1
|
Tenenhaus M, Rennekampff HO, Vassolas GA. Wearable biosensors for monitoring and as a predictive adjunct for patients at risk for ischemic cardiac-related injury. J Intern Med 2025; 297:437-447. [PMID: 39988463 DOI: 10.1111/joim.20073] [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: 02/25/2025]
Abstract
Despite increased attention and preventive efforts, the prevalence of major adverse cardiovascular events continues to rise, resulting in profound concerns for both the individual and the population at large. Rapidly evolving biotechnologies, micro-computerization, communication, and battery design have led to widespread commercial adoption, use, and dependence on smart devices, and, more recently, biosensors. Currently worn and carried, smart devices such as mobile phones and smart watches possess impressive computational and communication capabilities, monitoring a variety of biometrics such as heart rate, blood pressure, and cardiac rhythm. Several promising biomarkers have been identified that are expressed early in the development of cardiac injury. Biosensors that can assay multiple variants are now described, obviating the limitations generally attributed to dependence upon a single biomarker. Employing mathematical modeling along with intelligent learning capabilities complements and augments their potential value. Data derived from wearable multivariate biosensors linked to already worn smart devices can communicate information to protected settings with enhanced computational capability and cogency by evaluating relayed biometrics and early expressed biomarkers as well as trending data, improving sensitivity and specificity. Integrating intelligent learning capabilities can further power these efforts with beneficial impact on individuals and groups at risk, yielding great promise as monitoring and predictive adjuncts. Future derivations might, for those of particular concern, be linked to critical drug delivery and interventional systems.
Collapse
Affiliation(s)
| | - Hans Oliver Rennekampff
- Department of Plastic Surgery, Hand and Burn Surgery, Rhein Maas Klinikum, Wuerselen, Germany
| | | |
Collapse
|
2
|
Odeberg J, Halling A, Ringborn M, Freitag M, Persson ML, Vaara I, Råstam L, Odeberg H, Lindblad U. Markers of inflammation predicts long-term mortality in patients with acute coronary syndrome - a cohort study. BMC Cardiovasc Disord 2025; 25:190. [PMID: 40089663 PMCID: PMC11909928 DOI: 10.1186/s12872-025-04608-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Chronic low-grade inflammation is a well-known risk factor for coronary heart disease (CHD) and future cardiovascular events. Anti-inflammatory therapy can reduce the risk of ischemic cardiovascular disease (CVD) events following myocardial infarction (MI). However, it remains unknown to what extent inflammation at the time of an acute event predicts long-term outcomes. We explored whether routine blood measurements of inflammatory markers during an acute coronary syndrome (ACS) are predictive of long-term mortality. METHODS In a cohort of 5292 consecutive patients admitted to a coronary intensive care unit with suspected ACS over a four-year period in the Carlscrona Heart Attack Prognosis Study (CHAPS), 908 patients aged 30-74 years (644 men, 264 women) were diagnosed with MI (527) or unstable angina (UA) (381). A 10-year follow-up study was conducted using Swedish national registries, with total mortality and cardiac mortality as primary outcomes. RESULTS Long-term total and cardiac mortality were significantly associated with higher leukocyte counts (e.g., neutrophils, monocytes, p ≤ 0.001), higher levels of inflammatory biomarkers (e.g., C-reactive protein, Serum Amyloid A, fibrinogen, p ≤ 0.001), and elevated neutrophil-lymphocyte ratio (NLR) (p < 0.001) and monocyte-lymphocyte ratio (MLR) (p = 0.002), all measured at ACS admission. These associations were independent of ACS diagnosis. CONCLUSION Our results suggest that level of inflammation at ACS presentation-beyond its established role as a major CHD risk factor-also predicts long-term mortality following ACS. Notably, inflammation at the time of the event was a stronger predictor of long-term mortality than the acute event outcome itself. However, limitations include the observational study design, moderate sample size, and absence of modern high-sensitivity cardiac biomarkers and contemporary ACS management strategies in this cohort. The results should therefore be interpreted in the context of historical clinical practice. While our model-wise complete-case approach ensured consistency, missing data remains a potential source of bias. Future studies in larger, more contemporary cohorts are needed to validate these findings and refine risk stratification strategies.
Collapse
Affiliation(s)
- Jacob Odeberg
- Department of Protein Science, Science for Life Laboratory Stockholm, CBH, KTH Royal Institute of Technology, Stockholm, 100 44, Sweden.
- Department of Clinical Medicine, Faculty of Health Science, Arctic University of Tromsö (UiT), Tromsö, N 9037, Norway.
- Division of Internal Medicine, University Hospital North Norway (UNN), Tromsö, Norway.
- Department of Hematology, Coagulation Unit, Karolinska University Hospital, Stockholm, Sweden.
- Department of Medicine, Solna, Karolinska Institute, Stockholm, SE, 171 77, Sweden.
| | - Anders Halling
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Michael Ringborn
- Thoracic Center, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Michael Freitag
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Marie Louise Persson
- Department of Laboratory Medicine, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Ivar Vaara
- Department of Laboratory Medicine, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Lennart Råstam
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Håkan Odeberg
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Ulf Lindblad
- School of Public Health and Community Medicine/Primary Health Care, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
3
|
Yu H, Li X, Ning B, Feng L, Ren Y, Li S, Kang Y, Ma J, Zhao M. SIRT1: a potential therapeutic target for coronary heart disease combined with anxiety or depression. J Drug Target 2025; 33:328-340. [PMID: 39470049 DOI: 10.1080/1061186x.2024.2422882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Coronary heart disease (CHD) combined with anxiety or depression is increasingly receiving attention in the clinical field of cardiology, and exploring the comorbidity pathological mechanisms of cardiovascular disease combined with psychological disorders is a hot research topic for scholars in this field. Current research suggests that Silent Information Regulatory Factor 1 (SIRT1) may serve as a potential biomarker for the comorbidity mechanism and treatment of CHD with anxiety or depression. SIRT1 is considered a promising therapeutic target for CHD combined with anxiety or depression, with the ability to regulate inflammatory cytokine levels, alleviate oxidative stress damage, activate multiple signalling pathways, reduce platelet hyperresponsiveness, and exert neuroprotective and cardioprotective effects. In this comprehensive review, we deeply studied the structure, function, and mechanism of SIRT1, and discussed its protective effects in the cardiovascular and nervous system. The latest progress in the mechanism of SIRT1's role in CHD combined with anxiety or depression was emphasised, including its specific mechanisms in regulating inflammatory response, alleviating oxidative stress, and mediating various signalling pathways. In addition, this article also summarises the therapeutic potential of SIRT1 as a potential biomarker in patients with CHD combined with anxiety or depression.
Collapse
Affiliation(s)
- Hubin Yu
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xinping Li
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Bo Ning
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lanshuan Feng
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yaolong Ren
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Shilin Li
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yalong Kang
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Ma
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Air Force Military Medical University, Xi'an, China
| | - Mingjun Zhao
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
4
|
Loglo AD, Antwi PB, Abass KM, Osei-Mireku S, Amofa G, Ofori E, Adjei JK, Oppong MN, Phillips RO, Annan R, Engel B, Simmonds RE. Micronutrient-deficient diets and possible environmental enteric dysfunction in Buruli ulcer endemic communities in Ghana: Lower dietary diversity and reduced serum zinc and vitamin C implicate micronutrient status a possible susceptibility factor. PLoS Negl Trop Dis 2025; 19:e0012871. [PMID: 40072975 PMCID: PMC11902277 DOI: 10.1371/journal.pntd.0012871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The nutritional status of communities susceptible to Buruli ulcer (BU, a skin NTD caused by infection with Mycobacterium ulcerans) remains almost completely obscure. We have assessed the diets of BU patients vs. controls from the same BU-endemic communities, and compared their circulating biomarkers of nutrients and inflammation. METHODS/PRINCIPAL FINDINGS We investigated two cohorts of BU patients and controls. The first were administered food frequency and multi-pass 24-hour recall questionnaires to determine patterns of foods consumed, nutrient intake and nutrient adequacy. The second used archived serum samples collected as baseline to measure the circulating concentration of zinc, vitamin C, CRP, IL-1β, IFN-γ, TNF-α and IL-6. Stunted growth was more prevalent than expected (31%), while 18% of participants were underweight and most had inadequate intake of all micronutrients except for carbohydrate. BU patients had a lower intake of, selenium, vitamin B12 and zinc, and for selenium and vitamin B12 a higher proportion had dietary insufficiency (40% vs. 15% and 80% vs. 55%, respectively). In line with this, BU patients had significantly lower levels of zinc in their serum, and more had levels below the normal range (72% vs. 43%). Despite many participants having a good intake of vitamin C, serum levels were low, and lower amongst the BU patients. As expected, there was little evidence of systemic inflammation (CRP <0.6 mg/L). Elevated IL-6 levels were present in several participants suggesting that environmental enteric dysfunction may be prevalent in these communities, however this was similar in cases vs. controls. CONCLUSIONS/SIGNIFICANCE Diet and nutritional status may be a contributing factor to BU pathogenesis. Protein and the micronutrients zinc, selenium, vitamin B12 and vitamin C may be of particular importance. Nutritional interventions may have potential for both prophylaxis and treatment of BU, which may be a cost-effective approach to achieving the NTD Roadmap goals.
Collapse
Affiliation(s)
- Aloysius Dzigbordi Loglo
- Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Philemon Boasiako Antwi
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | | | | | | | - Jonathan Kofi Adjei
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Michael Ntiamoah Oppong
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Richard Odame Phillips
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Reginald Annan
- Department of Biochemistry and Biotechnology, College of Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Barbara Engel
- Department of Nutritional Sciences, School of Biosciences, University of Surrey, Guildford, United Kingdom
| | - Rachel E. Simmonds
- Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
5
|
Kang J, Moser DK, Lennie TA, Chung ML, Thomas DT, Biddle MJ. Diet Quality Mediates the Relationship Between Chronic Stress and Inflammation in Patients With Metabolic Syndrome. J Cardiovasc Nurs 2025; 40:124-132. [PMID: 38192030 PMCID: PMC11231055 DOI: 10.1097/jcn.0000000000001072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
BACKGROUND Chronic stress is associated with promotion of inflammation and development of metabolic syndrome, as well as deterioration of diet quality. Inflammation can be modified by changes in dietary intake. OBJECTIVE The aim of this study was to test the hypothesis that diet quality mediates the relationship of chronic stress with inflammation in patients with metabolic syndrome. METHODS Participants with metabolic syndrome (n = 73, 62 ± 12 years old, 71% female) completed questionnaires on chronic stress (Perceived Stress Scale-10) and diet quality (Healthy Eating Index-2020). The Perceived Stress Scale-10 was dichotomized. The Healthy Eating Index-2020 score was used as a continuous variable, and higher scores indicate better diet quality. Inflammation was assessed using plasma high-sensitivity C-reactive protein (log-transformed). We used PROCESS in SPSS to test the hypothesis. RESULTS Patients in the higher stress group had lower Healthy Eating Index-2020 scores (worse diet quality) than those in the lower stress group (57 ± 13 vs 64 ± 10, P = .01). Diet quality mediated the relationship between chronic stress and inflammation (indirect effect, 0.211; 95% bootstrap confidence interval, 0.006-0.496). Higher stress was associated with lower diet quality (effect, -7.152; 95% confidence interval, -13.168 to -1.137) that was associated with increased inflammation (effect, -0.030; 95% confidence interval, -0.052 to -0.007). CONCLUSIONS Our findings show the important role of diet quality in the relationship of chronic stress with inflammation in patients with metabolic syndrome. Healthcare providers should encourage patients with higher stress to improve diet quality, which can decrease inflammation.
Collapse
Affiliation(s)
- JungHee Kang
- College of Nursing, University of Kentucky, 2201 Regency Rd, Suite 403, Lexington, KY 40503, USA
| | - Debra K. Moser
- College of Nursing, University of Kentucky, 2201 Regency Rd, Suite 403, Lexington, KY 40503, USA
| | - Terry A. Lennie
- College of Nursing, University of Kentucky, 751 Rose Street, Lexington, KY 40536, USA
| | - Misook L. Chung
- College of Nursing, University of Kentucky, 2201 Regency Rd, Suite 403, Lexington, KY 40503, USA
| | - D. Travis Thomas
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, 214D, Wethington Building, Lexington, KY 40536, USA
| | - Martha J. Biddle
- College of Nursing, University of Kentucky, 2201 Regency Rd, Suite 403, Lexington, KY 40503, USA
| |
Collapse
|
6
|
Su Y, Martinkova A, O'Donnell E, Bailey SJ, Leicht CA. The acute effects of continuous and intermittent whole-body passive heating on cardiovascular disease risk indicators in healthy and young males and females. Eur J Appl Physiol 2025:10.1007/s00421-025-05718-0. [PMID: 39953332 DOI: 10.1007/s00421-025-05718-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE Heat therapy is recognised to promote cardiovascular health, and whilst most recent heat therapy investigations have focussed on continuous heat exposure, traditional sauna use often includes recovery periods. This study compared the acute effects of continuous versus intermittent whole-body heating on cardiovascular function markers in males and females. METHODS Twenty healthy participants (25 ± 3 years; 10 males, 10 females) were exposed to 2 passive heating regimens: continuous heating (CH) for 60 min and intermittent heating (IH) comprised of 3 × 20-min blocks interspersed by 15-min cooling breaks. Skin perfusion, blood pressure (BP), plasma nitrite, interleukins, body temperature, and thermal perceptual responses were assessed. RESULTS Greater increases in rectal temperature (Trec) (CH: 1.2 ± 0.1 °C; IH: 0.5 ± 0.1 °C), skin perfusion, systolic blood pressure (SBP), heart rate (HR), interleukin-6 (IL-6) and plasma nitrite were found in CH compared to IH (p ≤ 0.01), but the thermal perceptual response was more unfavourable during CH (p < 0.01). Females had higher skin perfusion and plasma nitrite concentrations (p ≤ 0.04), but lower brachial and central BP than males in both conditions (p ≤ 0.01). Furthermore, females reached a higher Trec and more unfavourable thermal perception in CH (p ≤ 0.02). CONCLUSION More pronounced cardiovascular responses were associated with higher Trec and discomfort. Females exhibited higher skin perfusion and plasma nitrite concentrations than males and reported less favourable thermal perception in CH, but not in IH.
Collapse
Affiliation(s)
- Yunuo Su
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Adela Martinkova
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Emma O'Donnell
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Christof A Leicht
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK.
| |
Collapse
|
7
|
Fukumoto T, Shimosawa T, Yakabe M, Yoshida S, Yoshida Y. Recent advances in biomarkers for senescence: Bridging basic research to clinic. Geriatr Gerontol Int 2025; 25:139-147. [PMID: 39754295 DOI: 10.1111/ggi.15054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/31/2024] [Accepted: 12/14/2024] [Indexed: 01/06/2025]
Abstract
In this review, we review the current status of biomarkers for aging and possible perspectives on anti-aging or rejuvenation from the standpoint of biomarkers. Aging is observed in all cells and organs, and we focused on research into senescence in the skin, musculoskeletal system, immune system, and cardiovascular system. Commonly used biomarkers include SA-βgal, cell-cycle markers, senescence-associated secretory phenotype (SASP) factors, damage-associated molecular patterns (DAMPs), and DNA-damage-related markers. In addition, each organ or cell has its specific markers. Generally speaking, a combination of biomarkers is required to define age-related changes. When considering the translation of basic research, biomarkers that are highly sensitive, highly specific, with validation and reliability as well as being non-invasive are optimal; however, currently reported markers do not fulfill the prerequisite for biomarkers. In addition, rodent models of aging do not necessarily represent human aging, and markers in rodent or cell models are not applicable in clinical settings. The prerequisite of clinically applicable biomarkers is that they provide useful information for clinical decision-making, such as predicting disease risk, diagnosing disease, monitoring disease progression, or guiding treatment decisions. Therefore, the development of non-invasive robust, reliable, and useful biomarkers in humans is necessary to develop anti-aging therapy for humans. Geriatr Gerontol Int 2025; 25: 139-147.
Collapse
Affiliation(s)
- Takeshi Fukumoto
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, Graduate School of Medicine, International University of Health and Welfare, Hyogo, Japan
| | - Mitsutaka Yakabe
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shota Yoshida
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yohko Yoshida
- Department of Advanced Senotherapeutics and Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Azami P, Mohammadzadeh S, Seirafi S, Razeghian-Jahromi I. A review of cutting-edge biomarkers for diagnosing coronary artery disease. Medicine (Baltimore) 2025; 104:e41377. [PMID: 39854741 PMCID: PMC11771658 DOI: 10.1097/md.0000000000041377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Chronic coronary artery disease (CAD) remains a significant global healthcare burden. Current risk assessment methods have notable limitations in early detection and risk stratification. Hence, there is an urgent need for innovative biomarkers that facilitate the premature CAD diagnosis, ultimately leading to reduction in associated morbidity and mortality rates. This review comprehensively examines recent advances in emerging biomarkers for CAD detection. Our analysis delves into various aspects of these biomarkers such as their mechanisms of action, roles in the pathophysiology of the disease, and different measurement techniques employed in clinical practice. Comparative assessment of biomarker performance between CAD patients and control groups was also presented relying on their sensitivity, specificity, and area under the curve at specific cutoff points. In this regard, prominent biomarkers including Tenascin-C, IL-37, PTX3, transthyretin, soluble interleukin-6 receptor α, and miR-15a are identified as having high diagnostic potential for chronic CAD that indeed showcase promising performance metrics. These findings underscore the role of novel biomarkers in enhancing CAD risk stratification and improving patient outcomes through early intervention. However, the pursuit of an ideal and inclusive biomarker continues due to the multifaceted nature of CAD. Future randomized controlled trials are essential to bridge the gap between research findings and clinical practice in order to augment the practical application of these biomarkers in routine healthcare settings.
Collapse
Affiliation(s)
- Pouria Azami
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Soroush Seirafi
- Department of Cardiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
9
|
Bonnefont-Rousselot D. [Lipoprotein-associated phospholipase A 2 (Lp-PLA 2): Relevant biomarker and therapeutic target?]. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:45-57. [PMID: 39241907 DOI: 10.1016/j.pharma.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Over the last fifteen years, numerous studies have sought to decipher the role of lipoprotein-associated phospholipase A2 (Lp-PLA2) in vascular inflammation-related diseases, notably atherosclerosis. Despite the disappointing results of clinical trials using the Lp-PLA2 inhibitor darapladib, new pathophysiological, epidemiological and genetic data have enabled the development of new inhibitors. Recent studies also show that Lp-PLA2 is involved in vascular inflammation-related diseases other than atherosclerosis (ischemic stroke, Alzheimer's disease and vascular dementia, diabetes, cancers…), and inhibition of Lp-PLA2 could have beneficial therapeutic in these diseases. This review aims to present new data on Lp-PLA2 and to evaluate its current interest as a biomarker but also as a therapeutic target.
Collapse
Affiliation(s)
- Dominique Bonnefont-Rousselot
- Service de biochimie métabolique, hôpitaux universitaires Pitié-Salpêtrière-Charles-Foix, AP-HP, 47-83, boulevard de l'Hôpital, 75651 Paris, France; Inserm, CNRS, UFR de pharmacie, UTCBS, université Paris Cité, Paris, France.
| |
Collapse
|
10
|
Gallo A, Le Goff W, Santos RD, Fichtner I, Carugo S, Corsini A, Sirtori C, Ruscica M. Hypercholesterolemia and inflammation-Cooperative cardiovascular risk factors. Eur J Clin Invest 2025; 55:e14326. [PMID: 39370572 PMCID: PMC11628670 DOI: 10.1111/eci.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Maintaining low concentrations of plasma low-density lipoprotein cholesterol (LDLc) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and delays the age at which mature atherosclerotic plaques develop. This substantially reduces the lifetime risk of atherosclerotic cardiovascular disease (ASCVD) events. In this context, plaque development and vulnerability result not only from lipid accumulation but also from inflammation. RESULTS Changes in the composition of immune cells, including macrophages, dendritic cells, T cells, B cells, mast cells and neutrophils, along with altered cytokine and chemokine release, disrupt the equilibrium between inflammation and anti-inflammatory mechanisms at plaque sites. Considering that it is not a competition between LDLc and inflammation, but instead that they are partners in crime, the present narrative review aims to give an overview of the main inflammatory molecular pathways linked to raised LDLc concentrations and to describe the impact of lipid-lowering approaches on the inflammatory and lipid burden. Although remarkable changes in LDLc are driven by the most recent lipid lowering combinations, the relative reduction in plasma C-reactive protein appears to be independent of the magnitude of LDLc lowering. CONCLUSION Identifying clinical biomarkers of inflammation (e.g. interleukin-6) and possible targets for therapy holds promise for monitoring and reducing the ASCVD burden in suitable patients.
Collapse
Affiliation(s)
- Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Wilfried Le Goff
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Raul D. Santos
- Academic Research Organization Hospital Israelita Albert Einstein and Lipid Clinic Heart Institute (InCor)University of Sao Paulo Medical School HospitalSao PauloBrazil
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Stefano Carugo
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Cesare Sirtori
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
11
|
Liu R, Bao J, Tang Y, Xu D, Shen L, Qin H. Changes in Treg cells and cytokines in the peripheral blood of patients with coronary artery disease combined with type 2 diabetes mellitus. Heart Lung 2025; 69:147-154. [PMID: 39426330 DOI: 10.1016/j.hrtlng.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Patients with coronary artery disease (CAD) combined with type 2 diabetes mellitus (T2DM) develop serious atherosclerotic and diffuse lesions. Inadequate numbers and the dysfunction of CD4+CD25+Foxp3+regulatory T lymphocytes (Treg cells) are common mechanisms underlying the immunopathological damage in CAD and T2DM. OBJECTIVES We aimed to explore Treg cell changes in patients with CAD complicated with T2DM and to investigate the association between Treg cells and the severity of CAD. METHODS A total of 257 participants were included in the study, divided into a healthy control group (HC, n = 63), CAD group (n = 106), and CAD complicated with T2DM group (CAD+T2DM, n = 88). Flow cytometry detected Treg cell levels, and serum IL-10, IL-6, and ELISA detected TGF-β. RESULTS The sample for this study consisted of 170 males and 87 females, with 88 (34.24 %) participants having diabetes and 169 (65.76 %) without diabetes. The proportion of circulating Treg cells was lower in the CAD and CAD+T2DM groups than in the HC group, and it was lower in the CAD+T2DM group than in the CAD group. The plasma levels of IL-10 and TGF-β were lower in the CAD than in the HC group, and the levels in the CAD+T2DM group were significantly lower than those in the CAD group. However, the plasma IL-6 level changed in the opposite direction. Gensini's score was negatively correlated with Treg cells (R = - 0.57, P < .05). Subgroup analyses and interaction analyses showed that the association of Treg with the Gensini score was robust. CONCLUSION The level of Treg cells was an independent protective factor for patients with CAD and T2DM and was negatively correlated with the Gensini score. Therefore, Treg cells may be used as therapeutic targets for CAD with T2DM patients.
Collapse
Affiliation(s)
- Runqi Liu
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Institute for Global Health, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Jinghui Bao
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Ying Tang
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Danyan Xu
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Li Shen
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Huali Qin
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
12
|
Bahrami P, Aromolaran KA, Aromolaran AS. Mechanistic Relevance of Ventricular Arrhythmias in Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2024; 25:13423. [PMID: 39769189 PMCID: PMC11677834 DOI: 10.3390/ijms252413423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing at an alarming rate worldwide, with limited effective therapeutic interventions in patients. Sudden cardiac death (SCD) and ventricular arrhythmias present substantial risks for the prognosis of these patients. Obesity is a risk factor for HFpEF and life-threatening arrhythmias. Obesity and its associated metabolic dysregulation, leading to metabolic syndrome, are an epidemic that poses a significant public health problem. More than one-third of the world population is overweight or obese, leading to an enhanced risk of incidence and mortality due to cardiovascular disease (CVD). Obesity predisposes patients to atrial fibrillation and ventricular and supraventricular arrhythmias-conditions that are caused by dysfunction in the electrical activity of the heart. To date, current therapeutic options for the cardiomyopathy of obesity are limited, suggesting that there is considerable room for the development of therapeutic interventions with novel mechanisms of action that will help normalize sinus rhythms in obese patients. Emerging candidates for modulation by obesity are cardiac ion channels and Ca-handling proteins. However, the underlying molecular mechanisms of the impact of obesity on these channels and Ca-handling proteins remain incompletely understood. Obesity is marked by the accumulation of adipose tissue, which is associated with a variety of adverse adaptations, including dyslipidemia (or abnormal systemic levels of free fatty acids), increased secretion of proinflammatory cytokines, fibrosis, hyperglycemia, and insulin resistance, which cause electrical remodeling and, thus, predispose patients to arrhythmias. Furthermore, adipose tissue is also associated with the accumulation of subcutaneous and visceral fat, which is marked by distinct signaling mechanisms. Thus, there may also be functional differences in the effects of the regional distribution of fat deposits on ion channel/Ca-handling protein expression. Evaluating alterations in their functional expression in obesity will lead to progress in the knowledge of the mechanisms responsible for obesity-related arrhythmias. These advances are likely to reveal new targets for pharmacological modulation. Understanding how obesity and related mechanisms lead to cardiac electrical remodeling is likely to have a significant medical and economic impact. Nevertheless, substantial knowledge gaps remain regarding HFpEF treatment, requiring further investigations to identify potential therapeutic targets. The objective of this study is to review cardiac ion channel/Ca-handling protein remodeling in the predisposition to metabolic HFpEF and arrhythmias. This review further highlights interleukin-6 (IL-6) as a potential target, cardiac bridging integrator 1 (cBIN1) as a promising gene therapy agent, and leukotriene B4 (LTB4) as an underappreciated pathway in future HFpEF management.
Collapse
Affiliation(s)
- Pegah Bahrami
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
| | - Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
13
|
Li Z, Miao L, Zhang T, Thomas AM, Li S. Causal relationship of inflammatory cytokines and serum metabolites in cerebral small vessel disease: a two-step Mendelian randomization study. Eur J Neurol 2024; 31:e16443. [PMID: 39150083 PMCID: PMC11555141 DOI: 10.1111/ene.16443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND AND PURPOSE The aim was to investigate the causal relationships of inflammatory cytokines and serum metabolites in cerebral small vessel disease (CSVD). METHODS Bidirectional Mendelian randomization was first conducted to screen inflammatory cytokines and serum metabolites that were associated with imaging features of CSVD, including white matter hyperintensities, recent small subcortical infarcts, cortical cerebral microinfarcts, cerebral microbleeds, lacunes and enlarged perivascular spaces. Sensitivity analyses were performed to evaluate the robustness and pleiotropy of these results. Subsequently, inflammatory cytokines and serum metabolites that were associated with CSVD were subjected to functional enrichment. Finally, mediation analysis was employed to investigate whether inflammatory cytokines or serum metabolites acted as an intermediary for the other in their causal relationship with CSVD. RESULTS Of the inflammatory cytokines, five were risk factors (e.g., tumour-necrosis-factor-related apoptosis-inducing ligand) and five (e.g., fibroblast growth factor 19) were protective factors for CSVD. Eleven serum metabolites that increased CSVD risk and 13 metabolites that decreased CSVD risk were also identified. The majority of these markers of CSVD susceptibility were lipid metabolites. Natural killer cell receptor sub-type 2B4 was determined to act as a mediating factor of an unidentified metabolite for the enlargement of perivascular spaces. CONCLUSION Several inflammatory cytokines and serum metabolites had causal relationships with imaging features of CSVD. A natural killer cell receptor mediated in part the promotional effect of a metabolite on perivascular space enlargement.
Collapse
Affiliation(s)
- Zidong Li
- Department of Neurology and Psychiatry, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Lu Miao
- Shanxi Academy of Medical SciencesThird Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Tianyi Zhang
- School of Basic Medical SciencesShandong UniversityJinanChina
| | - Aline M. Thomas
- Russell H. Morgan Department of Radiology and Radiological SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- Beijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
| |
Collapse
|
14
|
Wu Y, Li Z, Shi L, Zhu Y, Wang Y, Yan N, Yang Y, He S, Li J. Effects of leachate from disposable plastic takeout containers on the cardiovascular system after thermal contact. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117383. [PMID: 39591733 DOI: 10.1016/j.ecoenv.2024.117383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
The study investigated the cardiovascular effects of daily exposure to plastic products by simulating the oral heat exposure mode of disposable plastic takeout containers (DPTC) commonly used in society. Questionnaires were used to randomly choose 3179 people in order to examine any possible correlation between the frequency of plastic exposure and the risk of cardiovascular diseases (CVD). Additionally, Sprague-Dawley(SD) rats consumed leachate from DPTC exposed to boiling water for 1 minute,5 minutes and 15 minutes respectively, over three months. After intervention, fecal samples were taken for microbiota and metabolomics analysis, and rat cardiac tissue was studied by staining and electron microscopy. Serum parameters were tested to analyze cardiovascular system changes. The population-based plastic exposure questionnaire data revealed that high-frequency exposure to plastics is significantly associated with an increased risk of congestive heart failure, with an odds ratio of 1.13 (95 % CI: 1.03-1.24). Rat fecal analysis revealed that β diversity and composition of gut microbiota in experimental groups were changed. Inflammatory cell infiltration, mitochondrial swelling, and serum indicators of oxidative stress and inflammation were significantly elevated in the myocardium, without temporal differences observed. The study shows plastic exposure as a significant CVD risk factor regardless of duration. It leads to changes in myocardial tissue, gut microbiota, and metabolites, all closely tied to CVD.
Collapse
Affiliation(s)
- Yueping Wu
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Zhuoyuan Li
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Liping Shi
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Yongbin Zhu
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Yanrong Wang
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Ning Yan
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Yue Yang
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Shulan He
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Jiangping Li
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| |
Collapse
|
15
|
Muñoz-García N, Cordero A, Padro T, Mendieta G, Vilahur G, Flores E, Badimon L. First time ACS in patients with on-target lipid levels: Inflammation at admission and re-event rate at follow-up. Eur J Clin Invest 2024; 54:e14305. [PMID: 39159006 DOI: 10.1111/eci.14305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Dyslipidaemia, inflammation and elevated Lp(a) levels are associated with the progression of atherosclerosis. This study investigates whether patients with a first-time presentation of chest pain and on-target LDL-C levels and intermediate FRS/ESC-Score risks, display a high inflammatory burden linked to myocardial injury and whether inflammation at admission affects the re-event rate up to 6 years follow-up. METHODS Blind assessments of novel inflammatory markers such as Glycoprotein A and B via nuclear magnetic resonance (NMR), cytokines, hsCRP, Neutrophil-to-Lymphocyte ratio (NLR) and Lipoprotein(a) levels were examined. Out of 198 chest pain patients screened, 97 met the inclusion criteria at admission. RESULTS cTnI(+) patients (>61 ng/L) with elevated Lipoprotein(a), showed significantly increased levels of Glycoprotein A and B, hsCRP, IL-6, a high NLR and a reduced left ventricular ejection fraction (%) compared to cTnI(-) individuals. Those patients, with a higher inflammatory burden at hospital admission (hsCRP, IL-6, Glycoprotein A and B, and Lipoprotein(a)) had a higher re-event rate at follow-up. CONCLUSIONS Inflammation and Lipoprotein(a) levels were particularly prominent in patients presenting with reduced left ventricular ejection fraction. Notably, Glycoproteins A/B emerge as novel markers of inflammation in these patients. Our study highlights the significantly higher impact of inflammatory burden in patients with chest pain and high level of myocardial damage than in those with lower myocardial affectation, even when they all had lipid levels well controlled. Inflammation at the time of admission influenced the re-event rate over a follow-up period of up to 6 years.
Collapse
Affiliation(s)
- Natàlia Muñoz-García
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, Barcelona, Spain
- Medical School, Universtitat de Barcelona, Barcelona, Spain
| | - Alberto Cordero
- Cardiology Department, Hospital IMED Elche, Elche, Spain
- Unidad de Investigación en Cardiología. Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), València, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Teresa Padro
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Guiomar Mendieta
- Cardiology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Gemma Vilahur
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Emilio Flores
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departamento de Análisis Clínicos, Hospital Universitario de San Juan, Alicante, Spain
| | - Lina Badimon
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiovascular Research Chair, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
16
|
Mehta NN, deGoma E, Shapiro MD. IL-6 and Cardiovascular Risk: A Narrative Review. Curr Atheroscler Rep 2024; 27:12. [PMID: 39589436 PMCID: PMC11599326 DOI: 10.1007/s11883-024-01259-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE OF REVIEW The objective of this narrative review is to summarize data from recently published prospective observational studies that analyze the association between circulating interleukin-6 (IL-6) levels and cardiovascular clinical or imaging endpoints. RECENT FINDINGS Higher levels of IL-6 are associated with a higher risk of cardiovascular death, major adverse cardiovascular events, myocardial infarction, stroke, peripheral artery disease, and heart failure. Imaging studies have also shown an association between IL-6 and carotid intima-media thickness progression, carotid plaque progression, severity, and vulnerability. These observations have been consistent across a wide range of study populations and after adjusting for traditional and emerging risk factors including high-sensitivity C-reactive protein. Robust epidemiologic evidence supports IL-6 as a central mediator of cardiovascular risk along with human genetic studies and mechanistic experiments. Ongoing clinical studies are testing the therapeutic hypothesis of IL-6 inhibition in patients with atherosclerotic cardiovascular disease or heart failure.
Collapse
Affiliation(s)
- Nehal N Mehta
- The George Washington University School of Medicine, Washington, DC, USA
| | | | | |
Collapse
|
17
|
Stewart RAH, Robledo KP, Tonkin AM, Keech A, Kritharides L, Marschner I, Janus E, Thompson PL, Watts GF, Zeller T, White HD, Simes J. Plasma Protein Biomarkers and Long-Term Cardiovascular Mortality Risk in Patients With Chronic Coronary Heart Disease. J Am Heart Assoc 2024; 13:e034367. [PMID: 39450716 PMCID: PMC11935700 DOI: 10.1161/jaha.123.034367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/23/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Protein biomarkers that reflect different pathophysiological pathways have been associated with the risk of adverse cardiovascular events. However, it is uncertain whether these associations are sustained with increasing years after the biomarkers are measured. METHODS AND RESULTS In this cohort study, 7745 patients with coronary heart disease who participated in the LIPID (Long-Term Intervention With Pravastatin in Ischemic Disease) trial, BNP (B-type natriuretic peptide), troponin I, cystatin-C, C-reactive protein, d-dimer and midregional proadrenomedullin were measured at baseline and after 1 year. Discrimination of plasma biomarker concentrations for cardiovascular death were evaluated in landmark analyses from 1 year for the next 5 years of the randomized trial, and for 10 additional years after trial completion. All 6 biomarkers were associated with risk of cardiovascular death (n=1903) both during and after the clinical trial (each P<0.001). C-statistics for BNP were 0.706 and 0.704; cystatin-C, 0.686 and 0.693; troponin I, 0.686 and 0.689; C-reactive protein, 0.655 and 0.684; d-dimer, 0.670 and 0.679, and midregional adrenomedullin, 0.686 and 0.688, respectively. In multivariable models, adding all 6 biomarkers to models with clinical risk factors increased the C-statistic for cardiovascular death from 0.709 to 0.775 during the clinical trial, and from 0.713 to 0.751 during 10-year follow-up after the randomized trial (P<0.001 for both). CONCLUSIONS In patients with chronic coronary heart disease, biomarkers that reflect different pathophysiological pathways are associated with the risk of cardiovascular death for at least the next 15 years.
Collapse
Affiliation(s)
- Ralph A. H. Stewart
- Green Lane Cardiovascular Service, Auckland City Hospital, Te Toka Tumai, Te Whatu Ora—Health New ZealandAucklandNew Zealand
| | - Kristy P. Robledo
- Faculty Medicine and Health, NHMRC Clinical Trials CentreUniversity of Syndey and The Royal Prince Alfred HospitalCamperdownNSWAustralia
| | - Andrew M. Tonkin
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVICAustralia
| | - Anthony Keech
- Faculty Medicine and Health, NHMRC Clinical Trials CentreUniversity of Syndey and The Royal Prince Alfred HospitalCamperdownNSWAustralia
| | - Leonard Kritharides
- Department of Cardiology, Concord HospitalThe University of SydneyConcordNSWAustralia
- ANZAC Medical Research Institute, Faculty of Medicine, University of SydneyConcordNSWAustralia
| | - Ian Marschner
- Faculty Medicine and Health, NHMRC Clinical Trials CentreUniversity of Syndey and The Royal Prince Alfred HospitalCamperdownNSWAustralia
| | - Edward Janus
- Western Health Chronic Disease Alliance and Department of Medicine, Western Health—Melbourne Medical SchoolUniversity of MelbourneParkvilleVic3010Australia
| | - Peter L. Thompson
- School of Population and Global HealthThe University of Western AustraliaPerthWAAustralia
| | - Gerald F. Watts
- Medical SchoolThe University of Western AustraliaPerthWAAustralia
| | - Tanja Zeller
- University Heart Centre HamburgHamburgGermany
- Department of General and Interventional Cardiology, German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Campus ResearchHamburgGermany
| | - Harvey D. White
- Green Lane Cardiovascular Service, Auckland City Hospital, Te Toka Tumai, Te Whatu Ora—Health New ZealandAucklandNew Zealand
| | - John Simes
- Faculty Medicine and Health, NHMRC Clinical Trials CentreUniversity of Syndey and The Royal Prince Alfred HospitalCamperdownNSWAustralia
| |
Collapse
|
18
|
Boieriu AM, Dumitrel Luca C, Neculoiu CD, Ţînţ D. The impact of inflammatory and oxidative stress biomarkers on the sympathetic nervous system in severe coronary atherosclerosis. Front Cardiovasc Med 2024; 11:1480925. [PMID: 39469122 PMCID: PMC11513282 DOI: 10.3389/fcvm.2024.1480925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
Objective The present study aimed at evaluating the association between sympathetic nervous system activation (SNS) and the severity of coronary artery disease (CAD). In addition, we tested the hypothesis that inflammation and oxidative stress influence the SNS activation. Methods Adult patients with severe CAD scheduled for coronary artery bypass graft (CABG) surgery were enrolled. SYNTAX I score was calculated based on coronary angiography. Systemic activation of the SNS was estimated through circulating levels of norepinephrine (NE). Plasma levels of pro-inflammatory cytokines (IL 1β, IL 6 and HIF 1α) and oxidative stress molecules (SOD-1 and LOX-1) were obtained prior to surgery. Results Circulating NE levels were significantly correlated with the severity of CAD, as assessed by the SYNTAX I score (p 0.002; r 0.329). Elevated levels of circulating pro-inflammatory markers were significantly correlated with increased NE concentrations (for IL-1β: p < 0.001, r = 0.49; for IL-6 and NE: p = 0.003, r = 0.32; for HIF-1α and NE: p = 0.049, r = 0.21). Additionally, oxidative stress molecules were associated with circulating NE levels (for SOD-1 and NE: p = 0.016, r = 0.26; for LOX-1 and NE: p = 0.004, r = 0.31). Conclusion In patients with CAD referred for CABG, SNS activation, indicated by plasma NE levels, was correlated with disease severity as assessed by the SYNTAX I score, as well as with markers of inflammation and oxidative stress. This suggests that inflammation, oxidative stress, and SNS activation form an interconnected network, with each component influencing the others. It might be of interest to develop a scoring system including inflammation and oxidative stress markers to identify patients that require a more aggressive approach to lower inflammation, oxidative stress and modulate the sympathetic nervous system. This could be of use especially in the setting of a scheduled intervention -such as CABG surgery.
Collapse
Affiliation(s)
- Alexandra Maria Boieriu
- Faculty of Medicine, Transilvania University of Brasov, Brasov, Romania
- Department of Cardiology, Emergency County Hospital, Brasov, Romania
| | - Cezar Dumitrel Luca
- Faculty of Medicine, Transilvania University of Brasov, Brasov, Romania
- Cardiovascular Rehabilitation Hospital Covasna, Covasna, Romania
| | | | - Diana Ţînţ
- Faculty of Medicine, Transilvania University of Brasov, Brasov, Romania
- Department of Cardiology, ICCO Clinics, Brasov, Romania
| |
Collapse
|
19
|
Jiang W, Huang G, Du J, Yang H, Zhou S, Dai D, Tang K, Fang L, Wang X, Deng X. White blood cell counts can predict 4-year cardiovascular disease risk in patients with stable coronary heart disease: a prospective cohort study. Front Cardiovasc Med 2024; 11:1358378. [PMID: 39390990 PMCID: PMC11464350 DOI: 10.3389/fcvm.2024.1358378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Background The prevalence of cardiovascular disease has increased sharply in the Asian population, and evaluation of the risk of cardiovascular events with stable coronary heart disease remains challenging. The role of white blood cell (WBC) count in assisting clinical decision-making in this setting is still unclear. Objectives This study sought to evaluate the prognostic meaning of WBC count among patients with stable coronary heart disease. Methods This study included Asian participants (n = 1,933) from the prospective STABILITY (Stabilization of Atherosclerotic Plaque by Initiation of Darapladib Therapy) trial, which involved 15,828 patients with stable coronary heart disease with 3-5 years of follow-up on optimal secondary preventive treatment. WBC count was measured at baseline. Associations between WBC count and cardiovascular outcomes were evaluated by Cox regression analyses with multivariable adjustments. Hematologic emergencies in patients may introduce potential bias. Results In the lower WBC count quartiles, patients had lower-risk clinical profiles. Higher WBC counts were associated with greater event probabilities for cardiovascular death, major cardiovascular events, or all-cause death. In Cox regression models, WBC counts were an independent predictor of major adverse cardiovascular events (OR = 2.445, 95% CI 1.427-4.190, P = 0.001) for the primary outcomes. For the secondary outcomes, including the composite of all-cause death, cardiovascular death, myocardial infarction, stroke, and hospitalization for heart failure, WBC counts were significantly predictive of events with similar magnitude (OR = 1.716, 95% CI 1.169-2.521, P = 0.006). Conclusions In patients with stable coronary heart disease, higher WBC counts were associated with a heightened risk for the primary or secondary outcomes. Registration https://clinicaltrials.gov/; Unique identifier NCT00799903.
Collapse
Affiliation(s)
- Wencai Jiang
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| | - Gang Huang
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| | - Jinfeng Du
- Department of Hematology, Chongqing Emergency Medical Center, Chongqing, China
| | - Hanxuan Yang
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| | - Shiheng Zhou
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| | - Dayin Dai
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| | - Kai Tang
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| | - Lingxiao Fang
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| | - Xiao Wang
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| | - Xuejun Deng
- Department of Cardiology, Suining Central Hospital, Suining, Sichuan, China
| |
Collapse
|
20
|
Ou G, Cai H, Yao K, Qiu Z, Yang Y, Chen Y, Chen X. Exploring the therapeutic potential of interleukin-6 receptor blockade in cardiovascular disease treatment through Mendelian randomization. Sci Rep 2024; 14:21452. [PMID: 39271913 PMCID: PMC11399143 DOI: 10.1038/s41598-024-72195-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Interleukin-6 (IL-6) plays a crucial role in the pathogenesis of cardiovascular disease (CVD), and IL-6 receptor (IL-6R) blockade has emerged as a promising therapeutic option. However, their specific therapeutic effects in different types of CVDs remain unclear. This study aimed to assess the efficacy of IL-6R blockade in the management of various CVDs, including hypertension (HTN), coronary heart disease (CHD), myocardial infarction (MI), atrial fibrillation (AF), and heart failure (HF). The Mendelian randomization (MR) approach was utilized to investigate the therapeutic impact of IL-6R blockade on HTN, CHD, MI, AF, and HF based on the genome-wide association study (GWAS) summary statistics. MR-Egger intercept test, Cochran's Q test, and leave-one-out analysis were used for sensitivity analysis to verify the reliability of the MR results. The Bonferroni method was used to correct for bias caused by multiple comparisons. Inverse variance weighted (IVW) results demonstrated that IL-6R blockade significantly influenced CHD (odds ratio (OR) = 0.757, 95% confidence interval (CI): 0.690 - 0.832, P = 5.804 × 10-9) and MI (OR = 0.840, 95% CI: 0.744 - 0.949, P = 0.005). However, IL-6R blockade had no significant effect on HTN (OR = 1.015, 95% CI: 0.950 - 1.084, P = 0.663), AF (OR = 0.905, 95% CI: 0.800 - 1.025, P = 0.116) and HF (OR = 1.012, 95% CI: 0.921 - 1.113, P = 0.805). Genetically predicted IL-6R blockade was associated with a protective effect on CHD and MI, but not HTN, AF and HF. This study's findings offer valuable insights for tailoring IL-6R blockade treatment for different types of CVD, and serve as a reference for future research.
Collapse
Affiliation(s)
- Guangyang Ou
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Huzhi Cai
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Kunpeng Yao
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Zerui Qiu
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Yang Yang
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Yaowu Chen
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Xinyu Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China.
| |
Collapse
|
21
|
Gao M, Wen W, Li H, Zheng Y, Yun M, Meng J, Wang S, Wang B, Hu B, Mou T, Yu Y, Zhang X, Li X. Coronary sodium [ 18F]fluoride activity predicts outcomes post-CABG: a comparative evaluation with conventional metrics. Eur J Nucl Med Mol Imaging 2024; 51:3235-3251. [PMID: 38730084 DOI: 10.1007/s00259-024-06736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE The value of preoperative multidisciplinary approach remains inadequately delineated in forecasting postoperative outcomes of patients undergoing coronary artery bypass grafting (CABG). Herein, we aimed to ascertain the efficacy of multi-modality cardiac imaging in predicting post-CABG cardiovascular outcomes. METHODS Patients with triple coronary artery disease underwent cardiac sodium [18F]fluoride ([18F]NaF) positron emission tomography/computed tomography (PET/CT), coronary angiography, and CT-based coronary artery calcium scoring before CABG. The maximum coronary [18F]NaF activity (target-to-blood ratio [TBR]max) and the global coronary [18F]NaF activity (TBRglobal) was determined. The primary endpoint was perioperative myocardial infarction (PMI) within 7-day post-CABG. Secondary endpoint included major adverse cardiac and cerebrovascular events (MACCEs) and recurrent angina. RESULTS This prospective observational study examined 101 patients for a median of 40 months (interquartile range: 19-47 months). Both TBRmax (odds ratio [OR] = 1.445; p = 0.011) and TBRglobal (OR = 1.797; P = 0.018) were significant predictors of PMI. TBRmax>3.0 (area under the curve [AUC], 0.65; sensitivity, 75.0%; specificity, 56.8%; p = 0.036) increased PMI risk by 3.661-fold, independent of external confounders. Kaplan-Meier test revealed a decrease in MACCE survival rate concomitant with an escalating TBRmax. TBRmax>3.6 (AUC, 0.70; sensitivity, 76.9%; specificity, 73.9%; p = 0.017) increased MACCEs risk by 5.520-fold. Both TBRmax (hazard ratio [HR], 1.298; p = 0.004) and TBRglobal (HR = 1.335; p = 0.011) were significantly correlated with recurrent angina. No significant associations were found between CAC and SYNTAX scores and between PMI occurrence and long-term MACCEs. CONCLUSION Quantification of coronary microcalcification activity via [18F]NaF PET displayed a strong ability to predict early and long-term post-CABG cardiovascular outcomes, thereby outperforming conventional metrics of coronary macrocalcification burden and stenosis severity. TRIAL REGISTRATION The trial was registered with the Chinese Clinical Trial Committee (number: ChiCTR1900022527; URL: www.chictr.org.cn/showproj.html?proj=37933 ).
Collapse
Affiliation(s)
- Mingxin Gao
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Wanwan Wen
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Haiyang Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Yaqi Zheng
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Mingkai Yun
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Jingjing Meng
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Shipan Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Bolin Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Biao Hu
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Tiantian Mou
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Yang Yu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China.
| | - Xiaoli Zhang
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China.
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Chertow GM, Chang AM, Felker GM, Heise M, Velkoska E, Fellström B, Charytan DM, Clementi R, Gibson CM, Goodman SG, Jardine M, Levin A, Lokhnygina Y, Mears J, Mehran R, Stenvinkel P, Wang AYM, Wheeler DC, Zoccali C, Ridker PM, Mahaffey KW, Tricoci P, Wolf M. IL-6 inhibition with clazakizumab in patients receiving maintenance dialysis: a randomized phase 2b trial. Nat Med 2024; 30:2328-2336. [PMID: 38796655 PMCID: PMC11333272 DOI: 10.1038/s41591-024-03043-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/28/2024]
Abstract
Inflammation mediated by interleukin-6 (IL-6) is strongly associated with cardiovascular risk. Here we evaluated clazakizumab, a monoclonal antibody targeting the IL-6 ligand, in a phase 2b dose-finding study. Adults with cardiovascular disease and/or diabetes receiving maintenance dialysis with high-sensitivity C-reactive protein (hs-CRP) ≥ 2 mg l-1 at baseline were randomized to receive clazakizumab (2.5 mg, 5 mg or 10 mg, n = 32 per dose group) or placebo (n = 31) every 4 weeks. The primary endpoint was the change from baseline in hs-CRP to week 12, expressed as the geometric mean ratio. Clazakizumab treatment signficantly reduced serum hs-CRP concentrations at week 12 by 86%, 90% and 92% relative to placebo in patients randomized to 2.5 mg, 5 mg or 10 mg clazakizumab, respectively (all P < 0.0001), meeting the primary outcome. With regard to secondary endpoints, clazakizumab treatment reduced serum fibrinogen, amyloid A, secretory phospholipase A2, and lipoprotein(a) concentrations, as well as increased mean serum albumin concentrations at 12 weeks, relative to placebo. The proportion of patients who achieved hs-CRP < 2.0 mg l-1 was 79%, 82% and 79% in the 2.5 mg, 5 mg and 10 mg clazakizumab groups, respectively, compared with 0% of placebo-treated patients. With regard to safety, no cases of sustained grade 3 or 4 thrombocytopenia or neutropenia were observed. Serious infections were seen with similar frequency in the placebo, clazakizumab 2.5 mg and clazakizumab 5 mg groups, but were numerically more frequent in the clazakizumab 10 mg group. The results of this trial indicate that in patients receiving maintenance dialysis, clazakizumab reduced inflammatory biomarkers associated with cardiovascular events. ClinicalTrials.gov registration: NCT05485961 .
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Shaun G Goodman
- University of Toronto and University of Alberta, Edmonton, Alberta, Canada
| | - Meg Jardine
- University of Sydney, Sydney, New South Wales, Australia
| | - Adeera Levin
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Knopp T, Jung R, Wild J, Bochenek ML, Efentakis P, Lehmann A, Bieler T, Garlapati V, Richter C, Molitor M, Perius K, Finger S, Lagrange J, Ghasemi I, Zifkos K, Kommoss KS, Masri J, Reißig S, Randriamboavonjy V, Wunderlich T, Hövelmeyer N, Weber ANR, Mufazalov IA, Bosmann M, Bechmann I, Fleming I, Oelze M, Daiber A, Münzel T, Schäfer K, Wenzel P, Waisman A, Karbach S. Myeloid cell-derived interleukin-6 induces vascular dysfunction and vascular and systemic inflammation. EUROPEAN HEART JOURNAL OPEN 2024; 4:oeae046. [PMID: 39015379 PMCID: PMC11250217 DOI: 10.1093/ehjopen/oeae046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 11/29/2023] [Accepted: 05/03/2024] [Indexed: 07/18/2024]
Abstract
Aims The cytokine interleukin-6 (IL-6) plays a central role in the inflammation cascade as well as cardiovascular disease progression. Since myeloid cells are a primary source of IL-6 formation, we aimed to generate a mouse model to study the role of myeloid cell-derived IL-6 in vascular disease. Methods and results Interleukin-6-overexpressing (IL-6OE) mice were generated and crossed with LysM-Cre mice, to generate mice (LysM-IL-6OE mice) overexpressing the cytokine in myeloid cells. Eight- to 12-week-old LysM-IL-6OE mice spontaneously developed inflammatory colitis and significantly impaired endothelium-dependent aortic relaxation, increased aortic reactive oxygen species (ROS) formation, and vascular dysfunction in resistance vessels. The latter phenotype was associated with decreased survival. Vascular dysfunction was accompanied by a significant accumulation of neutrophils, monocytes, and macrophages in the aorta, increased myeloid cell reactivity (elevated ROS production), and vascular fibrosis associated with phenotypic changes in vascular smooth muscle cells. In addition to elevated Mcp1 and Cxcl1 mRNA levels, aortae from LysM-IL-6OE mice expressed higher levels of inducible NO synthase and endothelin-1, thus partially accounting for vascular dysfunction, whereas systemic blood pressure alterations were not observed. Bone marrow (BM) transplantation experiments revealed that vascular dysfunction and ROS formation were driven by BM cell-derived IL-6 in a dose-dependent manner. Conclusion Mice with conditional overexpression of IL-6 in myeloid cells show systemic and vascular inflammation as well as endothelial dysfunction. A decrease in circulating IL-6 levels by replacing IL-6-producing myeloid cells in the BM improved vascular dysfunction in this model, underpinning the relevant role of IL-6 in vascular disease.
Collapse
Affiliation(s)
- Tanja Knopp
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Rebecca Jung
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute of Molecular Medicine, University Medical Center Mainz, Mainz, Germany
| | - Johannes Wild
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhine-Main, Germany
| | - Magdalena L Bochenek
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhine-Main, Germany
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Annika Lehmann
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Tabea Bieler
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Venkata Garlapati
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Cindy Richter
- Institute of Anatomy, University Medical Center Leipzig, Leipzig, Germany
- Institute of Neuroradiology, University Medical Center, Leipzig, Germany
| | - Michael Molitor
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhine-Main, Germany
| | - Katharina Perius
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Jérémy Lagrange
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Iman Ghasemi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Konstantinos Zifkos
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katharina S Kommoss
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Joumana Masri
- Institute of Molecular Medicine, University Medical Center Mainz, Mainz, Germany
| | - Sonja Reißig
- Institute of Molecular Medicine, University Medical Center Mainz, Mainz, Germany
| | | | - Thomas Wunderlich
- Max Planck Institute for Metabolism Research Cologne, Cologne, Germany
| | - Nadine Hövelmeyer
- Institute of Molecular Medicine, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Alexander N R Weber
- Department of Innate Immunity, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Ilgiz A Mufazalov
- Institute of Molecular Medicine, University Medical Center Mainz, Mainz, Germany
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Department of Medicine, Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ingo Bechmann
- Institute of Anatomy, University Medical Center Leipzig, Leipzig, Germany
| | - Ingrid Fleming
- German Center for Cardiovascular Research (DZHK), Partner site Rhine-Main, Germany
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Matthias Oelze
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhine-Main, Germany
| | - Katrin Schäfer
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhine-Main, Germany
| | - Philip Wenzel
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhine-Main, Germany
| | - Ari Waisman
- Institute of Molecular Medicine, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Susanne Karbach
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhine-Main, Germany
| |
Collapse
|
24
|
Lake JE, Feng H, Hyatt AN, Miao H, Debroy P, Funderburg N, Ailstock K, Dobs A, Haberlen S, Magnani JW, Margolick JB, McGowan K, Palella FJ, Witt MD, Bhasin S, Budoff MJ, Post WS, Brown TT. Transgender Women With Suppressed Testosterone Display Lower Burden of Coronary Disease Than Matched Cisgender Men. J Endocr Soc 2024; 8:bvae120. [PMID: 38974987 PMCID: PMC11223995 DOI: 10.1210/jendso/bvae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Indexed: 07/09/2024] Open
Abstract
Context Cardiovascular disease (CVD) in transgender women (TW) may be affected by gender-affirming hormone therapy (GAHT) and HIV, but few data compare TW on contemporary GAHT to well-matched controls. Objective We compared CVD burden and biomarker profiles between TW and matched cisgender men (CM). Methods Adult TW on GAHT (n = 29) were recruited for a cross-sectional study (2018-2020). CM (n = 48) from the former Multicenter AIDS Cohort Study were matched 2:1 to TW on HIV serostatus, age ±5 years, race/ethnicity, BMI category and antiretroviral therapy (ART) type. Cardiac parameters were measured by CT and coronary atherosclerosis by coronary CT angiography; sex hormone and biomarker concentrations were measured centrally from stored samples. Results Overall, median age was 53 years and BMI 29 kg/m2; 69% were non-white. All participants with HIV (71%) had viral suppression on ART. Only 31% of TW had testosterone suppression (<50 ng/dL, TW-S). Traditional CVD risk factors were similar between groups, except that TW-S had higher BMI than TW with non-suppressed testosterone (TW-T). TW-S had no evidence of non-calcified coronary plaque or advanced coronary stenosis, whereas TW-T and CM had similar burden. TW had lower prevalence of any coronary plaque, calcified plaque and mixed plaque than CM, regardless of testosterone concentrations and HIV serostatus. Estradiol but not testosterone concentrations moderately and negatively correlated with the presence of coronary plaque and stenosis. Small sample size limited statistical power. Conclusion Older TW with suppressed total testosterone on GAHT had no CT evidence of non-calcified coronary plaque or advanced coronary stenosis. Longitudinal studies to understand relationships between GAHT and CVD risk in TW are needed.
Collapse
Affiliation(s)
- Jordan E Lake
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Han Feng
- Tulane Research and Innovation for Arrhythmia Discoveries-TRIAD Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ana N Hyatt
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Hongyu Miao
- Department of Statistics and College of Nursing, Florida State University, Tallahassee, FL 32306, USA
| | - Paula Debroy
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Nicholas Funderburg
- Division of Medical Laboratory Science, The Ohio State University, Columbus, OH 43210, USA
| | - Kate Ailstock
- Division of Medical Laboratory Science, The Ohio State University, Columbus, OH 43210, USA
| | - Adrian Dobs
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sabina Haberlen
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jared W Magnani
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Joseph B Margolick
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kate McGowan
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Frank J Palella
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mallory D Witt
- Department of Medicine, Lundquist Institute, Torrance, CA 90502, USA
| | - Shalender Bhasin
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Matthew J Budoff
- Department of Medicine, Lundquist Institute, Torrance, CA 90502, USA
| | - Wendy S Post
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Todd T Brown
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
25
|
Liu S, Ni S, Wang C, Yang K, Yang Y, Li L, Liu J, Wang Y, Qin Y, Zhang M. Association of serum cytokines with coronary chronic total occlusion and their role in predicting procedural outcomes. Hellenic J Cardiol 2024; 78:25-35. [PMID: 37652147 DOI: 10.1016/j.hjc.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Cytokines are strongly associated with coronary artery disease (CAD); however, few studies have explored the relevance of cytokines in coronary chronic total occlusion (CTO). This study aimed to clarify the association of cytokines with CTO and its procedural outcomes. METHODS A total of 526 patients with suspected CAD but not acute myocardial infarction were enrolled and divided into CTO (n = 122) and non-CTO (n = 404) groups based on coronary angiography. Furthermore, serum levels of 12 cytokines [Interleukin-1β (IL-1β), IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, IL-17, tumor necrosis factor-α (TNF-α), interferon-α (IFN-α), and IFN-γ] were measured for each patient. RESULTS Patients with CTO had higher rates of male (P = 0.001), smoking (P = 0.014), and diabetes (P = 0.008); higher levels of IL-6 (P < 0.001), total triglycerides (P = 0.020), serum creatine (P = 0.001), and high-sensitivity troponin I (P = 0.001); and lower IL-4 (P < 0.001), total cholesterol (P = 0.027), and high-density lipoprotein cholesterol (HDL-C) (P < 0.001) levels compared to those without CTO. IL-4 (OR = 0.216, 95%CI:0.135-0.345, P < 0.001), IL-6 (OR = 1.248, 95%CI:1.165-1.337, P < 0.001), and HDL-C (OR = 0.047, 95%CI:0.010-0.221, P < 0.001) were identified as independent predictors of CTO. And good predictive performance (AUC = 0.876) for CTO, with a sensitivity of 81.96% and specificity of 81.19%, could be achieved by combining these three predictors. Furthermore, patients with procedural success had younger age (P = 0.004) and lower serum IL-6 levels (P = 0.039) compared to those with procedural failure, and IL-6 levels (OR = 0.962, 95%CI: 0.931-0.995, P = 0.023) were associated with procedural success. CONCLUSION IL-4, IL-6, and HDL-C levels were strongly associated with CTO, and IL-6 also linked to procedural outcomes of CTO.
Collapse
Affiliation(s)
- Sheng Liu
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Siyao Ni
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengyang Wang
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Kexin Yang
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yunxiao Yang
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Surgical Center of Structural Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Li Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Xicheng District, Beijing, China
| | - Jinkai Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Wang
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanwen Qin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ming Zhang
- Center for Coronary Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Bay B, Vogel B, Sharma R, Sartori S, Leone PP, Nathani M, Oliva A, Smith KF, Hooda A, Sweeny J, Dangas G, Kini A, Krishnan P, Sharma SK, Mehran R. Inflammatory risk and clinical outcomes according to polyvascular atherosclerotic disease status in patients undergoing PCI. Clin Res Cardiol 2024:10.1007/s00392-024-02471-w. [PMID: 38900274 DOI: 10.1007/s00392-024-02471-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Individuals suffering from polyvascular atherosclerotic disease (PolyVD) face a higher likelihood of adverse cardiovascular events. Additionally, inflammation, assessed by high-sensitivity C-reactive protein (hsCRP), affects residual risk following percutaneous coronary intervention (PCI). We aimed to explore the interplay between PolyVD and hsCRP in terms of clinical outcomes after PCI. METHODS Patients undergoing PCI for chronic coronary disease at a tertiary center between January 2012 and February 2020 were included for the current analysis. PolyVD was defined by additional history of cerebrovascular and/or peripheral artery disease. HsCRP levels were defined as elevated when the measured baseline concentration was > 3 mg/L. The primary outcome of interest was major adverse cardiovascular events (MACE), a composite of all-cause mortality, spontaneous MI, or target vessel revascularization. RESULTS Overall, 10,359 participants were included in the current study, with 17.4% affected by PolyVD and 82.6% included in the non-PolyVD subgroup. Patients with PolyVD had higher hsCRP levels than those without. Among the PolyVD group, a larger proportion (33.6%) exhibited elevated hsCRP compared to the non-PolyVD group (24.7%). Patients with both PolyVD and elevated hsCRP levels had significantly higher adverse event rates than all other subgroups at 1-year follow-up. Furthermore, an independent association between elevated hsCRP and MACE was observed within the PolyVD population, while this was not the case for individuals without PolyVD. CONCLUSION A residual risk of adverse outcomes after PCI linked to inflammation appears to be present among individuals with PolyVD. This could help define further target populations for anti-inflammatory treatment options.
Collapse
Affiliation(s)
- Benjamin Bay
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Birgit Vogel
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Raman Sharma
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Samantha Sartori
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Pier Pasquale Leone
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Mashal Nathani
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Angelo Oliva
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Kenneth F Smith
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Amit Hooda
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Joseph Sweeny
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - George Dangas
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Annapoorna Kini
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Prakash Krishnan
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Samin K Sharma
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Roxana Mehran
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA.
| |
Collapse
|
27
|
Panova-Noeva M, Koeck T, Schoelch C, Schulz A, Prochaska JH, Michal M, Strauch K, Schuster AK, Lackner KJ, Münzel T, Hennige AM, Wild PS. Obesity-related inflammatory protein signature in cardiovascular clinical outcomes: results from the Gutenberg Health Study. Obesity (Silver Spring) 2024; 32:1198-1209. [PMID: 38664310 DOI: 10.1002/oby.24014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/23/2024] [Accepted: 02/14/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE The objective of this study was to investigate whether an obesity-related inflammatory protein signature (OIPS) is associated with adverse cardiovascular events. METHODS The Olink Target 96 Inflammation panel was performed in 6662 participants from the population-based Gutenberg Health Study (GHS). The OIPS was selected by a logistic regression model, and its association with cardiovascular outcomes was evaluated by Cox regression analysis. The GHS-derived OIPS was externally validated in the MyoVasc study. RESULTS The identified OIPS entailed 21 proteins involved in chemokine activity, tumor necrosis factor (TNF) receptor binding, and growth factor receptor binding. The signature revealed a novel positive association of axis inhibition protein 1 with obesity. The OIPS was associated with increased risk of all-cause and cardiac deaths, major adverse cardiovascular events, and incident coronary artery disease, independent of clinical covariates and established risk instruments. A BMI-stratified analysis confirmed the association of OIPS with increased death in those with obesity and overweight and with increased risk for coronary artery disease in those with obesity. The association of OIPS with increased risk of all-cause and cardiac deaths was validated in the MyoVasc cohort. CONCLUSIONS The OIPS showed a significant association with adverse clinical outcomes, particularly in those with overweight and obesity, and represents a promising tool for identifying patients at higher risk for worse cardiovascular outcomes.
Collapse
Affiliation(s)
- Marina Panova-Noeva
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
- Center for Thrombosis and Haemostasis, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Koeck
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Corinna Schoelch
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Andreas Schulz
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jürgen H Prochaska
- Center for Thrombosis and Haemostasis, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Matthias Michal
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Konstantin Strauch
- Institute for Medical Biometrics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander K Schuster
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Karl J Lackner
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Münzel
- Center for Thrombosis and Haemostasis, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
- Department of Cardiology-Cardiology I, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anita M Hennige
- Therapeutic Area CardioMetabolism & Respiratory, Boehringer Ingelheim International GmbH, Biberach, Germany
| | - Philipp S Wild
- Center for Thrombosis and Haemostasis, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| |
Collapse
|
28
|
Liu S, Yang R, Zuo Y, Qiao C, Jiang W, Cheng W, Wei W, Liu Z, Geng Y, Dong Y. The association of circulating systemic inflammation with premature death and the protective role of the Mediterranean diet: a large prospective cohort study of UK biobank. BMC Public Health 2024; 24:1449. [PMID: 39118094 PMCID: PMC11312373 DOI: 10.1186/s12889-024-18888-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 05/20/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Although previous studies have identified specific circulating inflammatory markers associated with the risk of mortality, they have often overlooked the broader impact of a comprehensive inflammatory response on health outcomes. This study aims to assess the association between circulating systemic inflammation and age-related hospitalization and premature death, as well as explore the potential mediating effects of various dietary patterns on these associations. METHODS A total of 448,574 participants enrolled in the UK Biobank study were included. Circulating C-reactive protein(CRP), white blood cell count(WBC), platelet count(Plt), and neutrophil/lymphocyte ratio(NLR) were measured, which were used to establish a weighted systemic inflammatory index of inflammation index(INFLA-Score). Dietary intake information was documented through 24-hour dietary recalls, and dietary pattern scores including Dietary Approaches to Stop Hypertension(DASH), Mediterranean(MED), and Healthy Eating Index-2020(HEI-2020) were calculated. Cox proportional hazards regression models were performed to assess the associations between INFLA-Score and age-related disease hospitalization, cause-specific and all-cause premature death. RESULTS During a median follow-up of 12.65 years, 23,784 premature deaths were documented. After adjusting for multiple covariates, higher levels of CRP, WBC, NLR, and INFLA-Score were significantly associated with increased risks of age-related disease hospitalization(HRCRP=1.19; 95%:1.17-1.21; HRWBC=1.17; 95%:1.15-1.19; HRNLR=1.18; 95%:1.16-1.20; HRINFLA-Score=1.19; 95%:1.17-1.21) and premature death(HRCRP=1.68; 95%:1.61-1.75; HRWBC=1.23; 95%:1.18-1.27; HRNLR=1.45; 95%:1.40-1.50; HRINFLA-Score=1.58; 95%:1.52-1.64). Compared to the lowest INFLA-Score group, the highest INFLA-Score group was associated with increased values of whole-body and organ-specific biological age, and had a shortened life expectancy of 2.96 (95% CI 2.53-3.41) and 4.14 (95% CI 3.75-4.56) years at the age of 60 years in women and men, respectively. Additionally, we observed no significant association of the INFLA-Score with aging-related hospitalization and premature death among participants who were more adhering to the Mediterranean (MED) dietary pattern(HRAging-related hospitalization=1.07; 95%:0.99-1.16;HRPremature death=1.19; 95%:0.96-1.47). CONCLUSION A higher INFLA-Score was correlated with an increased risk of age-related hospitalization and premature death. Nevertheless, adherence to a Mediterranean (MED) diet may mitigate these associations.
Collapse
Affiliation(s)
- ShiJian Liu
- Department of kidney, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Ruiming Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, 150081, China
| | - Yingdong Zuo
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, 150081, China
| | - Conghui Qiao
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, 150081, China
| | - Wenbo Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, 150081, China
| | - Weilun Cheng
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Wei Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, 150081, China
| | - Zijie Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, 150081, China
| | - Yiding Geng
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, 150081, China
| | - Ying Dong
- Department of Endocrinology and Metabolic Disease, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
29
|
Trujillo J, Alotaibi M, Seif N, Cai X, Larive B, Gassman J, Raphael KL, Cheung AK, Raj DS, Fried LF, Sprague SM, Block G, Chonchol M, Middleton JP, Wolf M, Ix JH, Prasad P, Isakova T, Srivastava A. Associations of Kidney Functional Magnetic Resonance Imaging Biomarkers with Markers of Inflammation in Individuals with CKD. KIDNEY360 2024; 5:681-689. [PMID: 38570905 PMCID: PMC11146641 DOI: 10.34067/kid.0000000000000437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Key Points Lower baseline apparent diffusion coefficient, indicative of greater cortical fibrosis, correlated with higher baseline concentrations of serum markers of inflammation. No association between baseline cortical R2* and baseline serum markers of inflammation were found. Baseline kidney functional magnetic resonance imaging biomarkers of fibrosis and oxygenation were not associated with changes in inflammatory markers over time, which may be due to small changes in kidney function in the study. Background Greater fibrosis and decreased oxygenation may amplify systemic inflammation, but data on the associations of kidney functional magnetic resonance imaging (fMRI) measurements of fibrosis (apparent diffusion coefficient [ADC]) and oxygenation (relaxation rate [R2*]) with systemic markers of inflammation are limited. Methods We evaluated associations of baseline kidney fMRI-derived ADC and R2* with baseline and follow-up serum IL-6 and C-reactive protein (CRP) in 127 participants from the CKD Optimal Management with Binders and NicotinamidE trial, a randomized, 12-month trial of nicotinamide and lanthanum carbonate versus placebo in individuals with CKD stages 3–4. Cross-sectional analyses of baseline kidney fMRI biomarkers and markers of inflammation used multivariable linear regression. Longitudinal analyses of baseline kidney fMRI biomarkers and change in markers of inflammation over time used linear mixed-effects models. Results Mean±SD eGFR, ADC, and R2* were 32.2±8.7 ml/min per 1.73 m2, 1.46±0.17×10−3 mm2/s, and 20.3±3.1 s−1, respectively. Median (interquartile range) IL-6 and CRP were 3.7 (2.4–4.9) pg/ml and 2.8 (1.2–6.3) mg/L, respectively. After multivariable adjustment, IL-6 and CRP were 13.1% and 27.3% higher per 1 SD decrease in baseline cortical ADC, respectively. Baseline cortical R2* did not have a significant association with IL-6 or CRP. Mean annual IL-6 and CRP slopes were 0.98 pg/ml per year and 0.91 mg/L per year, respectively. Baseline cortical ADC and R2* did not have significant associations with change in IL-6 or CRP over time. Conclusions Lower cortical ADC, suggestive of greater fibrosis, was associated with higher systemic inflammation. Baseline kidney fMRI biomarkers did not associate with changes in systemic markers of inflammation over time.
Collapse
Affiliation(s)
- Jacquelyn Trujillo
- The Graduate School, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Manal Alotaibi
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Department of Medicine, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Nay Seif
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xuan Cai
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Brett Larive
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio
| | - Jennifer Gassman
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio
| | - Kalani L. Raphael
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, Utah
| | - Alfred K. Cheung
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, Utah
| | - Dominic S. Raj
- Division of Renal Diseases and Hypertension, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Linda F. Fried
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Stuart M. Sprague
- Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois
| | | | - Michel Chonchol
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado
| | - John Paul Middleton
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Joachim H. Ix
- Renal Section, Department of Medicine, University of California San Diego School of Medicine, San Diego, California
| | - Pottumarthi Prasad
- Department of Radiology, NorthShore University HealthSystem, Evanston, Illinois
| | - Tamara Isakova
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Anand Srivastava
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
| |
Collapse
|
30
|
Blake SJ, Wolf Y, Boursi B, Lynn DJ. Role of the microbiota in response to and recovery from cancer therapy. Nat Rev Immunol 2024; 24:308-325. [PMID: 37932511 DOI: 10.1038/s41577-023-00951-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2023] [Indexed: 11/08/2023]
Abstract
Our understanding of how the microbiota affects the balance between response to and failure of cancer treatment by modulating the tumour microenvironment and systemic immune system has advanced rapidly in recent years. Microbiota-targeting interventions in patients with cancer are an area of intensive investigation. Promisingly, phase I-II clinical trials have shown that interventions such as faecal microbiota transplantation can overcome resistance to immune checkpoint blockade in patients with melanoma, improve therapeutic outcomes in treatment-naive patients and reduce therapy-induced immunotoxicities. Here, we synthesize the evidence showing that the microbiota is an important determinant of both cancer treatment efficacy and treatment-induced acute and long-term toxicity, and we discuss the complex and inter-related mechanisms involved. We also assess the potential of microbiota-targeting interventions, including bacterial engineering and phage therapy, to optimize the response to and recovery from cancer therapy.
Collapse
Affiliation(s)
- Stephen J Blake
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Yochai Wolf
- Ella Lemelbaum Institute for Immuno-oncology and Skin Cancer, Sheba Medical Center, Tel Hashomer, Israel
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ben Boursi
- School of Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Oncology, Sheba Medical Center, Tel Hashomer, Israel
- Center of Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Lynn
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
31
|
Matsuda S, Shintani T, Miyagawa T, Yumoto H, Komatsu Y, Dewake N, Iwata T, Nagano T, Morozumi T, Goto R, Kato S, Kitamura M, Shin K, Sekino S, Yamashita A, Yamashita K, Yoshimura A, Sugaya T, Takashiba S, Taguchi Y, Nemoto E, Nishi H, Mizuno N, Numabe Y, Kawaguchi H. Effect of Periodontal Treatment on Reducing Chronic Inflammation in Systemically Healthy Patients With Periodontal Disease. Am J Med 2024; 137:273-279.e2. [PMID: 37984772 DOI: 10.1016/j.amjmed.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND We determined the effects and an accurate marker of periodontal treatment on serum interleukin (IL)-6 and high-sensitivity C-reactive protein (HsCRP) levels in systemically healthy individuals with periodontal disease. METHODS This multicenter study included systemically healthy individuals with periodontal disease who received initial periodontal treatment and had no periodontal treatment history. Periodontal parameters, including periodontal inflamed surface area, masticatory efficiency, and periodontal disease classification; serum IL-6 and HsCRP levels; and serum immunoglobulin (Ig)G titers against periodontal pathogens were evaluated at baseline and after treatment. Subjects were classified as low or high responders (group) based on periodontal inflamed surface area changes. RESULTS There were 153 participants. Only periodontal inflamed surface area changes were markedly different between low and high responders. Periodontal treatment (time point) decreased both serum IL-6 and HsCRP levels. The interaction between group and time point was remarkable only for serum IL-6 levels. Changes in serum immunoglobulin (Ig)G titers against periodontal pathogens were not associated with IL-6 changes in high responders. We analyzed the indirect effect of serum anti-Porphyromonas gingivalis type 2 IgG titer changes using mediation analysis and found no significance. However, the direct effect of group (low or high responder) on IL-6 changes was considerable. CONCLUSIONS Periodontal treatment effectively decreased serum IL-6 levels, independent of periodontal pathogen infection, in systemically healthy individuals with periodontal disease.
Collapse
Affiliation(s)
- Shinji Matsuda
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | | | - Tsuyoshi Miyagawa
- Clinical Research Center in Hiroshima, Hiroshima University Hospital, Hiroshima, Japan
| | - Hiromichi Yumoto
- Department of Periodontology and Endodontology, Institute of Biomedical Sciences, Tokushima University Graduate school, Tokushima, Japan
| | - Yasutaka Komatsu
- Periodontal Clinic, Niigata University, Medical and Dental Hospital, Niigata, Japan
| | - Nanae Dewake
- Department of Operative Dentistry, Endodontology and Periodontology, School of Dentistry, Matsumoto Dental University, Nagano, Japan
| | - Takanori Iwata
- Department of Periodontology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takatoshi Nagano
- Department of Periodontology, Tsurumi University School of Dental Medicine, Kanagawa, Japan
| | - Toshiya Morozumi
- Department of Periodontology, Faculty of Dentistry, Kanagawa Dental University, Kanagawa, Japan; Department of Endodontics, The Nippon Dental University School of Life Dentistry at Niigata, Japan
| | - Ryoma Goto
- Department of Periodontology, School of Dentistry, Aichi Gakuin University, Aichi, Japan
| | - Satsuki Kato
- Health Sciences University of Hokkaido, School of Dentistry, Division of Periodontology and Endodontology, Department of Oral Rehabilitation, Hokkaido, Japan
| | - Masahiro Kitamura
- Graduate School of Dentistry, Department of Periodontology and Regenerative Dentistry, Osaka University, Osaka, Japan
| | - Kitetsu Shin
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Saitama, Japan
| | - Satoshi Sekino
- School of Life Dentistry Department of Periodontology, The Nippon Dental University, Tokyo, Japan
| | - Akiko Yamashita
- Section of Periodontology, Division of Oral Rehabilitation Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Keiko Yamashita
- Department of Periodontology, Tokyo Dental College, Tokyo, Japan
| | - Atsutoshi Yoshimura
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tsutomu Sugaya
- Department of Periodontology and Endodontology, Faculty of Dental Medicine, Hokkaido University, Hokkaido, Japan
| | - Shogo Takashiba
- Department of Pathophysiology-Periodontal Science, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoichiro Taguchi
- Faculty of Dentistry, Department of Periodontology, Osaka Dental University, Osaka, Japan
| | - Eiji Nemoto
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Hiromi Nishi
- Department of General Dentistry, Hiroshima University Hospital, Hiroshima, Japan
| | - Noriyoshi Mizuno
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukihiro Numabe
- School of Life Dentistry Department of Periodontology, The Nippon Dental University, Tokyo, Japan
| | - Hiroyuki Kawaguchi
- Department of General Dentistry, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
32
|
Wang C, Liu S, Yang Y, Kamronbek R, Ni S, Cheng Y, Zhou C, Yan H, Li L, Liu H, Wang Y, Qin Y, Yin C, Zhang M. Interleukin-4 and Interleukin-17 are associated with coronary artery disease. Clin Cardiol 2024; 47:e24188. [PMID: 38146141 PMCID: PMC10823557 DOI: 10.1002/clc.24188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/12/2023] [Accepted: 10/30/2023] [Indexed: 12/27/2023] Open
Abstract
INTRODUCTION The present study aimed to examine the correlation between serum cytokine levels and the incidence of coronary artery disease (CAD), a leading cause of mortality globally, which is known to have a strong association with inflammatory factors. The study further sought to determine the predictors of CAD to distinguish patients with coronary artery lesions from those suspected of having CAD. METHODS AND RESULTS In this study, 487 patients who underwent coronary angiography as a result of suspected CAD but without acute myocardial infarction (AMI) were recruited. The serum levels of the cytokines interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, IL-17, tumor necrosis factor-α, interferon (IFN)-α, and IFN-γ were measured using a multiplexed particle-based flow cytometric assay technique. The results of the study revealed that the levels of IL-4, IL-12p70, IL-17, IFN-α, and IFN-γ in the CAD group were significantly lower compared to those in the non-CAD group. Multivariate logistic regression analysis indicated that two serum cytokines (IL-4 and IL-17), one protective factor (high-density lipoprotein cholesterol [HDL-C]), and three risk factors (sex, smoking, and diabetes) were independently predictive of CAD. The receiver operating characteristic curve analysis showed that the combined use of these predictors in a multivariate model demonstrated good predictive performance for CAD, as evidenced by an area under the curve value of 0.826. CONCLUSION The results of the study indicated that serum IL-4 and IL-17 levels serve as independent predictors of CAD. The risk prediction model established in the research, which integrates these serum cytokines (IL-4 and IL-17) with relevant clinical risk factors (gender, smoking, and diabetes) and the protective factor HDL-C, holds the potential to differentiate patients with CAD from those suspected of having CAD but without AMI.
Collapse
Affiliation(s)
- Chenyang Wang
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Sheng Liu
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yunxiao Yang
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Raimov Kamronbek
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Siyao Ni
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yunjiu Cheng
- Department of Cardiology, Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Can Zhou
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Huiyuan Yan
- Department of CardiologyHangjinqi People's HospitalInner MongoliaChina
| | - Li Li
- Liver Research Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Hao Liu
- Department of OrthopedicsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yu Wang
- Key Laboratory of Upper Airway Dysfunction‐Related Cardiovascular Diseases, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel DiseaseCapital Medical UniversityBeijingChina
| | - Yanwen Qin
- Key Laboratory of Upper Airway Dysfunction‐Related Cardiovascular Diseases, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel DiseaseCapital Medical UniversityBeijingChina
| | - Chengqian Yin
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ming Zhang
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
33
|
White SC, Ruiz JM, Allison M, Uchino BN, Smith TW, Taylor DJ, Jones DR, Russell MA, Ansell EB, Smyth JM. Cardiovascular risk, social vigilance, and stress profiles of male law enforcement officers versus civilians. Health Psychol Open 2024; 11:20551029241244723. [PMID: 38586533 PMCID: PMC10996355 DOI: 10.1177/20551029241244723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024] Open
Abstract
This study examined the cardiovascular disease (CVD) risk profiles of male law enforcement officers (LEOs) and civilians. CVD risk profiles were based on data collected using traditional objective (e.g., resting BP, cholesterol), novel objective (e.g., ambulatory BP) and self-report measures (e.g., EMA social vigilance). A subset of male LEOs (n = 30, M age = 41.47, SD = 8.03) and male civilians (n = 120, M age = 40.73, SD = 13.52) from a larger study were included in analyses. Results indicated LEOs had significantly higher body mass index [BMI], 31.17 kg/m2 versus 28.87 kg/m2, and exhibited significantly higher trait and state social vigilance across multiple measures, whereas perceived stress was higher among civilians. Findings highlight the need for future research examining CVD risk associated with occupational health disparities, including attributes of individuals entering certain professions as well as experiential and environmental demands of the work.
Collapse
Affiliation(s)
- Shannon C White
- Department of Biobehavioral Health, Pennsylvania State University, USA
| | - John M Ruiz
- Department of Psychology, University of Arizona, USA
| | - Matthew Allison
- Department of Family and Preventative Medicine, University of California San Diego, USA
| | | | | | | | - Dusti R Jones
- Center for Health Outcomes and Population Equity (HOPE), University of Utah, USA
| | - Michael A Russell
- Department of Biobehavioral Health, Pennsylvania State University, USA
| | - Emily B Ansell
- Department of Biobehavioral Health, Pennsylvania State University, USA
| | - Joshua M Smyth
- Department of Biobehavioral Health, Pennsylvania State University, USA
| |
Collapse
|
34
|
Olszowka M, Hagström E, Hadziosmanovic N, Ljunggren M, Denchev S, Manolis A, Wallentin L, White HD, Stewart RAH, Held C. Excessive daytime sleepiness, morning tiredness, and prognostic biomarkers in patients with chronic coronary syndrome. Int J Cardiol 2024; 394:131395. [PMID: 37748524 DOI: 10.1016/j.ijcard.2023.131395] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Sleep-related breathing disorders (SRBD) are related to cardiovascular outcomes in patients with chronic coronary syndrome (CCS). Whether SRBD-related symptoms are associated with prognostic biomarkers in patients with CCS is not established. METHODS Associations between frequency (never/rarely, sometimes, often, always) of self-reported SRBD-related symptoms (excessive daytime sleepiness [EDS]; morning tiredness [MT]; loud snoring; multiple awakenings/night; gasping, choking, or apnea when asleep) and levels of biomarkers related to cardiovascular prognosis (high-sensitivity C-reactive protein [hs-CRP], interleukin 6 [IL-6], high-sensitivity cardiac troponin T [hs-cTnT], N-terminal pro B-type natriuretic peptide [NT-proBNP], cystatin C, growth differentiation factor 15 [GDF-15] and lipoprotein-associated phospholipase A2 activity) were assessed at baseline in 15,640 patients with CCS on optimal secondary preventive therapy in the STABILITY trial. Cross-sectional associations were assessed by adjusted linear regression models testing for trends with the never/rarely category serving as reference. RESULTS EDS was associated (geometric mean ratio, 95% confidence interval) with increased levels of IL-6 (often 1.07 [1.03-1.10], always 1.15 [1.10-1.21]), GDF-15 (often 1.03 [1.01-1.06], always 1.07 [1.03-1.11]), NT-proBNP (always 1.22 [1.12-1.33]), and hs-cTnT (always 1.07 [1.01-1.12]). MT was associated with increased levels of IL-6 (often 1.05 [1.01-1.09], always 1.09 [1.04-1.15]), and GDF-15 (always 1.06 [1.03-1.10]). All symptoms were to some degree associated with higher levels of hs-CRP and loud snoring was also associated with decreased levels of NT-proBNP and hs-cTnT. CONCLUSIONS In patients with CCS, stepwise increased frequency of SRBD-related symptoms, such as EDS and MT, were associated with gradually higher levels of IL-6 and GDF-15, each reflecting distinct pathophysiological pathways.
Collapse
Affiliation(s)
- Maciej Olszowka
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden.
| | - Emil Hagström
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | | | - Mirjam Ljunggren
- Department of Medical Sciences, Respiratory-, allergy- and sleep research, Uppsala University, Uppsala, Sweden
| | - Stefan Denchev
- Medical Institute of Ministry of Interior, Sofia, Bulgaria
| | | | - Lars Wallentin
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Harvey D White
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Ralph A H Stewart
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Claes Held
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
35
|
Garrity K, Gaw S, Blewitt A, Canon P, McCall P, McPeake J. Cardiac dysfunction in survivors of sepsis: a scoping review. Open Heart 2023; 10:e002454. [PMID: 38065588 PMCID: PMC10711866 DOI: 10.1136/openhrt-2023-002454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Sepsis is associated with an increased risk of adverse cardiovascular events in a magnitude comparable to other major cardiovascular risk factors. Sepsis is one of the most common reasons for intensive care admission and survivors often have significant functional limitations following discharge. However, it is not clear to what extent chronic cardiovascular dysfunction might mediate these functional impairments, or how we might screen and manage these patients at risk of chronic cardiovascular disease. We conducted a scoping review to map existing evidence and identify research gaps relating to cardiovascular dysfunction following sepsis. METHODS We conducted a systematic search of MEDLINE, Embase and CINAHL databases using a concept, context, population (CoCoPop) framework. Studies examining cardiovascular outcomes or symptoms following an episode of sepsis in adults were included. Data were mapped based on the population assessed, cardiovascular outcomes examined, inclusion of objective measures of cardiac dysfunction such as biomarkers or cardiovascular imaging, or whether cardiovascular symptoms or patient-reported functional outcomes measures were recorded. RESULTS We identified 11 210 articles of which 70 were eligible for full text review and 28 were included in final analysis. Across our dataset, a wide range of incident cardiovascular outcomes were reported in the literature including incidence of congestive heart failure (13/28), arrhythmia (6/28), myocardial infarction (24/28) or cardiovascular death or all-cause mortality (20/28). Only 39% (11/28) of articles reported objective measures of cardiovascular function and only one article related cardiovascular function to functional impairment via patient-reported outcome measures. CONCLUSION There are significant gaps in our understanding of cardiac dysfunction following sepsis . While the research highlights the strong association of sepsis with a variety of adverse cardiovascular outcomes, further prospective work is required to understand the mechanisms that mediate this phenomenon and how we can best identify and manage patients at risk.
Collapse
Affiliation(s)
- Kevin Garrity
- University of Glasgow, Glasgow, UK
- NHS Greater Glasgow and Clyde, Glasgow, UK
| | | | | | | | - Philip McCall
- University of Glasgow, Glasgow, UK
- NHS Golden Jubilee, Glasgow, UK
| | | |
Collapse
|
36
|
Gold ME, Woods E, Pobee D, Ibrahim R, Quyyumi AA. Multi-proteomic Biomarker Risk Scores for Predicting Risk and Guiding Therapy in Patients with Coronary Artery Disease. Curr Cardiol Rep 2023; 25:1811-1821. [PMID: 38079057 DOI: 10.1007/s11886-023-01995-3] [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] [Accepted: 11/08/2023] [Indexed: 01/26/2024]
Abstract
PURPOSE OF REVIEW Patients with established coronary artery disease (CAD) are at high residual risk for adverse events, despite guideline-based treatments. Herein, we aimed to determine whether risk scores based on multiple circulating biomarkers that represent activation of various pathophysiologically important pathways involved in atherosclerosis and myocardial dysfunction help identify those at greatest residual risk. RECENT FINDINGS Numerous circulating proteins, representing dysregulation of the pathways involved in the development and stability of coronary and myocardial diseases, have been identified. When aggregated together, biomarker risk scores (BRS) more accurately stratify patients with established CAD that may help target interventions in those individuals who are at elevated risk. Moreover, intensification of guideline-based therapies has been associated with parallel improvements in both BRS and outcomes, indicating that these risk scores may be employed clinically to target therapy. Multi-protein BRS are predictive of risk, independent of, and in addition to traditional risk factor assessments in patients with CAD. Those with elevated risk may benefit from optimization of therapies, and improvements in the BRS will identify those with improved outcomes.
Collapse
Affiliation(s)
- Matthew E Gold
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1760 Haygood Dr NE, Atlanta, GA, USA
| | - Edward Woods
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Darlington Pobee
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Rand Ibrahim
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1760 Haygood Dr NE, Atlanta, GA, USA.
| |
Collapse
|
37
|
Newman JD, Anthopolos R, Ruggles KV, Cornwell M, Reynolds HR, Bangalore S, Mavromatis K, Held C, Wallentin L, Kullo IJ, McManus B, Newby LKK, Rosenberg Y, Hochman JS, Maron DJ, Berger JS. Biomarkers and cardiovascular events in patients with stable coronary disease in the ISCHEMIA Trials. Am Heart J 2023; 266:61-73. [PMID: 37604357 PMCID: PMC10843480 DOI: 10.1016/j.ahj.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/23/2023]
Abstract
IMPORTANCE Biomarkers may improve prediction of cardiovascular events for patients with stable coronary artery disease (CAD), but their importance in addition to clinical tests of inducible ischemia and CAD severity is unknown. OBJECTIVES To evaluate the prognostic value of multiple biomarkers in stable outpatients with obstructive CAD and moderate or severe inducible ischemia. DESIGN AND SETTING The ISCHEMIA and ISCHEMIA CKD trials randomized 5,956 participants with CAD to invasive or conservative management from July 2012 to January 2018; 1,064 participated in the biorepository. MAIN OUTCOME MEASURES Primary outcome was cardiovascular death, myocardial infarction (MI), or hospitalization for unstable angina, heart failure, or resuscitated cardiac arrest. Secondary outcome was cardiovascular death or MI. Improvements in prediction were assessed by cause-specific hazard ratios (HR) and area under the receiver operating characteristics curve (AUC) for an interquartile increase in each biomarker, controlling for other biomarkers, in a base clinical model of risk factors, left ventricular ejection fraction (LVEF) and ischemia severity. Secondary analyses were performed among patients in whom core-lab confirmed severity of CAD was ascertained by computed cardiac tomographic angiography (CCTA). EXPOSURES Baseline levels of interleukin-6 (IL-6), high sensitivity troponin T (hsTnT), growth differentiation factor 15 (GDF-15), N-terminal pro-B-type natriuretic peptide (NT-proBNP), lipoprotein a (Lp[a]), high sensitivity C-reactive protein (hsCRP), Cystatin C, soluble CD 40 ligand (sCD40L), myeloperoxidase (MPO), and matrix metalloproteinase 3 (MMP3). RESULTS Among 757 biorepository participants, median (IQR) follow-up was 3 (2-5) years, age was 67 (61-72) years, and 144 (19%) were female; 508 had severity of CAD by CCTA available. In an adjusted multimarker model with hsTnT, GDF-15, NT-proBNP and sCD40L, the adjusted HR for the primary outcome per interquartile increase in each biomarker was 1.58 (95% CI 1.22, 2.205), 1.60 (95% CI 1.16, 2.20), 1.61 (95% 1.22, 2.14), and 1.46 (95% 1.12, 1.90), respectively. The adjusted multimarker model also improved prediction compared with the clinical model, increasing the AUC from 0.710 to 0.792 (P < .01) and 0.714 to 0.783 (P < .01) for the primary and secondary outcomes, respectively. Similar findings were observed after adjusting for core-lab confirmed atherosclerosis severity. CONCLUSIONS AND RELEVANCE Among ISCHEMIA biorepository participants, biomarkers of myocyte injury/distension, inflammation, and platelet activity improved cardiovascular event prediction in addition to risk factors, LVEF, and assessments of ischemia and atherosclerosis severity. These biomarkers may improve risk stratification for patients with stable CAD.
Collapse
Affiliation(s)
- Jonathan D Newman
- Department of Medicine, NYU Grossman School of Medicine, New York, NY.
| | - Rebecca Anthopolos
- Division of Biostatistics, Department of Population Health, NYU Langone Health, New York, NY
| | - Kelly V Ruggles
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | | | | | - Sripal Bangalore
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Kreton Mavromatis
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Claes Held
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Iftikar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Bruce McManus
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - L Kristin K Newby
- Division of Cardiology, Department of Medicine, Duke Clinical Research Institute, Durham, NC
| | - Yves Rosenberg
- Division of Cardiovascular Sciences, National Health Lung and Blood Institute, National Institute of Health, Bethesda, MD
| | - Judith S Hochman
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - David J Maron
- Department of Medicine, Stanford University, Stanford, CA
| | - Jeffrey S Berger
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
38
|
Meo L, Savarese M, Munno C, Mirabelli P, Ragno P, Leone O, Alfieri M. Circulating Biomarkers for Monitoring Chemotherapy-Induced Cardiotoxicity in Children. Pharmaceutics 2023; 15:2712. [PMID: 38140053 PMCID: PMC10747387 DOI: 10.3390/pharmaceutics15122712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Most commonly diagnosed cancer pathologies in the pediatric population comprise leukemias and cancers of the nervous system. The percentage of cancer survivors increased from approximatively 50% to 80% thanks to improvements in medical treatments and the introduction of new chemotherapies. However, as a consequence, heart disease has become the main cause of death in the children due to the cardiotoxicity induced by chemotherapy treatments. The use of different cardiovascular biomarkers, complementing data obtained from electrocardiogram, echocardiography cardiac imaging, and evaluation of clinical symptoms, is considered a routine in clinical diagnosis, prognosis, risk stratification, and differential diagnosis. Cardiac troponin and natriuretic peptides are the best-validated biomarkers broadly accepted in clinical practice for the diagnosis of acute coronary syndrome and heart failure, although many other biomarkers are used and several potential markers are currently under study and possibly will play a more prominent role in the future. Several studies have shown how the measurement of cardiac troponin (cTn) can be used for the early detection of heart damage in oncological patients treated with potentially cardiotoxic chemotherapeutic drugs. The advent of high sensitive methods (hs-cTnI or hs-cTnT) further improved the effectiveness of risk stratification and monitoring during treatment cycles.
Collapse
Affiliation(s)
- Luigia Meo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy; (L.M.); (P.R.)
| | - Maria Savarese
- Clinical Pathology, Santobono-Pausilipon Children’s Hospital, 80123 Naples, Italy; (M.S.); (C.M.); (O.L.)
| | - Carmen Munno
- Clinical Pathology, Santobono-Pausilipon Children’s Hospital, 80123 Naples, Italy; (M.S.); (C.M.); (O.L.)
| | - Peppino Mirabelli
- Clinical and Translational Research Unit, Santobono-Pausilipon Children’s Hospital, 80123 Naples, Italy;
| | - Pia Ragno
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy; (L.M.); (P.R.)
| | - Ornella Leone
- Clinical Pathology, Santobono-Pausilipon Children’s Hospital, 80123 Naples, Italy; (M.S.); (C.M.); (O.L.)
| | - Mariaevelina Alfieri
- Clinical Pathology, Santobono-Pausilipon Children’s Hospital, 80123 Naples, Italy; (M.S.); (C.M.); (O.L.)
| |
Collapse
|
39
|
Napoli G, Pergola V, Basile P, De Feo D, Bertrandino F, Baggiano A, Mushtaq S, Fusini L, Fazzari F, Carrabba N, Rabbat MG, Motta R, Ciccone MM, Pontone G, Guaricci AI. Epicardial and Pericoronary Adipose Tissue, Coronary Inflammation, and Acute Coronary Syndromes. J Clin Med 2023; 12:7212. [PMID: 38068263 PMCID: PMC10707039 DOI: 10.3390/jcm12237212] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/22/2023] [Accepted: 11/16/2023] [Indexed: 01/22/2025] Open
Abstract
Vascular inflammation is recognized as the primary trigger of acute coronary syndrome (ACS). However, current noninvasive methods are not capable of accurately detecting coronary inflammation. Epicardial adipose tissue (EAT) and pericoronary adipose tissue (PCAT), in addition to their role as an energy reserve system, have been found to contribute to the development and progression of coronary artery calcification, inflammation, and plaque vulnerability. They also participate in the vascular response during ischemia, sympathetic stimuli, and arrhythmia. As a result, the evaluation of EAT and PCAT using imaging techniques such as computed tomography (CT), cardiac magnetic resonance (CMR), and nuclear imaging has gained significant attention. PCAT-CT attenuation, which measures the average CT attenuation in Hounsfield units (HU) of the adipose tissue, reflects adipocyte differentiation/size and leukocyte infiltration. It is emerging as a marker of tissue inflammation and has shown prognostic value in coronary artery disease (CAD), being associated with plaque development, vulnerability, and rupture. In patients with acute myocardial infarction (AMI), an inflammatory pericoronary microenvironment promoted by dysfunctional EAT/PCAT has been demonstrated, and more recently, it has been associated with plaque rupture in non-ST-segment elevation myocardial infarction (NSTEMI). Endothelial dysfunction, known for its detrimental effects on coronary vessels and its association with plaque progression, is bidirectionally linked to PCAT. PCAT modulates the secretory profile of endothelial cells in response to inflammation and also plays a crucial role in regulating vascular tone in the coronary district. Consequently, dysregulated PCAT has been hypothesized to contribute to type 2 myocardial infarction with non-obstructive coronary arteries (MINOCA) and coronary vasculitis. Recently, quantitative measures of EAT derived from coronary CT angiography (CCTA) have been included in artificial intelligence (AI) models for cardiovascular risk stratification. These models have shown incremental utility in predicting major adverse cardiovascular events (MACEs) compared to plaque characteristics alone. Therefore, the analysis of PCAT and EAT, particularly through PCAT-CT attenuation, appears to be a safe, valuable, and sufficiently specific noninvasive method for accurately identifying coronary inflammation and subsequent high-risk plaque. These findings are supported by biopsy and in vivo evidence. Although speculative, these pieces of evidence open the door for a fascinating new strategy in cardiovascular risk stratification. The incorporation of PCAT and EAT analysis, mainly through PCAT-CT attenuation, could potentially lead to improved risk stratification and guide early targeted primary prevention and intensive secondary prevention in patients at higher risk of cardiac events.
Collapse
Affiliation(s)
- Gianluigi Napoli
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Valeria Pergola
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padua, 35122 Padua, Italy;
| | - Paolo Basile
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Daniele De Feo
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Fulvio Bertrandino
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Andrea Baggiano
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Saima Mushtaq
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Laura Fusini
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Fabio Fazzari
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Nazario Carrabba
- Department of Cardiothoracovascular Medicine, Azienda Ospedaliero Universitaria Careggi, 50134 Florence, Italy;
| | - Mark G. Rabbat
- Division of Cardiology, Loyola University of Chicago, Chicago, IL 60611, USA;
- Edward Hines Jr. VA Hospital, Hines, IL 60141, USA
| | - Raffaella Motta
- Radiology Unit, University Hospital of Padova, 35128 Padua, Italy;
| | - Marco Matteo Ciccone
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| | - Gianluca Pontone
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (A.B.); (S.M.); (L.F.); (F.F.); (G.P.)
| | - Andrea Igoren Guaricci
- University Cardiologic Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, 70124 Bari, Italy; (G.N.); (P.B.); (D.D.F.); (F.B.); (M.M.C.)
| |
Collapse
|
40
|
He J, Song C, Zhang R, Yuan S, Li J, Dou K. Discordance Between Neutrophil to Lymphocyte Ratio and High Sensitivity C-Reactive Protein to Predict Clinical Events in Patients with Stable Coronary Artery Disease: A Large-Scale Cohort Study. J Inflamm Res 2023; 16:5439-5450. [PMID: 38026249 PMCID: PMC10674642 DOI: 10.2147/jir.s428734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Neutrophil to lymphocyte ratio (NLR), a novel inflammatory biomarker, has been shown to positively predict prognosis independent of high-sensitivity C-reactive protein (hsCRP) in patients with coronary artery disease (CAD). This study aimed to use discordance analysis to evaluate the effectiveness of NLR and hsCRP to predict adverse events in patients with stable CAD. Patients and Methods This observational cohort study included 7827 consecutive CAD patients at Fuwai Hospital from March 2011 to April 2017. Discordant NLR with hsCRP was defined by the highest quartiles and medians. The primary endpoint was major adverse cardiovascular and cerebrovascular events (MACCEs), including cardiovascular death, non-fatal myocardial infarction, non-fatal stroke, and unplanned revascularization. Results During a median 36-month follow-up, 624 (8.0%) MACCEs occurred. Compared with the lowest NLR quartile, a significantly higher risk of MACCEs was observed in the highest NLR quartile after adjusting for confounding factors (hazard ratio [HR], 1.36; 95% confidence interval [CI], 1.09-1.71). High NLR and low hsCRP discordance were also associated with an increased risk of MACCEs in the fully adjusted model (HR, 1.39; 95% CI, 1.05-1.84). Conclusion This study demonstrated that discordantly elevated NLR levels were associated with a greater risk of adverse clinical events in patients with stable CAD, suggesting the potential clinical significance of NLR as a goal of inflammatory risk management.
Collapse
Affiliation(s)
- Jining He
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Beijing, People’s Republic of China
| | - Chenxi Song
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Beijing, People’s Republic of China
| | - Rui Zhang
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Beijing, People’s Republic of China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, People’s Republic of China
| | - Sheng Yuan
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Beijing, People’s Republic of China
| | - Jianjun Li
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Beijing, People’s Republic of China
| | - Kefei Dou
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Beijing, People’s Republic of China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, People’s Republic of China
| |
Collapse
|
41
|
Wang C, Chang L, Wang J, Xia L, Cao L, Wang W, Xu J, Gao H. Leptin and risk factors for atherosclerosis: A review. Medicine (Baltimore) 2023; 102:e36076. [PMID: 37986371 PMCID: PMC10659641 DOI: 10.1097/md.0000000000036076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023] Open
Abstract
Leptin is a hormone secreted primarily by adipose tissue. It regulates an organism's metabolism, energy balance, and body weight through a negative feedback mechanism. When a person or animal has low body fat and little energy, the leptin level in the body decreases, and conversely, when there is an excess of nutrients, the leptin level increases, giving a feeling of satiety. However, when leptin levels are abnormal (too high or too low) for a number of reasons, it can negatively affect your health, inducing inflammatory responses, obesity, and other problems. Many studies have shown that abnormal leptin levels, such as hyperleptinemia, are closely associated with common risk factors for atherosclerosis (AS). This review systematically states the relationship between leptin and common risk factors for AS (inflammation, obesity, diabetes mellitus, hypertension, and sleep disorders) and provides some new thoughts on the future direction of research on both. Because the abnormal level of leptin will have adverse effects on multiple atherosclerotic risk factors, how to regulate the leptin level of patients with AS, and whether we can treat and prevent AS by intervening the leptin level, these may be our new research directions in the future.
Collapse
Affiliation(s)
- Cheng Wang
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Liping Chang
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Jia Wang
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Libo Xia
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Liyuan Cao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Wei Wang
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jianwen Xu
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Huize Gao
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
42
|
Siegbahn A, Eriksson N, Assarsson E, Lundberg M, Ballagi A, Held C, Stewart RAH, White HD, Åberg M, Wallentin L. Development and validation of a quantitative Proximity Extension Assay instrument with 21 proteins associated with cardiovascular risk (CVD-21). PLoS One 2023; 18:e0293465. [PMID: 37963145 PMCID: PMC10645335 DOI: 10.1371/journal.pone.0293465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Treatment of cardiovascular diseases (CVD) is a substantial burden to healthcare systems worldwide. New tools are needed to improve precision of treatment by optimizing the balance between efficacy, safety, and cost. We developed a high-throughput multi-marker decision support instrument which simultaneously quantifies proteins associated with CVD. METHODS AND FINDINGS Candidate proteins independently associated with different clinical outcomes were selected from clinical studies by the screening of 368 circulating biomarkers. We then custom-designed a quantitative PEA-panel with 21 proteins (CVD-21) by including recombinant antigens as calibrator samples for normalization and absolute quantification of the proteins. The utility of the CVD-21 tool was evaluated in plasma samples from a case-control cohort of 4224 patients with chronic coronary syndrome (CCS) using multivariable Cox regression analyses and machine learning techniques. The assays in the CVD-21 tool gave good precision and high sensitivity with lower level of determination (LOD) between 0.03-0.7 pg/ml for five of the biomarkers. The dynamic range for the assays was sufficient to accurately quantify the biomarkers in the validation study except for troponin I, which in the modeling was replaced by high-sensitive cardiac troponin T (hs-TnT). We created seven different multimarker models, including a reference model with NT-proBNP, hs-TnT, GDF-15, IL-6, and cystatin C and one model with only clinical variables, for the comparison of the discriminative value of the CVD-21 tool. All models with biomarkers including hs-TnT provided similar discrimination for all outcomes, e.g. c-index between 0.68-0.86 and outperformed models using only clinical variables. Most important prognostic biomarkers were MMP-12, U-PAR, REN, VEGF-D, FGF-23, TFF3, ADM, and SCF. CONCLUSIONS The CVD-21 tool is the very first instrument which with PEA simultaneously quantifies 21 proteins with associations to different CVD. Novel pathophysiologic and prognostic information beyond that of established biomarkers were identified by a number of proteins.
Collapse
Affiliation(s)
- Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Niclas Eriksson
- 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
| | - Ralph A. H. Stewart
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Harvey D. White
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
Koenig W, Sager HB. Inflammation and cardiovascular disease: new epidemiologic data and their potential implications for anti-cytokine therapy. Eur J Prev Cardiol 2023; 30:1728-1730. [PMID: 37494726 DOI: 10.1093/eurjpc/zwad251] [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: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 07/28/2023]
Affiliation(s)
- Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Lazarettstr, 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Pettenkoferstr. 8a &9, 80336 Munich, Germany
| | - Hendrik B Sager
- Deutsches Herzzentrum München, Technische Universität München, Lazarettstr, 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Pettenkoferstr. 8a &9, 80336 Munich, Germany
| |
Collapse
|
44
|
Yao H, Pang Y, Chen Y, Si N, Wu C, Wang Z, Ren Y. Association Between Interleukin-6 Gene Polymorphism and Severity of Coronary Artery Disease in Patients with Diabetes. Diabetes Metab Syndr Obes 2023; 16:3599-3608. [PMID: 37964941 PMCID: PMC10642489 DOI: 10.2147/dmso.s427873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/15/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction Previous studies have identified diabetes as a risk factor for coronary heart disease. This study determined the correlation between the IL-6 gene -572 G/C polymorphism and the incidence and severity of coronary heart disease in patients with diabetes. Methods One hundred four patients with diabetes who were admitted to our hospital from January 2019 to December 2020 were retrospectively enrolled in the current study. These patients were divided into a diabetes only group (group A, 27 patients) and a diabetes complicated by coronary heart disease group (group B, 77 patients). Seventy patients in the latter group were further divided into low, medium, and high Syntax score groups based on coronary angiography results. A correlation analysis between IL-6, blood lipids, and the IL-6 -572 G/C gene levels was performed. Results The serum IL-6 level in patients with the IL-6-572G/C-GG genotype was higher than patients with the GC and CC genotypes. In patients with diabetes, the presence of the IL-6-572G/C-GG and GC genotypes was associated with a significantly increased risk of developing coronary heart disease. Patients with the IL-6-572G/C-GG genotype and diabetes were shown to have more severe coronary artery lesions compared to patients with the CC genotype. Moreover, the G allele of the IL-6-572G/C gene was linked to a higher risk of coronary heart disease and more severe coronary artery lesions in patients with diabetes compared to the C allele. Conclusion The IL-6-572G/C gene polymorphism is associated with the incidence and severity of coronary heart disease in patients with diabetes.
Collapse
Affiliation(s)
- Hongmei Yao
- Department of Cardiology, the First Hospital of Shanxi Medical University, TaiYuan City, 030001, People’s Republic of China
| | - Yongqiang Pang
- Department of Internal Medicine, Shanxi Medical University, TaiYuan City, 030001, People’s Republic of China
| | - Yubin Chen
- Department of Internal Medicine, Shanxi Medical University, TaiYuan City, 030001, People’s Republic of China
| | - Nilu Si
- Department of Internal Medicine, Shanxi Medical University, TaiYuan City, 030001, People’s Republic of China
| | - Chao Wu
- Department of Internal Medicine, Shanxi Medical University, TaiYuan City, 030001, People’s Republic of China
| | - Zijing Wang
- Department of Internal Medicine, Shanxi Medical University, TaiYuan City, 030001, People’s Republic of China
| | - Yi Ren
- Department of Endocrinology, the First Hospital of Shanxi Medical University, TaiYuan City, 030001, People’s Republic of China
| |
Collapse
|
45
|
Andreescu M. Recent Advances in Serum Biomarkers for Cardiological Risk Stratification and Insight into the Cardiac Management of the Patients With Hematological Malignancies Treated With Targeted Therapy. Cureus 2023; 15:e49696. [PMID: 38033434 PMCID: PMC10688222 DOI: 10.7759/cureus.49696] [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: 11/30/2023] [Indexed: 12/02/2023] Open
Abstract
Cardiovascular diseases (CVD) have emerged as a common and serious complication of cancer treatment, particularly in patients undergoing cardiotoxic therapies. Over the last few years, the medical community has become increasingly aware of the potential for cardiotoxicity resulting from cancer treatments involving chemotherapy, targeted therapies, and radiation therapy. This recognition is due to the significant risk of morbidity and mortality in cancer patients and survivors resulting from such treatment-induced cardiovascular damage. While the cardiotoxic effects of chemotherapy and targeted therapy have been discussed in medical literature, only a limited number of studies have explored the role of serum biomarkers in cardiological risk stratification. In recent years, serum biomarkers have emerged as a valuable tool for assessing and managing cardiotoxicity in patients with hematological malignancies. This review article provides a summary of the current state of knowledge on the usefulness of biomarkers in managing cardiotoxicity resulting from different targeted therapies throughout the cancer care continuum. Although cardiac biomarkers have demonstrated potential in identifying subclinical cardiotoxicity and tracking the response to cardioprotective treatments, further research is necessary to determine optimal biomarkers and surveillance strategies. The incorporation of cardiac biomarkers into clinical practice in patients undergoing targeted therapies could potentially lead to improved long-term cardiovascular outcomes in cancer patients and survivors.
Collapse
Affiliation(s)
- Mihaela Andreescu
- Department of Hematology, Colentina Clinical Hospital, Bucharest, ROU
- Department of Clinical Sciences, Hematology, Faculty of Medicine, Titu Maiorescu University of Bucharest, Bucharest, ROU
| |
Collapse
|
46
|
Della Corte V, Todaro F, Cataldi M, Tuttolomondo A. Atherosclerosis and Its Related Laboratory Biomarkers. Int J Mol Sci 2023; 24:15546. [PMID: 37958528 PMCID: PMC10649778 DOI: 10.3390/ijms242115546] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/15/2023] Open
Abstract
Atherosclerosis constitutes a persistent inflammatory ailment, serving as the predominant underlying condition for coronary artery disease (CAD), peripheral artery disease (PAD), and cerebrovascular disease. The progressive buildup of plaques within the walls of medium- and large-caliber arteries characterizes the atherosclerotic process. This accumulation results in significant narrowing that impedes blood flow, leading to critical tissue oxygen deficiency. Spontaneous blockage of thrombotic vessels can precipitate stroke and myocardial infarction, which are complications representing the primary global causes of mortality. Present-day models for predicting cardiovascular risk incorporate conventional risk factors to gauge the likelihood of cardiovascular events over a ten-year span. In recent times, researchers have identified serum biomarkers associated with an elevated risk of atherosclerotic events. Many of these biomarkers, whether used individually or in combination, have been integrated into risk prediction models to assess whether their inclusion enhances predictive accuracy. In this review, we have conducted a comprehensive analysis of the most recently published literature concerning serum biomarkers associated with atherosclerosis. We have explored the potential utility of incorporating these markers in guiding clinical decisions.
Collapse
|
47
|
Ponchia PI, Ahmed R, Farag M, Alkhalil M. Antiplatelet Therapy in End-stage Renal Disease Patients on Maintenance Dialysis: a State-of-the-art Review. Cardiovasc Drugs Ther 2023; 37:975-987. [PMID: 35867319 DOI: 10.1007/s10557-022-07366-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
Abstract
Patients with end-stage renal disease (ESRD) on maintenance dialysis have an increased risk of ischaemic events, such as recurrent myocardial infarction (MI) and stroke. Potent antiplatelet therapy may help mitigate this risk. Nonetheless, ERSD patients are also at increased risk of bleeding due to their complex vascular milieu, which limits the routine use of potent P2Y12 inhibitors. Moreover, these patients are often underrepresented or excluded from major clinical trials leaving a significant gap in existing knowledge. Understanding the mechanisms of this paradox may serve as a benchmark for the development of ESRD trials. The present review aims to provide an overview of the pathophysiological nature of increased bleeding and ischaemic risks in ERSD patients as well as summarize available evidence of antiplatelet use and propose new concepts to guide physicians in selecting appropriate drug regimes for this high-risk cohort.
Collapse
Affiliation(s)
| | | | - Mohamed Farag
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Mohammad Alkhalil
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK.
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, NE7 7DN, UK.
- Department of Cardiothoracic Services, Freeman Hospital, Freeman Road, Newcastle-upon-Tyne, NE7 7DN, UK.
| |
Collapse
|
48
|
Motawea KR, Elhalag RH, Rouzan SS, Talat NE, Reyad SM, Chébl P, Mohamed MS, Shah J. Cyclophilin C as a Novel Diagnostic and Prognostic Biomarker of Coronary Artery Diseases. A Systematic Review and Meta-Analysis. Curr Probl Cardiol 2023; 48:101812. [PMID: 37209796 DOI: 10.1016/j.cpcardiol.2023.101812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 05/22/2023]
Abstract
We aimed to perform a meta-analysis to investigate the value of Cyclophilin C as a diagnostic and prognostic biomarker in Coronary Artery Disease. PubMed, Web of Science, Scopus and Cochrane library databases were searched. The inclusion criteria were any randomized control trials or controlled observational studies that measured the levels of Cyclophilin C in Coronary Artery disease patients and healthy controls. We excluded case reports, case series, reviews, editorials and animal studies. After search of the literature, 4 studies were included in the meta-analysis with a total number of 454 individuals included in the study. The pooled analysis showed a significant association between CAD group and increased levels of Cyclophilin C (MD = 28.94, 95% confidence interval (CI) = 19.28-38.60, P-value < 0.00001). Subgroup analysis showed a significant association between acute and chronic CAD group with increased levels of cyclophilin c compared with the control group (MD = 35.98, 95% CI = 19.84-52.11, P-value < 0.0001) and (MD = 26.36, 95% CI = 21.87 to 30.85, P-value < 0.00001), respectively. The pooled effect estimate showed that the ROC area for the cyclophillin c as a diagnostic biomarker of CAD was (ROC= 0.880, 95% CI =0.844-0.917, P-value < 0.001). Our study revealed a significant association between acute and chronic coronary artery disease with increased levels of Cyclophilin C. Cyclophilin C could be used as a novel diagnostic and prognostic biomarker in acute and chronic CAD. More research is warranted to support our results.
Collapse
Affiliation(s)
- Karam R Motawea
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Rowan H Elhalag
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Samah S Rouzan
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | | - Sarraa M Reyad
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Pensée Chébl
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | | | |
Collapse
|
49
|
Brewster LM, Bain AR, Garcia VP, DeSouza NM, Tymko MM, Greiner JJ, Ainslie PN. Global REACH 2018: High Altitude-Related Circulating Extracellular Microvesicles Promote a Proinflammatory Endothelial Phenotype In Vitro. High Alt Med Biol 2023; 24:223-229. [PMID: 37504958 DOI: 10.1089/ham.2023.0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Brewster, L. Madden, Anthony R. Bain, Vinicius P. Garcia, Noah M. DeSouza, Michael M. Tymko, Jared J. Greiner, and Philip N. Ainslie. Global REACH 2018: high altitude-related circulating extracellular microvesicles promote a proinflammatory endothelial phenotype in vitro. High Alt Med Biol. 24:223-229, 2023. Introduction: Ascent to high altitude (HA) can induce vascular dysfunction by promoting a proinflammatory endothelial phenotype. Circulating microvesicles (MVs) can mediate the vascular endothelium and inflammation. It is unclear whether HA-related MVs are associated with endothelial inflammation. Objectives: We tested the hypothesis that MVs derived from ascent to HA induce a proinflammatory endothelial phenotype. Methods: Ten healthy adults (8 M/2 F; age: 28 ± 2 years) residing at sea level (SL) were studied before and 4-6 days after rapid ascent to HA (4,300 m). MVs were isolated and enumerated from plasma by centrifugation and flow cytometry. Human umbilical vein endothelial cells were treated with MVs collected from each subject at SL (MV-SL) and at HA (MV-HA). Results: Circulating MV number significantly increased at HA (26,637 ± 3,315 vs. 19,388 ± 1,699). Although intracellular expression of total nuclear factor kappa beta (NF-κB; 83.4 ± 6.7 arbitrary units [AU] vs. 90.2 ± 6.9 AU) was not affected, MV-HA resulted in ∼55% higher (p < 0.05) active NF-κB (129.6 ± 19.8 AU vs. 90.7 ± 10.5 AU) expression compared with MV-SL. In addition, MV-HA induced higher interleukin (IL)-6 (63.9 ± 3.9 pg/ml vs. 53.3 ± 3.6 pg/ml) and IL-8 (140.2 ± 3.6 pg/ml vs. 120.7 ± 3.8 pg/ml) release compared with MV-SL, which was blunted with NF-κB blockade. Conclusions: Circulating extracellular MVs increase at HA and induce endothelial inflammation, potentially contributing to altitude-related vascular dysfunction.
Collapse
Affiliation(s)
- L Madden Brewster
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, Faculty of Health and Social Development, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Anthony R Bain
- Department of Kinesiology, University of Windsor, Windsor, Ontario, Canada
| | - Vinicius P Garcia
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Noah M DeSouza
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Michael M Tymko
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, Faculty of Health and Social Development, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Jared J Greiner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Philip N Ainslie
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, Faculty of Health and Social Development, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| |
Collapse
|
50
|
Abubakar M, Rasool HF, Javed I, Raza S, Abang L, Hashim MMA, Saleem Z, Abdullah RM, Faraz MA, Hassan KM, Bhat RR. Comparative Roles of IL-1, IL-6, IL-10, IL-17, IL-18, 1L-22, IL-33, and IL-37 in Various Cardiovascular Diseases With Potential Insights for Targeted Immunotherapy. Cureus 2023; 15:e42494. [PMID: 37637634 PMCID: PMC10455045 DOI: 10.7759/cureus.42494] [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: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
In recent years, the study of interleukins (ILs), crucial cytokines involved in immune response and inflammation, has garnered significant attention within the sphere of cardiovascular diseases (CVDs). The research has provided insights into the involvement of ILs in diverse CVDs, including arrhythmias, myocardial infarction, atherosclerosis, and heart failure (HF). ILs have emerged as promising therapeutic targets for drug interventions through their involvement in disease development and progression. This comprehensive review provides a detailed overview of ILs, elucidating their functions within the immune system and offering insights into their specific contributions to various CVDs. Moreover, the article delves into the examination of current and potential drug therapies that selectively target ILs in the management of CVDs, presenting a comprehensive analysis of the advantages and disadvantages associated with these therapeutic approaches. A comprehensive literature review was conducted to investigate the involvement of ILs in CVDs. The relevant articles were searched on PubMed, PubMed Central, Medline, Cochrane, Google Scholar, and ScienceDirect databases. The search encompassed articles published from these databases' inception until July 12, 2023. We first examine generalized aspects of ILs, particularly CVDs. Then, we shift focus towards examining the direct impact of ILs on cardiac cells and tissue; on the immune system and inflammation; endothelial cells and vascular function; and finally, their interactions with other signaling pathways and molecules. Then, we discuss the molecular mechanisms of various ILs. Sequentially, we delve into a comprehensive analysis of the individualized role of each distinct IL in diverse CVDs, examining their specific contributions. Finally, we explore the potential for targeted drug therapy to modulate IL activity, aiming to enhance outcomes for patients burdened with CVD. The objective is the identification of gaps in current knowledge and highlight areas that require further investigation within the context of cardiovascular medicine. Through deepening our comprehension of the intricate involvement of ILs in CVDs and harnessing their potential for targeted drug therapy, novel treatment strategies can be devised, leading to improved patient outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
- Department of Internal Medicine, Siddique Sadiq Memorial Trust Hospital, Gujranwala, PAK
| | - Hafiz Fahad Rasool
- Department of Public Health, Nanjing Medical University School of Public Health, Nanjing, CHN
| | - Izzah Javed
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Saud Raza
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Lucy Abang
- Department of Biochemistry, All Saints University School of Medicine, Roseau, DMA
| | | | - Zartasha Saleem
- Department of Emergency Medicine, The University of Lahore Teaching Hospital, Lahore, PAK
| | | | - Muhammad Ahmad Faraz
- Department of Forensic Medicine, Post Graduate Medical Institute, Lahore General Hospital, Lahore, PAK
| | - Khawaja Mushammar Hassan
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Rakshita Ramesh Bhat
- Department of Medical Oncology, Mangalore Institute of Oncology, Mangalore, IND
- Department of Internal Medicine, Bangalore Medical College and Research Institute, Bangalore, IND
| |
Collapse
|