1
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, Dayer M, Alegría-Barrero E, De Sanctis JB, Parise Vasco JM, Riera Lizardo RJ, Nicola S, Martí-Amarista CE, Correa-Pérez A. Interleukin-receptor antagonist and tumour necrosis factor inhibitors for the primary and secondary prevention of atherosclerotic cardiovascular diseases. Cochrane Database Syst Rev 2024; 9:CD014741. [PMID: 39297531 PMCID: PMC11411914 DOI: 10.1002/14651858.cd014741.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ACVD) is worsened by chronic inflammatory diseases. Interleukin receptor antagonists (IL-RAs) and tumour necrosis factor-alpha (TNF) inhibitors have been studied to see if they can prevent cardiovascular events. OBJECTIVES The purpose of this study was to assess the clinical benefits and harms of IL-RAs and TNF inhibitors in the primary and secondary prevention of ACVD. SEARCH METHODS The Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, EBSCO CINAHL plus, and clinical trial registries for ongoing and unpublished studies were searched in February 2024. The reference lists of relevant studies, reviews, meta-analyses and health technology reports were searched to identify additional studies. No limitations on language, date of publication or study type were set. SELECTION CRITERIA RCTs that recruited people with and without pre-existing ACVD, comparing IL-RAs or TNF inhibitors versus placebo or usual care, were selected. The primary outcomes considered were all-cause mortality, myocardial infarction, unstable angina, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors, working independently at each step, selected studies, extracted data, assessed the risk of bias and used GRADE to judge the certainty of evidence. MAIN RESULTS We included 58 RCTs (22,053 participants; 21,308 analysed), comparing medication efficacy with placebo or usual care. Thirty-four trials focused on primary prevention and 24 on secondary prevention. The interventions included IL-1 RAs (anakinra, canakinumab), IL-6 RA (tocilizumab), TNF-inhibitors (etanercept, infliximab) compared with placebo or usual care. The certainty of evidence was low to very low due to biases and imprecision; all trials had a high risk of bias. Primary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality(RR 0.33, 95% CI 0.01 to 7.58, 1 trial), myocardial infarction (RR 0.71, 95% CI 0.04 to 12.48, I² = 39%, 2 trials), unstable angina (RR 0.24, 95% CI 0.03 to 2.11, I² = 0%, 2 trials), stroke (RR 2.42, 95% CI 0.12 to 50.15; 1 trial), adverse events (RR 0.85, 95% CI 0.59 to 1.22, I² = 54%, 3 trials), or infection (rate ratio 0.84, 95% 0.55 to 1.29, I² = 0%, 4 trials). Evidence is very uncertain about whether anakinra and cankinumab may reduce heart failure (RR 0.21, 95% CI 0.05 to 0.94, I² = 0%, 3 trials). Peripheral vascular disease (PVD) was not reported as an outcome. IL-6 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.68, 95% CI 0.12 to 3.74, I² = 30%, 3 trials), myocardial infarction (RR 0.27, 95% CI 0.04 to1.68, I² = 0%, 3 trials), heart failure (RR 1.02, 95% CI 0.11 to 9.63, I² = 0%, 2 trials), PVD (RR 2.94, 95% CI 0.12 to 71.47, 1 trial), stroke (RR 0.34, 95% CI 0.01 to 8.14, 1 trial), or any infection (rate ratio 1.10, 95% CI: 0.88 to 1.37, I2 = 18%, 5 trials). Adverse events may increase (RR 1.13, 95% CI 1.04 to 1.23, I² = 33%, 5 trials). No trial assessed unstable angina. TNF inhibitors The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.78, 95% CI 0.63 to 4.99, I² = 10%, 3 trials), myocardial infarction (RR 2.61, 95% CI 0.11 to 62.26, 1 trial), stroke (RR 0.46, 95% CI 0.08 to 2.80, I² = 0%; 3 trials), heart failure (RR 0.85, 95% CI 0.06 to 12.76, 1 trial). Adverse events may increase (RR 1.13, 95% CI 1.01 to 1.25, I² = 51%, 13 trials). No trial assessed unstable angina or PVD. Secondary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.94, 95% CI 0.84 to 1.06, I² = 0%, 8 trials), unstable angina (RR 0.88, 95% CI 0.65 to 1.19, I² = 0%, 3 trials), PVD (RR 0.85, 95% CI 0.19 to 3.73, I² = 38%, 3 trials), stroke (RR 0.94, 95% CI 0.74 to 1.2, I² = 0%; 7 trials), heart failure (RR 0.91, 95% 0.5 to 1.65, I² = 0%; 7 trials), or adverse events (RR 0.92, 95% CI 0.78 to 1.09, I² = 3%, 4 trials). There may be little to no difference between the groups in myocardial infarction (RR 0.88, 95% CI 0.0.75 to 1.04, I² = 0%, 6 trials). IL6-RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.09, 95% CI 0.61 to 1.96, I² = 0%, 2 trials), myocardial infarction (RR 0.46, 95% CI 0.07 to 3.04, I² = 45%, 3 trials), unstable angina (RR 0.33, 95% CI 0.01 to 8.02, 1 trial), stroke (RR 1.03, 95% CI 0.07 to 16.25, 1 trial), adverse events (RR 0.89, 95% CI 0.76 to 1.05, I² = 0%, 2 trials), or any infection (rate ratio 0.66, 95% CI 0.32 to 1.36, I² = 0%, 4 trials). No trial assessed PVD or heart failure. TNF inhibitors The evidence is very uncertain about the effect of the intervention on all-cause mortality (RR 1.16, 95% CI 0.69 to 1.95, I² = 47%, 5 trials), heart failure (RR 0.92, 95% 0.75 to 1.14, I² = 0%, 4 trials), or adverse events (RR 1.15, 95% CI 0.84 to 1.56, I² = 32%, 2 trials). No trial assessed myocardial infarction, unstable angina, PVD or stroke. Adverse events may be underestimated and benefits inflated due to inadequate reporting. AUTHORS' CONCLUSIONS This Cochrane review assessed the benefits and harms of using interleukin-receptor antagonists and tumour necrosis factor inhibitors for primary and secondary prevention of atherosclerotic diseases compared with placebo or usual care. However, the evidence for the predetermined outcomes was deemed low or very low certainty, so there is still a need to determine whether these interventions provide clinical benefits or cause harm from this perspective. In summary, the different biases and imprecision in the included studies limit their external validity and represent a limitation to determining the effectiveness of the intervention for both primary and secondary prevention of ACVD.
Collapse
Key Words
- humans
- angina, unstable
- angina, unstable/mortality
- angina, unstable/prevention & control
- antibodies, monoclonal, humanized
- antibodies, monoclonal, humanized/administration & dosage
- antibodies, monoclonal, humanized/adverse effects
- atherosclerosis
- atherosclerosis/mortality
- atherosclerosis/prevention & control
- bias
- cause of death
- myocardial infarction
- myocardial infarction/mortality
- myocardial infarction/prevention & control
- primary prevention
- primary prevention/methods
- randomized controlled trials as topic
- receptors, interleukin-1
- receptors, interleukin-1/antagonists & inhibitors
- secondary prevention
- secondary prevention/methods
- tumor necrosis factor-alpha
- tumor necrosis factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
- Facultad de Medicina (Centro Cochrane Madrid), Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia , Venezuela
| | - Mario A Gemmato-Valecillos
- Icahn School of Medicine at Mount Sinai/ NYCHH Elmhurst Hospital Center, 79-01 Broadway, Elmhurst, New York 11373, USA
| | | | - Mark Dayer
- Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland
- Faculty of Health, University of Plymouth, Plymouth, UK
| | | | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Palacky University, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | - Juan Marcos Parise Vasco
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Susana Nicola
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | | | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Hospital Pharmacy and Medical Devices Department, Hospital Central de la Defensa "Gómez Ulla" CSVE, Madrid, Spain
| |
Collapse
|
2
|
Cao J, Roth S, Zhang S, Kopczak A, Mami S, Asare Y, Georgakis MK, Messerer D, Horn A, Shemer R, Jacqmarcq C, Picot A, Green JP, Schlegl C, Li X, Tomas L, Dutsch A, Liman TG, Endres M, Wernsdorf SR, Fürle C, Carofiglio O, Zhu J, Brough D, Hornung V, Dichgans M, Vivien D, Schulz C, Dor Y, Tiedt S, Sager HB, Grosse GM, Liesz A. DNA-sensing inflammasomes cause recurrent atherosclerotic stroke. Nature 2024; 633:433-441. [PMID: 39112714 PMCID: PMC11390481 DOI: 10.1038/s41586-024-07803-4] [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: 01/28/2023] [Accepted: 07/09/2024] [Indexed: 08/17/2024]
Abstract
The risk of early recurrent events after stroke remains high despite currently established secondary prevention strategies1. Risk is particularly high in patients with atherosclerosis, with more than 10% of patients experiencing early recurrent events1,2. However, despite the enormous medical burden of this clinical phenomenon, the underlying mechanisms leading to increased vascular risk and recurrent stroke are largely unknown. Here, using a novel mouse model of stroke-induced recurrent ischaemia, we show that stroke leads to activation of the AIM2 inflammasome in vulnerable atherosclerotic plaques via an increase of circulating cell-free DNA. Enhanced plaque inflammation post-stroke results in plaque destabilization and atherothrombosis, finally leading to arterioarterial embolism and recurrent stroke within days after the index stroke. We confirm key steps of plaque destabilization also after experimental myocardial infarction and in carotid artery plaque samples from patients with acute stroke. Rapid neutrophil NETosis was identified as the main source of cell-free DNA after stroke and NET-DNA as the causative agent leading to AIM2 inflammasome activation. Neutralization of cell-free DNA by DNase treatment or inhibition of inflammasome activation reduced the rate of stroke recurrence after experimental stroke. Our findings present an explanation for the high recurrence rate after incident ischaemic events in patients with atherosclerosis. The detailed mechanisms uncovered here provide clinically uncharted therapeutic targets for which we show high efficacy to prevent recurrent events. Targeting DNA-mediated inflammasome activation after remote tissue injury represents a promising avenue for further clinical development in the prevention of early recurrent events.
Collapse
Affiliation(s)
- Jiayu Cao
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Stefan Roth
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany.
| | - Sijia Zhang
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Anna Kopczak
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Samira Mami
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Programme in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Denise Messerer
- Medizinische Klinik und Poliklinik I, LMU University Hospital, LMU Munich, Munich, Germany
| | - Amit Horn
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Ruth Shemer
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Charlene Jacqmarcq
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Audrey Picot
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Jack P Green
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK
| | - Christina Schlegl
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Xinghai Li
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Lukas Tomas
- Medizinische Klinik und Poliklinik I, LMU University Hospital, LMU Munich, Munich, Germany
| | - Alexander Dutsch
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Thomas G Liman
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Endres
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Saskia R Wernsdorf
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Christina Fürle
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Olga Carofiglio
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Jie Zhu
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - David Brough
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK
| | - Veit Hornung
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Denis Vivien
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
- Research Clinical Department, Caen Normandie University Hospital, Caen, France
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Immunopharmacology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Hendrik B Sager
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Gerrit M Grosse
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
3
|
Li C, Liu L, Li S, Liu YS. N 6-Methyladenosine in Vascular Aging and Related Diseases: Clinical Perspectives. Aging Dis 2024; 15:1447-1473. [PMID: 37815911 PMCID: PMC11272212 DOI: 10.14336/ad.2023.0924-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Aging leads to progressive deterioration of the structure and function of arteries, which eventually contributes to the development of vascular aging-related diseases. N6-methyladenosine (m6A) is the most prevalent modification in eukaryotic RNAs. This reversible m6A RNA modification is dynamically regulated by writers, erasers, and readers, playing a critical role in various physiological and pathological conditions by affecting almost all stages of the RNA life cycle. Recent studies have highlighted the involvement of m6A in vascular aging and related diseases, shedding light on its potential clinical significance. In this paper, we comprehensively discuss the current understanding of m6A in vascular aging and its clinical implications. We discuss the molecular insights into m6A and its association with clinical realities, emphasizing its significance in unraveling the mechanisms underlying vascular aging. Furthermore, we explore the possibility of m6A and its regulators as clinical indicators for early diagnosis and prognosis prediction and investigate the therapeutic potential of m6A-associated anti-aging approaches. We also examine the challenges and future directions in this field and highlight the necessity of integrating m6A knowledge into patient-centered care. Finally, we emphasize the need for multidisciplinary collaboration to advance the field of m6A research and its clinical application.
Collapse
Affiliation(s)
- Chen Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Le Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Ahmadi M, Askari VR, Shahri B, Mousavi Noghab SM, Jarahi L, Baradaran Rahimi V. Omega-3 fatty acids effectively mitigate high-sensitivity C-reactive protein (hs-CRP) biomarker of inflammation in acute myocardial infarction patients: a randomized, double-blind, placebo-controlled clinical trial. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03330-1. [PMID: 39073419 DOI: 10.1007/s00210-024-03330-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Myocardial infarction (MI) is considered an inflammatory disease and among the leading causes of death globally. An essential indicator of inflammation, high-sensitivity C-reactive protein (hs-CRP), is linked with the acute MI prognosis. We aimed to examine the impact of omega-3 polyunsaturated fatty acids (PUFAs) as an anti-inflammatory supplement on hs-CRP levels in acute MI patients. Sixty patients with acute MI participated in this randomized, placebo-controlled trial. For 30 days, patients were randomized to receive omega-3 PUFAs (2 g/day, N = 30) or placebo (N = 30) on top of guideline-directed medical therapy. An initial and endpoint measurement of hs-CRP was performed. We found that the hs-CRP levels in both omega-3 PUFAs and placebo groups remarkably decreased following 30 days of treatment (decreasing from 1.84 (2.3) and 1.3 (2.6) to 0.38 (0.54) and 0.63 (1.12) mg/dL, respectively; P < 0.001). Following the 30 days of treatment, the reducing impact of omega-3 PUFAs (↓ 1.54 (1.98) mg/dL) on hs-CRP was more robust than the placebo group (↓ 0.92 (1.57) mg/dL, P = 0.008). Furthermore, the WBC, cholesterol, LDL, and triglyceride levels were markedly decreased in omega-3 and placebo groups after 30 days of therapy (P < 0.001 for all). However, no remarkable differences were reported in the level of these parameters after 30 days of therapy between both studied groups. Our findings showed that omega-3 PUFAs decrease hs-CRP amounts in patients with acute MI. Omega-3 PUFA supplementation may be an appropriate candidate in patients with early-stage acute MI for inhibiting inflammation.
Collapse
Affiliation(s)
- Mostafa Ahmadi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahram Shahri
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdiar Mousavi Noghab
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Lida Jarahi
- Department of Community Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Tian L, Zhao S, Zhang R. ITIH4 is a predictor for coronary thrombus in coronary arteriography patients. Future Cardiol 2024; 20:547-554. [PMID: 39041488 PMCID: PMC11485701 DOI: 10.1080/14796678.2024.2377924] [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: 01/14/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Aim: To explore potential value of inter-alpha-trypsin inhibitor heavy chain-4 (ITIH4) for coronary artery disease (CAD) diagnosis.Patients & methods: We recruited the patients who received coronary arteriography (CAG) examination. The enzyme-linked immunosorbent assay was used to detect plasma ITIH4.Results: ITIH4 level was lower expression in CAD patients than that in patients of control group, and was negatively correlated with C-reactive protein (CRP). ITIH4 level is no differences between ST-elevated myocardial infarction (STEMI) and non-ST-elevated myocardial infarction (NSTEMI) patients. However, its expression was significantly correlated with D-Dimer and thrombin time, and the logistic analysis confirmed predictive value of ITIH4 for visible thrombus in coronary.Conclusion: ITIH4 may be a useful biomarker in CAD diagnosis, and to predict visible thrombus in coronary.
Collapse
Affiliation(s)
- Lei Tian
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Su Zhao
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Wang Y, Chen Y, Li B, Zhou Y, Guan J, Huang F, Wu J, Dong Y, Sun P, Tian X, Cai J, Ran F, Dai Q, Lv J. The antidepressant effect of Shexiang Baoxin Pills on myocardial infarction rats with depression may be achieved through the inhibition of the NLRP3 inflammasome pathway. Brain Behav 2024; 14:e3586. [PMID: 38970230 PMCID: PMC11226411 DOI: 10.1002/brb3.3586] [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: 08/13/2023] [Revised: 12/06/2023] [Accepted: 02/27/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Patients with myocardial infarction (MI) frequently experience a heightened incidence of depression, thereby increasing the risk of adverse cardiovascular events. Consequently, early detection and intervention in depressive symptoms among patients with MI are imperative. Shexiang Baoxin Pills (SBP), a Chinese patent medicine employed for the treatment of MI, exhibits diverse mechanisms targeting this condition. Nevertheless, its therapeutic efficacy on postmyocardial infarction depressive symptoms remains unclear. The aim of this study is to investigate the effectiveness and mechanism of SBP in managing depression during acute myocardial infarction (AMI). METHODS A rat model combining MI and depression was established, and the rats were randomly divided into four groups: the model (MOD) group, SBP group, Fluoxetine (FLX) group, and Sham group. After 28 days of drug intervention, cardiac function was assessed using echocardiography while behavior was evaluated through sucrose preference test (SPT), forced swimming test (FST), and open-field test (OFT). Additionally, levels of inflammatory factors in serum and hippocampus were measured along with NLRP3 inflammasome-related protein expression via Western blotting and immunofluorescence. RESULTS SBP can enhance cardiac function in rats with AMI and depression, while significantly ameliorating depressive-like behavior. Compared to the Sham group, levels of IL-1β, IL-18, TNF-α, and other inflammatory factors were markedly elevated in the MOD group. However, expressions of these inflammatory factors were reduced to varying degrees following treatment with SBP or FLX. Analysis of NLRP3 inflammasome-related proteins in the hippocampus revealed a significant upregulation of IL-1β, IL-18, NLRP3, ASC, caspase-1, and GSDMD in the MOD group; conversely, these measures were significantly attenuated after SBP intervention. CONCLUSION We have observed a significant amelioration in depression-like behavior upon SBP administration during the treatment of AMI, suggesting that this effect may be attributed to the inhibition of NLRP3-mediated pyroptosis. (The main findings are summarized in the graphical abstract in the supplementary file.).
Collapse
Affiliation(s)
- Yue Wang
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
| | - Yuwen Chen
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
| | - Bingqing Li
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
| | - Yilu Zhou
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
| | - Jing Guan
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
| | - Fanke Huang
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
| | - Jingjing Wu
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
| | - Yanyan Dong
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
- Institute of Cardiovascular and Cerebrovascular DiseasesChina Three Gorges University, Yichang, China
| | - Peiyuan Sun
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
- Institute of Cardiovascular and Cerebrovascular DiseasesChina Three Gorges University, Yichang, China
| | - Xue Tian
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
- Institute of Cardiovascular and Cerebrovascular DiseasesChina Three Gorges University, Yichang, China
| | - Jindan Cai
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
- Institute of Cardiovascular and Cerebrovascular DiseasesChina Three Gorges University, Yichang, China
| | - Feng Ran
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
- Institute of Cardiovascular and Cerebrovascular DiseasesChina Three Gorges University, Yichang, China
| | - Qiuting Dai
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
- Institute of Cardiovascular and Cerebrovascular DiseasesChina Three Gorges University, Yichang, China
| | - Jianfeng Lv
- Department of Cardiovascular MedicineAffiliated Renhe Hospital of China Three Gorges UniversityYichangChina
- Institute of Cardiovascular and Cerebrovascular DiseasesChina Three Gorges University, Yichang, China
| |
Collapse
|
7
|
Xu B, Wu Q, La R, Lu L, Abdu FA, Yin G, Zhang W, Ding W, Ling Y, He Z, Che W. Is systemic inflammation a missing link between cardiometabolic index with mortality? Evidence from a large population-based study. Cardiovasc Diabetol 2024; 23:212. [PMID: 38902748 PMCID: PMC11191290 DOI: 10.1186/s12933-024-02251-w] [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: 02/29/2024] [Accepted: 04/26/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND This study sought to elucidate the associations of cardiometabolic index (CMI), as a metabolism-related index, with all-cause and cardiovascular mortality among the older population. Utilizing data from the National Health and Nutrition Examination Survey (NHANES), we further explored the potential mediating effect of inflammation within these associations. METHODS A cohort of 3029 participants aged over 65 years old, spanning six NHANES cycles from 2005 to 2016, was enrolled and assessed. The primary endpoints of the study included all-cause mortality and cardiovascular mortality utilizing data from National Center for Health Statistics (NCHS). Cox regression model and subgroup analysis were conducted to assess the associations of CMI with all-cause and cardiovascular mortality. The mediating effect of inflammation-related indicators including leukocyte, neutrophil, lymphocyte, systemic immune-inflammation index (SII), neutrophil to lymphocyte ratio (NLR) were evaluated to investigate the potential mechanism of the associations between CMI and mortality through mediation package in R 4.2.2. RESULTS The mean CMI among the enrolled participants was 0.74±0.66, with an average age of 73.28±5.50 years. After an average follow-up period of 89.20 months, there were 1,015 instances of all-cause deaths and 348 cardiovascular deaths documented. In the multivariable-adjusted model, CMI was positively related to all-cause mortality (Hazard Ratio (HR)=1.11, 95% CI=1.01-1.21). Mediation analysis indicated that leukocytes and neutrophils mediated 6.6% and 13.9% of the association of CMI with all-cause mortality. CONCLUSION Elevated CMI is positively associated with all-cause mortality in the older adults. The association appeared to be partially mediated through inflammatory pathways, indicating that CMI may serve as a valuable indicator for poor prognosis among the older population.
Collapse
Affiliation(s)
- Bin Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
- Department of Cardiology, Zhongshan-Xuhui Hospital, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
| | - Qian Wu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China.
- Research Institute of Clinical Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea.
| | - Rui La
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Lingchen Lu
- Department of Pediatric Surgery and Rehabilitation, Kunshan Maternity and Children's Health Care Hospital, Kunshan, Jiangsu, China
| | - Fuad A Abdu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Guoqing Yin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Wen Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Wenquan Ding
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Yicheng Ling
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Zhiyuan He
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
8
|
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S, Liu C. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:130. [PMID: 38816371 PMCID: PMC11139930 DOI: 10.1038/s41392-024-01840-1] [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: 12/23/2023] [Revised: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 06/01/2024] Open
Abstract
The immune response holds a pivotal role in cardiovascular disease development. As multifunctional cells of the innate immune system, macrophages play an essential role in initial inflammatory response that occurs following cardiovascular injury, thereby inducing subsequent damage while also facilitating recovery. Meanwhile, the diverse phenotypes and phenotypic alterations of macrophages strongly associate with distinct types and severity of cardiovascular diseases, including coronary heart disease, valvular disease, myocarditis, cardiomyopathy, heart failure, atherosclerosis and aneurysm, which underscores the importance of investigating macrophage regulatory mechanisms within the context of specific diseases. Besides, recent strides in single-cell sequencing technologies have revealed macrophage heterogeneity, cell-cell interactions, and downstream mechanisms of therapeutic targets at a higher resolution, which brings new perspectives into macrophage-mediated mechanisms and potential therapeutic targets in cardiovascular diseases. Remarkably, myocardial fibrosis, a prevalent characteristic in most cardiac diseases, remains a formidable clinical challenge, necessitating a profound investigation into the impact of macrophages on myocardial fibrosis within the context of cardiac diseases. In this review, we systematically summarize the diverse phenotypic and functional plasticity of macrophages in regulatory mechanisms of cardiovascular diseases and unprecedented insights introduced by single-cell sequencing technologies, with a focus on different causes and characteristics of diseases, especially the relationship between inflammation and fibrosis in cardiac diseases (myocardial infarction, pressure overload, myocarditis, dilated cardiomyopathy, diabetic cardiomyopathy and cardiac aging) and the relationship between inflammation and vascular injury in vascular diseases (atherosclerosis and aneurysm). Finally, we also highlight the preclinical/clinical macrophage targeting strategies and translational implications.
Collapse
Affiliation(s)
- Runkai Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Hongrui Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Botao Tang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yukun Luo
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yufei Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Xin Zhong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Sifei Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Shengkang Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Canzhao Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| |
Collapse
|
9
|
McCutcheon K, Nqebelele U, Murray L, Thomas TS, Mpanya D, Tsabedze N. Cardiac and Renal Comorbidities in Aging People Living With HIV. Circ Res 2024; 134:1636-1660. [PMID: 38781295 PMCID: PMC11122746 DOI: 10.1161/circresaha.124.323948] [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: 05/25/2024]
Abstract
Contemporary World Health Organization data indicates that ≈39 million people are living with the human immunodeficiency virus. Of these, 24 million have been reported to have successfully accessed combination antiretroviral therapy. In 1996, the World Health Organization endorsed the widespread use of combination antiretroviral therapy, transforming human immunodeficiency virus infection from being a life-threatening disease to a chronic illness characterized by multiple comorbidities. The increased access to combination antiretroviral therapy has translated to people living with human immunodeficiency virus (PLWH) no longer having a reduced life expectancy. Although aging as a biological process increases exposure to oxidative stress and subsequent systemic inflammation, this effect is likely enhanced in PLWH as they age. This narrative review engages the intricate interplay between human immunodeficiency virus associated chronic inflammation, combination antiretroviral therapy, and cardiac and renal comorbidities development in aging PLWH. We examine the evolving demographic profile of PLWH, emphasizing the increasing prevalence of aging individuals within this population. A central focus of the review discusses the pathophysiological mechanisms that underpin the heightened susceptibility of PLWH to renal and cardiac diseases as they age.
Collapse
Affiliation(s)
| | - Unati Nqebelele
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, Western Cape, South Africa (U.N.)
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Western Cape, South Africa (U.N.)
| | - Lyle Murray
- Division of Infectious Diseases, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand and the Charlotte Maxeke Johannesburg Academic Hospital, South Africa (L.M.)
| | - Teressa Sumy Thomas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand and the Chris Hani Baragwanath Academic Hospital, Johannesburg, Gauteng, South Africa (T.S.T.)
| | - Dineo Mpanya
- Division of Cardiology, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa (D.M., N.T.)
| | - Nqoba Tsabedze
- Division of Cardiology, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa (D.M., N.T.)
| |
Collapse
|
10
|
Fularski P, Czarnik W, Dąbek B, Lisińska W, Radzioch E, Witkowska A, Młynarska E, Rysz J, Franczyk B. Broader Perspective on Atherosclerosis-Selected Risk Factors, Biomarkers, and Therapeutic Approach. Int J Mol Sci 2024; 25:5212. [PMID: 38791250 PMCID: PMC11121693 DOI: 10.3390/ijms25105212] [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: 03/30/2024] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) stands as the leading cause of mortality worldwide. At its core lies a progressive process of atherosclerosis, influenced by multiple factors. Among them, lifestyle-related factors are highlighted, with inadequate diet being one of the foremost, alongside factors such as cigarette smoking, low physical activity, and sleep deprivation. Another substantial group of risk factors comprises comorbidities. Amongst others, conditions such as hypertension, diabetes mellitus (DM), chronic kidney disease (CKD), or familial hypercholesterolemia (FH) are included here. Extremely significant in the context of halting progression is counteracting the mentioned risk factors, including through treatment of the underlying disease. What is more, in recent years, there has been increasing attention paid to perceiving atherosclerosis as an inflammation-related disease. Consequently, efforts are directed towards exploring new anti-inflammatory medications to limit ASCVD progression. Simultaneously, research is underway to identify biomarkers capable of providing insights into the ongoing process of atherosclerotic plaque formation. The aim of this study is to provide a broader perspective on ASCVD, particularly focusing on its characteristics, traditional and novel treatment methods, and biomarkers that can facilitate its early detection.
Collapse
Affiliation(s)
- Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Witold Czarnik
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Bartłomiej Dąbek
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewa Radzioch
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Alicja Witkowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
11
|
Neubauer-Geryk J, Wielicka M, Hoffmann M, Myśliwiec M, Bieniaszewski L. The Impact of Disease Duration on Microcirculatory Dysfunction in Young Patients with Uncomplicated Type 1 Diabetes. Biomedicines 2024; 12:1020. [PMID: 38790982 PMCID: PMC11117961 DOI: 10.3390/biomedicines12051020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
This study aimed to evaluate the earliest changes in the structure and function of the peripheral microcirculation using capillaroscopy and transcutaneous oxygen pressure measurement in children and adolescents with type 1 diabetes mellitus at baseline and during post-occlusive reactive hyperemia (PORH) in the function of diabetes duration. Sixty-seven patients with type 1 diabetes mellitus (T1D), aged 8 to 18 years, and twenty-eight age- and sex-matched healthy subjects were included in the analysis. Diabetic patients were divided into subgroups based on median disease duration. The subgroups differed in chronological age, lipid levels, and thyroid hormones. Capillaroscopy was performed twice: at baseline and then again after the PORH test. Transcutaneous oxygen pressure also was recorded under baseline conditions during and after the PORH test. Comparison of capillaroscopy and transcutaneous oxygen pressure parameters at rest and after the PORH showed no statistically significant difference between the subgroups. This remained true after adjusting for variables that differentiated the two subgroups. However, in the group of patients with long-standing diabetes, significant negative correlations were observed between the Coverage value after the PORH test and capillary reactivity with TcPO2_zero (biological zero). Significant positive correlations were also found between distance after the PORH test and TcPO2_zero. The results of our study indicate that in patients with a shorter duration of diabetes, the use of multiple tests provides a better characterization of the structure and function of microcirculation because the onset of dysfunction does not occur at the same time in all the tests.
Collapse
Affiliation(s)
- Jolanta Neubauer-Geryk
- Clinical Physiology Unit, Medical Simulation Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.W.); (L.B.)
| | - Melanie Wielicka
- Clinical Physiology Unit, Medical Simulation Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.W.); (L.B.)
| | - Magdalena Hoffmann
- Diabetology Outpatient Clinic, Non-Public Health Care Institution SAFMED, 83-000 Pruszcz Gdański, Poland;
| | - Małgorzata Myśliwiec
- Department of Pediatrics, Diabetology and Endocrinology, Medical University of Gdańsk, 80-211 Gdańsk, Poland;
| | - Leszek Bieniaszewski
- Clinical Physiology Unit, Medical Simulation Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.W.); (L.B.)
| |
Collapse
|
12
|
Aarstad HH, Moe SEE, Lybak S, Bruserud Ø, Tvedt THA, Aarstad HJ. Plasma IL-1 and IL-6 Family Cytokines with Soluble Receptor Levels at Diagnosis in Head and Neck Squamous Cell Carcinoma: High Levels Predict Decreased Five-Year Disease-Specific and Overall Survival. Cancers (Basel) 2024; 16:1484. [PMID: 38672565 PMCID: PMC11048558 DOI: 10.3390/cancers16081484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Activation of the acute-phase cascade (APC) has been correlated with outcomes in various cancers, including head and neck squamous cell carcinoma (HNSCC). Primary drivers of the APC are the cytokines within the interleukin-6 (IL-6) and IL-1 families. Plasma levels of IL-6 family cytokines/soluble receptors (IL-6, IL-27, IL-31, OSM, CNTF, soluble (s-)gp130, s-IL-6Rα) and IL-1 family members (IL-1RA, s-IL-33Rα) were determined at diagnosis for 87 human papillomavirus (HPV)-negative (-) HNSCC patients. We then studied the 5-year Disease-Specific Survival (DSS) and Overall Survival (OS). Increased plasma levels of IL-6 (p < 0.001/p < 0.001) (DSS/OS), IL-31 (p = 0.044/p = 0.07), IL-1RA (p = 0.004/p = 0.035), soluble (s)-IL-6Rα p = 0.022/p = 0.035), and s-gp130 (p = 0.007/p = 0.003) at diagnosis were predictors of both OS and DSS from HPV(-) HNSCC patients. The cytokine DSS/OS predictions were associated with TNM stage and smoking history, whereas the soluble receptors IL-6Rα, gp130, and IL33Rα more uniquely predicted DSS/OS. Clinically, IL-6 levels above 2.5 pg/mL yielded 75% specificity and 70% sensitivity for DSS. In conclusion, high plasma levels of IL-6, IL-31, and IL-1RA, as well as the soluble receptors IL-6Rα, gp130, and IL33Rα, predicted clinical outcome. This shows their potential as candidates for both general therapy and immune therapy stratification, as well as being future platforms for the development of new immunotherapy.
Collapse
Affiliation(s)
- Helene Hersvik Aarstad
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5009 Bergen, Norway; (H.H.A.); (S.E.E.M.); (S.L.)
- Department of Surgery, Haraldsplass Deaconal Hospital, 5009 Bergen, Norway
| | - Svein Erik Emblem Moe
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5009 Bergen, Norway; (H.H.A.); (S.E.E.M.); (S.L.)
| | - Stein Lybak
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5009 Bergen, Norway; (H.H.A.); (S.E.E.M.); (S.L.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5007 Bergen, Norway
| | - Øystein Bruserud
- Section for Haematology, Department of Medicine, Haukeland University Hospital, 5009 Bergen, Norway
| | | | - Hans Jørgen Aarstad
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5009 Bergen, Norway; (H.H.A.); (S.E.E.M.); (S.L.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5007 Bergen, Norway
| |
Collapse
|
13
|
Yu M, Gu J, Shi HS, Zhu ZF, Yang F, Yuan YF, Shuai XX, Wei YM, Cheng M, Yuan J, Xie T, Yang Y, Li DZ, Zhang M, Lu YX, Yang M, Zhou YC, Cheng X. No evidence of coronary plaque stabilization by allopurinol in patients with acute coronary syndrome. J Cardiovasc Comput Tomogr 2024; 18:195-202. [PMID: 38267335 DOI: 10.1016/j.jcct.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Allopurinol, a xanthine inhibitor that lowers uric acid concentration, has been proven to reduce inflammation and oxidative stress in patients with cardiovascular disease. However, it is unknown whether these beneficial effects translate into favorable plaque modification in acute coronary syndromes (ACS). This study aimed to investigate whether allopurinol could improve coronary plaque stabilization using coronary computed tomography angiography (CCTA). METHODS This was a prospective, single-center, randomized, double-blind clinical trial began in March 2019. A total of 162 ACS patients aged 18-80 years with a blood level of high-sensitivity C-reactive protein (hsCRP) > 2 mg/L were included. The subjects were randomly assigned in a 1:1 ratio to receive either allopurinol sustained-release capsules (at a dose of 0.25 g once daily) or placebo for 12 months. The plaque analysis was performed at CCTA. The primary efficacy endpoint was the change in low-attenuation plaque volume (LAPV) from baseline to the 12-month follow-up. RESULTS Among 162 patients, 54 in allopurinol group and 51 in placebo group completed the study. The median follow-up duration was 14 months in both groups. Compared with placebo, allopurinol therapy did not significantly alter LAPV (-13.4 ± 3.7 % vs. -17.8 ± 3.6 %, p = 0.390), intermediate attenuation plaque volume (-16.1 ± 3.0 % vs. -16.2 ± 2.9 %, p = 0.992), dense calcified plaque volume (12.2 ± 13.7 % vs. 9.7 ± 13.0 %, p = 0.894), total atheroma volume (-15.2 ± 3.2 % vs. -16.4 ± 3.1 %, p = 0.785), remodeling index (2.0 ± 3.9 % vs. 5.4 ± 3.8 %, p = 0.536) or hsCRP levels (-73.6 [-91.6-17.9] % vs. -81.2 [-95.4-47.7] %, p = 0.286). CONCLUSIONS Our findings suggest that allopurinol does not improve atherosclerotic plaque stability or inflammation in ACS.
Collapse
Affiliation(s)
- Miao Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jin Gu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, China
| | - He-Shui Shi
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, China
| | - Zheng-Feng Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Fen Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yuan-Fan Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xin-Xin Shuai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yu-Miao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jing Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Tian Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yong Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Da-Zhu Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Min Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yong-Xin Lu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Ming Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, China
| | - You-Cai Zhou
- Heilongjiang Aolida Ned Pharmaceutical Co.Ltd, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
14
|
Yu Y, Bian S, Jiang Y, Li B, Cui X, Ding S, Dai Z, Chen R, Zhong W, Yuan W. An Ex Vivo Aorta Culture Model to Study Vascular Cellular Senescence. Adv Biol (Weinh) 2024; 8:e2300140. [PMID: 38051940 DOI: 10.1002/adbi.202300140] [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: 04/09/2023] [Revised: 10/29/2023] [Indexed: 12/07/2023]
Abstract
Animal studies on vascular aging pose a few limitations. One of the most important reasons for this is the absence of a fast and efficient model of vascular tissue aging. In this study, ex vivo aortic culture and Matrigel subcutaneous implantation are combined to develop a new model for studying vascular cellular senescence. Eight-week-old C57BL/6J mice are used to obtain aortas. Bleomycin is used to induce aortas senescence in vitro. Then, aortas are transplanted to the acceptor mice with Matrigel. Senescence is evaluated using western blotting, quantitative polymerase chain reaction, and senescence-associated beta-galactosidase activity. Inflammatory cytokines are detected using Luminex Liquid Suspension Chip. RNA levels are analyzed by transcriptome sequencing. The results revealed that vessels in the bleomycin group exhibited significant senescence than those in the control group that can be enhanced by stripping vessel adventitia. The levels of cytokines such as interleukin (IL-2, IL-1β, and IL-6 increased significantly in the ex vivo model. Furthermore, transcriptome sequencing revealed 56 significantly differentially expressed genes (DEGs) in ex vivo model vessels compared with those in naturally aging aortas. In conclusion, this study introduces a cost-effective and time-saving vessel senescence model for vascular cellular senescence.
Collapse
Affiliation(s)
- Yijie Yu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Shihui Bian
- Department of Geriatrics, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Yu Jiang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Bo Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Xinggang Cui
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Shu Ding
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Zhiyin Dai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Rui Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Wei Zhong
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| |
Collapse
|
15
|
Tirandi A, Schiavetta E, Maioli E, Montecucco F, Liberale L. Inflammation as a cause of acute myocardial infarction in patients with myeloproliferative neoplasm. World J Cardiol 2024; 16:58-63. [PMID: 38456066 PMCID: PMC10915890 DOI: 10.4330/wjc.v16.i2.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/01/2024] [Accepted: 01/18/2024] [Indexed: 02/21/2024] Open
Abstract
Myeloproliferative neoplasms (MPN) are a group of diseases characterized by the clonal proliferation of hematopoietic progenitor or stem cells. They are clinically classifiable into four main diseases: chronic myeloid leukemia, essential thrombocythemia, polycythemia vera, and primary myelofibrosis. These pathologies are closely related to cardio- and cerebrovascular diseases due to the increased risk of arterial thrombosis, the most common underlying cause of acute myocardial infarction. Recent evidence shows that the classical Virchow triad (hypercoagulability, blood stasis, endothelial injury) might offer an explanation for such association. Indeed, patients with MPN might have a higher number and more reactive circulating platelets and leukocytes, a tendency toward blood stasis because of a high number of circulating red blood cells, endothelial injury or overactivation as a consequence of sustained inflammation caused by the neoplastic clonal cell. These abnormal cancer cells, especially when associated with the JAK2V617F mutation, tend to proliferate and secrete several inflammatory cytokines. This sustains a pro-inflammatory state throughout the body. The direct consequence is the induction of a pro-thrombotic state that acts as a determinant in favoring both venous and arterial thrombus formation. Clinically, MPN patients need to be carefully evaluated to be treated not only with cytoreductive treatments but also with cardiovascular protective strategies.
Collapse
Affiliation(s)
- Amedeo Tirandi
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Elisa Schiavetta
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Elia Maioli
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa 16132, Italy.
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa 16132, Italy
| |
Collapse
|
16
|
Jia X, He X, Huang C, Li J, Dong Z, Liu K. Protein translation: biological processes and therapeutic strategies for human diseases. Signal Transduct Target Ther 2024; 9:44. [PMID: 38388452 PMCID: PMC10884018 DOI: 10.1038/s41392-024-01749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Protein translation is a tightly regulated cellular process that is essential for gene expression and protein synthesis. The deregulation of this process is increasingly recognized as a critical factor in the pathogenesis of various human diseases. In this review, we discuss how deregulated translation can lead to aberrant protein synthesis, altered cellular functions, and disease progression. We explore the key mechanisms contributing to the deregulation of protein translation, including functional alterations in translation factors, tRNA, mRNA, and ribosome function. Deregulated translation leads to abnormal protein expression, disrupted cellular signaling, and perturbed cellular functions- all of which contribute to disease pathogenesis. The development of ribosome profiling techniques along with mass spectrometry-based proteomics, mRNA sequencing and single-cell approaches have opened new avenues for detecting diseases related to translation errors. Importantly, we highlight recent advances in therapies targeting translation-related disorders and their potential applications in neurodegenerative diseases, cancer, infectious diseases, and cardiovascular diseases. Moreover, the growing interest lies in targeted therapies aimed at restoring precise control over translation in diseased cells is discussed. In conclusion, this comprehensive review underscores the critical role of protein translation in disease and its potential as a therapeutic target. Advancements in understanding the molecular mechanisms of protein translation deregulation, coupled with the development of targeted therapies, offer promising avenues for improving disease outcomes in various human diseases. Additionally, it will unlock doors to the possibility of precision medicine by offering personalized therapies and a deeper understanding of the molecular underpinnings of diseases in the future.
Collapse
Affiliation(s)
- Xuechao Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Xinyu He
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Chuntian Huang
- Department of Pathology and Pathophysiology, Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
| | - Jian Li
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, 450052, China.
- Research Center for Basic Medicine Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, 450000, China.
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, 450052, China.
- Research Center for Basic Medicine Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, 450000, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, 450000, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450000, China.
| |
Collapse
|
17
|
Chen K, Liu Y, Xu B, Ye T, Chen L, Wu G, Zong G. Relationship between the lymphocyte to C‑reactive protein ratio and coronary artery disease severity. Exp Ther Med 2024; 27:60. [PMID: 38234629 PMCID: PMC10790159 DOI: 10.3892/etm.2023.12348] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/23/2023] [Indexed: 01/19/2024] Open
Abstract
Coronary atherosclerosis is a chronic systemic inflammatory disease. Laboratory parameters such as the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and systemic immune inflammation index (SII) have been used to assess inflammation degree and coronary artery disease (CAD) severity. The lymphocyte-to-C-reactive protein ratio (LCR) is a new SII. However, its relationship with CAD development and severity is unclear. A total of 1,107 patients (479 in control group, 628 in CAD group) underwent coronary angiography. The routine and biochemical indices of the venous blood of patients were assessed before coronary angiography. LCR, SII, NLR and PLR were calculated and statistical analyses were performed. Propensity score matching (PSM) and a logistic regression model were used to analyze the relationship between LCR and CAD. After the PSM, 384 pairs of patients with or without CAD were successfully matched. After the median binary classification of all indicators, uni- and multivariate logistic regression analyses showed that platelet count was an independent risk factor and LCR was an independent protective factor. Using the same method, in the coronary heart disease severity group, 212 pairs were successfully matched and NLR and PLR were independent risk factors, while LCR was an independent protective factor. In conclusion, LCR is an independent protective factor against CAD development and severity.
Collapse
Affiliation(s)
- Ke Chen
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Yehong Liu
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Baida Xu
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Ting Ye
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Liang Chen
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Gangyong Wu
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Gangjun Zong
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| |
Collapse
|
18
|
Caiazzo E, Sharma M, Rezig AOM, Morsy MI, Czesnikiewicz-Guzik M, Ialenti A, Sulicka-Grodzicka J, Pellicori P, Crouch SH, Schutte AE, Bruzzese D, Maffia P, Guzik TJ. Circulating cytokines and risk of developing hypertension: A systematic review and meta-analysis. Pharmacol Res 2024; 200:107050. [PMID: 38159784 DOI: 10.1016/j.phrs.2023.107050] [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: 09/10/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Immune responses play a significant role in hypertension, though the importance of key inflammatory mediators remains to be defined. We used a systematic literature review and meta-analysis to study the associations between key cytokines and incident hypertension. METHODS We performed a systematic search of Pubmed/Medline, Embase, Web of Science, and the Cochrane Central Register of Controlled Trials (CENTRAL), for peer-reviewed studies published up to August 2022. Incident hypertension was defined as systolic blood pressure ≥ 140 mmHg or diastolic blood pressure ≥ 90 mmHg and/or the use of antihypertensive medications. Random effects meta-analyses were used to calculate pooled hazard ratios (HRs)/risk ratios (RRs) and 95% confidence intervals by cytokine levels (highest vs. lowest quartile). RESULTS Only IL-6 and IL-1β levels have evidence allowing for quantitative evaluation concerning the onset of hypertension. Six studies (10406 participants, 2932 incident cases) examined the association of IL-6 with incident hypertension. The highest versus lowest quartile of circulating IL-6 was associated with a significant HR/RR of hypertension (1.61, 95% CI: 1.00 to 2.60; I2 =87%). After adjusting for potential confounders, including body mass index (BMI), HR/RR was no longer significant (HR/RR: 1.24; 95% CI, 0.96 to 1.61; I2 = 56%). About IL-1β, neither the crude (HR/RR: 1.03; 95% CI, 0.60 to 1.76; n = 2) nor multivariate analysis (HR/RR: 0.97, 95% CI, 0.60 to 1.56; n = 2) suggested a significant association with the risk of developing hypertension. CONCLUSIONS A limited number of studies suggest that higher IL-6, but not IL-1β, might be associated with the development of hypertension.
Collapse
Affiliation(s)
- Elisabetta Caiazzo
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Malvika Sharma
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Asma O M Rezig
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Moustafa I Morsy
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Marta Czesnikiewicz-Guzik
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Department of Periodontology, Prophylaxis and Oral Medicine, Jagiellonian University, Krakow, Poland
| | - Armando Ialenti
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Joanna Sulicka-Grodzicka
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Department of Rheumatology and Immunology, Jagiellonian University Medical College, Kracow, Poland
| | - Pierpaolo Pellicori
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Simone H Crouch
- SA MRC/Wits Developmental Pathways for Health Research Unit (DPHRU), School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa; MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa; The George Institute for Global Health, Sydney, Australia; School of Population Health, University of New South Wales, Sydney, Australia
| | - Dario Bruzzese
- Department of Public Health, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy; Africa-Europe Cluster of Research Excellence in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance (ARUA) & The Guild of European Research-intensive Universities, South Africa.
| | - Tomasz J Guzik
- Africa-Europe Cluster of Research Excellence in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance (ARUA) & The Guild of European Research-intensive Universities, South Africa; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; Department of Internal and Agricultural Medicine and Omicron Medical Genomics Laboratory, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
19
|
Pan W, Zhou G, Hu M, Li G, Zhang M, Yang H, Li K, Li J, Liu T, Wang Y, Jin J. Coenzyme Q10 mitigates macrophage mediated inflammation in heart following myocardial infarction via the NLRP3/IL1β pathway. BMC Cardiovasc Disord 2024; 24:76. [PMID: 38281937 PMCID: PMC10822151 DOI: 10.1186/s12872-024-03729-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: 04/27/2023] [Accepted: 01/15/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND The protective effect of Coenzyme Q10 (CoQ10) on the cardiovascular system has been reported, however, whether it can promote early recovery of cardiac function and alleviate cardiac remodeling after myocardial infarction (MI) remains to be elucidated. Whether CoQ10 may regulate the macrophage-mediated pro-inflammatory response after MI and its potential mechanism are worth further exploration. METHODS To determine the baseline plasma levels of CoQ10 by LC-MS/MS, healthy controls and MI patients (n = 11 each) with age- and gender-matched were randomly enrolled. Additional MI patients were consecutively enrolled and randomized into the blank control (n = 59) or CoQ10 group (n = 61). Follow-ups were performed at 1- and 3-month to assess cardiac function after percutaneous coronary intervention (PCI). In the animal study, mice were orally administered CoQ10/vehicle daily and were subjected to left anterior descending coronary artery (LAD) ligation or sham operation. Echocardiography and serum BNP measured by ELISA were analyzed to evaluate cardiac function. Masson staining and WGA staining were performed to analyze the myocardial fibrosis and cardiomyocyte hypertrophy, respectively. Immunofluorescence staining was performed to assess the infiltration of IL1β/ROS-positive macrophages into the ischemic myocardium. Flow cytometry was employed to analyze the recruitment of myeloid immune cells to the ischemic myocardium post-MI. The expression of inflammatory indicators was assessed through RNA-seq, qPCR, and western blotting (WB). RESULTS Compared to controls, MI patients showed a plasma deficiency of CoQ10 (0.76 ± 0.31 vs. 0.46 ± 0.10 µg/ml). CoQ10 supplementation significantly promoted the recovery of cardiac function in MI patients at 1 and 3 months after PCI. In mice study, compared to vehicle-treated MI mice, CoQ10-treated MI mice showed a favorable trend in survival rate (42.85% vs. 61.90%), as well as significantly alleviated cardiac dysfunction, myocardial fibrosis, and cardiac hypertrophy. Notably, CoQ10 administration significantly suppressed the recruitment of pro-inflammatory CCR2+ macrophages into infarct myocardium and their mediated inflammatory response, partially by attenuating the activation of the NLR family pyrin domain containing 3 (NLRP3)/Interleukin-1 beta (IL1β) signaling pathway. CONCLUSIONS These findings suggest that CoQ10 can significantly promote early recovery of cardiac function after MI. CoQ10 may function by inhibiting the recruitment of CCR2+ macrophages and suppressing the activation of the NLRP3/IL1β pathway in macrophages. TRIAL REGISTRATION Date of registration 09/04/2021 (number: ChiCTR2100045256).
Collapse
Affiliation(s)
- Wenxu Pan
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Guiquan Zhou
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Meiling Hu
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Gaoshan Li
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Mingle Zhang
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Hao Yang
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Kunyan Li
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jingwei Li
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Ting Liu
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Ying Wang
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Jun Jin
- Department of Cardiology, The Second Affiliated Hospital of Army Medical University, Chongqing, China.
| |
Collapse
|
20
|
Cui Y, Zhu Q, Hao H, Flaker GC, Liu Z. N-Acetylcysteine and Atherosclerosis: Promises and Challenges. Antioxidants (Basel) 2023; 12:2073. [PMID: 38136193 PMCID: PMC10741030 DOI: 10.3390/antiox12122073] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Atherosclerosis remains a leading cause of cardiovascular diseases. Although the mechanism for atherosclerosis is complex and has not been fully understood, inflammation and oxidative stress play a critical role in the development and progression of atherosclerosis. N-acetylcysteine (NAC) has been used as a mucolytic agent and an antidote for acetaminophen overdose with a well-established safety profile. NAC has antioxidant and anti-inflammatory effects through multiple mechanisms, including an increase in the intracellular glutathione level and an attenuation of the nuclear factor kappa-B mediated production of inflammatory cytokines like tumor necrosis factor-alpha and interleukins. Numerous animal studies have demonstrated that NAC significantly decreases the development and progression of atherosclerosis. However, the data on the outcomes of clinical studies in patients with atherosclerosis have been limited and inconsistent. The purpose of this review is to summarize the data on the effect of NAC on atherosclerosis from both pre-clinical and clinical studies and discuss the potential mechanisms of action of NAC on atherosclerosis, as well as challenges in the field.
Collapse
Affiliation(s)
- Yuqi Cui
- Department of Geriatrics, Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA;
| | - Qiang Zhu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Gregory C. Flaker
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
21
|
Zhu L, Tang Z, Hu R, Gu M, Yang Y. Ageing and Inflammation: What Happens in Periodontium? Bioengineering (Basel) 2023; 10:1274. [PMID: 38002398 PMCID: PMC10669535 DOI: 10.3390/bioengineering10111274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Periodontitis is a chronic inflammatory disease with a high incidence and severity in the elderly population, making it a significant public health concern. Ageing is a primary risk factor for the development of periodontitis, exacerbating alveolar bone loss and leading to tooth loss in the geriatric population. Despite extensive research, the precise molecular mechanisms underlying the relationship between ageing and periodontitis remain elusive. Understanding the intricate mechanisms that connect ageing and inflammation may help reveal new therapeutic targets and provide valuable options to tackle the challenges encountered by the rapidly expanding global ageing population. In this review, we highlight the latest scientific breakthroughs in the pathways by which inflammaging mediates the decline in periodontal function and triggers the onset of periodontitis. We also provide a comprehensive overview of the latest findings and discuss potential avenues for future research in this critical area of investigation.
Collapse
Affiliation(s)
| | | | | | | | - Yanqi Yang
- Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR 999077, China; (L.Z.); (Z.T.); (R.H.); (M.G.)
| |
Collapse
|
22
|
Tokuda K, Tanaka A, Tobe A, Shirai Y, Kurobe M, Kubota Y, Kunieda T, Miyazaki T, Mizutani K, Furusawa K, Ishii H, Murohara T. Impact of C-Reactive Protein on Long-Term Cardiac Events in Stable Coronary Artery Disease Patients with Chronic Kidney Disease. J Atheroscler Thromb 2023; 30:1635-1643. [PMID: 36908149 PMCID: PMC10627763 DOI: 10.5551/jat.64047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/21/2023] [Indexed: 03/13/2023] Open
Abstract
AIM Chronic inflammation is associated with atherosclerosis development. Chronic kidney disease (CKD) is an independent risk factor for cardiovascular events and is associated with chronic inflammation. We aimed to investigate the influence of C-reactive protein (CRP), an important marker of inflammation, on the clinical outcomes of patients with CKD and stable coronary artery disease (CAD) undergoing percutaneous coronary intervention (PCI). METHODS Among patients with stable CAD and CKD who underwent PCI, 516 patients whose CRP levels were available before the PCI procedure were identified. The patients were divided into two groups according to the CRP levels: those with CRP ≥ 2.0 mg/L (high-CRP group) and those with CRP <2.0 mg/L (low-CRP group). The primary endpoint of this study was the occurrence of major adverse cardiac events (MACE), defined as a composite of cardiac death, myocardial infarction, and unplanned revascularization. RESULTS Overall, the mean age of the patients was 72.5±9.7 years, and 20.7% were female. The median CRP level was 1.43 mg/L (0.6-4.9 mg/L). The median follow-up period was 3.6 years. The occurrence of MACE was significantly higher in the high-CRP group than in the low-CRP group (log-rank p<0.001). Notably, the incidence rate of cardiac death was significantly higher in the high-CRP group (log-rank p<0.001). According to the multivariable analysis, CRP level ≥ 2.0 mg/L was found to be a significant predictor of MACE (hazard ratio [HR]: 1.54, 95% confidence interval [CI]: 1.04-2.28, p=0.003), as well as estimated glomerular filtration rate (HR: 0.98, 95% CI: 0.97-0.99, p<0.01). CONCLUSION High-CRP levels adversely affect long-term cardiac events in patients with stable CAD and CKD.
Collapse
Affiliation(s)
- Kotaro Tokuda
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Akihito Tanaka
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Akihiro Tobe
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Yoshinori Shirai
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Masanari Kurobe
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Yoshiaki Kubota
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Takeshige Kunieda
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Tatsuya Miyazaki
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Koji Mizutani
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Kenji Furusawa
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| | - Hideki Ishii
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
- Department of Cardiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University graduate school of medicine, Nagoya, Japan
| |
Collapse
|
23
|
Tirandi A, Montecucco F, Liberale L. Heart and vessels 'on fire'. Eur J Clin Invest 2023; 53:e14052. [PMID: 37394695 DOI: 10.1111/eci.14052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/09/2023] [Accepted: 06/24/2023] [Indexed: 07/04/2023]
Affiliation(s)
- Amedeo Tirandi
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
24
|
Mengozzi A, de Ciuceis C, Dell'oro R, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Anyfanti P, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Mavraganis G, Montezano AC, Rios FJ, Winklewski PJ, Wolf J, Costantino S, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Triantafyllou A, Virdis A. The importance of microvascular inflammation in ageing and age-related diseases: a position paper from the ESH working group on small arteries, section of microvascular inflammation. J Hypertens 2023; 41:1521-1543. [PMID: 37382158 DOI: 10.1097/hjh.0000000000003503] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Microcirculation is pervasive and orchestrates a profound regulatory cross-talk with the surrounding tissue and organs. Similarly, it is one of the earliest biological systems targeted by environmental stressors and consequently involved in the development and progression of ageing and age-related disease. Microvascular dysfunction, if not targeted, leads to a steady derangement of the phenotype, which cumulates comorbidities and eventually results in a nonrescuable, very high-cardiovascular risk. Along the broad spectrum of pathologies, both shared and distinct molecular pathways and pathophysiological alteration are involved in the disruption of microvascular homeostasis, all pointing to microvascular inflammation as the putative primary culprit. This position paper explores the presence and the detrimental contribution of microvascular inflammation across the whole spectrum of chronic age-related diseases, which characterise the 21st-century healthcare landscape. The manuscript aims to strongly affirm the centrality of microvascular inflammation by recapitulating the current evidence and providing a clear synoptic view of the whole cardiometabolic derangement. Indeed, there is an urgent need for further mechanistic exploration to identify clear, very early or disease-specific molecular targets to provide an effective therapeutic strategy against the otherwise unstoppable rising prevalence of age-related diseases.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | - Raffaella Dell'oro
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - George Pavlidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Panagiota Anyfanti
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Livia L Camargo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine
- Center of Translational Medicine
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marcin Hellmann
- Department of Cardiac Diagnostics, Medical University, Gdansk, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Cardiovascular Science, University College London, London, UK
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Francesco J Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | | | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Ignatios Ikonomidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | | | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
- Division of Medicine, Spedali Civili di Brescia, Montichiari, Brescia, Italy
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site
- Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Areti Triantafyllou
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
25
|
Turkulainen E, Ihalainen J, Arvas M. Simulated effects of ferritin screening on C-reactive protein levels in recruited blood donors. Vox Sang 2023; 118:901-905. [PMID: 37622476 DOI: 10.1111/vox.13515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND AND OBJECTIVES Ferritin is commonly measured to evaluate iron stores in the body. Some countries have added or considered adding ferritin lower bounds to donor eligibility criteria. Ferritin is also elevated by inflammation. The main goal of this study is to estimate how different ferritin cut-offs would affect the proportion of donors with a C-reactive protein (CRP) level over 3 mg/L, which is the decision limit of the highest chronic cardiovascular risk. MATERIALS AND METHODS To simulate recruitment of new blood donors, we selected participants from two Finnish general population cohorts, namely FINRISK 1997 (n = 5369) and Health 2000 (n = 3278), that would likely fulfil the selection criteria of blood donation. We then calculated the proportion of individuals with high-sensitivity CRP values above 3 mg/L, over a range of ferritin values. RESULTS We found that for several ferritin cut-offs the proportion of potential donors with CRP > 3 mg/L would rise by a statistically significant amount. The trend was significant and similar for all subgroups but weaker for non-menstruating women as well as men. CONCLUSION Our results show that screening a population of potential blood donors with ferritin cut-offs raises the number of people with CRP > 3 mg/L within the blood donor population.
Collapse
Affiliation(s)
- Esa Turkulainen
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Jarkko Ihalainen
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Mikko Arvas
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| |
Collapse
|
26
|
Zhang H, Jiang S, Hao M, Li Y, Hu Z, Jiang X, Jin L, Wang X. Association of cardiometabolic multimorbidity with motoric cognitive risk syndrome in older adults. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12491. [PMID: 37937160 PMCID: PMC10626031 DOI: 10.1002/dad2.12491] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/01/2023] [Accepted: 09/25/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Motoric cognitive risk syndrome (MCR) is a predementia syndrome that is characterized by cognitive complaints and slow gait. Cardiometabolic multimorbidity (CMM) is associated with an increased risk of dementia. However, the relationship between CMM and MCR is still unclear. METHODS We included 4744 participants (aged 65+ years) without MCR at baseline from the National Health and Aging Trends Study (NHATS), who were followed-up from 2011 to 2018. CMM was defined as the presence of two or more cardiometabolic diseases (including diabetes mellitus, heart disease, and stroke). RESULTS CMM was significantly associated with an increased risk of MCR (hazard ratio [HR] 1.41, 95% confidence interval [CI] 1.13-1.75) in fully adjusted models. Consistent results were observed from stratified analyses of different subgroups. Increasing numbers of cardiometabolic diseases were dose-dependently associated with increased MCR risk (HR 1.33, 95% CI 1.20-1.48). DISCUSSION CMM is associated with an increased risk of MCR in older adults. HIGHLIGHTS Motoric cognitive risk syndrome (MCR) is a predementia syndrome characterized by slow gait speed and cognitive complaints.Cardiometabolic multimorbidity was associated with an increased MCR risk.An increased number of cardiometabolic diseases were dose-dependently associated with increased MCR risk.
Collapse
Affiliation(s)
- Hui Zhang
- Human Phenome InstituteZhangjiang Fudan International Innovation CentreFudan UniversityShanghaiChina
- Department of Vascular SurgeryShanghai Key Laboratory of Vascular Lesion Regulation and RemodelingShanghai Pudong HospitalFudan University Pudong Medical CenterShanghaiChina
| | - Shuai Jiang
- Department of Vascular SurgeryShanghai Key Laboratory of Vascular Lesion Regulation and RemodelingShanghai Pudong HospitalFudan University Pudong Medical CenterShanghaiChina
| | - Meng Hao
- Human Phenome InstituteZhangjiang Fudan International Innovation CentreFudan UniversityShanghaiChina
| | - Yi Li
- Human Phenome InstituteZhangjiang Fudan International Innovation CentreFudan UniversityShanghaiChina
| | - Zixin Hu
- Artificial Intelligence Innovation and Incubation InstituteFudan UniversityShanghaiChina
| | - Xiao‐Yan Jiang
- State Key Laboratory of CardiologyDepartment of Pathology and PathophysiologySchool of MedicineTongji UniversityShanghaiChina
| | - Li Jin
- Human Phenome InstituteZhangjiang Fudan International Innovation CentreFudan UniversityShanghaiChina
| | - Xiaofeng Wang
- Human Phenome InstituteZhangjiang Fudan International Innovation CentreFudan UniversityShanghaiChina
- National Clinical Research Centre for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
27
|
Wu Z, Zeng S, Wang X, Liu H, Sun H, Zhou X, Yang Q. Patterns of Circulating Microbiota during the Acute Phase Following ST-Segment Elevation Myocardial Infarction Predict Long-Term Cardiovascular Events. Int Heart J 2023; 64:551-561. [PMID: 37460320 DOI: 10.1536/ihj.22-672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Limited information exists regarding whether circulating microbiota could predict long-term clinical outcomes following ST-segment elevation myocardial infarction (STEMI). A total of 244 consecutive patients with STEMI were followed for 2.8 years, and 64 first major adverse cardiovascular events (MACEs) were recorded. Both microbiota abundance [Corynebacterium tuberculostearicum (HR, 1.28; 95% CI, 1.03-1.58) and Staphylococcus aureus (S. aureus) (HR, 1.16; 95% CI, 1.02-1.33) ] and microbiota clusters (Cluster 2 versus Cluster 1: HR, 1.84; 95% CI, 1.04-3.27) could independently predict MACE. Furthermore, a model based on established independent predictors alone was significantly improved by the addition of different microbiota patterns. In addition, CD14++CD16+ monocytes (Mon2) had a significant mediation effect on the microbiota patterns → MACE association. The present study demonstrated that the abundance and clusters of circulating microbiota are associated with future adverse cardiovascular events independent of traditional risk factors, which were partially mediated by an increase in Mon2.
Collapse
Affiliation(s)
- Zhaogui Wu
- Department of Cardiology, Tianjin Medical University General Hospital
| | - Shan Zeng
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Heart Center, Pingjin Hospital
| | - Xuezhu Wang
- Department of Cardiology, Tianjin Medical University General Hospital
| | - Hangkuan Liu
- Department of Cardiology, Tianjin Medical University General Hospital
| | - Haonan Sun
- Department of Cardiology, Tianjin Medical University General Hospital
| | - Xin Zhou
- Department of Cardiology, Tianjin Medical University General Hospital
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital
| |
Collapse
|
28
|
Guzik TJ, Channon KM. JCAD: a new GWAS target to reduce residual cardiovascular risk? Eur Heart J 2023; 44:1834-1836. [PMID: 36514298 PMCID: PMC10200022 DOI: 10.1093/eurheartj/ehac708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Tomasz J Guzik
- Centre for Cardiovascular Sciences, British Heart Foundation Centre of Research Excellence, Queen’s Medical Research Institute, Edinburgh Royal Infirmary, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
- Department of Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Keith M Channon
- Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
29
|
Tanase DM, Valasciuc E, Gosav EM, Ouatu A, Buliga-Finis ON, Floria M, Maranduca MA, Serban IL. Portrayal of NLRP3 Inflammasome in Atherosclerosis: Current Knowledge and Therapeutic Targets. Int J Mol Sci 2023; 24:ijms24098162. [PMID: 37175869 PMCID: PMC10179095 DOI: 10.3390/ijms24098162] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/26/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
We are witnessing the globalization of a specific type of arteriosclerosis with rising prevalence, incidence and an overall cardiovascular disease burden. Currently, atherosclerosis increasingly affects the younger generation as compared to previous decades. While early preventive medicine has seen improvements, research advances in laboratory and clinical investigation promise to provide us with novel diagnosis tools. Given the physio-pathological complexity and epigenetic patterns of atherosclerosis and the discovery of new molecules involved, the therapeutic field of atherosclerosis has room for substantial growth. Thus, the scientific community is currently investigating the role of nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a crucial component of the innate immune system in different inflammatory disorders. NLRP3 is activated by distinct factors and numerous cellular and molecular events which trigger NLRP3 inflammasome assembly with subsequent cleavage of pro-interleukin (IL)-1β and pro-IL-18 pathways via caspase-1 activation, eliciting endothelial dysfunction, promotion of oxidative stress and the inflammation process of atherosclerosis. In this review, we introduce the basic cellular and molecular mechanisms of NLRP3 inflammasome activation and its role in atherosclerosis. We also emphasize its promising therapeutic pharmaceutical potential.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Oana Nicoleta Buliga-Finis
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Minela Aida Maranduca
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Department of Morpho-Functional Sciences II, Discipline of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ionela Lacramioara Serban
- Department of Morpho-Functional Sciences II, Discipline of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
30
|
Cimmino G, Muscoli S, De Rosa S, Cesaro A, Perrone MA, Selvaggio S, Selvaggio G, Aimo A, Pedrinelli R, Mercuro G, Romeo F, Perrone Filardi P, Indolfi C, Coronelli M. Evolving concepts in the pathophysiology of atherosclerosis: from endothelial dysfunction to thrombus formation through multiple shades of inflammation. J Cardiovasc Med (Hagerstown) 2023; 24:e156-e167. [PMID: 37186566 DOI: 10.2459/jcm.0000000000001450] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Atherosclerosis is the anatomo-pathological substrate of most cardio, cerebro and vascular diseases such as acute and chronic coronary syndromes, stroke and peripheral artery diseases. The pathophysiology of atherosclerotic plaque and its complications are under continuous investigation. In the last 2 decades our understanding on the formation, progression and complication of the atherosclerotic lesion has greatly improved and the role of immunity and inflammation is now well documented and accepted. The conventional risk factors modulate endothelial function determining the switch to a proatherosclerotic phenotype. From this point, lipid accumulation with an imbalance from cholesterol influx and efflux, foam cells formation, T-cell activation, cytokines release and matrix-degrading enzymes production occur. Lesions with high inflammatory rate become vulnerable and prone to rupture. Once complicated, the intraplaque thrombogenic material, such as the tissue factor, is exposed to the flowing blood, thus inducing coagulation cascade activation, platelets aggregation and finally intravascular thrombus formation that leads to clinical manifestations of this disease. Nonconventional risk factors, such as gut microbiome, are emerging novel markers of atherosclerosis. Several data indicate that gut microbiota may play a causative role in formation, progression and complication of atherosclerotic lesions. The gut dysbiosis-related inflammation and gut microbiota-derived metabolites have been proposed as the main working hypothesis in contributing to disease formation and progression. The current evidence suggest that the conventional and nonconventional risk factors may modulate the degree of inflammation of the atherosclerotic lesion, thus influencing its final fate. Based on this hypothesis, targeting inflammation seems to be a promising approach to further improve our management of atherosclerotic-related diseases.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
| | | | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro
| | - Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Marco A Perrone
- Department of Cardiology and CardioLab, University of Rome Tor Vergata, Rome
| | | | | | - Alberto Aimo
- Fondazione Toscana Gabriele Monasterio
- Institute of Life Sciences, Scuola Superiore Sant'Anna
| | - Roberto Pedrinelli
- Critical Care Medicine-Cardiology Division, Department of Surgical, Medical and Molecular Pathology, University of Pisa, Pisa
| | - Giuseppe Mercuro
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi, Cagliari
| | | | - Pasquale Perrone Filardi
- Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli 'Federico II', Napoli
| | - Ciro Indolfi
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro
| | - Maurizio Coronelli
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
| |
Collapse
|
31
|
Zhou W, Wang C, Zhang B, Gou S. Hybrids of carbonic anhydrase and cyclooxygenase inhibitors attenuate cardiac hypoxic inflammatory injuries. Eur J Pharmacol 2023; 950:175751. [PMID: 37116562 DOI: 10.1016/j.ejphar.2023.175751] [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: 12/03/2022] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 04/30/2023]
Abstract
Cardiac inflammation is easily accompanied by hypoxia, while hypoxia-induced injury and microenvironmental variations limit the efficacy of common anti-inflammatory drugs. In order to effectively attenuate myocardial injury caused by hypoxic and inflammatory injury, we designed and synthesized a kind of anti-inflammatory compounds by coupling cyclooxygenase (COX) and carbonic anhydrase (CA) inhibitors, and evaluated the activity and their mechanism in vitro and in vivo. It was found that these compounds were structurally stable and had two enzymatic inhibition activities. By inhibiting the activity of overexpressed CA under hypoxia, the acidic microenvironment can be regulated to inhibit the hypoxic injury, in which the pH-dependent primary drug resistance can be overcome to improve the anti-inflammatory effect of the COX inhibitor. Consequently, this study provides a new strategy for the treatment of cardiac inflammation accompanied by hypoxia.
Collapse
Affiliation(s)
- Wen Zhou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, PR China
| | - Chunping Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Bin Zhang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, PR China.
| |
Collapse
|
32
|
Becerra CY, Wells RK, Kunihiro BP, Lee RH, Umeda L, Allan NP, Rubas NC, McCracken TA, Nunokawa CKL, Lee MH, Pidlaoan FGS, Phankitnirondorn K, Dye CK, Yamamoto BY, Peres R, Juarez R, Maunakea AK. Examining the immunoepigenetic-gut microbiome axis in the context of self-esteem among Native Hawaiians and other Pacific Islanders. Front Genet 2023; 14:1125217. [PMID: 37152987 PMCID: PMC10154580 DOI: 10.3389/fgene.2023.1125217] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/21/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction: Native Hawaiian and other Pacific Islander (NHPI) populations experience higher rates of immunometabolic diseases compared to other racial-ethnic groups in Hawaii. As annual NHPI mortality rates for suicide and type 2 diabetes mellitus (T2DM) exceed those of the state as a whole, understanding the social and biological mechanisms underlying these disparities are urgently needed to enable preventive strategies. Methods: A community-based approach was used to investigate the immunoepigenetic-gut microbiome axis in an NHPI-enriched cohort of Oahu residents (N = 68). Self-esteem (SE) data was collected using a modified Rosenberg self-esteem (SE) assessment as a proxy measure for mental wellbeing in consideration for cultural competency. T2DM status was evaluated using point-of-care A1c (%) tests. Stool samples were collected for 16s-based metagenomic sequencing analyses. Plasma from blood samples were isolated by density-gradient centrifugation. Peripheral blood mononuclear cells (PBMCs) were collected from the same samples and enriched for monocytes using negative selection techniques. Flow-cytometry was used for immunoprofiling assays. Monocyte DNA was extracted for Illumina EPIC array-based methylation analysis. Results: Compared to individuals with normal SE (NSE), those with low SE (LSE) exhibited significantly higher plasma concentrations (pg/ml) of proinflammatory cytokines IL-8 (p = 0.051) and TNF-α (p = 0.011). Metagenomic analysis revealed that the relative abundance (%) of specific gut bacteria significantly differed between SE groups - some of which directly correlated with SE scores. Gene ontology analysis revealed that 104 significantly differentially methylated loci (DML) between SE groups were preferentially located at genes involved in immunometabolic processes. Horvath clock analyses indicated epigenetic age (Epi-Age) deceleration in individuals with LSE and acceleration in individuals with NSE (p = 0.042), yet was not reproduced by other clocks. Discussion: These data reveal novel differences in the immunoepigenetic-gut microbiome axis with respect to SE, warranting further investigation into its relationship to brain activity and mental health in NHPI. Unexpected results from Epi-Age analyses warrant further investigation into the relationship between biological age and disparate health outcomes among the NHPI population. The modifiable component of epigenetic processes and the gut microbiome makes this axis an attractive target for potential therapeutics, biomarker discovery, and novel prevention strategies.
Collapse
Affiliation(s)
- Celyna Y Becerra
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
- IDeA Networks of Biomedical Research Excellence (INBRE), University of Hawaii at Manoa, Honolulu, HI, United States
| | - Riley K Wells
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Braden P Kunihiro
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
- IDeA Networks of Biomedical Research Excellence (INBRE), University of Hawaii at Manoa, Honolulu, HI, United States
| | - Rosa H Lee
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Lesley Umeda
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Nina P Allan
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Noelle C Rubas
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Trevor A McCracken
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Chandler K L Nunokawa
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Ming-Hao Lee
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Felix Gerard S Pidlaoan
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Krit Phankitnirondorn
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Christian K Dye
- Department of Environmental Health Sciences, Columbia University Irving Medical Center, NY, NY, United States
| | - Brennan Y Yamamoto
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Rafael Peres
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| | - Ruben Juarez
- Department of Economics, University of Hawaii at Manoa, Honolulu, HI, United States
- University of Hawaii Economic Research Organization (UHERO), University of Hawaii at Manoa, Honolulu, HI, United States
| | - Alika K Maunakea
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI, United States
| |
Collapse
|
33
|
Pugliese NR, Pellicori P, Filidei F, De Biase N, Maffia P, Guzik TJ, Masi S, Taddei S, Cleland JGF. Inflammatory pathways in heart failure with preserved left ventricular ejection fraction: implications for future interventions. Cardiovasc Res 2023; 118:3536-3555. [PMID: 36004819 PMCID: PMC9897694 DOI: 10.1093/cvr/cvac133] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
Many patients with symptoms and signs of heart failure have a left ventricular ejection fraction ≥50%, termed heart failure with preserved ejection fraction (HFpEF). HFpEF is a heterogeneous syndrome mainly affecting older people who have many other cardiac and non-cardiac conditions that often cast doubt on the origin of symptoms, such as breathlessness, or signs, such as peripheral oedema, rendering them neither sensitive nor specific to the diagnosis of HFpEF. Currently, management of HFpEF is mainly directed at controlling symptoms and treating comorbid conditions such as hypertension, atrial fibrillation, anaemia, and coronary artery disease. HFpEF is also characterized by a persistent increase in inflammatory biomarkers. Inflammation may be a key driver of the development and progression of HFpEF and many of its associated comorbidities. Detailed characterization of specific inflammatory pathways may provide insights into the pathophysiology of HFpEF and guide its future management. There is growing interest in novel therapies specifically designed to target deregulated inflammation in many therapeutic areas, including cardiovascular disease. However, large-scale clinical trials investigating the effectiveness of anti-inflammatory treatments in HFpEF are still lacking. In this manuscript, we review the role of inflammation in HFpEF and the possible implications for future trials.
Collapse
Affiliation(s)
| | - Pierpaolo Pellicori
- Robertson Institute of Biostatistics and Clinical Trials Unit, University of Glasgow, Glasgow G12 8QQ, UK
| | - Francesco Filidei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Nicolò De Biase
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples 80138, Italy
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Internal and Agricultural Medicine, Jagiellonian University, Collegium Medicum, Krakow 31-008, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - John G F Cleland
- Robertson Institute of Biostatistics and Clinical Trials Unit, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
34
|
Pan K, Xu C, Chen C, Chen S, Zhang Y, Ding X, Xu X, Lv Q. Soluble interleukin-2 receptor combined with interleukin-8 is a powerful predictor of future adverse cardiovascular events in patients with acute myocardial infarction. Front Cardiovasc Med 2023; 10:1110742. [PMID: 37139133 PMCID: PMC10150071 DOI: 10.3389/fcvm.2023.1110742] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/21/2023] [Indexed: 05/05/2023] Open
Abstract
Background Little is known about the role of interleukin (IL) in patients with acute myocardial infarction (MI), especially soluble IL-2 receptor (sIL-2R) and IL-8. We aim to evaluate, in MI patients, the predictive value of serum sIL-2R and IL-8 for future major adverse cardiovascular events (MACEs), and compare them with current biomarkers reflecting myocardial inflammation and injury. Methods This was a prospective, single-center cohort study. We measured serum concentrations of IL-1β, sIL-2R, IL-6, IL-8 and IL-10. Levels of current biomarkers for predicting MACEs were measured, including high-sensitivity C reactive protein, cardiac troponin T and N-terminal pro-brain natriuretic peptide. Clinical events were collected during 1-year and a median of 2.2 years (long-term) follow-up. Results Twenty-four patients (13.8%, 24/173) experienced MACEs during 1-year follow-up and 40 patients (23.1%, 40/173) during long-term follow-up. Of the five interleukins studied, only sIL-2R and IL-8 were independently associated with endpoints during 1-year or long-term follow-up. Patients with high sIL-2R or IL-8 levels (higher than the cutoff value) had a significantly higher risk of MACEs during 1-year (sIL-2R: HR 7.7, 3.3-18.0, p < 0.001; IL-8: HR 4.8, 2.1-10.7, p < 0.001) and long-term (sIL-2R: HR 7.7, 3.3-18.0, p < 0.001; IL-8: HR 4.8, 2.1-10.7, p < 0.001) follow-up. Receiver operator characteristic curve analysis regarding predictive accuracy for MACEs during 1-year follow-up showed that the area under the curve for sIL-2R, IL-8, sIL-2R combined with IL-8 was 0.66 (0.54-0.79, p = 0.011), 0.69 (0.56-0.82, p < 0.001) and 0.720 (0.59-0.85, p < 0.001), whose predictive value were superior to that of current biomarkers. The addition of sIL-2R combined with IL-8 to the existing prediction model resulted in a significant improvement in predictive power (p = 0.029), prompting a 20.8% increase in the proportion of correct classifications. Conclusions High serum sIL-2R combined with IL-8 levels was significantly associated with MACEs during follow-up in patients with MI, suggesting that sIL-2R combined with IL-8 may be a helpful biomarker for identifying the increased risk of new cardiovascular events. IL-2 and IL-8 would be promising therapeutic targets for anti-inflammatory therapy.
Collapse
Affiliation(s)
- Kunming Pan
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
| | - Chenqi Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Kidney Disease and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
| | - Can Chen
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shuqing Chen
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqian Zhang
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Kidney Disease and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Kidney Disease and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Correspondence: Qianzhou Lv Xialian Xu
| | - Qianzhou Lv
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
- Correspondence: Qianzhou Lv Xialian Xu
| |
Collapse
|
35
|
Carbone F, Elia E, Casula M, Bonaventura A, Bertolotto M, Minetti S, Artom N, Camici GG, Contini P, Pontremoli R, Viazzi F, Bertolini S, Pende A, Pisciotta L, Montecucco F, Liberale L. Among biomarkers of neutrophil activity, matrix metalloproteinases 8 independently predicts remission of metabolic syndrome. Nutr Metab Cardiovasc Dis 2023; 33:185-193. [PMID: 36411219 DOI: 10.1016/j.numecd.2022.10.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 09/24/2022] [Accepted: 10/24/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Inflammation due to the excess of nutrient intake plays an important role in the pathophysiology of metabolic syndrome (MetS). Here, the potential influence of neutrophils and their degranulation markers on MetS improvement upon dietary and behavioral counselling, has been investigated. Specifically, we aimed at investigating their role as potential predictors of metabolic syndrome improvements. METHODS AND RESULTS patients with MetS (n = 127) received behavioral and dietary recommendations before follow-up at 6 months. Serum levels of matrix metalloproteinases (MMP)8, MMP9, myeloperoxidase (MPO), tissue inhibitor of MMP (TIMP)-1, TIMP-2, TIMP-3 and resistin were tested at baseline. In the whole cohort, baseline levels of proinflammatory MMP8, MMP9 and MPO increased together with the number of MetS criteria. Seventy-three (57%) patients experienced a reduction in MetS-defining criteria at follow-up. With respect to those with no improvement, such individuals showed lower weight and waist circumference at enrolment, less frequent smoking habits, higher levels of triglycerides and lower circulating MMP8. At logistic regression analysis, baseline MMP8 showed negative predictive ability (odds ratio (OR) 0.979 [0.961-0.997]; p = 0.025) against MetS improvement. Such findings hold true even when included in the backward stepwise logistic regression model confirming MMP8 as an independent predictor (OR 0.970 [0.949-0.993]; p = 0.009). Receiver operating characteristic (ROC) curve confirmed the predictive ability of MMP8 combined in a model including baseline MetS criteria and waist circumference. Bootstrap resampling analysis internally validated our findings. CONCLUSION Improvement of MetS is independently associated with baseline low MMP-8 levels, suggesting a pivotal role for inflammation in metabolic alteration.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
| | - Edoardo Elia
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Matteo Casula
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Silvia Minetti
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Nathan Artom
- Department of Internal Medicine, Ospedale S. Paolo di Savona, 30 via Genova, 17110 Savona, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Paola Contini
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132 Genoa, Italy
| | - Roberto Pontremoli
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132 Genoa, Italy
| | - Francesca Viazzi
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132 Genoa, Italy
| | - Stefano Bertolini
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Aldo Pende
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132 Genoa, Italy
| | - Livia Pisciotta
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa, 10 Largo Benzi, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine, and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy.
| |
Collapse
|
36
|
Zhang H, Yang K, Chen F, Liu Q, Ni J, Cao W, Hua Y, He F, Liu Z, Li L, Fan G. Role of the CCL2-CCR2 axis in cardiovascular disease: Pathogenesis and clinical implications. Front Immunol 2022; 13:975367. [PMID: 36110847 PMCID: PMC9470149 DOI: 10.3389/fimmu.2022.975367] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
The CCL2-CCR2 axis is one of the major chemokine signaling pathways that has received special attention because of its function in the development and progression of cardiovascular disease. Numerous investigations have been performed over the past decades to explore the function of the CCL2-CCR2 signaling axis in cardiovascular disease. Laboratory data on the CCL2-CCR2 axis for cardiovascular disease have shown satisfactory outcomes, yet its clinical translation remains challenging. In this article, we describe the mechanisms of action of the CCL2-CCR2 axis in the development and evolution of cardiovascular diseases including heart failure, atherosclerosis and coronary atherosclerotic heart disease, hypertension and myocardial disease. Laboratory and clinical data on the use of the CCL2-CCR2 pathway as a targeted therapy for cardiovascular diseases are summarized. The potential of the CCL2-CCR2 axis in the treatment of cardiovascular diseases is explored.
Collapse
Affiliation(s)
- Haixia Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Ke Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qianqian Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Weilong Cao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng He
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, China
| | - Zhihao Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Lan Li, ; Guanwei Fan,
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, China
- *Correspondence: Lan Li, ; Guanwei Fan,
| |
Collapse
|
37
|
Lu S, Weiser-Evans MC. Lgals3-Transitioned Inflammatory Smooth Muscle Cells: Major Regulators of Atherosclerosis Progression and Inflammatory Cell Recruitment. Arterioscler Thromb Vasc Biol 2022; 42:957-959. [DOI: 10.1161/atvbaha.122.318009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sizhao Lu
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora. (S.L., M.C.M.W.-E.)
| | - Mary C.M. Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora. (S.L., M.C.M.W.-E.)
- Department of Medicine, School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora. (M.C.M.W.-E.)
- Department of Medicine, Cardiovascular Pulmonary Research Program, University of Colorado Anschutz Medical Campus, Aurora. (M.C.M.W.-E.)
| |
Collapse
|
38
|
Ruan B, Tang X, Guo W, Hu Y, Chen L. Synthesis and Biological Evaluation of Novel Phthalide Analogs-1,2,4-Oxadiazole Hybrids as Potential Anti-Inflammatory Agents. Chem Biodivers 2022; 19:e202200039. [PMID: 35794072 DOI: 10.1002/cbdv.202200039] [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: 01/14/2022] [Accepted: 06/29/2022] [Indexed: 11/10/2022]
Abstract
A series of novel pathalide-1,2,4-oxadiazole analogs were synthesized for discovering novel anti-inflammatory agents. After the assessment of their cytotoxicity in vitro, all compounds had been screened for their anti-inflammatory activity by evaluating their inhibitory effect on LPS-induced NO production in RAW 264.7 macrophages. SARs had been concluded, and finally compound E13 was found to be the most potent compound. This compound could also significantly decrease the production of iNOS and COX-2. Preliminary mechanism studies indicated that compound E13 could inhibit the TLR4/NF-κB and ERK/p38 signaling pathways. These findings indicate that E13 holds great potential to be a lead compound for discovering novel anti-inflammatory drugs.
Collapse
Affiliation(s)
- Banfeng Ruan
- Key Lab of Biofabrication of Anhui Higher Education, Hefei University, Hefei, 230601, P. R. China
| | - Xiaofei Tang
- Key Lab of Biofabrication of Anhui Higher Education, Hefei University, Hefei, 230601, P. R. China
| | - Weiyun Guo
- Key Lab of Biofabrication of Anhui Higher Education, Hefei University, Hefei, 230601, P. R. China
| | - Yong Hu
- Agro-products Processing Research Institute, Anhui Academy of Agricultural Sciences, Hefei, 230001, P. R. China
| | - Liuzeng Chen
- Key Lab of Biofabrication of Anhui Higher Education, Hefei University, Hefei, 230601, P. R. China
| |
Collapse
|
39
|
Casula M, Andreis A, Avondo S, Vaira MP, Imazio M. Colchicine for cardiovascular medicine: a systematic review and meta-analysis. Future Cardiol 2022; 18:647-659. [PMID: 35787150 DOI: 10.2217/fca-2020-0206] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Colchicine, a microtubule-disassembling (antitubulin) agent used for centuries for the treatment of gout and autoimmune diseases, is a drug of growing interest in the cardiovascular field. While in the last decades it has become cornerstone of pericarditis treatment, it has also emerged in the last few years as a promising drug in the management of coronary artery disease, atrial fibrillation and heart failure. This systematic review and meta-analysis aimed to assess the efficacy of colchicine in patients with cardiovascular diseases. Methods: Systematic search in electronic databases (MEDLINE/PubMed, Scopus, BioMed Central, the Cochrane Collaboration Database of Randomized Trials, ClinicalTrials.gov, EMBASE, Google Scholar) was performed to identify randomized controlled trials (RCTs) up to February 2021. Random-effects meta-analysis was performed to assess the risk of cardiovascular events, defined according to clinical setting. Results: Among 15,569 pooled patients from 21 RCTs, colchicine was superior to placebo in the reduction of cardiovascular events. In the setting of pericardial diseases, it was associated with a lower risk of recurrent pericarditis (17 vs 34%, RR = 0.50, 95% CI: 0.42-0.60, I2 = 10%). In other studies assessing coronary artery disease patients, colchicine was associated with a reduced risk of major adverse cardiovascular events (MACE) such as myocardial infarction, stroke, cardiovascular death, coronary revascularisation and hospitalization (6.3 vs 9%, RR = 0.67, 95% CI: 0.54-0.84, I2 = 55). Among patients with atrial fibrillation, it was associated with lower rates of recurrence (20 vs 30%, RR = 0.68, 95% CI: 0.58-0.81, I2 = 0). In the single RCT on heart failure, colchicine was not associated with improved NYHA class. Conclusion: Colchicine is a valuable anti-inflammatory agent for the prevention of cardiovascular events in patients with inflammatory cardiac conditions such as pericardial diseases, coronary artery disease and atrial fibrillation.
Collapse
Affiliation(s)
- Matteo Casula
- University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Alessandro Andreis
- University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Stefano Avondo
- University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Matteo Pio Vaira
- University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Massimo Imazio
- Cardiology, Cardiothoracic Department, University Hospital "Santa Maria della Misericordia", ASUFC, Udine, Italy
| |
Collapse
|
40
|
Jiao Y, Wang S, Jiang L, Sun X, Li J, Liu X, Yao X, Zhang C, Wang N, Deng H, Yang G. 2-undecanone protects against fine particles-induced heart inflammation via modulating Nrf2/HO-1 and NF-κB pathways. ENVIRONMENTAL TOXICOLOGY 2022; 37:1642-1652. [PMID: 35285579 DOI: 10.1002/tox.23513] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/26/2022] [Accepted: 03/06/2022] [Indexed: 05/20/2023]
Abstract
Exposure to air pollution has been closely associated with some cardiovascular disease. One of the mechanisms of PM2.5 -mediated heart injury may be to promote inflammation. We aim to investigate whether the main extract of Houttuynia cordata, 2-undecanone, can prevent the inflammation caused by PM2.5 , and to reveal the underlying mechanisms. The results showed that PM2.5 increased the expression of certain inflammatory cytokines, and caused oxidative damage in BALB/c mice and H9C2 cells. Supplementation with 2-undecanone attenuated this PM2.5 -induced inflammatory injury and oxidative damage. Further, we elucidated that the protective effect of 2-undecanone may be associated with NF-κB and Nrf2/HO-1 pathways. The NF-κB pathway was distinctly activated after treated by PM2.5 , which can be blocked by 2-undecanone, accompanied by increasing Nrf2 and HO-1 levels. To figure out the relationship between NF-κB and Nrf2/HO-1 pathways, we knocked down Nrf2 gene. NF-κB pathway proteins and downstream inflammatory cytokines were significantly increased after treatment with PM2.5 , while 2-undecanone could decrease expression of these proteins. In conclusion, it is possible that 2-undecanone can induce the expression of the antioxidant enzyme HO-1 by activating Nrf2, thereby reducing NF-κB pathway and inflammatory damage of mouse myocardium caused by PM2.5 exposure.
Collapse
Affiliation(s)
- Yuhang Jiao
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Shaopeng Wang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liping Jiang
- Liaoning Anti-degenerative Diseases Natural Products Engineering Technology Research Center, Dalian Medical University, Dalian, China
| | - Xiance Sun
- Liaoning Anti-degenerative Diseases Natural Products Engineering Technology Research Center, Dalian Medical University, Dalian, China
| | - Jing Li
- Department of Pathology, Dalian Medical University, Dalian, China
| | - Xiaofang Liu
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Xiaofeng Yao
- Liaoning Anti-degenerative Diseases Natural Products Engineering Technology Research Center, Dalian Medical University, Dalian, China
| | - Cong Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Ningning Wang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Haoyuan Deng
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Guang Yang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| |
Collapse
|
41
|
Inflammation, Lymphatics, and Cardiovascular Disease: Amplification by Chronic Kidney Disease. Curr Hypertens Rep 2022; 24:455-463. [PMID: 35727522 DOI: 10.1007/s11906-022-01206-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Kidney disease is a strong modulator of the composition and metabolism of the intestinal microbiome that produces toxins and inflammatory factors. The primary pathways for these harmful factors are blood vessels and nerves. Although lymphatic vessels are responsible for clearance of interstitial fluids, macromolecules, and cells, little is known about whether and how kidney injury impacts the intestinal lymphatic network. RECENT FINDINGS Kidney injury stimulates intestinal lymphangiogenesis, activates lymphatic endothelial cells, and increases mesenteric lymph flow. The mesenteric lymph of kidney-injured animals contains increased levels of cytokines, immune cells, isolevuglandin (IsoLG), a highly reactive dicarbonyl, and of apolipoprotein AI (apoAI). IsoLG is increased in the ileum of kidney injured animals, and intestinal epithelial cells exposed to myeloperoxidase produce more IsoLG. IsoLG-modified apoAI directly increases lymphatic vessel contractions and activates lymphatic endothelial cells. Inhibition of IsoLG by carbonyl scavenger treatment reduces intestinal lymphangiogenesis in kidney-injured animals. Research from our group and others suggests a novel mediator (IsoLG-modified apoAI) and a new pathway (intestinal lymphatic network) in the cross talk between kidneys and intestines and heart. Kidney injury activates intestinal lymphangiogenesis and increases lymphatic flow via mechanisms involving intestinally generated IsoLG. The data identify a new pathway in the kidney gut-heart axis and present a new target for kidney disease-induced intestinal disruptions that may lessen the major adverse consequence of kidney impairment, namely cardiovascular disease.
Collapse
|
42
|
Buelna-Chontal M, Bansal SS, Barrera-Chimal J, Liberale L. Editorial: Targeting Dysregulated Inflammation to Treat Cardiovascular Diseases. Front Cell Dev Biol 2022; 10:926086. [PMID: 35652092 PMCID: PMC9150769 DOI: 10.3389/fcell.2022.926086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Affiliation(s)
- Mabel Buelna-Chontal
- Department of Cardiovascular Biomedicine, National Institute of Cardiology, I.Ch., Mexico City, Mexico
| | - Shyam S Bansal
- Department of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Jonatan Barrera-Chimal
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, QC, Canada
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
43
|
Jarr KU, Kojima Y, Weissman IL, Leeper NJ. 2021 Jeffrey M. Hoeg Award Lecture: Defining the Role of Efferocytosis in Cardiovascular Disease: A Focus on the CD47 (Cluster of Differentiation 47) Axis. Arterioscler Thromb Vasc Biol 2022; 42:e145-e154. [PMID: 35387480 PMCID: PMC9183217 DOI: 10.1161/atvbaha.122.317049] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/21/2022] [Indexed: 01/09/2023]
Abstract
A key feature of atherogenesis is the accumulation of diseased and dying cells within the lesional necrotic core. While the burden of intraplaque apoptotic cells may be driven in part by an increase in programmed cell death, mounting evidence suggests that their presence may primarily be dictated by a defect in programmed cell removal, or efferocytosis. In this brief review, we will summarize the evidence suggesting that inflammation-dependent changes within the plaque render target cells inedible and reduce the appetite of lesional phagocytes. We will present the genetic causation studies, which indicate these phenomena promote lesion expansion and plaque vulnerability, and the interventional data which suggest that these processes can be reversed. Particular emphasis is provided related to the antiphagocytic CD47 (cluster of differentiation 47) do not eat me axis, which has emerged as a novel antiatherosclerotic translational target that is predicted to provide benefit independent of traditional cardiovascular risk factors.
Collapse
Affiliation(s)
- Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yoko Kojima
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Irving L. Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
44
|
Mechanistic insight into lysyl oxidase in vascular remodeling and angiogenesis. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
45
|
Inflammation, Aging, and Cardiovascular Disease: JACC Review Topic of the Week. J Am Coll Cardiol 2022; 79:837-847. [PMID: 35210039 PMCID: PMC8881676 DOI: 10.1016/j.jacc.2021.12.017] [Citation(s) in RCA: 149] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/08/2021] [Indexed: 01/19/2023]
Abstract
Aging and inflammation both contribute pivotally to cardiovascular (CV) and cerebrovascular disease, the leading causes of death and disability worldwide. The concept of inflamm-aging recognizes that low-grade inflammatory pathways observed in the elderly contribute to CV risk. Understanding the mechanisms that link inflammation and aging could reveal new therapeutic targets and offer options to cope with the growing aging population worldwide. This review reports recent scientific advances in the pathways through which inflamm-aging mediates age-dependent decline in CV function and disease onset and considers critically the translational potential of such concepts into everyday clinical practice.
Collapse
|
46
|
Wang Y, Gao L. Inflammation and Cardiovascular Disease Associated With Hemodialysis for End-Stage Renal Disease. Front Pharmacol 2022; 13:800950. [PMID: 35222026 PMCID: PMC8867697 DOI: 10.3389/fphar.2022.800950] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/10/2022] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD) and cardiac insufficiency often co-exist, particularly in uremic patients on hemodialysis (HD). The occurrence of abnormal renal function in patients with cardiac insufficiency is often indicative of a poor prognosis. It has long been established that in patients with cardiac insufficiency, poorer renal function tends to indicate poorer cardiac mechanics, including left atrial reserve strain, left ventricular longitudinal strain, and right ventricular free wall strain (Unger et al., Eur J Heart Fail, 2016, 18(1), 103-12). Similarly, patients with chronic kidney disease, particularly uremic patients on HD, often have cardiovascular complications in addition to abnormal endothelial function with volume overload, persistent inflammatory states, calcium overload, and imbalances in redox responses. Cardiac insufficiency due to uremia is therefore mainly due to multifaceted non-specific pathological changes rather than pure renal insufficiency. Several studies have shown that the risk of adverse cardiovascular events is greatly increased and persistent in all patients treated with HD, especially in those who have just started HD treatment. Inflammation, as an important intersection between CKD and cardiovascular disease, is involved in the development of cardiovascular complications in patients with CKD and is indicative of prognosis (Chan et al., Eur Heart J, 2021, 42(13), 1244-1253). Therefore, only by understanding the mechanisms underlying the sequential development of inflammation in CKD patients and breaking the vicious circle between inflammation-mediated renal and cardiac insufficiency is it possible to improve the prognosis of patients with end-stage renal disease (ESRD). This review highlights the mechanisms of inflammation and the oxidative stress that co-exists with inflammation in uremic patients on dialysis, as well as the mechanisms of cardiovascular complications in the inflammatory state, and provides clinical recommendations for the anti-inflammatory treatment of cardiovascular complications in such patients.
Collapse
Affiliation(s)
| | - Lu Gao
- Department of Cardiovascular Centre, The First Hospital of Jilin University, Jilin, China
| |
Collapse
|
47
|
Carbone F, Montecucco F, Liberale L. The Role of PCSK9 in Atherogenesis and Other
Inflammatory Diseases. Curr Med Chem 2022; 29:958-959. [PMID: 35227181 DOI: 10.2174/092986732906220218164509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine,
Department of Internal Medicine,
University of Genoa, 6 viale Benedetto,
XV, 16143 Genoa,
Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network 10 Largo Benzi 16132 Genoa,
Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine,
Department of Internal Medicine,
University of Genoa, 6 viale Benedetto,
XV, 16143 Genoa,
Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network 10 Largo Benzi ,16132 Genoa,
Italy
| | - Luca Liberale
- First Clinic of Internal Medicine,
Department of Internal Medicine,
University of Genoa, 6 viale Benedetto,
XV, 16143 Genoa,
Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa,
Italy
| |
Collapse
|
48
|
Andelova K, Bacova BS, Sykora M, Hlivak P, Barancik M, Tribulova N. Mechanisms Underlying Antiarrhythmic Properties of Cardioprotective Agents Impacting Inflammation and Oxidative Stress. Int J Mol Sci 2022; 23:1416. [PMID: 35163340 PMCID: PMC8835881 DOI: 10.3390/ijms23031416] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
The prevention of cardiac life-threatening ventricular fibrillation and stroke-provoking atrial fibrillation remains a serious global clinical issue, with ongoing need for novel approaches. Numerous experimental and clinical studies suggest that oxidative stress and inflammation are deleterious to cardiovascular health, and can increase heart susceptibility to arrhythmias. It is quite interesting, however, that various cardio-protective compounds with antiarrhythmic properties are potent anti-oxidative and anti-inflammatory agents. These most likely target the pro-arrhythmia primary mechanisms. This review and literature-based analysis presents a realistic view of antiarrhythmic efficacy and the molecular mechanisms of current pharmaceuticals in clinical use. These include the sodium-glucose cotransporter-2 inhibitors used in diabetes treatment, statins in dyslipidemia and naturally protective omega-3 fatty acids. This approach supports the hypothesis that prevention or attenuation of oxidative and inflammatory stress can abolish pro-arrhythmic factors and the development of an arrhythmia substrate. This could prove a powerful tool of reducing cardiac arrhythmia burden.
Collapse
Affiliation(s)
- Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Peter Hlivak
- Department of Arrhythmias and Pacing, National Institute of Cardiovascular Diseases, Pod Krásnou Hôrkou 1, 83348 Bratislava, Slovakia;
| | - Miroslav Barancik
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Narcis Tribulova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| |
Collapse
|
49
|
Wang C, Hu J. Influence of the Interaction Between Depressive Symptoms and Sleep Disorders on Cardiovascular Diseases Occurrence. Int J Gen Med 2022; 14:10327-10335. [PMID: 34992447 PMCID: PMC8713879 DOI: 10.2147/ijgm.s334894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/15/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Depressive symptoms and sleep disorders were independent risk factors for cardiovascular diseases (CVD). However, few studies have examined the combined effects of depressive symptoms and sleep disorders on CVD. We aimed to evaluate the association between depressive symptoms, sleep disorders and CVD occurrence. Methods Data on 30,398 participants were extracted from the National Health and Nutritional Examination Survey (NHANES) database (2005–2018). Univariate and multivariate analyses were used for assessing the association of depressive symptoms, sleep disorders, and CVD occurrence. Three indexes, including the relative excess risk of interaction (RERI), attributable proportion of interaction (API), and synergy index (SI), were used to analyze the interaction. Results Of 30,398 participants, 11,544 (37.98%) participants had CVD and 18,854 (62.02%) did not. Except for gender, the differences were significant between CVD and non-CVD participants in all variables (all P<0.001). Depressive symptoms [odds ratio (OR)=1.73; 95% confidence intervals (CI):1.57–1.91] and sleep disorders (OR=1.76; 95% CI:1.65–1.88) were associated with an increased risk of CVD after adjusting all confounders. Patients with both depressive symptoms and sleep disorders (OR=2.64; 95% CI:2.32–3.00) had a higher risk of CVD than those without. There may be a synergistic interaction between depression and sleep disorders on the CVD occurrence (SI=1.763; 95% CI:1.299–2.394), and the proportion of CVD caused by this interaction was 26.9% (API=0.269; 95% CI:0.148–0.389). In addition, only moderate depressive symptoms may interact with sleep disorders in the occurrence of CVD. Conclusion There may be a synergistic interaction between depressive symptoms and sleep disorders, and the synergistic interaction may increase the occurrence of CVD.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Vascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China
| | - Juan Hu
- Department of Vascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China
| |
Collapse
|
50
|
Alme KN, Ulvik A, Askim T, Assmus J, Mollnes TE, Naik M, Næss H, Saltvedt I, Ueland PM, Knapskog AB. Neopterin and kynurenic acid as predictors of stroke recurrence and mortality: a multicentre prospective cohort study on biomarkers of inflammation measured three months after ischemic stroke. BMC Neurol 2021; 21:476. [PMID: 34879833 PMCID: PMC8653541 DOI: 10.1186/s12883-021-02498-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic low-grade inflammation is associated with both ischemic stroke and sedentary behaviour. The aim of this study was to investigate the predictive abilities of biomarkers of inflammation and immune modulation associated with sedentary behaviour for ischemic stroke recurrence and mortality in a stroke population. METHODS Patients admitted to hospital for acute stroke were recruited to the prospective multicentre cohort study, the Norwegian Cognitive Impairment After Stroke (Nor-COAST) study, from May 2015 until March 2017. Patients with ischemic stroke, blood samples available from the three-month follow-up, and no stroke recurrence before the three-month follow-up were included. Serum was analysed for C-reactive protein (CRP) with high-sensitive technique, and plasma for interleukin-6 (IL-6), neopterin, pyridoxic acid ratio index (PAr-index: 4-pyridoxic acid: [pyrioxal+pyridoxal-5'-phosphate]) and kynurenic acid (KA). Ischemic stroke recurrence and death were identified by the Norwegian Stroke Registry and the Cause of Death Registry until 31 December 2018. RESULTS The study included 354 patients, 57% male, mean age 73 (SD 11) years, mean observation time 2.5 (SD 0.6) years, and median National Institute of Health Stroke Scale of 0 (IQR 1) at three months. CRP was associated with mortality (HR 1.40, CI 1.01, 1.96, p = 0.046), and neopterin was associated with the combined endpoint (recurrent ischemic stroke or death) (HR 1.52, CI 1.06, 2.20, p = 0.023), adjusted for age, sex, prior cerebrovascular disease, modified Rankin Scale, and creatinine. When adding neopterin and KA to the same model, KA was negatively associated (HR 0.57, CI 0.33, 0.97, p = 0.038), and neopterin was positively associated (HR 1.61, CI 1.02, 2.54, p = 0.040) with mortality. Patients with cardioembolic stroke at baseline had higher levels of inflammation at three months. CONCLUSION Neopterin might be a valuable prognostic biomarker in stroke patients. The use of KA as a measure of anti-inflammatory capacity should be investigated further. TRIAL REGISTRATION The study was registered at Clinicaltrials.gov ( NCT02650531 ). First posted on 08/01/2016.
Collapse
Affiliation(s)
- Katinka Nordheim Alme
- Institute of Clinical Medicine (K1), University of Bergen, Bergen, Norway. .,Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway.
| | | | - Torunn Askim
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Jörg Assmus
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway.,Research Laboratory, Nordland Hospital, Bodø, and K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Centre of Molecular Inflammation Research, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Mala Naik
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway.,Department of Clinical Science (K2), University of Bergen, Bergen, Norway
| | - Halvor Næss
- Institute of Clinical Medicine (K1), University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway.,Centre for age-related medicine, Stavanger University Hospital, Stavanger, Norway
| | - Ingvild Saltvedt
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Department of Geriatrics, Clinic of internal medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | | | - Anne-Brita Knapskog
- Department of Geriatric Medicine, Oslo University Hospital. Ullevaal, Oslo, Norway
| |
Collapse
|