151
|
Ciccarelli M, Dawson D, Falcao-Pires I, Giacca M, Hamdani N, Heymans S, Hooghiemstra A, Leeuwis A, Hermkens D, Tocchetti CG, van der Velden J, Zacchigna S, Thum T. Reciprocal organ interactions during heart failure: a position paper from the ESC Working Group on Myocardial Function. Cardiovasc Res 2021; 117:2416-2433. [PMID: 33483724 PMCID: PMC8562335 DOI: 10.1093/cvr/cvab009] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/20/2021] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Heart failure-either with reduced or preserved ejection fraction (HFrEF/HFpEF)-is a clinical syndrome of multifactorial and gender-dependent aetiology, indicating the insufficiency of the heart to pump blood adequately to maintain blood flow to meet the body's needs. Typical symptoms commonly include shortness of breath, excessive fatigue with impaired exercise capacity, and peripheral oedema, thereby alluding to the fact that heart failure is a syndrome that affects multiple organ systems. Patients suffering from progressed heart failure have a very limited life expectancy, lower than that of numerous cancer types. In this position paper, we provide an overview regarding interactions between the heart and other organ systems, the clinical evidence, underlying mechanisms, potential available or yet-to-establish animal models to study such interactions and finally discuss potential new drug interventions to be developed in the future. Our working group suggests that more experimental research is required to understand the individual molecular mechanisms underlying heart failure and reinforces the urgency for tailored therapeutic interventions that target not only the heart but also other related affected organ systems to effectively treat heart failure as a clinical syndrome that affects and involves multiple organs.
Collapse
Affiliation(s)
- Michele Ciccarelli
- University of Salerno, Department of Medicine, Surgery and Dentistry, Via S. Allende 1, 84081, Baronissi(Salerno), Italy
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2DZ, UK
| | - Inês Falcao-Pires
- Department of Surgery and Physiology, Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Mauro Giacca
- King’s College London, Molecular Medicine Laboratory, 125 Caldharbour Lane, London WC2R2LS, United Kingdom
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34129 Trieste, Italy
| | - Nazha Hamdani
- Department of Clinical Pharmacology and Molecular Cardiology, Institute of Physiology, Ruhr University Bochum, Universitätsstraße 150, D-44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Universitätsstraße 150, D-44801 Bochum, Germany
| | - Stéphane Heymans
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, Bus 911, 3000 Leuven, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
- ICIN-Netherlands Heart Institute, Holland Heart House, Moreelsepark 1, 3511 EP Utrecht, the Netherlands
| | - Astrid Hooghiemstra
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081HZ, Amsterdam, The Netherlands
- Department of Medical Humanities, Amsterdam Public Health Research Institute, Amsterdam UMC, Location VUmc, De Boelelaan 1089a, 1081HV, Amsterdam, The Netherlands
| | - Annebet Leeuwis
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081HZ, Amsterdam, The Netherlands
| | - Dorien Hermkens
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, the Netherlands
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, Amsterdam Cardiovascular Sciences, De Boelelaan 1118, 1081HZ Amsterdam, the Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34129 Trieste, Italy
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
- REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Nicolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
152
|
Pereira-Manfro W, de Lima G, Nogueira J, Portugal M, Milagres L, Bezerra F, Faerstein E, Koury J. Association between visceral/subcutaneous adipose tissue ratio and plasma inflammatory markers and score for cardiovascular risk prediction in a Brazilian cohort: Pró-Saúde Study. Braz J Med Biol Res 2021; 54:e11521. [PMID: 34730680 PMCID: PMC8555454 DOI: 10.1590/1414-431x2021e11521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Visceral adipose tissue (VAT) is associated with various metabolic disorders, and adipokines, secreted by adipose tissue, are involved in their pathogenesis. This study investigated associations between VAT/subcutaneous adipose tissue (SAT) ratio, inflammatory markers, and cardiovascular (CV) risk-score in adults. Plasma levels of adipokines, plasma lipid profile, blood pressure, and body composition (using dual-emission x-ray absorptiometry) were determined. CV risk-score based on the American College of Cardiology and the American Heart Association (ACC/AHA) score was calculated in a sample of 309 Brazilian civil servants aged <60 years. Participants' VAT/SAT ratio were categorized into quartiles. Among males, plasma leptin (2.8 ng/mL) and C reactive protein (CRP) (0.2 mg/dL) (P<0.05) levels were higher at P75 and P50 than P5, and the highest calculated CV risk-score was observed at P75 (7.1%). Among females, higher plasma adiponectin levels were observed at P25 (54.3 ng/mL) compared with P75 (36 ng/mL) (P<0.05). Higher plasma CRP levels were observed at P75 (0.4 mg/dL) compared with P5 (0.1 mg/dL) (P<0.05). Higher CV risk-score was observed at P75 (2.0%) compared with P5 (0.7%). In both sexes, VAT and VAT/SAT ratio were directly associated with plasma leptin, CRP, and CV risk-score, and inversely associated with adiponectin; SAT was directly associated with plasma leptin and CRP (P<0.01); interleukin (IL)-10 and CRP were directly associated with adiponectin and leptin, respectively (P<0.05). Among men only, IL-10 (inversely) and CRP (directly) were associated with CV risk-score (P=0.02). Our results strengthened the relevance of the VAT/SAT ratio in cardiovascular risk.
Collapse
Affiliation(s)
- W.F. Pereira-Manfro
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - G.R. de Lima
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - J.F. Nogueira
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - M.R.C. Portugal
- Instituto de Nutrição, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
- Departamento de Nutrição, Centro Universitário de Volta Redonda - UniFOA, Volta Redonda, RJ, Brasil
| | - L.G. Milagres
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - F.F. Bezerra
- Instituto de Nutrição, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - E. Faerstein
- Instituto de Medicina Social, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - J.C. Koury
- Instituto de Nutrição, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
153
|
Miri Y, Leander K, Eriksson P, Gigante B, Ziegler L. Interleukin 6 trans-signalling and the risk of future cardiovascular events in men and women. Open Heart 2021; 8:openhrt-2021-001694. [PMID: 34635574 PMCID: PMC8506880 DOI: 10.1136/openhrt-2021-001694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/23/2021] [Indexed: 11/25/2022] Open
Abstract
Objective Pro-inflammatory interleukin 6 (IL6) trans-signalling is associated with increased risk of cardiovascular events (CVEs). Whether this association exists for both sexes is, however, uncertain. Hence, we analysed the risk of CVE associated with IL6 trans-signalling in men and women and investigated if potential interaction between IL6 trans-signalling and sex affects the risk. Methods In a prospective cohort of 60-year-old men and women without cardiovascular disease (men=2039, women=2193), subjects were followed for 20 years. To assess the IL6 trans-signalling activity, the proportion between the active binary and inactive ternary IL6 complexes, the binary/ternary ratio (B/T ratio), was estimated. CVE (myocardial infarction, angina pectoris and ischaemic stroke, n=629) risk was analysed with Cox regression, presented as HRs with 95% CIs. B/T ratio was dichotomised, with levels >median representing IL6 trans-signalling. Interaction was analysed on the additive scale and expressed as the synergy index (S). Analyses were adjusted for cardiovascular risk factors. Results B/T ratio >median was associated with increased CVE risk in men (HR 1.63; 95% CI 1.32 to 2.01), but not in women (HR 1.21; 95% CI 0.93 to 1.57). There was a significant synergistic interaction (S=1.98; 95% CI 1.15 to 3.42) between the B/T ratio and male sex, the combination increasing the risk by 88%. Conclusions Our results suggest differential susceptibility to inflammation mediated by IL6 trans-signalling and subsequent CVE in men and women. The B/T ratio could be considered as a novel biomarker for cardiovascular risk in men, but not in women.
Collapse
Affiliation(s)
- Yasmin Miri
- Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Leander
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per Eriksson
- Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bruna Gigante
- Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Louise Ziegler
- Department of Clinical Sciences Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
154
|
Lutgens E, Joffre J, van Os B, Ait-Oufella H. Targeting cytokines and immune checkpoints in atherosclerosis with monoclonal antibodies. Atherosclerosis 2021; 335:98-109. [PMID: 34593238 DOI: 10.1016/j.atherosclerosis.2021.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/24/2021] [Accepted: 09/21/2021] [Indexed: 11/15/2022]
Abstract
Over the past fifteen years, treatments using monoclonal antibodies specifically targeting cytokines have been developed to treat chronic inflammatory diseases, including rheumatoid arthritis or psoriasis, both associated with increased cardiovascular risk. The cardiovascular impact of these therapies allows us to validate the clinical relevance of the knowledge acquired from experimental studies about the role of cytokines in atherosclerosis. Several clinical studies have confirmed the protective effects of anti-TNFα and anti-IL-6R monoclonal antibodies against athero-thrombotic cardiovascular risk in patients with chronic inflammatory diseases. Yet, caution is needed since anti-TNFα treatment can aggravate chronic heart failure. More recently, the CANTOS study showed for the first time that an anti-inflammatory treatment using anti-IL-1β monoclonal antibody in coronary artery disease patients significantly reduced cardiovascular events. The effects of IL-23/IL-17 axis blockade on cardiovascular risk in patients with psoriasis or arthritis remain controversial. Several monoclonal antibodies targeting costimulatory molecules have also been developed, a direct way to confirm their involvement in atherothrombotic cardiovascular diseases. Blocking the CD28-CD80/86 axis with Abatacept has been shown to reduce cardiovascular risk. In contrast, the treatment of cancer patients with antibodies blocking immune checkpoint inhibitory receptors, such as CTLA-4, PD1, or PDL1, could worsen the risk of atherothrombotic events. In the future, cardiologists will be increasingly solicited to assess the cardiovascular risk of patients suffering from chronic inflammatory diseases or cancer and participate in choosing the most appropriate treatment. At the same time, immunomodulatory approaches directly targeting cardiovascular diseases will be developed as a complement to the usual treatment strategies.
Collapse
Affiliation(s)
- Esther Lutgens
- Department of Medical Biochemistry Experimental Vascular Biology, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstraße 8a & 9, 80336, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Pettenkoferstraße 8a & 9, 80336, Munich, Germany.
| | - Jeremie Joffre
- Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France; Université de Paris, Inserm U970, Paris Cardiovascular Research Center, Paris, France; Department of Anesthesia and Perioperative Care, UCSF School of Medicine, San Francisco, CA, USA
| | - Bram van Os
- Department of Medical Biochemistry Experimental Vascular Biology, Amsterdam, the Netherlands
| | - Hafid Ait-Oufella
- Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France; Université de Paris, Inserm U970, Paris Cardiovascular Research Center, Paris, France.
| |
Collapse
|
155
|
Langer K, Cohen RA, Porges EC, Williamson JB, Woods AJ. Circulating Cytokines Predict 1H-Proton MRS Cerebral Metabolites in Healthy Older Adults. Front Aging Neurosci 2021; 13:690923. [PMID: 34489672 PMCID: PMC8416992 DOI: 10.3389/fnagi.2021.690923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 07/05/2021] [Indexed: 11/29/2022] Open
Abstract
Background: Changes in both circulating cytokines and neurochemical concentrations have been observed in aging. Patterns of change across these factors are associated with age-related pathologies, including neurodegenerative disease. More evidence to define patterns of change that are characteristic of healthy aging is needed, as is an investigation into how age-related changes in blood cytokines and brain neurochemicals may relate to one another in a healthy older adult population. Methods: Single voxel 1H-proton magnetic resonance spectroscopy was collected in medial frontal and medial parietal regions. Phosphocholine and glycerophosphocholine (Cho), myo-inositol (MI), N-acetylaspertate and N-acetylasperglutamate (NAA), creatine and phosphocreatine (Cr), and glutamate and glutamine (Glx) were measured in a sample of 83 healthy, cognitively normal adults aged 52–89. Blood data were collected to quantify 12 cytokines: interleukins (IL-) 2, 5, 6, 7, 8, 10, 12, 13, IL-1 β, tumor necrosis factor α (TNF-α), interferon γ (IFN-γ), and IL-17 α. Correlation analyses were performed to assess age relationships between each of these factors. Backward linear regressions were performed. Cytokine data and age were used as predictors of each cerebrospinal fluid (CSF)-corrected metabolite concentration in both voxels. Results: Associations were identified between a variety of cytokines and concentrations of frontal NAA, Cr, and Glx, and of parietal MI, Cho, NAA, and Cr. In the frontal voxel, NAA was predicted by more IL-1B and less TNF-α, Cr by less TNF-α and more IL-5, and Glx by less TNF-α. In the parietal voxel, MI was predicted by more IL-10 and IL-8 and less IL-2, Cho by more TNF-α and less IL-2, NAA by more IL-1B and TNF-α and less IL-13, IL-2, and IL-7, and Cr by more IL-10 and less IL-2. Conclusions: Associations were identified between circulating cytokines and neurometabolite concentrations in this sample of older adults. The present results serve as the initial evidence of relationships between circulating cytokines and neurophysiology. Findings invite further investigation to understand the physiological consequences of aging, and how peripheral inflammatory markers may relate to neurochemical concentrations in healthy aging.
Collapse
Affiliation(s)
- Kailey Langer
- Center for Cognitive Aging and Memory, Clinical Translational Research Program, College of Medicine, University of Florida, Gainesville, FL, United States.,Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Ronald A Cohen
- Center for Cognitive Aging and Memory, Clinical Translational Research Program, College of Medicine, University of Florida, Gainesville, FL, United States.,Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Eric C Porges
- Center for Cognitive Aging and Memory, Clinical Translational Research Program, College of Medicine, University of Florida, Gainesville, FL, United States.,Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - John B Williamson
- Center for Cognitive Aging and Memory, Clinical Translational Research Program, College of Medicine, University of Florida, Gainesville, FL, United States.,Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Adam J Woods
- Center for Cognitive Aging and Memory, Clinical Translational Research Program, College of Medicine, University of Florida, Gainesville, FL, United States.,Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| |
Collapse
|
156
|
Ellulu MS, Naser IA, Abuhajar SM, Najim AA. Determination of risk factors associated with inflammation in hypertensive patients with type-2 diabetes mellitus in a Palestinian Diabetes Study. Curr Med Res Opin 2021; 37:1451-1459. [PMID: 34125641 DOI: 10.1080/03007995.2021.1941826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To determine the risk factors associated with inflammation in hypertensive patients with type-2 diabetes mellitus. METHODS A total of 164 hypertensive patients with type 2 diabetes patients aged 38-60 years were selected from 7 primary healthcare centers in Gaza city, Palestine. Interview questionnaire were employed to collect data related to age, gender, smoking habits, and physical activity pattern. Laboratory biochemical tests included fasting blood glucose (FBG), triglyceride (TG), total cholesterol (TC), interleukin 6 (IL-6), high sensitive C reactive protein (hs-CRP), and adiponectin were estimated in all patients. RESULTS The study involved 118 (72%) women and 46 (28%) men; the mean of age was 53.7 ± 0.46 years. A tertile of inflammation feature with hs-CRP was developed. The highest tertile of hs-CRP was significantly associated with women, higher obesity indices, metabolic dysregulation involving lipid profile markers, FBG and blood pressure, IL-6, and lower adiponectin. After adjusting for age, gender, smoking habits, and physical activity; the risk factor of high level of hs-CRP were the increased body mass index [OR: 1.17, p = .018], IL-6 [OR: 2.22, p = .025] and FBG [OR: 1.01, p = .007], as well as reduced adiponectin [OR: 0.81, p = .002]. CONCLUSION The inflammation state was affected by obesity and had been related to altered adipokines levels of IL-6 and adiponectin, as well as affected by the glycemic control, as evidenced by higher serum level of FBG.
Collapse
Affiliation(s)
- Mohammed S Ellulu
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Al-Azhar University of Gaza (AUG), Gaza City, State of Palestine
| | - Ihab A Naser
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Al-Azhar University of Gaza (AUG), Gaza City, State of Palestine
| | - Sahar M Abuhajar
- Master Program of Clinical Nutrition, Al Azhar University - Gaza (AUG), Gaza City, State of Palestine
| | - Ahmed A Najim
- Department of Nursing, Faculty of Applied Medical Sciences, Al-Azhar University of Gaza (AUG), Gaza City, State of Palestine
| |
Collapse
|
157
|
Soliman AM, Das S, Mahakkanukrauh P. Inflammatory Molecular Mediators and Pathways Involved in Vascular Aging and Stroke: A Comprehensive Review. Curr Med Chem 2021; 29:5522-5542. [PMID: 34488579 DOI: 10.2174/0929867328666210901122359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/01/2021] [Accepted: 07/23/2021] [Indexed: 11/22/2022]
Abstract
There is an increase in the incidence of cardiovascular diseases with aging and it is one of the leading causes of death worldwide. The main cardiovascular pathologies include atherosclerosis, stroke, myocardial infarction, hypertension and stroke. Chronic inflammation is one of the significant contributors to the age-related vascular diseases. Therefore, it is important to understand the molecular mechanisms of the persistent inflammatory conditions occurring in the blood vessels as well as the signaling pathways involved. Herein, we performed an extant search of literature involving PubMed, ISI, WoS and Scopus databases for retrieving all relevant articles with the most recent findings illustrating the potential role of various inflammatory mediators along with their proposed activated pathways in the pathogenesis and progression of vascular aging. We also highlight the major pathways contributing to age-related vascular disorders. The outlined molecular mechanisms, pathways and mediators of vascular aging represent potential drug targets that can be utilized to inhibit and/or slow the pathogenesis and progression of vascular aging.
Collapse
Affiliation(s)
- Amro M Soliman
- Department of Biological Sciences-Physiology, Cell and Developmental Biology, University of Alberta, Edmonton, AB T6G 2R3. Canada
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, P.C. 123, Al Khoud, Muscat. Oman
| | - Pasuk Mahakkanukrauh
- Department of Anatomy & Excellence center of Osteology Research and Training, Cadaveric Surgical and Training Center, Chiang Mai University, Chiang Mai 50200. Thailand
| |
Collapse
|
158
|
Status of biomarkers for the identification of stable or vulnerable plaques in atherosclerosis. Clin Sci (Lond) 2021; 135:1981-1997. [PMID: 34414413 DOI: 10.1042/cs20210417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is a systemic inflammation of the arteries characterized by atherosclerotic plaque due to the accumulation of lipids, inflammatory cells, apoptotic cells, calcium and extracellular matrix (ECM) proteins. Stable plaques present a chronic inflammatory infiltration, whereas vulnerable plaques present an 'active' inflammation involved in the thinning of the fibrous cap that predisposes to plaque rupture. Several complex biological cellular processes lead plaques to evolve from stable to vulnerable predisposing them to rupture and thrombosis. In this review, we analyze some emerging circulating biomarkers related to inflammation, ECM and lipid infiltration, angiogenesis, metalloproteinases and microRNA (miRNA), as possible diagnostic and prognostic indicators of plaque vulnerability.
Collapse
|
159
|
Restivo V, Candiloro S, Daidone M, Norrito R, Cataldi M, Minutolo G, Caracci F, Fasano S, Ciccia F, Casuccio A, Tuttolomondo A. Systematic review and meta-analysis of cardiovascular risk in rheumatological disease: Symptomatic and non-symptomatic events in rheumatoid arthritis and systemic lupus erythematosus. Autoimmun Rev 2021; 21:102925. [PMID: 34454117 DOI: 10.1016/j.autrev.2021.102925] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 08/22/2021] [Indexed: 12/14/2022]
Abstract
Although each autoimmune disease is associated with specific tissue or organ damage, rheumatic diseases share a pro-inflammatory pattern that might increase cardiovascular risk. Retrospective and prospective studies on patients affected by systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) highlighted the concept of "accelerated atherosclerosis". Therefore, the purpose of this systematic review and meta-analysis is the assessment of symptomatic or asymptomatic cardiovascular events among patients with rheumatic diseases as RA and SLE. The literature research obtained all manuscripts published in the English language between 2015 and 2019 for a total of 2355 manuscripts. After selection through inclusion and exclusion criteria, four articles examined cardiovascular risk in RA patients, 8 in SLE patients, and 2 in RA and SLE patients. Patients with SLE had a RR of 1.98 (95% CI: 1.18-3.31) of symptomatic cardiovascular events compared to the unexposed cohort. The meta-regression analysis showed that younger patient (age per year increase β = -0.12 95%CI: -0.20, -0.4), belonging to studies conducted in continent different from America (β = -0.89; -95% CI: 1.67, -0.10), after 2000 (β = 0.87; 95% CI: 0.09, 1.65) and with a higher quality score 0.80 (95% CI: 0.31, 1.29) had a higher risk of cardiovascular events. In patients with RA, the RR of cardiovascular events was 1.55 (95% CI: 1.18-2.02). These data are helpful to implement cardiovascular preventive strategies among people suffering from rheumatologic diseases to decrease the incidence of cardiovascular events. However, these implementation needs to build a higher network between rheumatologists and primary care healthcare workers to furnish the same information to patients and monitor their preventive practice compliance.
Collapse
Affiliation(s)
- Vincenzo Restivo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy.
| | - Stefania Candiloro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Mario Daidone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Rosario Norrito
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Marco Cataldi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Giuseppa Minutolo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Francesca Caracci
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Serena Fasano
- Division of Rheumatology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesco Ciccia
- Division of Rheumatology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alessandra Casuccio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
160
|
Time Restricted Feeding Reduces Inflammation and Cortisol Response to a Firegrounds Test in Professional Firefighters. J Occup Environ Med 2021; 63:441-447. [PMID: 33928938 DOI: 10.1097/jom.0000000000002169] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Firefighters are at a heightened risk for developing cardiovascular disease. The purpose of this study was to determine if time restricted feeding (TRF) can improve the stress/inflammatory response to a simulated firegrounds test (FGT) in professional firefighters. METHODS Thirteen firefighters participated in an 8-week TRF intervention (14:10 [fasting:feeding]) protocol and completed a FGT before and after the intervention. Blood lactate, heart rate, salivary C-reactive protein (CRP), interleukin-6 (IL-6), interleukin 1-β (IL-1β), and cortisol were measured pre and post FGT. RESULTS Following TRF, the salivary cortisol response to the FGT was significantly (P < 0.05) reduced. Salivary IL-6 and IL-1β were also significantly lower, and CRP was higher following the intervention. CONCLUSIONS These findings demonstrate lower inflammation and reduced stress response to FGT following TRF and may suggest implications in terms of cardiometabolic benefits for firefighters.
Collapse
|
161
|
Batra G, Ghukasyan Lakic T, Lindbäck J, Held C, White HD, Stewart RAH, Koenig W, Cannon CP, Budaj A, Hagström E, Siegbahn A, Wallentin L. Interleukin 6 and Cardiovascular Outcomes in Patients With Chronic Kidney Disease and Chronic Coronary Syndrome. JAMA Cardiol 2021; 6:1440-1445. [PMID: 34431970 DOI: 10.1001/jamacardio.2021.3079] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Importance Inflammation promotes cardiovascular disease and anti-inflammatory treatment reduces cardiovascular events in patients with chronic coronary syndrome. Chronic kidney disease (CKD) is a risk factor for cardiovascular disease. It is unclear how inflammation mediated by interleukin 6 (IL-6) in patients with CKD is linked to cardiovascular disease. Objective To investigate associations between IL-6 and cardiovascular outcomes in patients with chronic coronary syndrome in association with kidney function. Design, Setting, and Participants This multicenter cohort study included patients enrolled at 663 centers in 39 countries with chronic coronary syndrome who were included in the Stabilization of Atherosclerotic Plaque by Initiation of Darapladib Therapy (STABILITY) trial. Patients were enrolled between December 2008 and April 2010 and were followed up for a median length of 3.7 years. Analysis in this substudy began September 2020. Exposures Exposures were IL-6 and creatinine estimated glomerular filtration rates (eGFR), which were collected at baseline. Associations between continuous and categorical levels (<2.0 ng/L vs ≥2.0 ng/L) of IL-6 and cardiovascular outcomes were tested in association with eGFR cutoffs (normal eGFR level [≥90 mL/min/1.73 m2], mildly decreased eGFR level [60-90 mL/min/1.73 m2], and moderately to severely decreased eGFR level [<60 mL/min/1.73 m2]). Main Outcomes and Measures Main outcome was major adverse cardiovascular events (MACE), a composite of cardiovascular death, myocardial infarction, and stroke. Results This substudy of the STABILITY trial included 14 611 patients with available IL-6 levels at baseline. The median (interquartile range) age was 65 (59-71) years, and 2700 (18.5%) were female. During follow-up, MACE occurred in 1459 individuals (10.0%). Higher levels of IL-6 were in continuous models independently associated with risk of MACE (P < .001) in all CKD strata. Using predefined strata, elevated IL-6 level (≥2.0 vs <2.0 ng/L) was associated with increased risk of MACE at normal kidney function (2.9% vs 1.9% events/y [hazard ratio, 1.35; 95% CI, 1.02-1.78]), mild CKD (3.3% vs 1.9% [hazard ratio, 1.57; 95% CI, 1.35-1.83]), and moderate to severe CKD (5.0% vs 2.9% [hazard ratio, 1.60; 95% CI, 1.28-1.99]). Conclusions and Relevance In patients with chronic coronary syndrome, elevated levels of IL-6 were associated with risk of MACE in all CKD strata. Thus, IL-6 and CKD stage may help when identifying patients with chronic coronary syndrome for anti-inflammatory treatment.
Collapse
Affiliation(s)
- Gorav Batra
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | | | - Johan Lindbäck
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Claes Held
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Harvey D White
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
| | - Ralph A H Stewart
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
| | - Wolfgang Koenig
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany.,Deutsches Herzzentrum München, Technische Universität München, DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | | | - Andrzej Budaj
- Department of Cardiology, Centre of Postgraduate Medical Education, Grochowski Hospital, Warsaw, Poland
| | - Emil Hagström
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Agneta Siegbahn
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
162
|
Ćurić ŽB, Masle AM, Kibel A, Selthofer-Relatić K, Stupin A, Mihaljević Z, Jukić I, Stupin M, Matić A, Kozina N, Šušnjara P, Juranić B, Kolobarić N, Šerić V, Drenjančević I. Effects of n-3 Polyunsaturated Fatty Acid-Enriched Hen Egg Consumption on the Inflammatory Biomarkers and Microvascular Function in Patients with Acute and Chronic Coronary Syndrome—A Randomized Study. BIOLOGY 2021; 10:biology10080774. [PMID: 34440006 PMCID: PMC8389665 DOI: 10.3390/biology10080774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/29/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022]
Abstract
Simple Summary There is a strong potential of n-3 polyunsaturated fatty acid (n-3 PUFA) consumption to reduce cardiovascular risk and prevent adverse outcomes in existing cardiovascular diseases. This study aimed to test the effect of n-3 PUFA supplementation in the form of enriched hen eggs on serum lipid and free fatty acid profiles, inflammatory and oxidative stress biomarkers, and microvascular reactivity in patients with acute and chronic coronary artery disease. Consumption of three n-3 PUFA-enriched hen eggs for three weeks had a favorable effect on serum free fatty acid profile (a lower n-6/n-3 PUFA ratio) and mild anti-inflammatory effects but did not significantly affect microvascular reactivity in patients with coronary artery disease. Because consumption of both regular and n-3 PUFA eggs had no negative effects on any of the measured biological and functional vascular parameters, the results of the present study indicate that eggs can be safely consumed in the daily diet of patients with coronary artery disease. Abstract This study aimed to test the effect of n-3 polyunsaturated fatty acid (PUFA)-enriched hen egg consumption on serum lipid and free fatty acid profiles, inflammatory and oxidative stress biomarkers, and microvascular reactivity in patients with coronary artery disease (CAD). Forty CAD patients participated in this study. Of those, 20 patients had acute CAD (Ac-CAD), and 20 patients had chronic CAD (Ch-CAD). The control group (N = 20) consumed three regular hen eggs/daily (249 mg n-3 PUFAs/day), and the n-3 PUFAs group (N = 20) consumed three n-3 PUFA-enriched hen eggs/daily (1053 g n-3 PUFAs/day) for 3 weeks. Serum n-3 PUFA concentration significantly increased (in all CAD patients), while LDL cholesterol and IL-6 (in Ac-CAD patients), and hsCRP and IL-1a (in all CAD patients) significantly decreased in the n-3 PUFAs group. Glutathione peroxidase (GPx) activity significantly decreased, and forearm skin microvascular reactivity in response to vascular occlusion (postocclusive reactive hyperemia (PORH)) remained unchanged in both the n-3 PUFAs and control groups in total CAD, Ac-CAD, and Ch-CAD patients. Potentially, n-3 PUFA-enriched hen eggs can change the free fatty acid profile to a more favorable lower n6/n3 ratio, and to exhibit mild anti-inflammatory effects but not to affect microvascular reactivity in CAD patients.
Collapse
Affiliation(s)
- Željka Breškić Ćurić
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Internal Medicine, General Hospital Vinkovci, HR-32100 Vinkovci, Croatia
| | - Ana Marija Masle
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Rheumatology, Clinical Immunology and Allergology, Osijek University Hospital, HR-31000 Osijek, Croatia
| | - Aleksandar Kibel
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
- Department for Cardiovascular Disease, Osijek University Hospital, HR-31000 Osijek, Croatia
| | - Kristina Selthofer-Relatić
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department for Cardiovascular Disease, Osijek University Hospital, HR-31000 Osijek, Croatia
- Department of Internal Medicine, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
| | - Ana Stupin
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
- Department of Pathophysiology, Physiology and Immunology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
- Correspondence: (A.S.); (I.D.)
| | - Zrinka Mihaljević
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
| | - Ivana Jukić
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
| | - Marko Stupin
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
- Department for Cardiovascular Disease, Osijek University Hospital, HR-31000 Osijek, Croatia
| | - Anita Matić
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
| | - Nataša Kozina
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
| | - Petar Šušnjara
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
| | - Brankica Juranić
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department for Cardiovascular Disease, Osijek University Hospital, HR-31000 Osijek, Croatia
- Department of Nursing and Palliative Medicine, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
| | - Nikolina Kolobarić
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
| | - Vatroslav Šerić
- Department of Clinical Laboratory Diagnostics, Osijek University Hospital, HR-31000 Osijek, Croatia;
| | - Ines Drenjančević
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (A.M.M.); (A.K.); (K.S.-R.); (Z.M.); (I.J.); (M.S.); (A.M.); (N.K.); (P.Š.); (B.J.); (N.K.)
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, HR-31000 Osijek, Croatia
- Correspondence: (A.S.); (I.D.)
| |
Collapse
|
163
|
Akbar N, Forteath C, Hussain MS, Reyskens K, Belch JJF, Lang CC, Mordi IR, Bhalraam U, Arthur JSC, Khan F. Mitogen and Stress-Activated Kinases 1 and 2 Mediate Endothelial Dysfunction. Int J Mol Sci 2021; 22:ijms22168655. [PMID: 34445361 PMCID: PMC8395442 DOI: 10.3390/ijms22168655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
Inflammation promotes endothelial dysfunction, but the underlying mechanisms remain poorly defined in vivo. Using translational vascular function testing in myocardial infarction patients, a situation where inflammation is prevalent, and knock-out (KO) mouse models we demonstrate a role for mitogen-activated-protein-kinases (MAPKs) in endothelial dysfunction. Myocardial infarction significantly lowers mitogen and stress kinase 1/2 (MSK1/2) expression in peripheral blood mononuclear cells and diminished endothelial function. To further understand the role of MSK1/2 in vascular function we developed in vivo animal models to assess vascular responses to vasoactive drugs using laser Doppler imaging. Genetic deficiency of MSK1/2 in mice increased plasma levels of pro-inflammatory cytokines and promoted endothelial dysfunction, through attenuated production of nitric oxide (NO), which were further exacerbated by cholesterol feeding. MSK1/2 are activated by toll-like receptors through MyD88. MyD88 KO mice showed preserved endothelial function and reduced plasma cytokine expression, despite significant hypercholesterolemia. MSK1/2 kinases interact with MAPK-activated proteins 2/3 (MAPKAP2/3), which limit cytokine synthesis. Cholesterol-fed MAPKAP2/3 KO mice showed reduced plasma cytokine expression and preservation of endothelial function. MSK1/2 plays a significant role in the development of endothelial dysfunction and may provide a novel target for intervention to reduce vascular inflammation. Activation of MSK1/2 could reduce pro-inflammatory responses and preserve endothelial vasodilator function before development of significant vascular disease.
Collapse
Affiliation(s)
- Naveed Akbar
- The Institute of Cardiovascular Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; (C.F.); (M.S.H.); (J.J.F.B.); (U.B.)
- Correspondence: (N.A.); (F.K.); Tel.: +44-13-8238-3531 (F.K.); Fax: +44-13-8263-2333 (F.K.)
| | - Calum Forteath
- The Institute of Cardiovascular Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; (C.F.); (M.S.H.); (J.J.F.B.); (U.B.)
| | - Muhammad S. Hussain
- The Institute of Cardiovascular Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; (C.F.); (M.S.H.); (J.J.F.B.); (U.B.)
| | - Kathleen Reyskens
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK; (K.R.); (J.S.C.A.)
| | - Jill J. F. Belch
- The Institute of Cardiovascular Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; (C.F.); (M.S.H.); (J.J.F.B.); (U.B.)
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; (C.C.L.); (I.R.M.)
| | - Ify R. Mordi
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; (C.C.L.); (I.R.M.)
| | - U Bhalraam
- The Institute of Cardiovascular Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; (C.F.); (M.S.H.); (J.J.F.B.); (U.B.)
| | - J. Simon C. Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK; (K.R.); (J.S.C.A.)
| | - Faisel Khan
- The Institute of Cardiovascular Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; (C.F.); (M.S.H.); (J.J.F.B.); (U.B.)
- Correspondence: (N.A.); (F.K.); Tel.: +44-13-8238-3531 (F.K.); Fax: +44-13-8263-2333 (F.K.)
| |
Collapse
|
164
|
Chen E, Debrosse R, Ham PJ, Hoffer LC, Leigh AKK, Destin M. Effects of social support in an academic context on low-grade inflammation in high school students. J Behav Med 2021; 44:803-810. [PMID: 34363145 DOI: 10.1007/s10865-021-00241-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 06/22/2021] [Indexed: 01/03/2023]
Abstract
Bolstering academic motivation is a high priority in school settings, but some evidence suggests this could take a toll on students' physical health. To address this, this study compared the effects of an experimental manipulation of academic motivation alone (AM) to academic motivation enhanced with social support (SS + AM) on markers of inflammation in a sample of 80 high school 9th graders. Outcomes included low-grade inflammation: C-reactive protein (CRP) and interleukin-6 (IL-6); a motivation measure; and grade point average (GPA), taken at baseline and follow-up (beginning and end of school year, respectively). Students in the SS + AM condition had lower levels of inflammation at follow-up (covarying baseline levels) compared to those in the AM condition. The two groups were equivalent on motivation and GPA at follow-up. This preliminary study suggests that incorporating social support into academic motivation programs has the potential to benefit inflammatory markers in young people while allowing them to maintain positive academic outcomes.
Collapse
Affiliation(s)
- Edith Chen
- Northwestern University, 2029 Sheridan Road, Evanston, IL, 60208, USA. .,Department of Psychology, Northwestern University, Evanston, IL, USA. .,Institute for Policy Research, Northwestern University, Evanston, IL, USA.
| | - Régine Debrosse
- School of Social Work, McGill University, Montreal, QC, Canada
| | - Paula J Ham
- University of Illinois at Chicago, Chicago, IL, USA
| | - Lauren C Hoffer
- Northwestern University, 2029 Sheridan Road, Evanston, IL, 60208, USA.,Department of Psychology, Northwestern University, Evanston, IL, USA.,Institute for Policy Research, Northwestern University, Evanston, IL, USA
| | - Adam K K Leigh
- Northwestern University, 2029 Sheridan Road, Evanston, IL, 60208, USA.,Department of Psychology, Northwestern University, Evanston, IL, USA.,Institute for Policy Research, Northwestern University, Evanston, IL, USA
| | - Mesmin Destin
- Northwestern University, 2029 Sheridan Road, Evanston, IL, 60208, USA.,Department of Psychology, Northwestern University, Evanston, IL, USA.,School of Education and Social Policy, Northwestern University Evanston, Evanston, IL, USA.,Institute for Policy Research, Northwestern University, Evanston, IL, USA
| |
Collapse
|
165
|
Ahmad Ibrahim O, Ahmad AB, Nigm DA, Hussien AN, Mohammad Ibrahim WH. Subclinical atherosclerotic predictive value of inflammatory markers in thalassemia intermedia patients. Expert Rev Hematol 2021; 14:669-677. [PMID: 34296962 DOI: 10.1080/17474086.2021.1959316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND A high incidence of thromboembolic events is observed in thalassemia patients. This study investigated the relationship between carotid intima-media thickness (CIMT) and lipid profile, iron metabolic indices (IMI), and inflammatory markers in β-thalassemia intermedia (β- TI) patients. PATIENTS AND METHODS Forty-five β-TI patients at Assiut University Hospital and 34 healthy individuals were enrolled in the study. We measured Lipid profile, IMI, high sensitive CRP (Hs-CRP), and interleukin-6 (IL-6) and compared the results between both groups. We used CIMT measurement as a marker for subclinical atherosclerosis. We used both univariate and multivariate analyses to test relations and independent predictors of CIMT. RESULTS β-TI patients had higher CIMT (P = 0.000). CIMT was positively correlated with absolute neutrophil count (ANC) (r = 0.320, p = 0.032), ferritin (r = 0.544, p = 0.000), Hs-CRP (r = 0.603, p = 0.000), and IL-6 (r = 0.520, p = 0.000). Hs-CRP was an independent predictor of CIMT (p = 0.000). Hs-CRP cut off value of 60.4 ug/dl has sensitivity of 63.3% and specificity of 93.3% in predicting premature atherosclerosis. CONCLUSION β-TI patients had higher CIMT despite the protective lipid profile. Hs-CRP was an independent predictor of CIMT.
Collapse
Affiliation(s)
- Osama Ahmad Ibrahim
- Department of Internal Medicine, Hematology Unit, Assiut University Hospital, Assiut, Egypt
| | - Ahmad B Ahmad
- Department of Internal Medicine, Cardiology and Critical Care Unit, Assiut University Hospital, Assiut, Egypt
| | - Dalia Ahmad Nigm
- Department of Clinical Pathology, Assiut University Hospital, Assiut, Egypt
| | - Asmaa Nady Hussien
- Department of Internal Medicine, Hematology Unit, Assiut University Hospital, Assiut, Egypt
| | | |
Collapse
|
166
|
Degroote C, von Känel R, Thomas L, Zuccarella-Hackl C, Pruessner JC, Wiest R, Wirtz PH. Acute Stress-Induced Blood Lipid Reactivity in Hypertensive and Normotensive Men and Prospective Associations with Future Cardiovascular Risk. J Clin Med 2021; 10:jcm10153400. [PMID: 34362177 PMCID: PMC8347098 DOI: 10.3390/jcm10153400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Hyperreactivity to stress may be one explanation for the increased risk of cardiovascular disease (CVD) in individuals with essential hypertension. We investigated blood lipid reactivity to the Montreal Imaging Stress Task (MIST), a psychosocial stressor, in hypertensive and normotensive men and tested for prospective associations with biological risk factors. Fifty-six otherwise healthy and medication-free hypertensive and normotensive men underwent the MIST. We repeatedly measured cortisol and blood lipid profiles (total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides (TG)) immediately before and up to 1 h after stress. Lipid levels were corrected for stress hemoconcentration. Thirty-five participants completed follow-up assessment 2.9 ± 0.12 (SEM) years later. CVD risk was assessed by prospective changes in TC/HDL-C ratio, IL-6, D-dimer, and HbA1c from baseline to follow-up. The MIST induced significant changes in all parameters except TC (p-values ≤ 0.043). Compared with normotensives, hypertensives had higher TC/HDL-C-ratio and TG (p-values ≤ 0.049) stress responses. Blood lipid stress reactivity predicted future cardiovascular risk (p = 0.036) with increases in HbA1c (ß = 0.34, p = 0.046), IL-6 (ß = 0.31, p = 0.075), and D-dimer (ß = 0.33, p = 0.050). Our results suggest that the greater blood lipid reactivity to psychosocial stress in hypertensives, the greater their future biological CVD risk. This points to lipid stress reactivity as a potential mechanism through which stress might increase CVD risk in essential hypertension.
Collapse
Affiliation(s)
- Cathy Degroote
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany; (C.D.); (L.T.)
| | - Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (R.v.K.); (C.Z.-H.)
| | - Livia Thomas
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany; (C.D.); (L.T.)
| | - Claudia Zuccarella-Hackl
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (R.v.K.); (C.Z.-H.)
| | - Jens C. Pruessner
- Clinical Neuropsychology, University of Konstanz, 78457 Konstanz, Germany;
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, 78457 Konstanz, Germany
| | - Roland Wiest
- Support Center of Advanced Neuroimaging, Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, University of Bern, 3010 Bern, Switzerland;
| | - Petra H. Wirtz
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany; (C.D.); (L.T.)
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, 78457 Konstanz, Germany
- Correspondence: ; Tel.: +49-7531-88-3742
| |
Collapse
|
167
|
Fowler J, Tsui MTK, Chavez J, Khan S, Ahmed H, Smith L, Jia Z. Methyl mercury triggers endothelial leukocyte adhesion and increases expression of cell adhesion molecules and chemokines. Exp Biol Med (Maywood) 2021; 246:2522-2532. [PMID: 34308659 DOI: 10.1177/15353702211033812] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of morbidity, mortality, and health care costs in the USA, and around the world. Among the various risk factors of cardiovascular disease, environmental and dietary exposures to methyl mercury, a highly toxic metal traditionally labeled as a neurotoxin, have been epidemiologically linked to human cardiovascular disease development. However, its role in development and promotion of atherosclerosis, an initial step in more immediately life-threatening cardiovascular diseases, remains unclear. This study was conducted to examine the role that methyl mercury plays in the adhesion of monocytes to human microvascular endothelial cells (HMEC-1), and the underlying mechanisms. Methyl mercury treatment significantly induced the adhesion of monocyte to HMEC-1 endothelial cells, a critical step in atherosclerosis, while also upregulating the expression of proinflammatory cytokines interleukin-6, interleukin-8. Further, methyl mercury treatment also upregulated the chemotactic cytokine monocyte chemoattractant protein-1 and intercellular adhesion molecule-1. These molecules are imperative for the firm adhesion of leukocytes to endothelial cells. Additionally, our results further demonstrated that methyl mercury stimulated a significant increase in NF-κB activation. These findings suggest that NF-κB signaling pathway activation by methyl mercury is an important factor in the binding of monocytes to endothelial cells. Finally, by using flow cytometric analysis, methyl mercury treatment caused a significant increase in necrotic cell death only at higher concentrations without initiating apoptosis. This study provides new insights into the molecular actions of methyl mercury that can lead to endothelial dysfunction, inflammation, and subsequent atherosclerotic development.
Collapse
Affiliation(s)
- Joshua Fowler
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Martin Tsz-Ki Tsui
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA.,School of Life Sciences, Chinese University of Hong Kong, Hong Kong SAR 00000, China
| | - Jessica Chavez
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Safeera Khan
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Hassan Ahmed
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Lena Smith
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27410, USA
| |
Collapse
|
168
|
Effects of atorvastatin doses on serum level of procalcitonin and predictors for major adverse cardiovascular events in patients with acute myocardial infarction: a pilot study and post hoc analysis. Coron Artery Dis 2021; 31:e87-e93. [PMID: 34292180 DOI: 10.1097/mca.0000000000001084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Inflammation plays an important role in acute myocardial infarction (AMI). Procalcitonin levels rise in response to proinflammatory stimuli. This study aimed to investigate the effects of different doses of atorvastatin on the serum inflammatory profiles, especially procalcitonin and major adverse cardiovascular events (MACEs) in patients with AMI during hospitalization. METHODS The patients who were admitted to the Coronary Care Unit of The Third Medical Center of PLA General Hospital (Beijing, China) between January 2015 and December 2015 with a diagnosis of AMI were enrolled, and randomized to atorvastatin 20 mg/day postoperatively (20-mg group), 40 mg/day postoperatively (40-mg group) and 80 mg preoperatively+40 mg/day postoperatively (80/40-mg group). Serum procalcitonin and high-sensitivity C-reactive protein (hs-CRP) were evaluated before and at 1 and 3 days after percutaneous coronary intervention (PCI). RESULTS A total of 112 patients with AMI (23 women and 89 men) were prospectively eligible for the study. There were no significant differences in most clinical data among the three groups. The 80/40-mg group showed significantly reduced serum procalcitonin levels at 1 and 3 days after PCI (P < 0.001) and reduced hs-CRP levels at 3 days P = 0.001) compared with 20-mg and 40-mg groups. Serum procalcitonin (OR, 4.593; 95% CI, 1.476-8.387; P = 0.005), hs-CRP (OR, 1.149; 95% CI, 1.012-1.338; P = 0.018), highly sensitive cardiac troponin T (OR, 1.255; 95% CI, 1.004-1.569, P = 0.009) and Gensini score (OR, 1.022; 95% CI, 1.045-1.062; P = 0.013) were independently associated with MACEs during hospitalization. CONCLUSION The use of atorvastatin 80 mg before and 40 mg/day after PCI in patients with AMI can effectively reduce serum inflammatory factors. procalcitonin and hs-CRP were independently associated with in-hospital MACEs.
Collapse
|
169
|
Deroissart J, Porsch F, Koller T, Binder CJ. Anti-inflammatory and Immunomodulatory Therapies in Atherosclerosis. Handb Exp Pharmacol 2021; 270:359-404. [PMID: 34251531 DOI: 10.1007/164_2021_505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hypercholesterolemia is a major risk factor in atherosclerosis development and lipid-lowering drugs (i.e., statins) remain the treatment of choice. Despite effective reduction of LDL cholesterol in patients, a residual cardiovascular risk persists in some individuals, highlighting the need for further therapeutic intervention. Recently, the CANTOS trial paved the way toward the development of specific therapies targeting inflammation, a key feature in atherosclerosis progression. The pre-existence of multiple drugs modulating both innate and adaptive immune responses has significantly accelerated the number of translational studies applying these drugs to atherosclerosis. Additional preclinical research has led to the discovery of new therapeutic targets, offering promising perspectives for the treatment and prevention of atherosclerosis. Currently, both drugs with selective targeting and broad unspecific anti-inflammatory effects have been tested. In this chapter, we aim to give an overview of current advances in immunomodulatory treatment approaches for atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Justine Deroissart
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Koller
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
170
|
Relf RL, Lee BJ, Eichhorn G, Flint MS, Beale L, Maxwell N. Thermoregulation is not impaired in breast cancer survivors during moderate-intensity exercise performed in warm and hot environments. Physiol Rep 2021; 9:e14968. [PMID: 34291605 PMCID: PMC8295682 DOI: 10.14814/phy2.14968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 11/30/2022] Open
Abstract
This study aimed to assess how female breast cancer survivors (BCS) respond physiologically, hematologically, and perceptually to exercise under heat stress compared to females with no history of breast cancer (CON). Twenty-one females (9 BCS and 12 CON [age; 54 ± 7 years, stature; 167 ± 6 cm, body mass; 68.1 ± 7.62 kg, and body fat; 30.9 ± 3.8%]) completed a warm (25℃, 50% relative humidity, RH) and hot (35℃, 50%RH) trial in a repeated-measures crossover design. Trials consisted of 30 min of rest, 30 min of walking at 4 metabolic equivalents, and a 6-minute walk test (6MWT). Physiological measurements (core temperature (Tre ), skin temperature (Tskin ), heart rate (HR), and sweat analysis) and perceptual rating scales (ratings of perceived exertion, thermal sensation [whole body and localized], and thermal comfort) were taken at 5- and 10-min intervals throughout, respectively. Venous blood samples were taken before and after to assess; IL-6, IL-10, CRP, IFN-γ, and TGF-β1 . All physiological markers were higher during the 35 versus 25℃ trial; Tre (~0.25℃, p = 0.002), Tskin (~3.8℃, p < 0.001), HR (~12 beats·min-1 , p = 0.023), and whole-body sweat rate (~0.4 L·hr-1 , p < 0.001), with no difference observed between groups in either condition (p > 0.05). Both groups covered a greater 6MWT distance in 25 versus 35℃ (by ~200 m; p = 0.003). Nevertheless, the control group covered more distance than BCS, regardless of environmental temperature (by ~400 m, p = 0.03). Thermoregulation was not disadvantaged in BCS compared to controls during moderate-intensity exercise under heat stress. However, self-paced exercise performance was reduced for BCS regardless of environmental temperature.
Collapse
Affiliation(s)
- Rebecca L. Relf
- Environmental Extremes LaboratoryUniversity of BrightonEastbourneUK
| | - Ben J. Lee
- Occupational and Environmental Physiology GroupCoventry UniversityCoventryUK
| | - Gregor Eichhorn
- Environmental Extremes LaboratoryUniversity of BrightonEastbourneUK
| | - Melanie S. Flint
- Cancer Stress LaboratoryUniversity of BrightonMoulsecoombUK
- Centre for Stress and Age‐Related DiseaseBrightonEast SussexUK
| | - Louisa Beale
- Environmental Extremes LaboratoryUniversity of BrightonEastbourneUK
| | - Neil Maxwell
- Environmental Extremes LaboratoryUniversity of BrightonEastbourneUK
| |
Collapse
|
171
|
Ekholm M, Kahan T. The Impact of the Renin-Angiotensin-Aldosterone System on Inflammation, Coagulation, and Atherothrombotic Complications, and to Aggravated COVID-19. Front Pharmacol 2021; 12:640185. [PMID: 34220496 PMCID: PMC8245685 DOI: 10.3389/fphar.2021.640185] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is considered a disease caused by a chronic inflammation, associated with endothelial dysfunction, and several mediators of inflammation are up-regulated in subjects with atherosclerotic disease. Healthy, intact endothelium exhibits an antithrombotic, protective surface between the vascular lumen and vascular smooth muscle cells in the vessel wall. Oxidative stress is an imbalance between anti- and prooxidants, with a subsequent increase of reactive oxygen species, leading to tissue damage. The renin-angiotensin-aldosterone system is of vital importance in the pathobiology of vascular disease. Convincing data indicate that angiotensin II accelerates hypertension and augments the production of reactive oxygen species. This leads to the generation of a proinflammatory phenotype in human endothelial and vascular smooth muscle cells by the up-regulation of adhesion molecules, chemokines and cytokines. In addition, angiotensin II also seems to increase thrombin generation, possibly via a direct impact on tissue factor. However, the mechanism of cross-talk between inflammation and haemostasis can also contribute to prothrombotic states in inflammatory environments. Thus, blocking of the renin-angiotensin-aldosterone system might be an approach to reduce both inflammatory and thrombotic complications in high-risk patients. During COVID-19, the renin-angiotensin-aldosterone system may be activated. The levels of angiotensin II could contribute to the ongoing inflammation, which might result in a cytokine storm, a complication that significantly impairs prognosis. At the outbreak of COVID-19 concerns were raised about the use of angiotensin converting enzyme inhibitors and angiotensin receptor blocker drugs in patients with COVID-19 and hypertension or other cardiovascular comorbidities. However, the present evidence is in favor of continuing to use of these drugs. Based on experimental evidence, blocking the renin-angiotensin-aldosterone system might even exert a potentially protective influence in the setting of COVID-19.
Collapse
Affiliation(s)
- M Ekholm
- Karolinska Institutet, Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Stockholm, Sweden
| | - T Kahan
- Karolinska Institutet, Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Stockholm, Sweden
| |
Collapse
|
172
|
Bleil ME, Spieker SJ, Booth-LaForce C. Targeting Parenting Quality to Reduce Early Life Adversity Impacts on Lifespan Cardiometabolic Risk. Front Psychol 2021; 12:678946. [PMID: 34149571 PMCID: PMC8211431 DOI: 10.3389/fpsyg.2021.678946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/12/2021] [Indexed: 11/16/2022] Open
Abstract
Mounting evidence that early life adversity (ELA) exposures confer risk for cardiometabolic disease over the lifespan motivated this narrative review to examine parenting quality as a potential intervention target to reduce ELA exposures or mitigate their impact as a way of reducing or preventing cardiometabolic disease. We describe findings from the limited number of family-based intervention studies in ELA-exposed children that have tested parenting impacts on cardiometabolic health outcomes. We then describe the implications of this work and make recommendations for future research that will move this field forward.
Collapse
Affiliation(s)
- Maria E. Bleil
- Child, Family, and Population Health Nursing, University of Washington, Seattle, WA, United States
| | | | | |
Collapse
|
173
|
Ridker PM, Devalaraja M, Baeres FMM, Engelmann MDM, Hovingh GK, Ivkovic M, Lo L, Kling D, Pergola P, Raj D, Libby P, Davidson M. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 2021; 397:2060-2069. [PMID: 34015342 DOI: 10.1016/s0140-6736(21)00520-1] [Citation(s) in RCA: 372] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND IL-6 has emerged as a pivotal factor in atherothrombosis. Yet, the safety and efficacy of IL-6 inhibition among individuals at high atherosclerotic risk but without a systemic inflammatory disorder is unknown. We therefore addressed whether ziltivekimab, a fully human monoclonal antibody directed against the IL-6 ligand, safely and effectively reduces biomarkers of inflammation and thrombosis among patients with high cardiovascular risk. We focused on individuals with elevated high-sensitivity CRP and chronic kidney disease, a group with substantial unmet clinical need in whom previous studies in inflammation inhibition have shown efficacy for cardiovascular event reduction. METHODS RESCUE is a randomised, double-blind, phase 2 trial done at 40 clinical sites in the USA. Inclusion criteria were age 18 years or older, moderate to severe chronic kidney disease, and high-sensitivity CRP of at least 2 mg/L. Participants were randomly allocated (1:1:1:1) to subcutaneous administration of placebo or ziltivekimab 7·5 mg, 15 mg, or 30 mg every 4 weeks up to 24 weeks. The primary outcome was percentage change from baseline in high-sensitivity CRP after 12 weeks of treatment with ziltivekimab compared with placebo, with additional biomarker and safety data collected over 24 weeks of treatment. Primary analyses were done in the intention-to-treat population. Safety was assessed in all patients who received at least one dose of assigned treatment. The trial is registered with ClinicalTrials.gov, NCT03926117. FINDINGS Between June 17, 2019, and Jan 14, 2020, 264 participants were enrolled into the trial, of whom 66 were randomly assigned to each of the four treatment groups. At 12 weeks after randomisation, median high-sensitivity CRP levels were reduced by 77% for the 7·5 mg group, 88% for the 15 mg group, and 92% for the 30 mg group compared with 4% for the placebo group. As such, the median pairwise differences in percentage change in high-sensitivity CRP between the ziltivekimab and placebo groups, after aligning for strata, were -66·2% for the 7·5 mg group, -77·7% for the 15 mg group, and -87·8% for the 30 mg group (all p<0·0001). Effects were stable over the 24-week treatment period. Dose-dependent reductions were also observed for fibrinogen, serum amyloid A, haptoglobin, secretory phospholipase A2, and lipoprotein(a). Ziltivekimab was well tolerated, did not affect the total cholesterol to HDL cholesterol ratio, and there were no serious injection-site reactions, sustained grade 3 or 4 neutropenia or thrombocytopenia. INTERPRETATION Ziltivekimab markedly reduced biomarkers of inflammation and thrombosis relevant to atherosclerosis. On the basis of these data, a large-scale cardiovascular outcomes trial will investigate the effect of ziltivekimab in patients with chronic kidney disease, increased high-sensitivity CRP, and established cardiovascular disease. FUNDING Novo Nordisk.
Collapse
Affiliation(s)
- Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | | - Larry Lo
- Corvidia Therapeutics, Waltham, MA, USA
| | | | | | - Dominic Raj
- Division of Kidney Diseases and Hypertension, George Washington University, Washington, DC, USA
| | - Peter Libby
- the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
174
|
Ji E, Lee S. Antibody-Based Therapeutics for Atherosclerosis and Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms22115770. [PMID: 34071276 PMCID: PMC8199089 DOI: 10.3390/ijms22115770] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide, and its prevalence is increasing due to the aging of societies. Atherosclerosis, a type of chronic inflammatory disease that occurs in arteries, is considered to be the main cause of cardiovascular diseases such as ischemic heart disease or stroke. In addition, the inflammatory response caused by atherosclerosis confers a significant effect on chronic inflammatory diseases such as psoriasis and rheumatic arthritis. Here, we review the mechanism of action of the main causes of atherosclerosis such as plasma LDL level and inflammation; furthermore, we review the recent findings on the preclinical and clinical effects of antibodies that reduce the LDL level and those that neutralize the cytokines involved in inflammation. The apolipoprotein B autoantibody and anti-PCSK9 antibody reduced the level of LDL and plaques in animal studies, but failed to significantly reduce carotid inflammation plaques in clinical trials. The monoclonal antibodies against PCSK9 (alirocumab, evolocumab), which are used as a treatment for hyperlipidemia, lowered cholesterol levels and the incidence of cardiovascular diseases. Antibodies that neutralize inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-17, and IL-12/23) have shown promising but contradictory results and thus warrant further research.
Collapse
Affiliation(s)
- Eunhye Ji
- Division of Cardiology, Heart Institute, Asan Medical Center, Seoul 05505, Korea;
| | - Sahmin Lee
- Division of Cardiology, Heart Institute, Asan Medical Center, Seoul 05505, Korea;
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul 05505, Korea
- Correspondence:
| |
Collapse
|
175
|
Ridker PM, MacFadyen JG, Glynn RJ, Bradwin G, Hasan AA, Rifai N. Comparison of interleukin-6, C-reactive protein, and low-density lipoprotein cholesterol as biomarkers of residual risk in contemporary practice: secondary analyses from the Cardiovascular Inflammation Reduction Trial. Eur Heart J 2021; 41:2952-2961. [PMID: 32221587 DOI: 10.1093/eurheartj/ehaa160] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/20/2020] [Accepted: 03/02/2020] [Indexed: 12/24/2022] Open
Abstract
AIMS In epidemiologic cohorts initiated >30 years ago, inflammatory biomarkers, such as interleukin-6 (IL-6) and high-sensitivity C-reactive protein (hsCRP) were shown to independently predict future cardiovascular events with a magnitude of effect comparable to that of low-density lipoprotein cholesterol (LDLC). Whether aggressive contemporary therapy for atherosclerosis has altered these relationships is unknown yet has major implications for future drug development. METHODS AND RESULTS Interleukin-6, hsCRP, and LDLC were measured at baseline in up to 4168 North American patients enrolled in the contemporary Cardiovascular Inflammation Reduction Trial with prior myocardial infarction or multivessel coronary disease who additionally had diabetes or metabolic syndrome and were followed for a period of up to 5 years for incident major recurrent cardiovascular events and all-cause mortality. Three-quarters of the cohort were previously revascularized and the great majority was taking statins, angiotensin blocking agents, beta-blockers, and antithrombotic agents. Participants were randomly allocated to low-dose methotrexate 15 mg weekly or to placebo. Randomized use of methotrexate had no effect on event rates nor plasma levels of IL-6, hsCRP, or LDL over time. Yet, baseline levels of IL-6, hsCRP, and LDLC were all predictors of major recurrent cardiovascular events; adjusted hazard ratios [HR; 95% confidence interval (CI)] for the lowest to highest baseline quartiles of IL-6 were 1.0 (referent), 1.66 (1.18-2.35), 1.92 (1.36-2.70), and 2.11 (1.49-2.99; P < 0.0001), while adjusted HRs for increasing quartiles of hsCRP were 1.0 (referent), 1.28 (0.92-1.79), 1.73 (1.25-2.38), and 1.79 (1.28-2.50; P < 0.0001) and adjusted HRs for increasing quartiles of LDLC were 1.0 (referent), 1.12 (0.78-1.62), 1.25 (0.87-1.79), and 2.38 (1.72-3.30; P < 0.0001). Effect estimates were not statistically different in these analyses for comparisons between IL-6, hsCRP, or LDLC, although IL-6 was the strongest predictor of all-cause mortality. The highest absolute risks were observed among those with elevated levels of both cholesterol and inflammation [HR 6.4 (95% CI 2.9-14.1) for those in the top quartiles of baseline IL-6 and LDLC, HR 4.9 (95% CI 2.6-9.4) for those in the top quartiles of baseline hsCRP and LDLC, both P < 0.0001]. CONCLUSION Despite aggressive contemporary secondary prevention efforts, the relationships between inflammation, cholesterol, and cardiovascular risk are largely unchanged from those described two decades ago. These data are consistent with the hypothesis that future treatments for atherosclerosis may require a combination of inflammation inhibition and additional cholesterol reduction. CLINICAL TRIAL ClinicalTrials.gov NCT01594333.
Collapse
Affiliation(s)
- Paul M Ridker
- Division of Preventive Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Brigham and Women's Hospital, Boston, MA 02215, USA.,Cardiovascular Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Jean G MacFadyen
- Division of Preventive Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Robert J Glynn
- Division of Preventive Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Gary Bradwin
- Department of Laboratory Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Ahmed A Hasan
- National Heart Lung and Blood Institute, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Nader Rifai
- Department of Laboratory Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
176
|
Ridker PM, MacFadyen JG, Thuren T, Libby P. Residual inflammatory risk associated with interleukin-18 and interleukin-6 after successful interleukin-1β inhibition with canakinumab: further rationale for the development of targeted anti-cytokine therapies for the treatment of atherothrombosis. Eur Heart J 2021; 41:2153-2163. [PMID: 31504417 DOI: 10.1093/eurheartj/ehz542] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/05/2019] [Accepted: 07/26/2019] [Indexed: 11/15/2022] Open
Abstract
AIMS The Canakinumab Antiinflammatory Thrombosis Outcomes Study (CANTOS) established that targeting inflammation with interleukin-1β (IL-1β) inhibition can significantly reduce cardiovascular (CV) event rates in the absence of any beneficial effects on cholesterol. Yet, CANTOS participants treated with both high-intensity statins and canakinumab remain at considerable risk for recurrent CV events. Both interleukin-18 (IL-18, which like IL-1β requires the NLRP3 inflammasome for activation) and interleukin-6 (IL-6, a pro-inflammatory cytokine downstream of IL-1) may contribute to the recurrent events that occur even on canakinumab therapy, and thus represent novel targets for treating atherothrombosis. METHODS AND RESULTS Plasma samples from 4848 stable post-myocardial infarction patients who were assigned to active IL-1β inhibition or placebo within CANTOS underwent measurement of IL-18 and IL-6 both before and after initiation of canakinumab using validated ELISA. All participants were followed over a median 3.7-year period (maximum 5 years) for recurrent major adverse cardiovascular events (MACE) and for all-cause mortality. Compared to placebo, canakinumab significantly reduced IL-6 levels in a dose-dependent manner yielding placebo-subtracted median percent reductions in IL-6 at 3 months of 24.8%, 36.3%, and 43.2% for the 50, 150, and 300 mg doses, respectively (all P-values <0.001). By contrast, no dose of canakinumab significantly altered IL-18 levels measured at 3 months (all effects <1%, all P-values > 0.05). Yet, despite these differential plasma effects, either baseline and on-treatment levels of IL-18 or IL-6 associated with rates of future CV events. For example, for MACE, each tertile increase in IL-18 measured 3 months after canakinumab initiation associated with a 15% increase in risk [95% confidence interval (CI) 3-29%, P = 0.016], while each tertile increase in IL-6 measured 3 months after canakinumab initiation associated with a 42% increase in risk (95% CI 26-59%, P < 0.0001). Similar effects were observed for MACE-plus, CV death, all-cause mortality, and the for the combination endpoint of all vascular events inclusive of revascularization procedures and hospitalization for congestive heart failure. In baseline as well as on-treatment analyses, risks were highest among those with the highest levels of both IL-18 and IL-6. CONCLUSION There remains substantial residual inflammatory risk related to both IL-18 and IL-6 after IL-1β inhibition with canakinumab These data support further pharmacologic development of therapies for atherothrombosis that target IL-18 or IL-6 signalling, or that can simultaneously inhibit both IL-1β and IL-18 (such as NLRP3 inflammasome inhibitors). CLINICAL TRIAL REGISTRATION ClinicalTrials.gov NCT01327846.
Collapse
Affiliation(s)
- Paul M Ridker
- Department of Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02215, USA.,Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Jean G MacFadyen
- Department of Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02215, USA
| | - Tom Thuren
- Novartis Pharmaceutical Corporation, One Health Plaza, East Hanover, NJ 07936, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
177
|
Kerola AM, Rollefstad S, Semb AG. Atherosclerotic Cardiovascular Disease in Rheumatoid Arthritis: Impact of Inflammation and Antirheumatic Treatment. Eur Cardiol 2021; 16:e18. [PMID: 34040652 PMCID: PMC8145075 DOI: 10.15420/ecr.2020.44] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
Patients with rheumatoid arthritis (RA) are at approximately 1.5-fold risk of atherosclerotic cardiovascular disease (CVD) compared with the general population, a phenomenon resulting from combined effects of traditional CVD risk factors and systemic inflammation. Rheumatoid synovitis and unstable atherosclerotic plaques share common inflammatory mechanisms, such as expression of proinflammatory cytokines interleukin (IL)-1, tumour necrosis factor (TNF)-α and IL-6. RA patients are undertreated in terms of CVD prevention, and structured CVD prevention programmes are warranted. Alongside management of traditional risk factors, suppressing systemic inflammation with antirheumatic medication is fundamental for the reduction of CVD risk among this high-risk patient group. Many antirheumatic drugs, especially methotrexate, TNF-α-inhibitors and IL-6-inhibitors are associated with reduced risk of CVD in observational studies among RA patients, but randomised controlled trials with hard CVD endpoints are lacking. In patients without rheumatic disease, anti-inflammatory therapies targeting nucleotide-binding oligomerisation domain, leucine-rich repeat and pyrin domain-containing protein 3 inflammasome and the IL-1/IL-6 pathway arise as potential therapies after an atherosclerotic CVD event.
Collapse
Affiliation(s)
- Anne Mirjam Kerola
- Preventive Cardio-Rheuma Clinic, Division of Rheumatology and Research, Diakonhjemmet Hospital Oslo, Norway.,Department of Rheumatology, Päijät-Häme Joint Authority for Health and Wellbeing Lahti, Finland
| | - Silvia Rollefstad
- Preventive Cardio-Rheuma Clinic, Division of Rheumatology and Research, Diakonhjemmet Hospital Oslo, Norway
| | - Anne Grete Semb
- Preventive Cardio-Rheuma Clinic, Division of Rheumatology and Research, Diakonhjemmet Hospital Oslo, Norway
| |
Collapse
|
178
|
Schunk SJ, Kleber ME, März W, Pang S, Zewinger S, Triem S, Ege P, Reichert MC, Krawczyk M, Weber SN, Jaumann I, Schmit D, Sarakpi T, Wagenpfeil S, Kramann R, Boerwinkle E, Ballantyne CM, Grove ML, Tragante V, Pilbrow AP, Richards AM, Cameron VA, Doughty RN, Dubé MP, Tardif JC, Feroz-Zada Y, Sun M, Liu C, Ko YA, Quyyumi AA, Hartiala JA, Tang WHW, Hazen SL, Allayee H, McDonough CW, Gong Y, Cooper-DeHoff RM, Johnson JA, Scholz M, Teren A, Burkhardt R, Martinsson A, Smith JG, Wallentin L, James SK, Eriksson N, White H, Held C, Waterworth D, Trompet S, Jukema JW, Ford I, Stott DJ, Sattar N, Cresci S, Spertus JA, Campbell H, Tierling S, Walter J, Ampofo E, Niemeyer BA, Lipp P, Schunkert H, Böhm M, Koenig W, Fliser D, Laufs U, Speer T, eQTLGen consortium, BIOS consortium. Genetically determined NLRP3 inflammasome activation associates with systemic inflammation and cardiovascular mortality. Eur Heart J 2021; 42:1742-1756. [PMID: 33748830 PMCID: PMC8244638 DOI: 10.1093/eurheartj/ehab107] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/19/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
AIMS Inflammation plays an important role in cardiovascular disease (CVD) development. The NOD-like receptor protein-3 (NLRP3) inflammasome contributes to the development of atherosclerosis in animal models. Components of the NLRP3 inflammasome pathway such as interleukin-1β can therapeutically be targeted. Associations of genetically determined inflammasome-mediated systemic inflammation with CVD and mortality in humans are unknown. METHODS AND RESULTS We explored the association of genetic NLRP3 variants with prevalent CVD and cardiovascular mortality in 538 167 subjects on the individual participant level in an explorative gene-centric approach without performing multiple testing. Functional relevance of single-nucleotide polymorphisms on NLRP3 inflammasome activation has been evaluated in monocyte-enriched peripheral blood mononuclear cells (PBMCs). Genetic analyses identified the highly prevalent (minor allele frequency 39.9%) intronic NLRP3 variant rs10754555 to affect NLRP3 gene expression. rs10754555 carriers showed significantly higher C-reactive protein and serum amyloid A plasma levels. Carriers of the G allele showed higher NLRP3 inflammasome activation in isolated human PBMCs. In carriers of the rs10754555 variant, the prevalence of coronary artery disease was significantly higher as compared to non-carriers with a significant interaction between rs10754555 and age. Importantly, rs10754555 carriers had significantly higher risk for cardiovascular mortality during follow-up. Inflammasome inducers (e.g. urate, triglycerides, apolipoprotein C3) modulated the association between rs10754555 and mortality. CONCLUSION The NLRP3 intronic variant rs10754555 is associated with increased systemic inflammation, inflammasome activation, prevalent coronary artery disease, and mortality. This study provides evidence for a substantial role of genetically driven systemic inflammation in CVD and highlights the NLRP3 inflammasome as a therapeutic target.
Collapse
Affiliation(s)
- Stefan J Schunk
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - Marcus E Kleber
- Vth Department of Medicine, University Heidelberg, Mannheim Medical Faculty, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Harrlachweg 1, 68163 Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine, University Heidelberg, Mannheim Medical Faculty, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Clinical Institute of Medical and Laboratory Diagnostics, Medical University Graz, Auenbruggerpl. 2, 8036 Graz, Austria
- Synlab Academy, Synlab Holding GmbH, Harrlachweg 1, 68163 Mannheim, Germany
| | - Shichao Pang
- Kardiologie, Deutsches Herzzentrum München, Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany
| | - Stephen Zewinger
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - Sarah Triem
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - Philipp Ege
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - Matthias C Reichert
- Department of Medicine II, Saarland University Medical Center, Kirrberger Straße, 66424 Homburg, Germany
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Kirrberger Straße, 66424 Homburg, Germany
- Laboratory of Metabolic Liver Diseases, Centre for Preclinical Research, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, ul. Banacha 1B, CePT, 02-097 Warsaw, Poland
| | - Susanne N Weber
- Department of Medicine II, Saarland University Medical Center, Kirrberger Straße, 66424 Homburg, Germany
| | - Isabella Jaumann
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - David Schmit
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - Tamim Sarakpi
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - Stefan Wagenpfeil
- Institute of Medical Biometry, Epidemiology & Medical Informatics, Saarland University Campus Homburg/Saar, Kirrberger Straße, 66424 Homburg/Saar, Germany
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Pauwelsstrasse 30 52074 Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, RWTH, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, 1200 Pressler Street, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, BCM226, Houston, TX 77030, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
- Center of Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, 6565 Fannin St, Houston, TX 77030, USA
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, 1200 Pressler Street, Houston, TX 77030, USA
| | - Vinicius Tragante
- Department of Cardiology, Heart and Lungs Division, UMC Utrecht, Heidelberglaan 100 3584 CX Utrecht, Netherlands
| | - Anna P Pilbrow
- The Christchurch Heart Institute, University of Otago Christchurch, 2 Riccarton Avenue, Christchurch Central City, Christchurch 8011, New Zealand
| | - A Mark Richards
- The Christchurch Heart Institute, University of Otago Christchurch, 2 Riccarton Avenue, Christchurch Central City, Christchurch 8011, New Zealand
| | - Vicky A Cameron
- The Christchurch Heart Institute, University of Otago Christchurch, 2 Riccarton Avenue, Christchurch Central City, Christchurch 8011, New Zealand
| | - Robert N Doughty
- Heart Health Research Group, University of Auckland, Level 2 / 22-30 Park Ave, Grafton, Auckland, New Zealand
| | - Marie-Pierre Dubé
- Montreal Heart Institute, 5000 Rue Bélanger, Montreal QC H1T 1C8, Canada
- Faculty of Medicine, Université der Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, 5000 Rue Bélanger, Montreal QC H1T 1C8, Canada
- Faculty of Medicine, Université der Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada
| | | | - Maxine Sun
- Faculty of Medicine, Université der Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada
| | - Chang Liu
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, 1462 Clifton Road NE, Atlanta, GA 30322, USA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Healthy, Emory University, 1518 Clifton Road NE, Atlanta, GA 30322, USA
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, 1462 Clifton Road NE, Atlanta, GA 30322, USA
| | - Jaana A Hartiala
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, 2001 N. Soto St. Los Angeles, CA 90033, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, NB 21, Cleveland, OH 44195, USA
| | - Stanley L Hazen
- Department of Cardiovascular Medicine, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, NB 21, Cleveland, OH 44195, USA
| | - Hooman Allayee
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, 2001 N. Soto St. Los Angeles, CA 90033, USA
| | - Caitrin W McDonough
- Department of Pharmacotherapy and Translational Research, University of Florida, College of Pharmacy, 1225 Center Drive, HPNP Building, Gainesville, FL 32610-0486, USA
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research, University of Florida, College of Pharmacy, 1225 Center Drive, HPNP Building, Gainesville, FL 32610-0486, USA
| | - Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research, University of Florida, College of Pharmacy, 1225 Center Drive, HPNP Building, Gainesville, FL 32610-0486, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research, University of Florida, College of Pharmacy, 1225 Center Drive, HPNP Building, Gainesville, FL 32610-0486, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Härtelstraße 16-18, 04107 Leipzig, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, Härtelstraße 16-18, 04107 Leipzig, Germany
| | - Andrej Teren
- LIFE Research Center for Civilization Diseases, University of Leipzig, Härtelstraße 16-18, 04107 Leipzig, Germany
- Heart Center Leipzig, Strümpellstraße 39, 04289 Leipzig, Germany
| | - Ralph Burkhardt
- LIFE Research Center for Civilization Diseases, University of Leipzig, Härtelstraße 16-18, 04107 Leipzig, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg,Germany
| | - Andreas Martinsson
- Department of Cardiology, Sahlgrenska University Hospital, Blå stråket 5, 413 45 Göteborg, Sweden
| | - J Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University and Skane University Hospital, BMC F12, 221 84 Lund, Sweden
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska sjukhuset Entrance 40, 751 85 Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds Väg 38, 751 85 Uppsala, Sweden
| | - Stefan K James
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska sjukhuset Entrance 40, 751 85 Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds Väg 38, 751 85 Uppsala, Sweden
| | - Niclas Eriksson
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska sjukhuset Entrance 40, 751 85 Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds Väg 38, 751 85 Uppsala, Sweden
| | - Harvey White
- Green Lane Cardiovascular Service, Auckland City Hospital, 2 Park Road, Grafton, Auckland 1023, New Zealand
| | - Claes Held
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska sjukhuset Entrance 40, 751 85 Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds Väg 38, 751 85 Uppsala, Sweden
| | - Dawn Waterworth
- Genetics, GlaxoSmithKline, 709 Swedeland Rd, King of Prussia, PA 19406, USA
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Cernter, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Netherlands Heart Institute, Moreelsepark 1, 3511 EP Utrecht, The Netherlands
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Boyd Orr Building University Avenue, Glasgow G12 8QQ, UK
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Naveed Sattar
- BHF Glasgow Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA UK
| | - Sharon Cresci
- Washington University School of Medicine, 2300 I St NW, Washington, DC 20052, USA
- Department of Medicine & Genetics, Campus Box 8232, 4515 McKinley Ave., St. Louis, MO 63110, USA
| | - John A Spertus
- Saint Luke's Mid America Heart Institute and University of Missouri-Kansas City, 4401 Wornall Rd, Kansas City, MO 64111, USA
| | - Hannah Campbell
- Washington University School of Medicine, 2300 I St NW, Washington, DC 20052, USA
- Department of Medicine & Genetics, Campus Box 8232, 4515 McKinley Ave., St. Louis, MO 63110, USA
| | - Sascha Tierling
- Faculty of Natural Sciences and Technology, Department of Genetics/Epigenetics, Saarland University, Postfach 151150, 66041 Saarbrücken, Germany
| | - Jörn Walter
- Faculty of Natural Sciences and Technology, Department of Genetics/Epigenetics, Saarland University, Postfach 151150, 66041 Saarbrücken, Germany
| | - Emmanuel Ampofo
- Institute of Clinical & Experimental Surgery, Saarland University, Kirrberger Straße, 66424 Homburg/Saar, Germany
| | - Barbara A Niemeyer
- Molecular Biophysics, CIPMM, Saarland University, Kirrberger Straße, 66424 Homburg/Saar, Germany
| | - Peter Lipp
- Center for Molecular Signaling (PZMS), Institute for Molecular Cell Biology, Research Center for Molecular Imaging and Screening, Medical Faculty, Saarland University, Kirrberger Straße, 66424 Homburg, Germany
| | - Heribert Schunkert
- Kardiologie, Deutsches Herzzentrum München, Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany
- Partner Site Munich Heart Alliance, German Centre of Cardiovascular Research (DZHK), Ismaninger Straße 22, 81675 Munich, Germany
| | - Michael Böhm
- Department of Internal Medicine III, Cardiology, Angiology, and Intensive Care Medicine, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - Wolfgang Koenig
- Kardiologie, Deutsches Herzzentrum München, Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany
- Partner Site Munich Heart Alliance, German Centre of Cardiovascular Research (DZHK), Ismaninger Straße 22, 81675 Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Helmholtzstr. 22, 89081 Ulm, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
| | - Ulrich Laufs
- Department of Cardiology, University Medical Center Leipzig, Liebigstraße 20, Leipzig, Germany
| | - Thimoteus Speer
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Hospital, Kirrberger Strasse, Building 41, 66424 Homburg/Saar, Germany
- Translational Cardio-Renal Medicine, Saarland University, Kirrberger Straße, 66424 Homburg/Saar, Germany
| | | | | |
Collapse
|
179
|
Rahvar AH, Riesel M, Graf T, Harbeck B. Cardiovascular outcome in patients with adrenal insufficiency-a therapeutic dilemma. Endocrine 2021; 72:582-585. [PMID: 33400177 DOI: 10.1007/s12020-020-02571-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/22/2020] [Indexed: 10/22/2022]
Affiliation(s)
- Amir-Hossein Rahvar
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Tobias Graf
- Department of Cardiology, University Hospital Luebeck, Luebeck, Germany
| | - Birgit Harbeck
- University of Luebeck, Luebeck, Germany.
- MVZ Amedes Experts, Endocrinology, Hamburg, Germany.
| |
Collapse
|
180
|
Zanella I, Biasiotto G, Castelli F, Calza S, Carriero C, Degli Antoni M, Focà E, Quiros-Roldan E. Descriptive modification of inflammatory markers in HIV patients after cART initiation according to gender, smoking habit, CMV infection, BMI and serum lipids. Cytokine 2021; 143:155547. [PMID: 33931289 DOI: 10.1016/j.cyto.2021.155547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/03/2021] [Accepted: 04/15/2021] [Indexed: 09/30/2022]
Abstract
Persistent inflammation, despite anti-retroviral therapy (ART), is an independent predictor of mortality and comorbidities in HIV infection. Multiple factors, including lifestyle and chronic viral coinfections, may contribute. Several of these factors are also associated with a chronic inflammation in the general population. Little is known about the degree to which these factors influence inflammation in HIV infection, particularly within the first year of ART. The purpose of this study was to distinguish the effects of factors (gender, body mass index, cholesterol and triglyceride levels, smoke habit and cytomegalovirus seropositivity), known to contribute to inflammation, on inflammation biomarkers over the first year of ART in HIV-infected patients. Linear mixed model analysis revealed significant biomarker decreases [soluble CD14 (s-CD14), soluble CD163 (s-CD163) and D-dimer (DD)], or increases [C Reactive Protein (CRP) and interleukin-6 (IL-6)] over time in the whole cohort, differences in most categories (genders for IL-6, smoke habit for s-CD14, cytomegalovirus infection for s-CD163 and IL-6) and in some category × time interactions [gender for interleukin-7 (IL-7)], cytomegalovirus infection for s-CD14 and cholesterol levels for s-CD14 and Tumor Necrosis Factor α (TNF-α)]. This explorative longitudinal study suggests further investigations on targeting inflammation pathophysiology in HIV-infected patients on ART.
Collapse
Affiliation(s)
- Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia and Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Italy.
| | - Giorgio Biasiotto
- Department of Molecular and Translational Medicine, University of Brescia and Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Italy
| | - Francesco Castelli
- University Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, Italy
| | - Stefano Calza
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Canio Carriero
- University Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, Italy
| | - Melania Degli Antoni
- University Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, Italy
| | - Emanuele Focà
- University Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, Italy
| | - Eugenia Quiros-Roldan
- University Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, Italy
| |
Collapse
|
181
|
Vinuesa A, Pomilio C, Gregosa A, Bentivegna M, Presa J, Bellotto M, Saravia F, Beauquis J. Inflammation and Insulin Resistance as Risk Factors and Potential Therapeutic Targets for Alzheimer's Disease. Front Neurosci 2021; 15:653651. [PMID: 33967682 PMCID: PMC8102834 DOI: 10.3389/fnins.2021.653651] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Overnutrition and modern diets containing high proportions of saturated fat are among the major factors contributing to a low-grade state of inflammation, hyperglycemia and dyslipidemia. In the last decades, the global rise of type 2 diabetes and obesity prevalence has elicited a great interest in understanding how changes in metabolic function lead to an increased risk for premature brain aging and the development of neurodegenerative disorders such as Alzheimer's disease (AD). Cognitive impairment and decreased neurogenic capacity could be a consequence of metabolic disturbances. In these scenarios, the interplay between inflammation and insulin resistance could represent a potential therapeutic target to prevent or ameliorate neurodegeneration and cognitive impairment. The present review aims to provide an update on the impact of metabolic stress pathways on AD with a focus on inflammation and insulin resistance as risk factors and therapeutic targets.
Collapse
Affiliation(s)
- Angeles Vinuesa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Pomilio
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Amal Gregosa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melisa Bentivegna
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jessica Presa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melina Bellotto
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Saravia
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Beauquis
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
182
|
Physical Aggression and Coronary Artery Calcification: A North Texas Healthy Heart Study. Int J Behav Med 2021; 29:14-24. [PMID: 33880713 DOI: 10.1007/s12529-021-09989-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND The purpose of this study was to determine the association between aspects of hostility and coronary artery calcification (CAC) scores. Specifically, analyses differentiated between subtypes of hostility and their relation to CAC. METHODS A sample of 571 patients aged 45 or older with no history of cardiovascular disease completed assessments of demographic, psychosocial, and medical history, along with a radiological CAC determination. Logistic regression was used to determine the association between hostility and CAC. Hostility was measured using the Aggression Questionnaire, which measured total aggression and how aggression is manifested on four scales: Physical, Verbal, Anger, and Hostility Aggression. RESULTS Regression analyses indicated that only the physical aggression parameter was related to CAC: a 5% increase in odds of CAC presence was indicated for every point increase in physical aggression. The association remained significant in adjusted analyses. Other factors associated with CAC in adjusted analyses included: age, gender, race/ethnicity, BMI, and dyslipidemia. CONCLUSIONS Psychosocial factors, such as physical aggression, are emerging factors that need to be considered in cardiovascular risk stratification.
Collapse
|
183
|
Libby P. Targeting Inflammatory Pathways in Cardiovascular Disease: The Inflammasome, Interleukin-1, Interleukin-6 and Beyond. Cells 2021; 10:951. [PMID: 33924019 PMCID: PMC8073599 DOI: 10.3390/cells10040951] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 02/08/2023] Open
Abstract
Recent clinical trials have now firmly established that inflammation participates causally in human atherosclerosis. These observations point the way toward novel treatments that add to established therapies to help stem the growing global epidemic of cardiovascular disease. Fortunately, we now have a number of actionable targets whose clinical exploration will help achieve the goal of optimizing beneficial effects while avoiding undue interference with host defenses or other unwanted actions. This review aims to furnish the foundation for this quest by critical evaluation of the current state of anti-inflammatory interventions within close reach of clinical application, with a primary focus on innate immunity. In particular, this paper highlights the pathway from the inflammasome, through interleukin (IL)-1 to IL-6 supported by a promising body of pre-clinical, clinical, and human genetic data. This paper also considers the use of biomarkers to guide allocation of anti-inflammatory therapies as a step toward realizing the promise of precision medicine. The validation of decades of experimental work and association studies in humans by recent clinical investigations provides a strong impetus for further efforts to target inflammation in atherosclerosis to address the considerable risk that remains despite current therapies.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
184
|
Shaya GE, Leucker TM, Jones SR, Martin SS, Toth PP. Coronary heart disease risk: Low-density lipoprotein and beyond. Trends Cardiovasc Med 2021; 32:181-194. [PMID: 33872757 DOI: 10.1016/j.tcm.2021.04.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 01/06/2023]
Abstract
Coronary heart disease (CHD) is the leading cause of morbidity and mortality world-wide and has been characterized as a chronic immunoinflammatory, fibroproliferative disease fueled by lipids. Great advances have been made in elucidating the complex mechanistic interactions among risk factors associated with CHD, yielding abundant success towards preventive measures and the development of pharmaceuticals to prevent and treat CHD via attenuation of lipoprotein-mediated risk. However, significant residual risk remains. Several potentially modifiable CHD risk factors ostensibly contributing to this residual risk have since come to the fore, including systemic inflammation, diabetes mellitus, high-density lipoprotein, plasma triglycerides (TG) and remnant lipoproteins (RLP), lipoprotein(a) (Lp[a]), and vascular endothelial dysfunction (ED). Herein, we summarize the body of evidence implicating each of these risk factors in residual CHD risk.
Collapse
Affiliation(s)
- Gabriel E Shaya
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA
| | - Thorsten M Leucker
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA
| | - Steven R Jones
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA
| | - Seth S Martin
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA
| | - Peter P Toth
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA; Community Hospital General Medical Center, Sterling, IL, USA.
| |
Collapse
|
185
|
Benz AP, Aeschbacher S, Krisai P, Moschovitis G, Blum S, Meyre P, Blum MR, Rodondi N, Di Valentino M, Kobza R, De Perna ML, Bonati LH, Beer JH, Kühne M, Osswald S, Conen D. Biomarkers of Inflammation and Risk of Hospitalization for Heart Failure in Patients With Atrial Fibrillation. J Am Heart Assoc 2021; 10:e019168. [PMID: 33843247 PMCID: PMC8174180 DOI: 10.1161/jaha.120.019168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Hospitalization for heart failure (HF) is very common in patients with atrial fibrillation (AF). We hypothesized that biomarkers of inflammation can identify patients with AF at increased risk of this important complication. Methods and Results Patients with established AF were prospectively enrolled. Levels of hs‐CRP (high‐sensitivity C‐reactive protein) and interleukin‐6 were measured from plasma samples obtained at baseline. We calculated an inflammation score ranging from 0 to 4 (1 point for each biomarker between the 50th and 75th percentile, 2 points for each biomarker above the 75th percentile). Individual associations of biomarkers and the inflammation score with HF hospitalization were obtained from multivariable Cox proportional hazards models. A total of 3784 patients with AF (median age 72 years, 24% prior HF) were followed for a median of 4.0 years. The median (interquartile range) plasma levels of hs‐CRP and interleukin‐6 were 1.64 (0.81–3.69) mg/L and 3.42 (2.14–5.60) pg/mL, respectively. The overall incidence of HF hospitalization was 3.04 per 100 person‐years and increased from 1.34 to 7.31 per 100 person‐years across inflammation score categories. After multivariable adjustment, both biomarkers were significantly associated with the risk of HF hospitalization (per increase in 1 SD, adjusted hazard ratio [HR], 1.22; 95% CI, 1.11–1.34 for log‐transformed hs‐CRP; adjusted HR, 1.48; 95% CI, 1.35–1.62 for log‐transformed interleukin‐6). Similar results were obtained for the inflammation score (highest versus lowest score, adjusted HR, 2.43; 95% CI, 1.80–3.30; P value for trend <0.001). Conclusions Biomarkers of inflammation strongly predicted HF hospitalization in a large, contemporary sample of patients with AF. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT02105844.
Collapse
Affiliation(s)
- Alexander P Benz
- Population Health Research Institute McMaster University Hamilton Canada
| | - Stefanie Aeschbacher
- Division of Cardiology Department of Medicine University Hospital Basel Basel Switzerland.,Cardiovascular Research Institute Basel University Hospital BaselUniversity of Basel Switzerland
| | - Philipp Krisai
- Cardiovascular Research Institute Basel University Hospital BaselUniversity of Basel Switzerland.,Electrophysiology and Ablation Unit and L'Institut de Rythmologie et Modélisation Cardiaque (LIRYC) Centre Hospitalier Universitaire de Bordeaux Bordeaux-Pessac France
| | - Giorgio Moschovitis
- Population Health Research Institute McMaster University Hamilton Canada.,Division of Cardiology Ente Ospedaliero Cantonale (EOC)Ospedale Regionale di Lugano Lugano Ticino Switzerland
| | - Steffen Blum
- Division of Cardiology Department of Medicine University Hospital Basel Basel Switzerland.,Cardiovascular Research Institute Basel University Hospital BaselUniversity of Basel Switzerland
| | - Pascal Meyre
- Division of Cardiology Department of Medicine University Hospital Basel Basel Switzerland.,Cardiovascular Research Institute Basel University Hospital BaselUniversity of Basel Switzerland
| | - Manuel R Blum
- Institute of Primary Health Care (BIHAM) University of Bern Switzerland.,Department of General Internal Medicine, Inselspital Bern University HospitalUniversity of Bern Switzerland
| | - Nicolas Rodondi
- Institute of Primary Health Care (BIHAM) University of Bern Switzerland.,Department of General Internal Medicine, Inselspital Bern University HospitalUniversity of Bern Switzerland
| | - Marcello Di Valentino
- Division of Cardiology Ente Ospedaliero Cantonale (EOC)Ospedale San Giovanni Bellinzona Bellinzona Ticino Switzerland.,Biomedical Sciences Università della Svizzera Italiana Lugano Switzerland
| | - Richard Kobza
- Division of Cardiology Luzerner Kantonsspital Luzern Switzerland
| | - Maria Luisa De Perna
- Division of Cardiology Ente Ospedaliero Cantonale (EOC)Ospedale Regionale di Lugano Lugano Ticino Switzerland
| | - Leo H Bonati
- Department of Neurology and Stroke Center University Hospital Basel Basel Switzerland
| | - Jürg H Beer
- Department of Medicine Cantonal Hospital of Baden Switzerland.,Center for Molecular Cardiology University of Zurich Switzerland
| | - Michael Kühne
- Division of Cardiology Department of Medicine University Hospital Basel Basel Switzerland.,Cardiovascular Research Institute Basel University Hospital BaselUniversity of Basel Switzerland
| | - Stefan Osswald
- Division of Cardiology Department of Medicine University Hospital Basel Basel Switzerland.,Cardiovascular Research Institute Basel University Hospital BaselUniversity of Basel Switzerland
| | - David Conen
- Population Health Research Institute McMaster University Hamilton Canada.,Cardiovascular Research Institute Basel University Hospital BaselUniversity of Basel Switzerland
| | | |
Collapse
|
186
|
Abstract
OBJECTIVES The purpose of this study was to determine the inflammatory response to a 12-hour wildfire suppression shift, in firefighters attending the "Black Saturday" natural disaster. METHODS Thirty-eight male volunteer firefighters provided venous blood samples before and after a 12-hour firefighting shift. Pre- to post-shift changes in pro-inflammatory (Interleukin [IL]-1β, IL-2, IL-6, IL-8, IL-12P70, granulocyte macrophage-colony stimulating factor [GM-CSF], tumor necrosis factor-alpha [TNF-α], interferon-gamma [IFNγ]), and anti-inflammatory (IL-4, IL-5, IL-7, IL-10, IL-13) cytokines were measured with paired sample t tests, or Wilcoxon t tests for non-parametric data. RESULTS Interleukin (IL)-6 (P = 0.003) and IL-8 (P = 0.017) were significantly increased following 12-hours of wildfire suppression. There was also a significant decrease in IL-10 (P = 0.021). CONCLUSIONS The observed acute inflammatory response may have resulted from multiple stressors including physical exertion, thermal strain, or smoke inhalation experienced during the shift, and may be a necessary response for the body to adapt to stressor exposure.
Collapse
|
187
|
Ridker PM. Inhibiting Interleukin-6 to Reduce Cardiovascular Event Rates: A Next Step for Atherothrombosis Treatment and Prevention. J Am Coll Cardiol 2021; 77:1856-1858. [PMID: 33858621 DOI: 10.1016/j.jacc.2021.02.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/20/2022]
Affiliation(s)
- Paul M Ridker
- Center for Cardiovascular Disease Prevention and the Divisions of Preventive Medicine and Cardiovascular Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
188
|
Mazaheri-Tehrani E, Mohraz M, Nasi M, Chester J, De Gaetano A, Lo Tartaro D, SeyedAlinaghi S, Gholami M, De Biasi S, Gibellini L, Mattioli AV, Pinti M, Mussini C, Cossarizza A. NLRP3 and IL-1β Gene Expression Is Elevated in Monocytes From HIV-Treated Patients With Neurocognitive Disorders. J Acquir Immune Defic Syndr 2021; 86:496-499. [PMID: 33273212 DOI: 10.1097/qai.0000000000002588] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/23/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Systemic immune activation and inflammation in chronic HIV infection are driving factors of non-AIDS-related events, including neurocognitive impairment. The role of inflammasome in monocytes from patients with HIV infection has been extensively studied, but its association with the extent of neurocognitive dysfunction has been poorly investigated. METHODS We enrolled 79 HIV-positive patients; 44 with varying levels of HIV-associated neurocognitive disorder (HAND) and 35 without and 8 healthy donors. HAND subtypes included asymptomatic neurocognitive impairment (asymptomatic neurocognitive impairment; n = 19), mild neurocognitive disorder (MND; n = 17), and HIV-associated dementia (n = 8). We quantified plasmatic concentrations of proinflammatory cytokines (TNF-α, IL-6, IL-17A, IL-1β, and IFN-γ) for all HIV patients, and the mRNA expression of genes involved in the inflammasome activity (NLRP3, PYCARD, NAIP, AIM2, IL-1β, and IL-18) in monocytes of a subgroup of 28 HIV patients and 8 healthy donors. RESULTS HIV patients' plasma concentrations of IFN-γ, IL-1β, and IL-17A were undetectable. Levels of TNF-α and IL-6 were similar among the HIV patient groups. A trend toward an increased expression of inflammasome genes according to neurocognitive disorder severity was observed. Of note, the NLRP3 mRNA relative expression was higher in MND compared with other groups, and IL-1β was lower in MND than HIV-associated dementia patients. CONCLUSIONS Changes in inflammasome components in circulating monocytes according to different HAND severity suggest that NLRP3 may be a possible biomarker or target to better understand and treat the link between systemic inflammation and neurocognitive impairment in HIV infection.
Collapse
Affiliation(s)
- Elham Mazaheri-Tehrani
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Minoo Mohraz
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Johanna Chester
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna De Gaetano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - SeyedAhmad SeyedAlinaghi
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Gholami
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Vittoria Mattioli
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- National Institute for Cardiovascular Research-INRC, Bologna, Italy; and
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
- National Institute for Cardiovascular Research-INRC, Bologna, Italy; and
| |
Collapse
|
189
|
Ahn SB, Kamath KS, Mohamedali A, Noor Z, Wu JX, Pascovici D, Adhikari S, Cheruku HR, Guillemin GJ, McKay MJ, Nice EC, Baker MS. Use of a Recombinant Biomarker Protein DDA Library Increases DIA Coverage of Low Abundance Plasma Proteins. J Proteome Res 2021; 20:2374-2389. [PMID: 33752330 DOI: 10.1021/acs.jproteome.0c00898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Credible detection and quantification of low abundance proteins from human blood plasma is a major challenge in precision medicine biomarker discovery when using mass spectrometry (MS). In this proof-of-concept study, we employed a mixture of selected recombinant proteins in DDA libraries to subsequently identify (not quantify) cancer-associated low abundance plasma proteins using SWATH/DIA. The exemplar DDA recombinant protein spectral library (rPSL) was derived from tryptic digestion of 36 recombinant human proteins that had been previously implicated as possible cancer biomarkers from both our own and other studies. The rPSL was then used to identify proteins from nondepleted colorectal cancer (CRC) EDTA plasmas by SWATH-MS. Most (32/36) of the proteins used in the rPSL were reliably identified from CRC plasma samples, including 8 proteins (i.e., BTC, CXCL10, IL1B, IL6, ITGB6, TGFα, TNF, TP53) not previously detected using high-stringency protein inference MS according to PeptideAtlas. The rPSL SWATH-MS protocol was compared to DDA-MS using MARS-depleted and postdigestion peptide fractionated plasmas (here referred to as a human plasma DDA library). Of the 32 proteins identified using rPSL SWATH, only 12 could be identified using DDA-MS. The 20 additional proteins exclusively identified using the rPSL SWATH approach were almost exclusively lower abundance (i.e., <10 ng/mL) proteins. To mitigate justified FDR concerns, and to replicate a more typical library creation approach, the DDA rPSL library was merged with a human plasma DDA library and SWATH identification repeated using such a merged library. The majority (33/36) of the low abundance plasma proteins added from the rPSL were still able to be identified using such a merged library when high-stringency HPP Guidelines v3.0 protein inference criteria were applied to our data set. The MS data set has been deposited to ProteomeXchange Consortium via the PRIDE partner repository (PXD022361).
Collapse
Affiliation(s)
- Seong Beom Ahn
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Karthik S Kamath
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Abidali Mohamedali
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Zainab Noor
- ProCan, Children's Medical Research Institute, The University of Sydney, Westmead, Newtown, NSW 2042, Australia
| | - Jemma X Wu
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Dana Pascovici
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Subash Adhikari
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Harish R Cheruku
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Gilles J Guillemin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Matthew J McKay
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Mark S Baker
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| |
Collapse
|
190
|
Kremer JM. Methotrexate and Cardiovascular Disease in Patients With Rheumatoid Arthritis: Insights and Novel Speculations. J Rheumatol 2021; 48:793-795. [PMID: 33722953 DOI: 10.3899/jrheum.201415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Joel M Kremer
- J.M. Kremer, MD, Pfaff Family Professor of Medicine, The Department of Medicine, Division of Rheumatology, Albany Medical College, Albany, New York, USA.
| |
Collapse
|
191
|
Jung R, Wild J, Ringen J, Karbach S, Wenzel P. Innate Immune Mechanisms of Arterial Hypertension and Autoimmune Disease. Am J Hypertens 2021; 34:143-153. [PMID: 32930786 DOI: 10.1093/ajh/hpaa145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/15/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
The immune system is indispensable in the development of vascular dysfunction and hypertension. The interplay between immune cells and the vasculature, kidneys, heart, and blood pressure regulating nuclei in the central nervous system results in a complex and closely interwoven relationship of the immune system with arterial hypertension. A better understanding of this interplay is necessary for optimized and individualized antihypertensive therapy. Our review article focuses on innate cells in hypertension and to what extent they impact on development and preservation of elevated blood pressure. Moreover, we address the association of hypertension with chronic autoimmune diseases. The latter are ideally suited to learn about immune-mediated mechanisms in cardiovascular disease leading to high blood pressure.
Collapse
Affiliation(s)
- Rebecca Jung
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Johannes Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Julia Ringen
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Susanne Karbach
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Germany
| |
Collapse
|
192
|
Saeed M, Tapia G, Ariansen I, Stene LC, Seljeflot I, Tell GS, Skrivarhaug T, Joner G. Serum Galectin-3 and Subsequent Risk of Coronary Heart Disease in Subjects With Childhood-Onset Type 1 Diabetes: A Cohort Study. Diabetes Care 2021; 44:810-816. [PMID: 33408220 PMCID: PMC7896257 DOI: 10.2337/dc20-1712] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/07/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To study whether serum galectin-3 and other biomarkers of inflammation predict coronary heart disease (CHD) in subjects with long-standing childhood-onset type 1 diabetes. RESEARCH DESIGN AND METHODS A population-based nationwide cohort of 299 subjects with type 1 diabetes diagnosed in Norway at <15 years of age during 1973-1982 was examined in 2002-2003 at a mean age of 33 years (range 21-44), with mean diabetes duration of 24 years (range 19-30). Subjects were followed through 31 December 2017 for their first CHD event registered by a hospitalization or cause of death using nationwide registries. Stored serum samples were available for 296 subjects and analyzed for interleukin-6 (IL-6), IL-6 receptor, IL-18, hs-CRP, matrix metalloproteinase-9, tissue inhibitor of metalloproteinase-1 (TIMP-1), galectin-3, and high-sensitivity troponin T. Adjusted hazard ratios (aHRs) for CHD per SD increase in biomarker were estimated using Cox regression. RESULTS Of 295 subjects, 40 (13.6%) had a documented CHD event during a mean follow-up of 14.4 years (range 0.5-16). IL-6 (aHR 1.32 [95% CI 1.07-1.63]), galectin-3 (aHR 1.44 [95% CI 1.09-1.80]), and TIMP-1 (aHR 1.37 [95% CI 1.04-1.81]) were significant predictors of CHD after adjustment for conventional risk factors. CONCLUSIONS Galectin-3 was significantly associated with future CHD in subjects with type 1 diabetes, and if the results are replicated in larger studies, it may aid in prediction together with conventional risk factors for CHD.
Collapse
Affiliation(s)
- Maryam Saeed
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway .,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - German Tapia
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, Oslo, Norway
| | - Inger Ariansen
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, Oslo, Norway
| | - Lars C Stene
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, Oslo, Norway
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital, Oslo, Norway
| | - Grethe S Tell
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, Oslo, Norway.,Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Torild Skrivarhaug
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Geir Joner
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
193
|
Wang YZ, Ngowi EE, Wang D, Qi HW, Jing MR, Zhang YX, Cai CB, He QL, Khattak S, Khan NH, Jiang QY, Ji XY, Wu DD. The Potential of Hydrogen Sulfide Donors in Treating Cardiovascular Diseases. Int J Mol Sci 2021; 22:2194. [PMID: 33672103 PMCID: PMC7927090 DOI: 10.3390/ijms22042194] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 02/08/2023] Open
Abstract
Hydrogen sulfide (H2S) has long been considered as a toxic gas, but as research progressed, the idea has been updated and it has now been shown to have potent protective effects at reasonable concentrations. H2S is an endogenous gas signaling molecule in mammals and is produced by specific enzymes in different cell types. An increasing number of studies indicate that H2S plays an important role in cardiovascular homeostasis, and in most cases, H2S has been reported to be downregulated in cardiovascular diseases (CVDs). Similarly, in preclinical studies, H2S has been shown to prevent CVDs and improve heart function after heart failure. Recently, many H2S donors have been synthesized and tested in cellular and animal models. Moreover, numerous molecular mechanisms have been proposed to demonstrate the effects of these donors. In this review, we will provide an update on the role of H2S in cardiovascular activities and its involvement in pathological states, with a special focus on the roles of exogenous H2S in cardiac protection.
Collapse
Affiliation(s)
- Yi-Zhen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Department of Biological Sciences, Faculty of Science, Dar es Salaam University College of Education, Dar es Salaam 2329, Tanzania
| | - Di Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Hui-Wen Qi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Mi-Rong Jing
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Chun-Bo Cai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Qing-Lin He
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- School of Nursing and Health, Henan University, Kaifeng 475004, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng 475004, China
- School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng 475004, China
- School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- School of Stomatology, Henan University, Kaifeng 475004, China
| |
Collapse
|
194
|
Ma J, Chen X. Anti-inflammatory Therapy for Coronary Atherosclerotic Heart Disease: Unanswered Questions Behind Existing Successes. Front Cardiovasc Med 2021; 7:631398. [PMID: 33598482 PMCID: PMC7882495 DOI: 10.3389/fcvm.2020.631398] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/21/2020] [Indexed: 02/05/2023] Open
Abstract
Coronary atherosclerotic heart disease is a serious threat to human health. The results of the Canakinumab Anti-Inflammatory Thrombosis Outcome Study published in 2017 put an end to the perennial debate about the anti-inflammatory treatment of coronary atherosclerotic heart disease. In addition to interleukin 1β monoclonal antibody, interleukin 6 receptor antagonists and colchicine have also shown exciting results in clinical trials within the last 3 years. However, behind these successes, questions remain that need to be addressed. In this review, we summarize the successes and existing doubts of interleukin 1β antibodies, interleukin 6 receptor antagonists, and colchicine in the anti-inflammatory treatment of coronary atherosclerotic heart disease.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China.,Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, China
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
195
|
Silvis MJM, Demkes EJ, Fiolet ATL, Dekker M, Bosch L, van Hout GPJ, Timmers L, de Kleijn DPV. Immunomodulation of the NLRP3 Inflammasome in Atherosclerosis, Coronary Artery Disease, and Acute Myocardial Infarction. J Cardiovasc Transl Res 2021; 14:23-34. [PMID: 32648087 PMCID: PMC7892681 DOI: 10.1007/s12265-020-10049-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality and morbidity worldwide. Atherosclerosis is responsible for the majority of cardiovascular disorders with inflammation as one of its driving processes. The nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, responsible for the release of the pro-inflammatory cytokines, interleukin-1β (IL-1β), and interleukin-18 (IL-18), has been studied extensively and showed to play a pivotal role in the progression of atherosclerosis, coronary artery disease (CAD), and myocardial ischemia reperfusion (I/R) injury. Both the NLRP3 inflammasome and its downstream cytokines, IL-1ß and IL-18, could therefore be promising targets in cardiovascular disease. This review summarizes the role of the NLRP3 inflammasome in atherosclerosis, CAD, and myocardial I/R injury. Furthermore, the current therapeutic approaches targeting the NLRP3 inflammasome and its downstream signaling cascade in atherosclerosis, CAD, and myocardial I/R injury are discussed.
Collapse
Affiliation(s)
- Max J M Silvis
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3508, GA, Utrecht, The Netherlands.
| | - Evelyne J Demkes
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Aernoud T L Fiolet
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3508, GA, Utrecht, The Netherlands
| | - Mirthe Dekker
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Cardiology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Lena Bosch
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3508, GA, Utrecht, The Netherlands
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerardus P J van Hout
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3508, GA, Utrecht, The Netherlands
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leo Timmers
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, The Netherlands
| | | |
Collapse
|
196
|
Zuk AM, Liberda EN, Tsuji LJS. Examining chronic inflammatory markers on blood pressure measures in the presence of vitamin D insufficiency among indigenous cree adults: results from the cross-sectional Multi-Community Environment-and-Health Study in Eeyou Istchee, Quebec, Canada. BMJ Open 2021; 11:e043166. [PMID: 33504558 PMCID: PMC7843349 DOI: 10.1136/bmjopen-2020-043166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE High blood pressure (BP) is a risk factor for cardiovascular disease. Examining the role of inflammatory mediators on BP is important since vitamin D (VD) is a modifiable risk factor, which possibly modulates inflammatory cytokines. This study simulated what are known as average 'controlled direct effects (CDE)' of inflammatory markers, C reactive protein (CRP), tumour necrosis factor-α (TNF-α), and interlukin-6 (IL-6) on continuous BP measures, while fixing VD, an intermediate variable to specific level. DESIGN Cross-sectional study. SETTING We analysed data from the Multi-Community Environment-and-Health Study, 2005-2009, conducted in Eeyou Istchee, Quebec, Canada. PARTICIPANTS This study recruited 1425 study Indigenous Cree participants from seven Cree communities. Only adults with serum VD levels, inflammatory markers and BP measures were included in this data analysis. PRIMARY AND SECONDARY OUTCOMES MEASURES Inflammatory markers examined the top 25th exposure percentiles. VD 'insufficiency' (ie, 25-hydroxyvitamin-D levels<50 nmol/L) defined by the Institute of Medicine. CDE for each inflammatory marker in the presence and absence of population VD insufficiency simulated the average direct effect change for systolic and diastolic BP (SBP and DBP) measures. All models were adjusted for exposure-and-mediator outcome relationship. RESULTS Among 161 participants, 97 (60 %) were female. The prevalence of VD insufficiency was 32%. CDE estimates show in the presence and absence of population vitamin D insufficiency, inflammatory markers have a slightly different association on BP. TNF-α significantly and inversely associated with SBP in the presence of vitamin D insufficiency, fully adjusted model β = -13.61 (95% CI -24.42 to -2.80); however, TNF-α was not associated with SBP in the absence of vitamin D insufficiency. CRP, IL-6 were also not significantly associated with BP measures, although the magnitude of association was greater for those with elevated inflammation and VD insufficiency. CONCLUSION This novel analysis shows in the presence of VD insufficiency, inflammation (particularly TNF-α) may affect SBP. Additional research is needed to elucidate these findings, and the temporal relationship between these variables.
Collapse
Affiliation(s)
- Aleksandra M Zuk
- Department of Physical and Environmental Sciences, University of Toronto, Toronto, Ontario, Canada
- School of Nursing, Queen's University, Kingston, Ontario, Canada
| | - Eric N Liberda
- School of Occupational and Public Health, Ryerson University, Toronto, Ontario, Canada
| | - Leonard J S Tsuji
- Department of Physical and Environmental Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
197
|
Wild J, Wenzel P. Myeloid cells, tissue homeostasis, and anatomical barriers as innate immune effectors in arterial hypertension. J Mol Med (Berl) 2021; 99:315-326. [PMID: 33443617 PMCID: PMC7899956 DOI: 10.1007/s00109-020-02019-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 11/18/2020] [Indexed: 11/29/2022]
Abstract
Although essential hypertension affects a large proportion of the human population and is one of the key drivers of cardiovascular mortality worldwide, we still do not have a complete understanding of its pathophysiology. More than 50 years ago, the immune system has been identified as an important part of the pathogenesis of arterial hypertension. An exceeding variety of recent publications deals with the interplay between the numerous different components of the immune system and mechanisms of arterial hypertension and has substantially contributed to our understanding of the role of immunity and inflammation in the pathogenesis of the disease. In this review, we focus on myeloid cells and anatomical barriers as particular aspects of innate immunity in arterial hypertension. Since it represents a first line of defense protecting against pathogens and maintaining tissue homeostasis, innate immunity provides many mechanistic hinge points in the area of hypertension.
Collapse
Affiliation(s)
- Johannes Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Cardiology - Cardiology I and CTH Professorship "Vascular Inflammation", University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,German Center for Cardiovascular Research (DZHK) - Partner site RheinMain, Berlin, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany. .,Center for Cardiology - Cardiology I and CTH Professorship "Vascular Inflammation", University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany. .,German Center for Cardiovascular Research (DZHK) - Partner site RheinMain, Berlin, Germany.
| |
Collapse
|
198
|
Scholtz B, Vo Minh D, Kiss C, Tar I, Kumar A, Tőzsér J, Csősz É, Márton I. Examination of Oral Squamous Cell Carcinoma and Precancerous Lesions Using Proximity Extension Assay and Salivary RNA Quantification. Biomedicines 2020; 8:biomedicines8120610. [PMID: 33327496 PMCID: PMC7764999 DOI: 10.3390/biomedicines8120610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022] Open
Abstract
Saliva is an easy-to access body fluid with high diagnostic potential. The utilization of saliva for oral cancer diagnosis can be an attractive possibility. Besides the oral cancer, it is important to better understand the precancerous lesions such as oral lichen planus (OLP) and leukoplakia (OLK). In order to examine the changes of salivary proteins in controls, patients with oral cancer, and patients with precancerous conditions, proximity extension assay was utilized. Some proteins and functions were characteristic to the examined groups and can serve as a starting point for further biomarker studies. The different nature of OLK and OLP was demonstrated, showing the malignant transformation and the inflammation as the prominent biological processes in the OLK and OLP, respectively. The salivary level of IL6 was verified using quantitative ELISA and the mRNA level was also studied. Elevated IL6 levels could be detected in precancerous groups compared to controls.
Collapse
Affiliation(s)
- Beáta Scholtz
- Genomic Medicine and Bioinformatic Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Doan Vo Minh
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.V.M.); (A.K.)
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Csongor Kiss
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Ildikó Tar
- Department of Oral Medicine, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary;
| | - Ajneesh Kumar
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.V.M.); (A.K.)
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- Doctoral School of Molecular Cell and Immune Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Tőzsér
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Éva Csősz
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.V.M.); (A.K.)
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- Correspondence: (É.C.); (I.M.); Tel.: +36-52-416432 (É.C.)
| | - Ildikó Márton
- Department of Restorative Dentistry, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: (É.C.); (I.M.); Tel.: +36-52-416432 (É.C.)
| |
Collapse
|
199
|
Platelet-derived extracellular vesicles to target plaque inflammation for effective anti-atherosclerotic therapy. J Control Release 2020; 329:445-453. [PMID: 33285103 DOI: 10.1016/j.jconrel.2020.11.064] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a kind of chronic inflammatory diseases characterized by dysfunction of local immune responses. Here we engineer platelet-derived extracellular vesicles (PEVs) to load MCC950, an NLRP3-inflammasome inhibitor, for atherosclerosis-targeted therapy. PEVs which are readily collected from the activated platelets selectively bind multiple cell types associated with the formation of atherosclerotic plaque in vivo. Intravenous administration of MCC950-PEVs could significantly reduce the formation of atherosclerotic plaques, lower the local inflammation and inhibit proliferation of macrophages and T cells at the plaque site compared with free drug administration in ApoE-KO mice. Our strategy suggests the promise of PEVs for targeted drug delivery for treatment of atherosclerosis.
Collapse
|
200
|
Tang H, Cheng Z, Li N, Mao S, Ma R, He H, Niu Z, Chen X, Xiang H. The short- and long-term associations of particulate matter with inflammation and blood coagulation markers: A meta-analysis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 267:115630. [PMID: 33254709 PMCID: PMC7687019 DOI: 10.1016/j.envpol.2020.115630] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 05/16/2023]
Abstract
Inflammation and the coagulation cascade are considered to be the potential mechanisms of ambient particulate matter (PM) exposure-induced adverse cardiovascular events. Tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-8 (IL-8), and fibrinogen are arguably the four most commonly assayed markers to reflect the relationships of PM with inflammation and blood coagulation. This review summarized and quantitatively analyzed the existing studies reporting short- and long-term associations of PM2.5(PM with an aerodynamic diameter ≤2.5 μm)/PM10 (PM with an aerodynamic diameter≤10 μm) with important inflammation and blood coagulation markers (TNF-α, IL-6, IL-8, fibrinogen). We reviewed relevant studies published up to July 2020, using three English databases (PubMed, Web of Science, Embase) and two Chinese databases (Wang-Fang, China National Knowledge Infrastructure). The OHAT tool, with some modification, was applied to evaluate risk of bias. Meta-analyses were conducted with random-effects models for calculating the pooled estimate of markers. To assess the potential effect modifiers and the source of heterogeneity, we conducted subgroup analyses and meta-regression analyses where appropriate. The assessment and correction of publication bias were based on Begg's and Egger's test and "trim-and-fill" analysis. We identified 44 eligible studies. For short-term PM exposure, the percent change of a 10 μg/m3 PM2.5 increase on TNF-α and fibrinogen was 3.51% (95% confidence interval (CI): 1.21%, 5.81%) and 0.54% (95% confidence interval (CI): 0.21%, 0.86%) respectively. We also found a significant short-term association between PM10 and fibrinogen (percent change = 0.17%, 95% CI: 0.04%, 0.29%). Overall analysis showed that long-term associations of fibrinogen with PM2.5 and PM10 were not significant. Subgroup analysis showed that long-term associations of fibrinogen with PM2.5 and PM10 were significant only found in studies conducted in Asia. Our findings support significant short-term associations of PM with TNF-α and fibrinogen. Future epidemiological studies should address the role long-term PM exposure plays in inflammation and blood coagulation markers level change.
Collapse
Affiliation(s)
- Hong Tang
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Zilu Cheng
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 122# Luoshi Road, Wuhan, China
| | - Na Li
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Shuyuan Mao
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Runxue Ma
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Haijun He
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Zhiping Niu
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Xiaolu Chen
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Hao Xiang
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China.
| |
Collapse
|