon
- DAMA- University of Udine, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| | - Giovanni Cimmino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| | - Luca Camparini
- Molecular Cardiology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Giulio Ciucci
- Molecular Cardiology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata and University of Trieste, Trieste, Italy
| | - Francesco S. Loffredo
- Molecular Cardiology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata and University of Trieste, Trieste, Italy
| |
Collapse
|
30
|
Bernelot Moens SJ, Neele AE, Kroon J, van der Valk FM, Van den Bossche J, Hoeksema MA, Hoogeveen RM, Schnitzler JG, Baccara-Dinet MT, Manvelian G, de Winther MP, Stroes ES. PCSK9 monoclonal antibodies reverse the pro-inflammatory profile of monocytes in familial hypercholesterolaemia. Eur Heart J 2017; 38:1584-1593. [DOI: 10.1093/eurheartj/ehx002] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 01/02/2017] [Indexed: 12/26/2022] Open
Affiliation(s)
| | - Annette E. Neele
- Experimental Vascular Biology, Medical Biochemistry, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Fleur M. van der Valk
- Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jan Van den Bossche
- Experimental Vascular Biology, Medical Biochemistry, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Marten A. Hoeksema
- Experimental Vascular Biology, Medical Biochemistry, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Renate M. Hoogeveen
- Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Johan G. Schnitzler
- Experimental Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Marie T. Baccara-Dinet
- Sanofi, Clinical Development, R&D, 371 Rue du Professeur Blayac, 34080, Montpellier, France
| | - Garen Manvelian
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Menno P.J. de Winther
- Experimental Vascular Biology, Medical Biochemistry, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstraße 8a & 9, 80336 Munich, Germany
| | - Erik S.G. Stroes
- Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Merlini E, Tincati C, Biasin M, Saulle I, Cazzaniga FA, d'Arminio Monforte A, Cappione AJ, Snyder-Cappione J, Clerici M, Marchetti GC. Stimulation of PBMC and Monocyte-Derived Macrophages via Toll-Like Receptor Activates Innate Immune Pathways in HIV-Infected Patients on Virally Suppressive Combination Antiretroviral Therapy. Front Immunol 2016; 7:614. [PMID: 28066424 PMCID: PMC5165253 DOI: 10.3389/fimmu.2016.00614] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/05/2016] [Indexed: 12/25/2022] Open
Abstract
In HIV-infected, combination antiretroviral therapy (cART)-treated patients, immune activation and microbial translocation persist and associate with inadequate CD4 recovery and morbidity/mortality. We analyzed whether alterations in the toll-like receptor (TLR) pathway could be responsible for the immune hyperactivation seen in these patients. PBMC/monocyte-derived macrophages (MDMs) of 28 HIV+ untreated and 35 cART-treated patients with HIV-RNA < 40 cp/mL [20 Full Responders (FRs): CD4 ≥ 350; 15 Immunological Non-Responders (INRs): CD4 < 350], as well as of 16 healthy controls were stimulated with a panel of TLR agonists. We measured: CD4/CD8/CD14/CD38/HLA-DR/Ki67/AnnexinV/CD69/TLR4/8 (Flow Cytometry); PBMC expression of 84 TLR pathway genes (qPCR); PBMC/MDM cytokine release (Multiplex); and plasma lipopolysaccharide (LPS)/sCD14 (LAL/ELISA). PBMC/MDM from cART patients responded weakly to LPS stimulation but released high amounts of pro-inflammatory cytokines. MDM from these patients were characterized by a reduced expression of HLA-DR+ MDM and failed to expand activated HLA-DR+ CD38+ T-lymphocytes. PBMC/MDM from cART patients responded more robustly to ssRNA stimulation; this resulted in a significant expansion of activated CD38 + CD8 and the release of amounts of pro-inflammatory cytokines comparable to those seen in untreated viremic patients. Despite greater constitutive TLR pathway gene expression, PBMC from INRs seemed to upregulate only type I IFN genes following TLR stimulation, whereas PBMC from full responders showed a broader response. Systemic exposure to microbial antigens drives immune activation during cART by triggering TLRs. Bacterial stimulation modifies MDM function/pro-inflammatory profile in cART patients without affecting T-lymphocytes; this suggests translocating bacteria as selective stimulus to chronic innate activation during cART. High constitutive TLR activation is seen in patients lacking CD4 recovery, suggesting an exhausted immune milieu, anergic to further antigen encounters.
Collapse
Affiliation(s)
- Esther Merlini
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| | - Camilla Tincati
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences - "L. Sacco", University of Milan , Milan , Italy
| | - Irma Saulle
- Department of Biomedical and Clinical Sciences - "L. Sacco", University of Milan , Milan , Italy
| | - Federico Angelo Cazzaniga
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| | - Antonella d'Arminio Monforte
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| | | | | | - Mario Clerici
- Department of Physiopathology and Transplants, University of Milan, Milan, Italy; Don C. Gnocchi Foundation, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giulia Carla Marchetti
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| |
Collapse
|
32
|
Pais P, Villar A, Rull S. Impact of a Proprietary Standardized Olive Fruit Extract (SOFE) on Cardio-Ankle Vascular Index, Visual Analog Scale and C-Reactive Protein Assessments in Subjects with Arterial Stiffness Risk. Drugs R D 2016; 16:355-368. [PMID: 27798770 PMCID: PMC5114205 DOI: 10.1007/s40268-016-0147-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION The chemical components of olive fruit provide a wide range of cardiovascular benefits. Arterial stiffness is an important cardiovascular risk factor and can be assessed using the Cardio-Ankle Vascular Index (CAVI). OBJECTIVE The objective of this study was to assess the impact of a proprietary standardized olive fruit extract (SOFE) in subjects at risk for arterial stiffness. METHODS Twelve of 36 subjects were assigned to each of the following groups for this 11-day, double-blind, placebo-controlled study: Group 1: 250 mg SOFE-50 mg active ingredient, hydroxytyrosol (dosage achieved with two capsules per day); Group 2: 500 mg SOFE-100 mg active ingredient, hydroxytyrosol (dosage achieved with two capsules per day); and Group 3: placebo. RESULTS All three groups showed a decrease in CAVI scores, although no significant between-group differences were observed. Group 2 had the largest reduction, with mean CAVI scores decreasing from 11.02 to 8.91. Group 2 showed the strongest response in visual analog scale (VAS) energy intensity (11.71% increase). Results for C-reactive protein (CRP) blood levels showed no changes of statistical significance between groups. Mean triglyceride levels from Group 2 decreased by 21.64%, the most significant change among all three groups. CONCLUSIONS SOFE, an olive fruit extract containing many of the phytochemicals shown to provide cardiovascular benefits, was safe and well-tolerated. SOFE 500 mg had a greater effect on CAVI scores, suggesting improved arterial elasticity, and a clear reduction in triglycerides compared with placebo and SOFE 250 mg. The decreased CAVI scores suggest that increasing high-density lipoprotein cholesterol and lowering triglycerides with SOFE could potentially reduce patients' risk of developing atherosclerosis. Although more studies are needed, positive cardiovascular health trends, including improved vessel elasticity and positive triglyceride effects, were evident with SOFE.
Collapse
Affiliation(s)
- Pilar Pais
- Euromed, C/Rec de Dalt, 21-23, 08100 Mollet del Vallès, Barcelona, Spain
| | - Agustin Villar
- Euromed, C/Rec de Dalt, 21-23, 08100 Mollet del Vallès, Barcelona, Spain.
| | - Santiago Rull
- Euromed, C/Rec de Dalt, 21-23, 08100 Mollet del Vallès, Barcelona, Spain
| |
Collapse
|
33
|
Sahebkar A, Di Giosia P, Stamerra CA, Grassi D, Pedone C, Ferretti G, Bacchetti T, Ferri C, Giorgini P. Effect of monoclonal antibodies to PCSK9 on high-sensitivity C-reactive protein levels: a meta-analysis of 16 randomized controlled treatment arms. Br J Clin Pharmacol 2016; 81:1175-90. [PMID: 26861255 DOI: 10.1111/bcp.12905] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/31/2016] [Accepted: 02/08/2016] [Indexed: 12/21/2022] Open
Abstract
AIMS Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are an emerging class of low-density lipoprotein cholesterol (LDL-C)-lowering agents. In spite of their known effects on lipids, the impact of these drugs on systemic inflammation is less known. We aimed to investigate the effect of PCSK9 inhibitors on high-sensitivity C-reactive protein (hs-CRP) levels through a meta-analysis of randomized controlled trials (RCTs). METHODS A systematic literature search of Medline, SCOPUS and Google Scholar was conducted up to December 2015 to identify RCTs assessing changes in hs-CRP concentrations during treatment with PCSK9 inhibitors. Quantitative data synthesis was performed using a random-effects model, with weighed mean difference (WMD) and 95% confidence interval (CI) as summary statistics. RESULTS Sixteen treatment arms, with a total of 2546 participants, were included. Random-effects meta-analysis did not show any significant effect of PCSK9 inhibitors on hs-CRP levels (WMD: 0.002 mg l(-1) , CI: -0.017, 0.021; P = 0.807; I(2) = 37.26%). This effect size was robust, not sensitive to any single study, and not affected by the type of PCSK9 inhibitor (evolocumab: WMD: 0.002 mg l(-1) , CI: -0.02, 0.02; P = 0.855; alirocumab WMD: 0.15 mg l(-1) , CI: -0.11, 0.40; P = 0.259; I(2) = 0%), or dosing frequency (biweekly: WMD: 0.13 mg l(-1) , CI: -0.20, 0.46; P = 0.433; I(2) = 55.19%; monthly: WMD: 0.003 mg l(-1) , CI: -0.01, 0.01; P = 0.59; I(2) = 0%). Random-effects meta-regression did not suggest any association of changes in hs-CRP levels with changes in plasma LDL-C concentrations (P = 0.697) or cumulative dosage of the drug (P = 0.980). CONCLUSIONS This meta-analysis of RCTs did not suggest an effect of PCSK9 inhibitors on hs-CRP concentrations.
Collapse
Affiliation(s)
- Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Research Centre, Royal Perth Hospital, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Paolo Di Giosia
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Cosimo Andrea Stamerra
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Davide Grassi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Claudio Pedone
- Area di Geriatria, Università Campus Biomedico di Roma, Rome, Italy
| | - Gianna Ferretti
- Dipartimento di Scienze cliniche Specialistiche ed Odontostomatologiche (DISCO), Università Politecnica delle Marche, Ancona, Italy
| | - Tiziana Bacchetti
- Dipartimento di Scienze della Vita e dell'Ambiente (DISVA), Università Politecnica delle Marche, Ancona, Italy
| | - Claudio Ferri
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paolo Giorgini
- Department of Life, Health & Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
34
|
Domene C, Jorgensen C, Abbasi SW. A perspective on structural and computational work on collagen. Phys Chem Chem Phys 2016; 18:24802-24811. [DOI: 10.1039/c6cp03403a] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Collagen is the single most abundant protein in the extracellular matrix in the animal kingdom, with remarkable structural and functional diversity and regarded one of the most useful biomaterials.
Collapse
Affiliation(s)
- Carmen Domene
- Department of Chemistry
- King's College London
- UK
- Chemistry Research Laboratory
- University of Oxford
| | | | | |
Collapse
|