201
|
Adamstein NH, Cornel JH, Davidson M, Libby P, de Remigis A, Jensen C, Ekström K, Ridker PM. Association of Interleukin 6 Inhibition With Ziltivekimab and the Neutrophil-Lymphocyte Ratio: A Secondary Analysis of the RESCUE Clinical Trial. JAMA Cardiol 2023; 8:177-181. [PMID: 36449307 PMCID: PMC9713672 DOI: 10.1001/jamacardio.2022.4277] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Importance The neutrophil-lymphocyte ratio (NLR) independently predicts atherosclerotic events and is a potential biomarker for residual inflammatory risk. Interleukin (IL) 1β inhibition reduces the NLR, but whether inhibition of IL-6, a cytokine downstream of IL-1, also lowers the NLR is uncertain. Objective To evaluate whether ziltivekimab, a therapeutic monoclonal antibody targeting the IL-6 ligand, associates with a lower NLR compared with placebo. Design, Setting, and Participants This was an exploratory post hoc analysis of Trial to Evaluate Reduction in Inflammation in Patients With Advanced Chronic Renal Disease Utilizing Antibody Mediated IL-6 Inhibition (RESCUE), a double-blind, randomized, placebo-controlled, phase 2 trial conducted from June 17, 2019, to January 14, 2020, with 24 weeks of follow-up. Participants were enrolled at 40 sites in the US and included adults aged 18 or older with moderate to severe chronic kidney disease and high-sensitivity C-reactive protein levels of 2 mg/L or greater. Data were analyzed from September 28, 2021, to October 2, 2022. Interventions Participants were randomly assigned equally to placebo or ziltivekimab, 7.5 mg, 15 mg, or 30 mg, subcutaneously every 4 weeks. Main Outcomes and Measures The primary outcome was the change in the NLR at 12 weeks. Results A total of 264 participants (median [IQR] age, 68 [60-75] years; 135 men [51%]; 129 women [49%]) were enrolled, of which 187 (71%) had diabetes, and 126 (48%) had known atherosclerosis. The median (IQR) change in the NLR at 12 weeks was 1.56% (IQR, -15.7% to 20.0%), -13.5% (IQR, -31.6% to 3.20%), -14.3% (IQR, -26.9% to 4.62%), and -22.4% (IQR, -33.3% to -4.27%) in the placebo, 7.5-mg, 15-mg, and 30-mg groups, respectively. The estimated treatment difference compared with placebo was -14.6% (95% CI, -24.8% to -4.81%; P = .004), -15.3% (95% CI, -25.2% to -5.10%; P = .004), and -23.6% (95% CI, -33.2% to -14.2%; P < .001) in the 7.5-mg, 15-mg, and 30-mg groups, respectively. A similar reduction in the absolute neutrophil count was observed. Conclusions and Relevance Results of this post hoc analysis of the RESCUE trial show that IL-6 ligand inhibition with ziltivekimab associates with a lower NLR, suggesting that it may disrupt multiple atherogenic inflammatory pathways, including those mediated by the myeloid cell compartment. The NLR may have use in monitoring ziltivekimab's efficacy should it be introduced into clinical practice.
Collapse
Affiliation(s)
- Nicholas H Adamstein
- Center for Cardiovascular Disease Prevention, Divisions of Preventive Medicine and Cardiovascular Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jan Hein Cornel
- Radboud University Medical Center, Nijmegen, the Netherlands
| | - Michael Davidson
- Section of Cardiology, Department of Medicine, University of Chicago Medicine, Pritzker School of Medicine, Chicago, Illinois
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Divisions of Preventive Medicine and Cardiovascular Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
202
|
Associations Between Dysfunctional Thoughts, Leisure Activities, and IL-6 in Caregivers of Family Members With Dementia. Psychosom Med 2023; 85:175-181. [PMID: 36516289 DOI: 10.1097/psy.0000000000001158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Dementia caregiving is associated with negative physical health consequences, including inflammation processes. The objective of this study was to analyze the associations between dysfunctional thoughts, frequency of leisure activities, and interleukin 6 (IL-6) in a sample of dementia family caregivers. METHODS One hundred forty dementia caregivers participated in this cross-sectional study. The relationships among caregivers' dysfunctional thoughts, leisure activities, and IL-6 were adjusted for demographic characteristics, stressors, and physical and mental health indicators in a linear regression analysis. RESULTS Higher levels of dysfunctional thoughts ( t = -2.02, p = .045) were significantly associated with lower frequency of leisure activities. In turn, lower frequency of leisure activities was significantly associated with higher levels of IL-6 ( t = -2.03, p = .045). Dysfunctional thoughts were no longer significantly associated with IL-6 levels when both dysfunctional thoughts and leisure activities were included in the same model ( t = 1.78, p = .076). A significant indirect effect was found for the association between higher levels of dysfunctional thoughts and higher levels of IL-6 (standardized indirect effect = 0.036, bootstrap standard error = 0.026, 95% confidence interval = 0.0001-0.1000) through its association with fewer leisure activities. CONCLUSIONS Our findings suggest that the direct effect of caregivers' dysfunctional thoughts on IL-6 may be mediated by the impact on caregivers' frequency of leisure activities. Results suggest that training caregivers in reducing dysfunctional thoughts to thereby increase leisure activities may be useful in reducing inflammation.
Collapse
|
203
|
Wheless L, Pike MM, Chen HC, Yu Z, Tao R, Bick A, Chung CP, Robinson-Cohen C, Hung A. Genetic Determinants of IL-6 Levels and Risk of ESKD. KIDNEY360 2023; 4:241-244. [PMID: 36821615 PMCID: PMC10103241 DOI: 10.34067/kid.0003332022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
Genetically predicted IL-6 levels are associated with risk of ESKD. Therapeutic modulation of IL-6 could potentially reduce the risk of ESKD.
Collapse
Affiliation(s)
- Lee Wheless
- Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| | - Mindy M. Pike
- Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| | - Hua-Chang Chen
- Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| | - Zhihong Yu
- Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| | - Ran Tao
- Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| | - Alexander Bick
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| | - Cecilia P. Chung
- Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| | - Cassianne Robinson-Cohen
- Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| | - Adriana Hung
- Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Health System VA Medical Center, Nashville, Tennessee
| |
Collapse
|
204
|
Weber B, Liao KP. Evidence for Biologic Drug Modifying Anti-Rheumatoid Drugs and Association with Cardiovascular Disease Risk Mitigation in Inflammatory Arthritis. Rheum Dis Clin North Am 2023; 49:165-178. [PMID: 36424023 PMCID: PMC10250044 DOI: 10.1016/j.rdc.2022.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Systemic auto-immune inflammatory arthritides are associated with increased cardiovascular (CV) risk compared to those without these conditions, and is a leading cause of morbidity and mortality. Newer biologic drug modifying antirheumatoid drugs (bDMARD) and small molecules have transformed treatment paradigms enabling tighter control of disease activity and in some cases, remission. There is evidence to suggest that the majority of bDMARDs may also reduce cardiovascular risk, although prospective interventional data remain sparse. Additionally, recent results raise concern for treatments targeting specific pathways that may negatively affect cardiovascular risk. This review will cover key biologic pathways targeted in rheumatoid arthritis, psoriatic arthritis, and spondyloarthropathies.
Collapse
Affiliation(s)
- Brittany Weber
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; Cardiovascular Imaging Program, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Katherine P Liao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
205
|
Could uric acid to high density lipoprotein-cholesterol ratio be considered as a marker of hemodialysis sufficiency? JOURNAL OF CONTEMPORARY MEDICINE 2023. [DOI: 10.16899/jcm.1223641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Aim: Hemodialysis (HD) is one of the most widely utilized renal replacement therapies in individuals with end-stage chronic kidney disease (CKD). The purpose of this study was to compare the Uric acid to HDL cholesterol ratio (UHR) levels of well-treated HD patients to those of those who had inadequate HD therapy.
Materials and Methods: Data on HD patients were acquired from patient files and the institution's database. A URR value of more than 70% designated the sufficient HD group, whereas less than 70% defined the insufficient HD group. Furthermore, laboratory data, such as the UHR of the study groups, were compared.
Results: The UHRs for adequate and inadequate HD were 0.160±04 and 0.20±0.07, respectively. The UHR of patients with insufficient HD was substantially greater than that of the subjects with sufficient HD (p=0.004). Besides, UHR was substantially and positively connected with urea before HD (r=0.37, p=0.001), urea after HD (r=0.39, p=0.001), serum creatinine before HD (r=0.48, p0.001), serum creatinine after HD (r=0.45, p0.001), and negatively correlated with URR (r=-0.29, p=0.008), according to correlation analyses. In individuals with chronic renal disease, a UHR value higher than 0.16 exhibited 67% sensitivity and 57% specificity in detecting inadequate HD.
Conclusion: We propose that UHR, in addition to URR, might be used to determine HS sufficiency in CKD patients undergoing HD therapy.
Collapse
|
206
|
Lopez-Sendon J, Fernández-Velasco M. New play for an old actress: A Cinderella in cardiovascular disease. Int J Cardiol 2023; 371:76-77. [PMID: 36115437 DOI: 10.1016/j.ijcard.2022.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/08/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Jose Lopez-Sendon
- IdiPAZ Research Institute, La Paz University Hospital, Universidad Autonoma de Madrid, Madrid, Spain.
| | | |
Collapse
|
207
|
Meng Q, Liu H, Liu J, Pang Y, Liu Q. Advances in immunotherapy modalities for atherosclerosis. Front Pharmacol 2023; 13:1079185. [PMID: 36703734 PMCID: PMC9871313 DOI: 10.3389/fphar.2022.1079185] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. Atherosclerosis is the pathological basis of atherosclerotic cardiovascular disease (ASCVD). Atherosclerosis is now understood to be a long-term immune-mediated inflammatory condition brought on by a complicated chain of factors, including endothelial dysfunction, lipid deposits in the artery wall, and monocyte-derived macrophage infiltration, in which both innate immunity and adaptive immunity play an indispensable role. Recent studies have shown that atherosclerosis can be alleviated by inducing a protective immune response through certain auto-antigens or exogenous antigens. Some clinical trials have also demonstrated that atherosclerotic is associated with the presence of immune cells and immune factors in the body. Therefore, immunotherapy is expected to be a new preventive and curative measure for atherosclerosis. In this review, we provide a summary overview of recent progress in the research of immune mechanisms of atherosclerosis and targeted therapeutic pathways.
Collapse
Affiliation(s)
- Qingwen Meng
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou, China,Deparment of Cardiovascular, The First Affiliated Hospital of Hainan Medical University, Haikou, China,Hainan Provincial Key Laboratory of Tropical Brain Research and Transformation, Hainan Medical University, Haikou, China
| | - Huajiang Liu
- Deparment of Cardiovascular, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jinteng Liu
- School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Yangyang Pang
- School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Qibing Liu
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou, China,School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China,*Correspondence: Qibing Liu,
| |
Collapse
|
208
|
Inflammageing and Cardiovascular System: Focus on Cardiokines and Cardiac-Specific Biomarkers. Int J Mol Sci 2023; 24:ijms24010844. [PMID: 36614282 PMCID: PMC9820990 DOI: 10.3390/ijms24010844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
The term "inflammageing" was introduced in 2000, with the aim of describing the chronic inflammatory state typical of elderly individuals, which is characterized by a combination of elevated levels of inflammatory biomarkers, a high burden of comorbidities, an elevated risk of disability, frailty, and premature death. Inflammageing is a hallmark of various cardiovascular diseases, including atherosclerosis, hypertension, and rapid progression to heart failure. The great experimental and clinical evidence accumulated in recent years has clearly demonstrated that early detection and counteraction of inflammageing is a promising strategy not only to prevent cardiovascular disease, but also to slow down the progressive decline of health that occurs with ageing. It is conceivable that beneficial effects of counteracting inflammageing should be most effective if implemented in the early stages, when the compensatory capacity of the organism is not completely exhausted. Early interventions and treatments require early diagnosis using reliable and cost-effective biomarkers. Indeed, recent clinical studies have demonstrated that cardiac-specific biomarkers (i.e., cardiac natriuretic peptides and cardiac troponins) are able to identify, even in the general population, the individuals at highest risk of progression to heart failure. However, further clinical studies are needed to better understand the usefulness and cost/benefit ratio of cardiac-specific biomarkers as potential targets in preventive and therapeutic strategies for early detection and counteraction of inflammageing mechanisms and in this way slowing the progressive decline of health that occurs with ageing.
Collapse
|
209
|
Fathieh S, Grieve SM, Negishi K, Figtree GA. Potential Biological Mediators of Myocardial and Vascular Complications of Air Pollution-A State-of-the-Art Review. Heart Lung Circ 2023; 32:26-42. [PMID: 36585310 DOI: 10.1016/j.hlc.2022.11.014] [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: 03/09/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 12/29/2022]
Abstract
Ambient air pollution is recognised globally as a significant contributor to the burden of cardiovascular diseases. The evidence from both human and animal studies supporting the cardiovascular impact of exposure to air pollution has grown substantially, implicating numerous pathophysiological pathways and related signalling mediators. In this review, we summarise the list of activated mediators for each pathway that lead to myocardial and vascular injury in response to air pollutants. We performed a systematic search of multiple databases, including articles between 1990 and Jan 2022, summarising the evidence for activated pathways in response to each significant air pollutant. Particulate matter <2.5 μm (PM2.5) was the most studied pollutant, followed by particulate matter between 2.5 μm-10 μm (PM10), nitrogen dioxide (NO2) and ozone (O3). Key pathogenic pathways that emerged included activation of systemic and local inflammation, oxidative stress, endothelial dysfunction, and autonomic dysfunction. We looked at how potential mediators of each of these pathways were linked to both cardiovascular disease and air pollution and included the overlapping mediators. This review illustrates the complex relationship between air pollution and cardiovascular diseases, and discusses challenges in moving beyond associations, towards understanding causal contributions of specific pathways and markers that may inform us regarding an individual's exposure, response, and likely risk.
Collapse
Affiliation(s)
- Sina Fathieh
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Stuart M Grieve
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Radiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Kazuaki Negishi
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia; Department of Cardiology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan; Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia.
| |
Collapse
|
210
|
Wang D, Tan Z, Yang J, Li L, Li H, Zhang H, Liu H, Liu Y, Wang L, Li Q, Guo H. Perfluorooctane sulfonate promotes atherosclerosis by modulating M1 polarization of macrophages through the NF-κB pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114384. [PMID: 36512850 DOI: 10.1016/j.ecoenv.2022.114384] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/27/2022] [Accepted: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a widely used and distributed perfluorinated compounds and is reported to be harmful to cardiovascular health; however, the direct association between PFOS exposure and atherosclerosis and the underlying mechanisms remain unknown. Therefore, this study aimed to investigate the effects of PFOS exposure on the atherosclerosis progression and the underlying mechanisms. PFOS was administered through oral gavage to apolipoprotein E-deficient (ApoE-/-) mice for 12 weeks. PFOS exposure significantly increased pulse wave velocity (PWV) and intima-media thickness (IMT), increased aortic plaque burden and vulnerability, and elevated serum lipid and inflammatory cytokine levels. PFOS promoted aortic and RAW264.7 M1 macrophage polarization, which increased the secretion of nitric oxide synthase (iNOS) and pro-inflammatory factors (tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], and interleukin-1β [IL-1β]), and suppressed M2 macrophage polarization, which decreased the expression of CD206, arginine I (Arg-1), and interleukin-10 (IL-10). Moreover, PFOS activated nuclear factor-kappa B (NF-κB) in the aorta and macrophages. BAY11-7082 was used to inhibit NF-κB-alleviated M1 macrophage polarization and the inflammatory response induced by PFOS in RAW264.7 macrophages. Our results are the first to reveal the acceleratory effect of PFOS on the atherosclerosis progression in ApoE-/- mice, which is associated with the NF-κB activation of macrophages to M1 polarization to induce inflammation.
Collapse
Affiliation(s)
- Dan Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Zhenzhen Tan
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Jing Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Longfei Li
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Haoran Li
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, China
| | - Heqiong Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Yi Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Lei Wang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang, China
| | - Qian Li
- Department of Physiology, Hebei Medical University, Shijiazhuang, China.
| | - Huicai Guo
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China.
| |
Collapse
|
211
|
Innate Immunity in Cardiovascular Diseases-Identification of Novel Molecular Players and Targets. J Clin Med 2023; 12:jcm12010335. [PMID: 36615135 PMCID: PMC9821340 DOI: 10.3390/jcm12010335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
During the past few years, unexpected developments have driven studies in the field of clinical immunology. One driver of immense impact was the outbreak of a pandemic caused by the novel virus SARS-CoV-2. Excellent recent reviews address diverse aspects of immunological re-search into cardiovascular diseases. Here, we specifically focus on selected studies taking advantage of advanced state-of-the-art molecular genetic methods ranging from genome-wide epi/transcriptome mapping and variant scanning to optogenetics and chemogenetics. First, we discuss the emerging clinical relevance of advanced diagnostics for cardiovascular diseases, including those associated with COVID-19-with a focus on the role of inflammation in cardiomyopathies and arrhythmias. Second, we consider newly identified immunological interactions at organ and system levels which affect cardiovascular pathogenesis. Thus, studies into immune influences arising from the intestinal system are moving towards therapeutic exploitation. Further, powerful new research tools have enabled novel insight into brain-immune system interactions at unprecedented resolution. This latter line of investigation emphasizes the strength of influence of emotional stress-acting through defined brain regions-upon viral and cardiovascular disorders. Several challenges need to be overcome before the full impact of these far-reaching new findings will hit the clinical arena.
Collapse
|
212
|
Jiang Z, Qu H, Chen K, Gao Z. Beneficial effects of folic acid on inflammatory markers in the patients with metabolic syndrome: Meta-analysis and meta-regression of data from 511 participants in 10 randomized controlled trials. Crit Rev Food Sci Nutr 2022; 64:5450-5461. [PMID: 36576260 DOI: 10.1080/10408398.2022.2154743] [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] [Indexed: 12/29/2022]
Abstract
Previous clinical studies on the anti-inflammatory effects of folic acid (FA) in patients with metabolic syndrome (MetS) have shown controversial results. This study aimed to synthesize the evidence on the effect of FA on inflammatory marker levels in MetS patients. We screened PubMed, Embase, Medline, and the Cochrane Library (from inception to March 2022) to identify relevant randomized controlled trials (RCTs). DerSimonian and Laird random effects were used to estimate the pooled weighted mean difference (WMD) with 95% confidence interval (CI). Funnel plot, Egger's test, and the Begg-Mazumdar correlation test was used to assess publication bias. Subgroup analysis, meta-regression and sensitivity analysis were performed to find out possible sources of between-study heterogeneity. Ten RCTs with a total of 511 participants were included. The analysis showed that FA reduced high sensitivity C-reactive protein (hs-CRP) (WMD, -0.94; 95% CI, -1.56 to -0.32; P = 0.00), interleukin-6 (IL-6) (WMD, -0.39; 95% CI, -0.51 to -0.28; P = 0.00), and tumor necrosis factor-alpha (TNF-α) (WMD, -1.28; 95% CI, -1.88 to -0.68; P = 0.00), but did not decrease the C-reactive protein (CRP) (WMD, 0.10; 95% CI, -0.13 to 0.33; P = 0.38). Sensitivity analysis, subgroup analysis, and meta-regression showed that the effect sizes remained stable. Our findings suggest that FA supplementation could reduce inflammatory markers, such as hs-CRP, IL-6, TNF-α in patients with MetS. This study is registered with PROSPERO (CRD42021223843).
Collapse
Affiliation(s)
- Zhonghui Jiang
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Hua Qu
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Keji Chen
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zhuye Gao
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| |
Collapse
|
213
|
Liu W, Ma R, Lu S, Wen Y, Li H, Wang J, Sun B. Acid-Resistant Mesoporous Metal-Organic Frameworks as Carriers for Targeted Hypoglycemic Peptide Delivery: Peptide Encapsulation, Release, and Bioactivity. ACS APPLIED MATERIALS & INTERFACES 2022; 14:55447-55457. [PMID: 36478454 DOI: 10.1021/acsami.2c18452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Oral administration of bioactive peptides with α-glucosidase inhibitory activities is a promising strategy for diabetes mellitus. The wheat germ peptide Leu-Asp-Leu-Gln-Arg (LDLQR) has been previously proven to inhibit the activity of α-glucosidase efficiently. However, it is still difficult to transport the peptide to the intestine completely due to the harsh condition of the stomach. Herein, an acid-resistant zirconium-based metal-organic framework, NU-1000, was used to immobilize LDLQR with a high encapsulation capacity (92.72%) and encapsulation efficiency (44.08%) in only 10 min. The in vitro release results showed that the acid-stable NU-1000 not only effectively protected LDLQR from degradation in the presence of stomach acid and pepsin effectively but also ensured the release of encapsulated LDLQR under simulated intestinal conditions. Furthermore, LDLQR@NU-1000 could slow down the elevated blood sugar caused by maltose in mice and the area under blood sugar curve decreased by almost 20% when compared with the control group. The inflammatory factor (IL-1β, IL-6) in vivo and cell growth in vitro were almost the same between NU-1000 treatment and normal control groups. This study indicates NU-1000 is a promising vehicle for targeted peptide-based bioactive delivery to the small intestine.
Collapse
Affiliation(s)
- Weiwei Liu
- China-Canada Joint Laboratory of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing100048, China
| | - Ruolan Ma
- China-Canada Joint Laboratory of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing100048, China
| | - Shiyi Lu
- China-Canada Joint Laboratory of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing100048, China
| | - Yangyang Wen
- College of Chemistry and Materials Engineering, Beijing Technology and Business University (BTBU), Beijing100048, China
| | - Hongyan Li
- China-Canada Joint Laboratory of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing100048, China
| | - Jing Wang
- China-Canada Joint Laboratory of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing100048, China
| | - Baoguo Sun
- China-Canada Joint Laboratory of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing100048, China
| |
Collapse
|
214
|
Sex-specific features of optical coherence tomography detected plaque vulnerability related to clinical outcomes: insights from the CLIMA study. Int J Cardiovasc Imaging 2022; 39:873-881. [PMID: 36534217 DOI: 10.1007/s10554-022-02775-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE To investigate the different impact of optical coherence tomography (OCT)-derived vulnerable plaque features on future adverse events (AEs) according to the biological sex. METHODS The prospective multicenter CLIMA study (ClinicalTrials.gov: NCT02883088) enrolled 1003 patients with OCT plaque analysis of non-treated coronary plaques located in the proximal left anterior descending artery. Sex-specific differences in plaque composition and vulnerable features were described. We investigated the incidence of AEs, including cardiac death, any myocardial infarction and target vessel revascularization at 1-year. RESULTS Among 1003 patients, 24.6% were women. Women were older and more frequently affected by chronic kidney disease. Dyslipidemia, prior MI and smoking habit were more common in men. At OCT analysis, women had shorter plaque length (p < 0.001), ticker fibrous cap (p = 0.001), smaller maximum lipid arc (p = 0.019), lower macrophage infiltration (p < 0.001) and intra-plaque layered tissue (p = 0.007). During follow-up, 65 AEs were registered. The presence of a thin fibrous cap and a large macrophage infiltration (> 67°) predicted AEs in both sexes. The presence of macrophages (HR 3.38, p = 0.018) and a small minimum lumen area (HR 4.97, p = 0.002) were associated with AEs in women but not in men, while a large lipid arc (> 180°) was associated with AEs in men (HR 2.56, p = 0.003) but not in women. CONCLUSION This subanalysis of the CLIMA study investigated for the first-time sex-specific OCT features of plaque vulnerability associated with AEs. Local inflammation was associated with AEs in women and a large lipid arc was predictive in men. OCT may help develop sex-specific risk stratification strategies.
Collapse
|
215
|
Huang B, Lang X, Li X. The role of IL-6/JAK2/STAT3 signaling pathway in cancers. Front Oncol 2022; 12:1023177. [PMID: 36591515 PMCID: PMC9800921 DOI: 10.3389/fonc.2022.1023177] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine involved in immune regulation. It can activate janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signaling pathway. As one of the important signal transduction pathways in cells, JAK2/STAT3 signaling pathway plays a critical role in cell proliferation and differentiation by affecting the activation state of downstream effector molecules. The activation of JAK2/STAT3 signaling pathway is involved in tumorigenesis and development. It contributes to the formation of tumor inflammatory microenvironment and is closely related to the occurrence and development of many human tumors. This article focuses on the relationship between IL-6/JAK2/STAT3 signaling pathway and liver cancer, breast cancer, colorectal cancer, gastric cancer, lung cancer, pancreatic cancer and ovarian cancer, hoping to provide references for the research of cancer treatment targeting key molecules in IL-6/JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Bei Huang
- Operational Management Office, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaoling Lang
- Operational Management Office, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China,*Correspondence: Xiaoling Lang, ; Xihong Li,
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China,Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China,*Correspondence: Xiaoling Lang, ; Xihong Li,
| |
Collapse
|
216
|
Anand S, Azam Ansari M, Kumaraswamy Sukrutha S, Alomary MN, Anwar Khan A, Elderdery AY. Resolvins Lipid Mediators: Potential Therapeutic Targets in Alzheimer and Parkinson Disease. Neuroscience 2022; 507:139-148. [PMID: 36372297 DOI: 10.1016/j.neuroscience.2022.11.001] [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: 07/27/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
Inflammation and resolution are highly programmed processes involving a plethora of immune cells. Lipid mediators synthesized from arachidonic acid metabolism play a pivotal role in orchestrating the signaling cascades in the game of inflammation. The majority of the studies carried out so far on inflammation were aimed at inhibiting the generation of inflammatory molecules, whereas recent research has shifted more towards understanding the resolution of inflammation. Owing to chronic inflammation as evident in neuropathophysiology, the resolution of inflammation together with the class of lipid mediators actively involved in its regulation has attracted the attention of the scientific community as therapeutic targets. Both omega-three polyunsaturated fatty acids, eicosapentaenoic acid and docosahexaenoic acid, orchestrate a vital regulatory role in inflammation development. Resolvins derived from these fatty acids comprise the D-and E-series resolvins. A growing body of evidence using in vitro and in vivo models has revealed the pro-resolving and anti-inflammatory potential of resolvins. This systematic review sheds light on the synthesis, specialized receptors, and resolution of inflammation mediated by resolvins in Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Santosh Anand
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institutes for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia.
| | - Sambamurthy Kumaraswamy Sukrutha
- Department of Microbiology, Biotechnology and Food Technology, Jnana Bharathi Campus, Bangalore University, Bengaluru, Karnataka, India
| | - Mohammad N Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Anmar Anwar Khan
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abozer Y Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Saudi Arabia
| |
Collapse
|
217
|
Ridker PM, Tuttle KR, Perkovic V, Libby P, MacFadyen JG. Inflammation drives residual risk in chronic kidney disease: a CANTOS substudy. Eur Heart J 2022; 43:4832-4844. [PMID: 35943897 DOI: 10.1093/eurheartj/ehac444] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/09/2022] [Accepted: 07/27/2022] [Indexed: 01/12/2023] Open
Abstract
AIMS Hyperlipidaemia and inflammation jointly contribute to atherosclerotic disease. Yet, after the initiation of statin therapy, the relative contributions of these processes may differ in patient groups, such as those with and without impaired kidney function. METHODS AND RESULTS Among 9151 stable statin-treated post-myocardial infarction patients participating in the CANTOS trial, the contributions of residual cholesterol risk and residual inflammatory risk were evaluated as determinants of recurrent major adverse cardiovascular events (MACE) and total mortality, stratified by baseline estimated glomerular filtration rate (eGFR) above or below 60 mL/min/1.73 m2 using the race agnostic CKD-EPI 2021 formula (all participants had eGFR > 30 mL/min/1.73 m2). Analyses of residual inflammatory risk focused on high-sensitivity C-reactive protein (hsCRP) and interleukin-6 (IL-6) while analyses of residual cholesterol risk focused on LDL-cholesterol (LDL-C) and non-HDL-cholesterol (non-HDL-C). Participants were followed for a period of up to 5 years (median 3.7 years). Median baseline levels of LDL-C and hsCRP were 81 mg/dL and 4.2 mg/L. Among participants with eGFR ≥ 60 mL/min/1.73 m2, increasing quartiles of plasma hsCRP, IL-6, LDL-C, and non-HDL-C all positively associated with risks of recurrent MACE [hazard ratios (HR) comparing the top to bottom quartile for hsCRP 1.45; for IL-6 2.48; for LDL-C 1.64; and for non-HDL-C 1.68] (all P < 0.0001). By contrast, among those with eGFR < 60 mL/min/1.73 m2, increasing quartiles of hsCRP and IL-6 significantly predicted recurrent MACE [HR comparing the top to bottom quartile for hsCRP 1.50 (P = 0.021); for IL-6 1.84 (P = 0.048)], whereas increasing quartiles of LDL-C and non-HDL-C did not [HR comparing the top to bottom quartile for LDL-C 1.04 (P = 0.80); for non-HDL-C 0.98 (P = 0.88)]. The predictive utility of hsCRP and IL-6 in the setting of eGFR < 60 mL/min/1.73 m2 remained significant after adjustment for a wide range of potential confounding factors including age, sex, smoking status, blood pressure, body mass index, and diabetes. For the endpoint of total mortality, both hsCRP (HR 1.77, P = 0.0021) and IL-6 (HR 2.15, P = 0.015) were significant predictors among those with eGFR < 60 mL/min/1.73 m2, whereas LDL-C (HR 0.91, P = 0.56) and non-HDL-C (HR 0.85, P = 0.31) were not. Similar effects were observed in analyses stratified by the albumin to creatinine ratio rather than eGFR. CONCLUSION Among atherosclerosis patients with impaired kidney function already aggressively treated with statin therapy, residual inflammatory risk plays a substantial role in determining the risk of recurrent cardiovascular events. These data have implications for risk stratification of individuals with chronic kidney disease and for the development of novel agents that target inflammatory processes in this high-risk group of patients. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov: NCT01327846.
Collapse
Affiliation(s)
- Paul M Ridker
- Cardiovascular Disease Prevention, Division of Preventive Medicine, Brigham and Women's Hospital, 900 Commonwealth Avenue, Boston, MA 02215, USA.,Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Vlado Perkovic
- Royal North Shore Hospital, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jean G MacFadyen
- Cardiovascular Disease Prevention, Division of Preventive Medicine, Brigham and Women's Hospital, 900 Commonwealth Avenue, Boston, MA 02215, USA
| |
Collapse
|
218
|
Lipoprotein(a), high-sensitivity C-reactive protein, and cardiovascular risk in patients undergoing percutaneous coronary intervention. Atherosclerosis 2022; 363:109-116. [PMID: 36357218 DOI: 10.1016/j.atherosclerosis.2022.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS In patients with coronary artery disease (CAD) undergoing percutaneous coronary intervention (PCI), the effects of high-sensitivity C-reactive protein (hsCRP) on Lipoprotein(a) (Lp(a))-associated cardiovascular risk remains unclear. This study aimed to investigate the independent and combined association of Lp(a) and hsCRP with cardiovascular events in this specific population. METHODS A total of 10,424 patients with measurements of both Lp(a) and hsCRP were included in this prospective cohort study. Cox proportional hazards models and Kaplan-Meier analysis were performed to evaluate the relationship between Lp(a), hsCRP and adverse cardiac and cerebrovascular events (MACCE; all-cause death, myocardial infarction, ischemic stroke and revascularization). RESULTS During 5 years of follow-up, 2140 (20.5%) MACCE occurred. Elevated Lp(a) and hsCRP levels were associated with increased risks of MACCE (p<0.05). Notably, there might be a significant interaction between Lp(a) and hsCRP (P for interaction = 0.019). In the setting of hsCRP≥2 mg/L, significant higher risk of MACCE was observed with Lp(a) 15-29.9 mg/dL (HR: 1.18; 95% CI 1.01-1.39) and Lp(a) ≥30 mg/dL (HR: 1.20; 95% CI 1.04-1.39), whereas such association was attenuated when hsCRP was <2 mg/L with Lp(a) 15-29.9 mg/dL (HR: 0.94; 95% CI 0.80-1.10) and Lp(a) ≥30 mg/dL (HR: 1.12; 95% CI 0.98-1.28). Moreover, when Lp(a) and hsCRP were combined for risk stratification, patients with dual elevation of these two biomarkers had a significant higher risk of MACCE compared with the reference group (Lp(a) < 15 mg/dL and hsCRp<2 mg/L) (p<0.05). CONCLUSIONS In patients with CAD undergoing PCI, high Lp(a) level was associated with worse outcomes, and this association might be stronger in those with elevated hsCRP concomitantly. Evaluation of Lp(a) and hsCRP together may help identify high-risk individuals for targeted intervention in clinical utility.
Collapse
|
219
|
Li Z, Zhang R, Mu H, Zhang W, Zeng J, Li H, Wang S, Zhao X, Chen W, Dong J, Yang R. Oral Administration of Branched-Chain Amino Acids Attenuates Atherosclerosis by Inhibiting the Inflammatory Response and Regulating the Gut Microbiota in ApoE-Deficient Mice. Nutrients 2022; 14:5065. [PMID: 36501095 PMCID: PMC9739883 DOI: 10.3390/nu14235065] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that serves as a common pathogenic underpinning for various cardiovascular diseases. Although high circulating branched-chain amino acid (BCAA) levels may represent a risk factor for AS, it is unclear whether dietary BCAA supplementation causes elevated levels of circulating BCAAs and hence influences AS, and the related mechanisms are not well understood. Here, ApoE-deficient mice (ApoE-/-) were fed a diet supplemented with or without BCAAs to investigate the effects of BCAAs on AS and determine potential related mechanisms. In this study, compared with the high-fat diet (HFD), high-fat diet supplemented with BCAAs (HFB) reduced the atherosclerotic lesion area and caused a significant decrease in serum cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels. BCAA supplementation suppressed the systemic inflammatory response by reducing macrophage infiltration; lowering serum levels of inflammatory factors, including monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6); and suppressing inflammatory related signaling pathways. Furthermore, BCAA supplementation altered the gut bacterial beta diversity and composition, especially reducing harmful bacteria and increasing probiotic bacteria, along with increasing bile acid (BA) excretion. In addition, the levels of total BAs, primary BAs, 12α-hydroxylated bile acids (12α-OH BAs) and non-12α-hydroxylated bile acids (non-12α-OH BAs) in cecal and colonic contents were increased in the HFB group of mice compared with the HFD group. Overall, these data indicate that dietary BCAA supplementation can attenuate atherosclerosis induced by HFD in ApoE-/- mice through improved dyslipidemia and inflammation, mechanisms involving the intestinal microbiota, and promotion of BA excretion.
Collapse
Affiliation(s)
- Ziyun Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Ranran Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Hongna Mu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Wenduo Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jie Zeng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Hongxia Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Siming Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Xianghui Zhao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Wenxiang Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Jun Dong
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Ruiyue Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| |
Collapse
|
220
|
The intracellular signaling pathways governing macrophage activation and function in human atherosclerosis. Biochem Soc Trans 2022; 50:1673-1682. [DOI: 10.1042/bst20220441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by lipid accumulation and plaque formation in arterial vessel walls. Atherosclerotic plaques narrow the arterial lumen to increase the risk of heart attacks, ischemic stroke and peripheral vascular disease, which are major and worldwide health and economic burdens. Macrophage accumulation within plaques is characteristic of all stages of atherosclerosis and their presence is a potential marker of disease activity and plaque stability. Macrophages engulf lipids and modified lipoproteins to form foam cells that express pro-inflammatory and chemotactic effector molecules, stress inducing factors and reactive oxygen species. They control plaque stability and rupture through secretion of metalloproteinases and extracellular matrix degradation. Although macrophages can worsen disease by propagating inflammation, they can stabilize atherosclerotic plaques through tissue remodeling, promoting the formation of a fibrous cap, clearing apoptotic cells to prevent necrotic core formation and through vascular repair. In atherosclerosis, macrophages respond to dyslipidaemia, cytokines, dying cells, metabolic factors, lipids, physical stimuli and epigenetic factors and exhibit heterogeneity in their activation depending on the stimuli they receive. Understanding these signals and the pathways driving macrophage function within developing and established plaques and how they can be pharmacologically modulated, represents a strategy for the prevention and treatment of atherosclerosis. This review focusses on the current understanding of factors controlling macrophage heterogeneity and function in atherosclerosis. Particular attention is given to the macrophage intracellular signaling pathways and transcription factors activated by biochemical and biophysical stimuli within plaques, and how they are integrated to regulate plaque formation and stability.
Collapse
|
221
|
Chen Z, Jiang F, Yang M, Yang J. Relationship between CRP gene polymorphisms and ischemic stroke risk: A systematic review and meta-analysis. Open Life Sci 2022; 17:1519-1530. [DOI: 10.1515/biol-2022-0505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/28/2022] [Accepted: 09/04/2022] [Indexed: 11/17/2022] Open
Abstract
Abstract
Ischemic stroke (IS), usually caused due to an abrupt blockage of an artery, is the leading cause of disability and the second leading cause of death worldwide. The association of the C-reactive protein (CRP) gene (s3093059 T/C and rs1205 C/T) polymorphisms and IS susceptibility has been widely studied, but the results remain inconsistent. Our study aimed to assess the association between CRP gene (s3093059 T/C and rs1205 C/T) polymorphisms and IS risk. PubMed, Embase, Cochrane Library, Web of Science, China National Knowledge Infrastructure, and WanFang databases were searched up to April 2022 to identify eligible studies. The Newcastle-Ottawa scale (NOS) score was calculated to assess study quality. The odd ratios (ORs) with a 95% confidence interval (CI) were calculated to assess the association between CRP gene (rs3093059 T/C and rs1205 C/T) polymorphisms and IS risk. Eighteen case–control studies with 6339 cases and 29580 controls were identified. We found that CRP (s3093059 T/C and rs1205 C/T) polymorphism was not significantly associated with the risk of IS in any genetic model (recessive model: OR 1.00, 95% CI 0.79–1.26; OR 1.06, 95% CI 0.90–1.25). When stratified analysis by country, genotype method, source of controls, and NOS score, still no statistically significant association was found. Our study indicated that the CRP (rs3093059 T/C and rs1205 C/T) polymorphisms were not associated with the susceptibility to IS.
Collapse
Affiliation(s)
- Zhizhi Chen
- Department of Neurology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital , Quzhou 324000 , Zhejiang , China
| | - Feifei Jiang
- Department of Neurology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital , Quzhou 324000 , Zhejiang , China
| | - Ming Yang
- Department of Neurology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital , Quzhou 324000 , Zhejiang , China
| | - Jie Yang
- Department of Rehabilitation Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital , 100 Minjiang Road , Quzhou 324000 , Zhejiang , China
| |
Collapse
|
222
|
Nasonov EL, Feist E. The prospects of interleukin-6 inhibition in rheumatoid arthritis: Olokizumab (novel monoclonal antibodies to IL-6). RHEUMATOLOGY SCIENCE AND PRACTICE 2022. [DOI: 10.47360/1995-4484-2022-505-518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic immune-mediated rheumatic diseases (IMRDs) manifested with progressive destruction of joints, systemic inflammation of visceral organs and a wide range of co-morbidities associated with chronic inflammation. Among the cytokines involved in the pathogenesis of RA and certain other IMRDs, the role of interleukin (IL) 6 is of special interest. The introduction of mAbs tocilizumab (TCZ) and later sarilumab (SAR), both blocking the receptor of this cytokine, into clinical practice was an important achievement in the treatment of IIRDs at the beginning of the 21st century. As a novel approach in the treatment of RA, the humanized mAb against IL-6 olokizumab (OKZ) is in development by the Russian company R-PHARM under the license agreement with UCB Pharma. The review examines new data on efficacy and safety of OKZ in RA and the prospects of its use in rheumatology
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology; I.M. Sechenov First Moscow State Medical University of the Ministry of Health Care of Russian Federation (Sechenov University)
| | - Eugen Feist
- Department of Rheumatology, Helios Clinic VogelsangGommern, cooperation partner of the Otto-vonGuericke University Magdeburg
| |
Collapse
|
223
|
McAlpine CS, Kiss MG, Zuraikat FM, Cheek D, Schiroli G, Amatullah H, Huynh P, Bhatti MZ, Wong LP, Yates AG, Poller WC, Mindur JE, Chan CT, Janssen H, Downey J, Singh S, Sadreyev RI, Nahrendorf M, Jeffrey KL, Scadden DT, Naxerova K, St-Onge MP, Swirski FK. Sleep exerts lasting effects on hematopoietic stem cell function and diversity. J Exp Med 2022; 219:213487. [PMID: 36129517 PMCID: PMC9499822 DOI: 10.1084/jem.20220081] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/21/2022] [Accepted: 08/22/2022] [Indexed: 01/21/2023] Open
Abstract
A sleepless night may feel awful in its aftermath, but sleep's revitalizing powers are substantial, perpetuating the idea that convalescent sleep is a consequence-free physiological reset. Although recent studies have shown that catch-up sleep insufficiently neutralizes the negative effects of sleep debt, the mechanisms that control prolonged effects of sleep disruption are not understood. Here, we show that sleep interruption restructures the epigenome of hematopoietic stem and progenitor cells (HSPCs) and increases their proliferation, thus reducing hematopoietic clonal diversity through accelerated genetic drift. Sleep fragmentation exerts a lasting influence on the HSPC epigenome, skewing commitment toward a myeloid fate and priming cells for exaggerated inflammatory bursts. Combining hematopoietic clonal tracking with mathematical modeling, we infer that sleep preserves clonal diversity by limiting neutral drift. In humans, sleep restriction alters the HSPC epigenome and activates hematopoiesis. These findings show that sleep slows decay of the hematopoietic system by calibrating the hematopoietic epigenome, constraining inflammatory output, and maintaining clonal diversity.
Collapse
Affiliation(s)
- Cameron S. McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Cameron S. McAlpine:
| | - Máté G. Kiss
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Faris M. Zuraikat
- Sleep Center of Excellence, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - David Cheek
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Giulia Schiroli
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| | - Hajera Amatullah
- Division of Gastroenterology and Center for the Study of Inflammatory Bowel Disease, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Pacific Huynh
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mehreen Z. Bhatti
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Lai-Ping Wong
- Department of Molecular Biology, Massachusetts General Hospital and Department of Genetics, Harvard Medical School, Boston, MA
| | - Abi G. Yates
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Wolfram C. Poller
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - John E. Mindur
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Christopher T. Chan
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Henrike Janssen
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Jeffrey Downey
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Sumnima Singh
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital and Department of Genetics, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Matthias Nahrendorf
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Kate L. Jeffrey
- Division of Gastroenterology and Center for the Study of Inflammatory Bowel Disease, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - David T. Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| | - Kamila Naxerova
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Marie-Pierre St-Onge
- Sleep Center of Excellence, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Marie-Pierre St-Onge:
| | - Filip K. Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Correspondence to Filip K. Swirski:
| |
Collapse
|
224
|
Koshino A, Schechter M, Sen T, Vart P, Neuen BL, Neal B, Arnott C, Perkovic V, Ridker PM, Tuttle KR, Hansen MK, Heerspink HJL. Interleukin-6 and Cardiovascular and Kidney Outcomes in Patients With Type 2 Diabetes: New Insights From CANVAS. Diabetes Care 2022; 45:2644-2652. [PMID: 36134918 PMCID: PMC9862371 DOI: 10.2337/dc22-0866] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/11/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The inflammatory cytokine interleukin-6 (IL-6) is associated with cardiovascular (CV) and kidney outcomes in various populations. However, data in patients with type 2 diabetes are limited. We assessed the association of IL-6 with CV and kidney outcomes in the Canagliflozin Cardiovascular Assessment Study (CANVAS) and determined the effect of canagliflozin on IL-6. RESEARCH DESIGN AND METHODS Patients with type 2 diabetes at high CV risk were randomly assigned to canagliflozin or placebo. Plasma IL-6 was measured at baseline and years 1, 3, and 6. The composite CV outcome was nonfatal myocardial infarction, nonfatal stroke, or CV death; the composite kidney outcome was sustained ≥40% estimated glomerular filtration rate decline, end-stage kidney disease, or kidney-related death. Multivariable-adjusted Cox proportional hazards regression was used to estimate the associations between IL-6 and the outcomes. The effect of canagliflozin on IL-6 over time was assessed with a repeated-measures mixed-effects model. RESULTS The geometric mean IL-6 at baseline, available in 3,503 (80.2%) participants, was 1.7 pg/mL. Each doubling of baseline IL-6 was associated with 14% (95% CI 4, 24) and 21% (95% CI 1, 45) increased risk of CV and kidney outcomes, respectively. Over 6 years, IL-6 increased by 5.8% (95% CI 3.4, 8.3) in the placebo group. Canagliflozin modestly attenuated the IL-6 increase (absolute percentage difference vs. placebo 4.4% [95% CI 1.3, 9.9; P = 0.01]). At year 1, each 25% lower level of IL-6 compared with baseline was associated with 7% (95% CI 1, 22) and 14% (95% CI 5, 22) lower risks for the CV and kidney outcome, respectively. CONCLUSIONS In patients with type 2 diabetes at high CV risk, baseline IL-6 and its 1-year change were associated with CV and kidney outcomes. The effect of IL-6-lowering therapy on CV, kidney, and safety outcomes remains to be tested.
Collapse
Affiliation(s)
- Akihiko Koshino
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Nephrology and Laboratory Medicine, Kanazawa University, Ishikawa, Japan
| | - Meir Schechter
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Diabetes Unit, Department of Endocrinology and Metabolism, Hadassah Medical Center, Jerusalem, Israel.,Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Taha Sen
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Priya Vart
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Bruce Neal
- The George Institute for Global Health, Sydney, Australia
| | - Clare Arnott
- The George Institute for Global Health, Sydney, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | | | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Katherine R Tuttle
- Providence Medical Research Center, Providence Health Care, and Nephrology Division, University of Washington, Spokane, WA
| | | | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,The George Institute for Global Health, Sydney, Australia.,University of New South Wales, Sydney, Australia
| |
Collapse
|
225
|
Yang Y, Ding X, Chen F, Wu X, Chen Y, Zhang Q, Cao J, Wang J, Dai Y. Inhibition Effects of Nippostrongylus brasiliensis and Its Derivatives against Atherosclerosis in ApoE-/- Mice through Anti-Inflammatory Response. Pathogens 2022; 11:pathogens11101208. [PMID: 36297265 PMCID: PMC9610917 DOI: 10.3390/pathogens11101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis (AS) is a dominant and growing cause of death and disability worldwide that involves inflammation from its inception to the emergence of complications. Studies have demonstrated that intervention with helminth infections or derived products could modulate the host immune response and effectively prevent or mitigate the onset and progression of inflammation-related diseases. Therefore, to understand the molecular mechanisms underlying the development of atherosclerosis, we intervened in ApoE-/- mice maintained on a high-fat diet with Nippostrongylus brasiliensis (N. brasiliensis) infection and immunized with its derived products. We found that N. brasiliensis infection and its derived proteins had suitable protective effects both in the initial and progressive stages of atherosclerosis, effectively reducing aortic arch plaque areas and liver lipid contents and downregulating serum LDL levels, which may be associated with the significant upregulation of serum anti-inflammatory cytokines (IL-10 and IL-4) and the down-regulation of proinflammatory cytokines (TNF-α and IFN-γ) in the serum. In conclusion, these data highlighted the effective regulatory role of N. brasiliensis and its derived proteins in the development and progression of atherosclerosis. This could provide a promising new avenue for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yougui Yang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Xin Ding
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Fuzhong Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaomin Wu
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei 230601, China
| | - Yuying Chen
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Qiang Zhang
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Jun Cao
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Junhong Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Correspondence: (J.W.); (Y.D.)
| | - Yang Dai
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
- Correspondence: (J.W.); (Y.D.)
| |
Collapse
|
226
|
Kalyuzhin VV, Teplyakov AT, Bespalova ID, Kalyuzhina EV, Terentyeva NN, Grakova EV, Kopeva KV, Usov VY, Garganeeva NP, Pavlenko OA, Gorelova YV, Teteneva AV. Promising directions in the treatment of chronic heart failure: improving old or developing new ones? BULLETIN OF SIBERIAN MEDICINE 2022. [DOI: 10.20538/1682-0363-2022-3-181-197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Unprecedented advances of recent decades in clinical pharmacology, cardiac surgery, arrhythmology, and cardiac pacing have significantly improved the prognosis in patients with chronic heart failure (CHF). However, unfortunately, heart failure continues to be associated with high mortality. The solution to this problem consists in simultaneous comprehensive use in clinical practice of all relevant capabilities of continuously improving methods of heart failure treatment proven to be effective in randomized controlled trials (especially when confirmed by the results of studies in real clinical practice), on the one hand, and in development and implementation of innovative approaches to CHF treatment, on the other hand. This is especially relevant for CHF patients with mildly reduced and preserved left ventricular ejection fraction, as poor evidence base for the possibility of improving the prognosis in such patients cannot justify inaction and leaving them without hope of a clinical improvement in their condition. The lecture consistently covers the general principles of CHF treatment and a set of measures aimed at inotropic stimulation and unloading (neurohormonal, volumetric, hemodynamic, and immune) of the heart and outlines some promising areas of disease-modifying therapy.
Collapse
Affiliation(s)
| | - A. T. Teplyakov
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | | | | | | | - E. V. Grakova
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | - K. V. Kopeva
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | - V. Yu. Usov
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | | | | | | | | |
Collapse
|
227
|
Liu S, Zhang X, Zhou Q. Efficacy of Periodontal Endodontics Combined with Diode Laser (DL) Therapy on Severe Periodontitis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5689900. [PMID: 36285164 PMCID: PMC9588349 DOI: 10.1155/2022/5689900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/17/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022]
Abstract
Background For a long time, the impact of severe periodontitis on the pulp has been the focus of periodontal clinical research. Whether the teeth with severe periodontitis should be treated with pulp has also become the focus of clinical research. Aims To explore the effect of periodontal endodontic therapy combined with DL therapy on severe periodontitis. Materials and Methods The clinical data of 100 patients with severe periodontitis from January 2020 to July 2022 were selected and included in the retrospective study. According to the different retrieval treatment methods, they were divided into the control group and the treatment group with 50 cases in each group. The control group received periodontal endodontic treatment, and the treatment group received DL treatment on the basis of the control group. The differences in periodontal probing depth (PD), toothache degree, bleeding index (BI), inflammatory factors, plaque index (PLI), and attachment loss (AL) between the two groups were compared and analyzed. Results After 3 months of treatment, the bleeding index (BI), plaque index (PLI), and periodontal probing depth (PD) of the treatment group were significantly lower than those of the control group, and the difference was statistically significant (P < 0.05). The attachment loss (AL) of the group was not significantly different from that of the control group (P > 0.05). Before treatment, there was no significant difference in the levels of inflammatory factors between the two groups (P > 0.05). After 3 months of treatment, the levels of IL-6 and CRP in the treatment group were significantly lower than those in the control group, and the difference was statistically significant (P < 0.05). Before treatment, there was no significant difference in the levels of inflammatory factors between the two groups (P > 0.05). After 3 days of treatment, the VAS score of the treatment group was significantly lower than that of the control group, and the difference was statistically significant (P < 0.05).After treatment, there were no complications during follow-up in the two groups. Conclusion The application of DL treatment has a significant effect, which can promote the healing of periodontal tissue, reduce the depth of periodontal pockets, and reduce the degree of toothache, thereby providing a reference for clinical treatment.
Collapse
Affiliation(s)
- Sijia Liu
- Wuhan Hospital of Traditional Chinese Medicine, Department of Stomatology, No. 303 Sixin Avenue, Hanyang, Wuhan 430000, Hubei, China
| | - Xu Zhang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology (Stomatology), No. 116 Yangyuan Street, Wuchang District, Wuhan 430063, Hubei, China
| | - Quanying Zhou
- Wuhan 9th Hospital, Department Stomatology, No. 20, Jilin Street, Qingshan District, Wuhan 430080, Hubei, China
| |
Collapse
|
228
|
Long H, Lin H, Zheng P, Hou L, Zhang M, Lin S, Yin K, Zhao G. WTAP mediates the anti-inflammatory effect of Astragalus mongholicus polysaccharide on THP-1 macrophages. Front Pharmacol 2022; 13:1023878. [PMID: 36278233 PMCID: PMC9585178 DOI: 10.3389/fphar.2022.1023878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Background:Astragalus mongholicus polysaccharides (APS) have anti-inflammatory, antioxidant and immunomodulatory effects. Recent studies have demonstrated the epigenetic regulation of N6-methyladenosine (m6A) in the development of inflammation. However, the effect of APS on m6A modification is unclear. Here, for the first time, we investigate the mechanism of m6A modification in APS regulation of THP-1 macrophage inflammation. Methods: We treated LPS-induced THP-1 macrophages with APS at different concentrations and times, and detected IL-6 mRNA and protein levels by quantitative real-time PCR (qRT-PCR) and western blot, respectively. The m6A modification level was detected by m6A quantification kit. The proteins that regulate m6A modification were screened by western blot. Wilms’ tumor 1-associating protein (WTAP) was overexpressed in APS-treated THP-1 macrophages and the m6A modification level and IL-6 expressions were detected. Results: These findings confirmed that APS significantly abolished LPS-induced IL-6 levels in THP-1 macrophages. Meanwhile, APS reduced m6A modification levels and WTAP gene expression in THP-1 macrophages. Further overexpression of WTAP can significantly reverse APS-induced m6A modification level and IL-6 expression. Mechanistically, APS regulates IL-6 expression through WTAP-mediated p65 nuclear translocation. Conclusion: Overall, our study suggested that WTAP mediates the anti-inflammatory effect of APS by regulating m6A modification levels in THP-1 macrophages. This study reveals a new dimension of APS regulation of inflammation at the epigenetic level.
Collapse
Affiliation(s)
- Haijiao Long
- Xiangya Hospital, Central South University, Changsha, China
- The Sixth Affiliated Hospital, Guangzhou Medical University, Qingyuan, China
| | - Haiyue Lin
- The Sixth Affiliated Hospital, Guangzhou Medical University, Qingyuan, China
| | - Pan Zheng
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Lianjie Hou
- The Sixth Affiliated Hospital, Guangzhou Medical University, Qingyuan, China
| | - Ming Zhang
- The Sixth Affiliated Hospital, Guangzhou Medical University, Qingyuan, China
| | - Shuyun Lin
- The Sixth Affiliated Hospital, Guangzhou Medical University, Qingyuan, China
| | - Kai Yin
- Department of Cardiology, The Second Afliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, China
- *Correspondence: Kai Yin, ; Guojun Zhao,
| | - Guojun Zhao
- Xiangya Hospital, Central South University, Changsha, China
- The Sixth Affiliated Hospital, Guangzhou Medical University, Qingyuan, China
- *Correspondence: Kai Yin, ; Guojun Zhao,
| |
Collapse
|
229
|
Hetherington I, Totary-Jain H. Anti-atherosclerotic therapies: Milestones, challenges, and emerging innovations. Mol Ther 2022; 30:3106-3117. [PMID: 36065464 PMCID: PMC9552812 DOI: 10.1016/j.ymthe.2022.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
Atherosclerosis is the main underlying pathology for many cardiovascular diseases (CVDs), which are the leading cause of death globally and represent a serious health crisis. Atherosclerosis is a chronic condition that can lead to myocardial infarction, ischemic cardiomyopathy, stroke, and peripheral arterial disease. Elevated plasma lipids, hypertension, and high glucose are the major risk factors for developing atherosclerotic plaques. To date, most pharmacological therapies aim to control these risk factors, but they do not target the plaque-causing cells themselves. In patients with acute coronary syndromes, surgical revascularization with percutaneous coronary intervention has greatly reduced mortality rates. However, stent thrombosis and neo-atherosclerosis have emerged as major safety concerns of drug eluting stents due to delayed re-endothelialization. This review summarizes the major milestones, strengths, and limitations of current anti-atherosclerotic therapies. It provides an overview of the recent discoveries and emerging game-changing technologies in the fields of nanomedicine, mRNA therapeutics, and gene editing that have the potential to revolutionize CVD clinical practice by steering it toward precision medicine.
Collapse
Affiliation(s)
- Isabella Hetherington
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC08, 2170, Tampa, FL 33612, USA
| | - Hana Totary-Jain
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC08, 2170, Tampa, FL 33612, USA.
| |
Collapse
|
230
|
Jia X, Bai X, Yang X, Wang L, Lu Y, Zhu L, Zhao Y, Cheng W, Shu M, Mei Q, Jin S. VCAM-1-binding peptide targeted cationic liposomes containing NLRP3 siRNA to modulate LDL transcytosis as a novel therapy for experimental atherosclerosis. Metabolism 2022; 135:155274. [PMID: 35917895 DOI: 10.1016/j.metabol.2022.155274] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Activation of NLRP3 inflammasome accelerates the formation of atherosclerotic plaques. Here, we evaluated the effects of inflammation on the expression of the NLRP3 inflammasome in endothelial cells (ECs). METHODS The effect of TNF-α on transcytosis of LDL was measured. VCAM-1 binding peptide targeting cationic liposomes (PCLs) were prepared as siRNA vectors. Methylated NLRP3 siRNA was encapsulated into the PCLs to knock down NLRP3 in vitro and in vivo. In rats with partial carotid ligation, TNF-α-induced LDL retention in the carotid artery endothelium was observed. In ApoE-/- mice, NLRP3 siRNA-PCLs were injected intravenously to observe their effect on the formation of atherosclerosis. RESULTS Our results showed that TNF-α upregulated NLRP3 in ECs, promoting the assembly of the NLRP3 inflammasome and processing of pro-IL-1β into IL-1β. Moreover, TNF-α accelerated LDL transcytosis in ECs. Knockdown of NLRP3 prevented TNF-α-induced NLPR3 inflammasome/IL-1β signaling and LDL transcytosis. Using optimized cationic liposomes to encapsulate methylated NLRP3 siRNA, resulting in targeting of VCAM-1-expressing ECs, to knockdown NLRP3, TNF-α-induced NLRP3 inflammasome activation and LDL transcytosis were prevented. Using the partial carotid ligation as an atherosclerosis rat model, we found that local administration of NLRP3 siRNA-PCLs efficiently knocked down NLPR3 expression in the carotid endothelium and dramatically attenuated the deposition of atherogenic LDL in carotid ECs in TNF-α-challenged rats. Furthermore, NLRP3 siRNA-PCLs were injected intravenously in ApoE-/- mice, resulting in reduced plaque formation. CONCLUSION These findings established a novel strategy for targeting the NLRP3 inflammasome using NLRP3 siRNA-PCLs to interrupt LDL transcytosis, representing a potential novel therapy for atherosclerosis.
Collapse
Affiliation(s)
- Xiong Jia
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China; Department of Cardiovascular Surgery, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Xiangli Bai
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Xiaoyan Yang
- Department of Pharmacology, the Key Laboratory of Drug Target Researches and Pharmacodynamics Evaluation of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling Wang
- Department of Pharmacology, the Key Laboratory of Drug Target Researches and Pharmacodynamics Evaluation of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yajing Lu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Lin Zhu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Ying Zhao
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Wenzhuo Cheng
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Meng Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China; Department of Pharmacology, the Key Laboratory of Drug Target Researches and Pharmacodynamics Evaluation of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
231
|
Park A, Ryder S, Sevigny M, Monden KR, Battaglino RA, Nguyen N, Goldstein R, Morse LR. Association between weekly exercise minutes and resting IL-6 in adults with chronic spinal cord injury: findings from the fracture risk after spinal cord injury exercise study. Spinal Cord 2022; 60:917-921. [PMID: 35840744 DOI: 10.1038/s41393-022-00833-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 11/08/2022]
Abstract
STUDY DESIGN Cross-sectional study. OBJECTIVE To assess associations between weekly aerobic exercise minutes and resting interleukin-6 (IL-6), C-reactive protein (CRP), or leptin levels in adults with chronic spinal cord injury (SCI). SETTING Three hundred and forty-four community-dwelling men and women with SCI duration of > 1 year. METHODS CRP, IL-6, and leptin levels were quantified by ultra-sensitive enzyme-linked immunoassay. Smoking, medication use, comorbidities, and aerobic exercise minutes per week were assessed by self-reported questionnaire. Body composition was determined by whole-body dual-energy X-ray absorptiometry. Generalized linear models were used to assess associations. RESULTS In multivariable modeling, resting IL-6 levels were 0.001 pg/mL lower for every 1 min of weekly aerobic exercise. IL-6 levels increased with increasing android-to-gynoid fat ratio, in active/ever smokers compared to never smokers, and in individuals with skin pressure injuries compared to those without. IL-6 levels were lower in active ibuprofen users compared to nonusers. We found no association between weekly exercise minutes and CRP or leptin when designing similar models. CONCLUSIONS Increasing aerobic exercise minutes is associated with lower IL-6 levels in adults with chronic SCI when considering body composition, smoking, skin pressure injuries, and ibuprofen use. CRP and leptin did not demonstrate an association with exercise when considering the similar variables. The use of these biomarkers in assessing the therapeutic value of future exercise-related interventions will be paramount for meaningful health improvement among those with SCI. Although a large, prospective dataset, this cross-sectional study cannot assign causation. Future prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Andrew Park
- Department of Physical Medicine and Rehabilitation, University of Colorado School of Medicine, Aurora, CO, USA
- Craig Rehabilitation Hospital, Englewood, CO, USA
| | - Stephanie Ryder
- Department of Physical Medicine and Rehabilitation, University of Colorado School of Medicine, Aurora, CO, USA
- Rocky Mountain Regional Veteran Affairs SCI/D, Aurora, CO, USA
| | | | - Kimberley R Monden
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Ricardo A Battaglino
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Nguyen Nguyen
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Richard Goldstein
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Leslie R Morse
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, MN, USA.
| |
Collapse
|
232
|
Kurti SP, Wisseman WS, Miller ME, Frick HL, Malin SK, Emerson SR, Edwards DA, Edwards ES. Acute Exercise and the Systemic and Airway Inflammatory Response to a High-Fat Meal in Young and Older Adults. Metabolites 2022; 12:metabo12090853. [PMID: 36144256 PMCID: PMC9505738 DOI: 10.3390/metabo12090853] [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: 07/16/2022] [Revised: 08/25/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
The purpose of the present study was to determine fasting and high-fat meal (HFM)-induced post-prandial systemic inflammation and airway inflammation (exhaled nitric oxide (eNO)) in older adults (OAs) compared to younger adults (YAs) before and after acute exercise. Twelve YAs (23.3 ± 3.9 y n = 5 M/7 F) and 12 OAs (67.7 ± 6 y, n = 8 M/4 F) completed two HFM challenges. After an overnight fast, participants underwent an HFM session or pre-prandial exercise (EX, 65% VO2Peak to expend 75% of the caloric content of the HFM) plus HFM (EX + HFM) in a randomized order. Systemic inflammatory cytokines were collected at 0, 3, and 6 h, while eNO was determined at 0, 2, and 4 h after the HFM (12 kcal/kg body weight: 61% fat, 35% CHO, 4% PRO). TNF-α was higher in OAs compared to YAs (p = 0.005) and decreased across time from baseline to 6 h post-HFM (p = 0.007). In response to the HFM, IL-6 decreased from 0 to 3 h but increased at 6 h regardless of age or exercise (p = 0.018). IL-8 or IL-1β did not change over the HFM by age or exercise (p > 0.05). eNO was also elevated in OAs compared to YAs (p = 0.003) but was not altered by exercise (p = 0.108). There was a trend, however, towards significance post-prandially in OAs and YAs from 0 to 2 h (p = 0.072). TNF-α and eNO are higher in OAs compared to YAs but are not elevated more in OAs post-prandially compared to YAs. Primary systemic inflammatory cytokines and eNO were not modified by acute exercise prior to an HFM.
Collapse
Affiliation(s)
- Stephanie P. Kurti
- Human Performance Laboratory, Department of Kinesiology, James Madison University, Harrisonburg, VA 22807, USA
- Morrison Bruce Center, James Madison University, Harrisonburg, VA 22807, USA
- Correspondence:
| | - William S. Wisseman
- Human Performance Laboratory, Department of Kinesiology, James Madison University, Harrisonburg, VA 22807, USA
| | - Molly E. Miller
- Human Performance Laboratory, Department of Kinesiology, James Madison University, Harrisonburg, VA 22807, USA
| | - Hannah L. Frick
- Human Performance Laboratory, Department of Kinesiology, James Madison University, Harrisonburg, VA 22807, USA
- Morrison Bruce Center, James Madison University, Harrisonburg, VA 22807, USA
| | - Steven K. Malin
- Department of Kinesiology and Health, Division of Endocrinology, Metabolism and Nutrition, Rutgers University, Piscataway, NJ 08854, USA
| | - Sam R. Emerson
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - David A. Edwards
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22904, USA
| | - Elizabeth S. Edwards
- Human Performance Laboratory, Department of Kinesiology, James Madison University, Harrisonburg, VA 22807, USA
- Morrison Bruce Center, James Madison University, Harrisonburg, VA 22807, USA
| |
Collapse
|
233
|
Targeting innate immunity-driven inflammation in CKD and cardiovascular disease. Nat Rev Nephrol 2022; 18:762-778. [PMID: 36064794 DOI: 10.1038/s41581-022-00621-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 11/08/2022]
Abstract
Mortality among patients with chronic kidney disease (CKD) is largely a consequence of cardiovascular disease (CVD) and is a particular concern given the increasing prevalence of CKD. Sterile inflammation triggered by activation of the innate immune system is an important driver of both CKD and associated CVD. Several endogenous mediators, including lipoproteins, crystals such as silica, urate and cholesterol crystals, or compounds released from dying cells interact with pattern recognition receptors expressed on a variety of different cell types, leading to the release of pro-inflammatory cytokines. Disturbed regulation of the haematopoietic system by damage-associated molecular patterns, or as a consequence of clonal haematopoiesis or trained innate immunity, also contributes to the development of inflammation. In observational and genetic association studies, inflammation is linked to the progression of CKD and cardiovascular events. In 2017, the CANTOS trial of canakinumab provided evidence that inhibiting inflammation driven by NLRP3-IL-1-IL-6-mediated signalling significantly reduced cardiovascular event rates in individuals with and without CKD. Other approaches to target innate immune pathways are now under investigation for their ability to reduce cardiovascular events and slow disease progression among patients with atherosclerosis and stage 3 and 4 CKD. This Review summarizes current understanding of the role of inflammation in the pathogenesis of CKD and its associated CVD, and how this knowledge may translate into novel therapeutics.
Collapse
|
234
|
Biomarkers for Non-Invasive Stratification of Coronary Artery Disease and Prognostic Impact on Long-Term Survival in Patients with Stable Coronary Heart Disease. Nutrients 2022; 14:nu14163433. [PMID: 36014939 PMCID: PMC9413764 DOI: 10.3390/nu14163433] [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: 06/14/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Knowledge about cardiac and inflammatory biomarkers in patients with stable coronary artery disease (CAD) is limited. To address this, we analyzed 3072 patients (36% female) with a median follow-up of 10 years in the Leipzig LIFE Heart Study with suspected CAD with coronary angiography. Selected biomarkers included troponin T (hsTNT), N-terminal pro B-type natriuretic peptide (NT-proBNP), copeptin, C-reactive protein (hsCRP), and interleukin-6 (IL-6). Patients were stratified by CAD severity: CAD0 (no sclerosis), CAD1 (non-obstructive, i.e., stenosis < 50%), and CAD2 (≥one stenosis ≥ 50%). Group comparison (GC) included GC1: CAD0 + 1 vs. CAD2; GC2: CAD0 vs. CAD1 + 2. CAD0, CAD1, and CAD2 were apparent in 1271, 631, and 1170 patients, respectively. Adjusted for classical risk factors, hs-cTnT, NT-proBNP, and IL-6 differed significantly in both GC and hsCRP only in GC2. After multivariate analysis, hs-cTnT, NT-proBNP, and IL-6 remained significant in GC1. In GC2, hs-cTnT (p < 0.001) and copeptin (p = 0.014) reached significance. Ten-year survival in groups CAD0, CAD1, and CAD2 was 88.3%, 77.3%, and 72.4%. Incorporation of hs-cTnT, NT-proBNP, copeptin, and IL-6 improved risk prediction (p < 0.001). The studied cardiac and inflammatory biomarkers enable fast and precise non-invasive identification of mortality risk in CAD patients, allowing the tailoring of primary and secondary CAD prevention.
Collapse
|
235
|
Georgakis MK, Malik R, Richardson TG, Howson JMM, Anderson CD, Burgess S, Hovingh GK, Dichgans M, Gill D. Associations of genetically predicted IL-6 signaling with cardiovascular disease risk across population subgroups. BMC Med 2022; 20:245. [PMID: 35948913 PMCID: PMC9367072 DOI: 10.1186/s12916-022-02446-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/20/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Interleukin 6 (IL-6) signaling is being investigated as a therapeutic target for atherosclerotic cardiovascular disease (CVD). While changes in circulating high-sensitivity C-reactive protein (hsCRP) are used as a marker of IL-6 signaling, it is not known whether there is effect heterogeneity in relation to baseline hsCRP levels or other cardiovascular risk factors. The aim of this study was to explore the association of genetically predicted IL-6 signaling with CVD risk across populations stratified by baseline hsCRP levels and cardiovascular risk factors. METHODS Among 397,060 White British UK Biobank participants without known CVD at baseline, we calculated a genetic risk score for IL-6 receptor (IL-6R)-mediated signaling, composed of 26 variants at the IL6R gene locus. We then applied linear and non-linear Mendelian randomization analyses exploring associations with a combined endpoint of incident coronary artery disease, ischemic stroke, peripheral artery disease, aortic aneurysm, and cardiovascular death stratifying by baseline hsCRP levels and cardiovascular risk factors. RESULTS The study participants (median age 59 years, 53.9% females) were followed-up for a median of 8.8 years, over which time a total of 46,033 incident cardiovascular events occurred. Genetically predicted IL-6R-mediated signaling activity was associated with higher CVD risk (hazard ratio per 1-mg/dL increment in absolute hsCRP levels: 1.11, 95% CI: 1.06-1.17). The increase in CVD risk was linearly related to baseline absolute hsCRP levels. There was no evidence of heterogeneity in the association of genetically predicted IL-6R-mediated signaling with CVD risk when stratifying the population by sex, age, body mass index, estimated glomerular filtration rate, or systolic blood pressure, but there was evidence of greater associations in individuals with low-density lipoprotein cholesterol ≥ 160 mg/dL. CONCLUSIONS Any benefit of inhibiting IL-6 signaling for CVD risk reduction is likely to be proportional to absolute reductions in hsCRP levels. Therapeutic inhibition of IL-6 signaling for CVD risk reduction should therefore prioritize those individuals with the highest baseline levels of hsCRP.
Collapse
Affiliation(s)
- Marios K Georgakis
- Center for Genomic Medicine, Massachusetts General Hospital, Richard B. Simches Research Center, 185 Cambridge Street, CPZN 6818, Boston, MA, 02114, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, MA, USA.
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | | | | | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Richard B. Simches Research Center, 185 Cambridge Street, CPZN 6818, Boston, MA, 02114, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Stephen Burgess
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Global Chief Medical Office, Novo Nordisk, Copenhagen, Denmark
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Dipender Gill
- Genetics Department, Novo Nordisk Research Centre, Oxford, UK.
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK.
- Department of Epidemiology and Biostatistics, School of Public Health, Medical School Building, St Mary's Hospital, Imperial College London, London, W2 1PG, UK.
- Clinical Pharmacology and Therapeutics Section, Institute for Infection and Immunity, St George's, University of London, London, UK.
| |
Collapse
|
236
|
Neutrophils and Neutrophil Extracellular Traps in Cardiovascular Disease: An Overview and Potential Therapeutic Approaches. Biomedicines 2022; 10:biomedicines10081850. [PMID: 36009397 PMCID: PMC9405087 DOI: 10.3390/biomedicines10081850] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/18/2022] [Accepted: 07/23/2022] [Indexed: 11/17/2022] Open
Abstract
Recent advances in pharmacotherapy have markedly improved the prognosis of cardiovascular disease (CVD) but have not completely conquered it. Therapies targeting the NOD-like receptor family pyrin domain containing 3 inflammasome and its downstream cytokines have proven effective in the secondary prevention of cardiovascular events, suggesting that inflammation is a target for treating residual risk in CVD. Neutrophil-induced inflammation has long been recognized as important in the pathogenesis of CVD. Circadian rhythm-related and disease-specific microenvironment changes give rise to neutrophil diversity. Neutrophils are primed by various stimuli, such as chemokines, cytokines, and damage-related molecular patterns, and the activated neutrophils contribute to the inflammatory response in CVD through degranulation, phagocytosis, reactive oxygen species generation, and the release of neutrophil extracellular traps (NETs). In particular, NETs promote immunothrombosis through the interaction with vascular endothelial cells and platelets and are implicated in the development of various types of CVD, such as acute coronary syndrome, deep vein thrombosis, and heart failure. NETs are promising candidates for anti-inflammatory therapy in CVD, and their efficacy has already been demonstrated in various animal models of the disease; however, they have yet to be clinically applied in humans. This narrative review discusses the diversity and complexity of neutrophils in the trajectory of CVD, the therapeutic potential of targeting NETs, and the related clinical issues.
Collapse
|
237
|
Matsumoto K, Suzuki K, Yoshida H, Magi M, Kaneko Y, Takeuchi T. Longitudinal monitoring of circulating immune cell phenotypes in large vessel vasculitis. Autoimmun Rev 2022; 21:103160. [PMID: 35926769 DOI: 10.1016/j.autrev.2022.103160] [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: 07/17/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022]
Abstract
Giant cell arteritis (GCA) and Takayasu arteritis (TAK) are two types of primary large vessel vasculitis (LVV). LVV is an intractable, rare disease with a high relapse rate. Disease progression in asymptomatic patients is an important issue in the clinical management of LVV. Useful biomarkers associated with clinical phenotypes, disease activity, and prognosis may be present in peripheral blood. In this review, we focused on peripheral leukocyte counts, surface markers, functions, and gene expression in LVV patients. In particular, we explored longitudinal changes in circulating immune cell phenotypes during the active phase of the disease and during treatment. The numbers and phenotypes of leukocytes in the peripheral blood were different between LVV and healthy controls, GCA and TAK, LVV in active versus treatment phases, and LVV in treatment responders versus non-responders. Therefore, biomarkers obtained from peripheral blood immune cells may be useful for longitudinal monitoring of disease activity in LVV.
Collapse
Affiliation(s)
- Kotaro Matsumoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | - Mayu Magi
- Chugai Pharmaceutical Co. Ltd., Kanagawa, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
238
|
Matz I, Pappritz K, Springer J, Van Linthout S. Left ventricle- and skeletal muscle-derived fibroblasts exhibit a differential inflammatory and metabolic responsiveness to interleukin-6. Front Immunol 2022; 13:947267. [PMID: 35967380 PMCID: PMC9366145 DOI: 10.3389/fimmu.2022.947267] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Interleukin-6 (IL-6) is an important player in chronic inflammation associated with heart failure and tumor-induced cachexia. Fibroblasts are salient mediators of both inflammation and fibrosis. Whereas the general outcome of IL-6 on the heart’s function and muscle wasting has been intensively studied, the influence of IL-6 on fibroblasts of the heart and skeletal muscle (SM) has not been analyzed so far. We illustrate that SM-derived fibroblasts exhibit higher basal mRNA expression of α-SMA, extracellular matrix molecules (collagen1a1/3a1/5a1), and chemokines (CCL2, CCL7, and CX3CL1) as compared to the left ventricle (LV)-derived fibroblasts. IL-6 drives the transdifferentiation of fibroblasts into myofibroblasts as indicated by an increase in α-SMA expression and upregulates NLRP3 inflammasome activity in both LV- and SM-derived fibroblasts. IL-6 increases the release of CCL7 to CX3CL1 in the supernatant of SM-derived fibroblasts associated with the attraction of more pro(Ly6Chi) versus anti(Ly6Clo) inflammatory monocytes as compared to unstimulated fibroblasts. IL-6-stimulated LV-derived fibroblasts attract less Ly6Chi to Ly6Clo monocytes compared to IL-6-stimulated SM-derived fibroblasts. In addition, SM-derived fibroblasts have a higher mitochondrial energy turnover and lower glycolytic activity versus LV-derived fibroblasts under basal and IL-6 conditions. In conclusion, IL-6 modulates the inflammatory and metabolic phenotype of LV- and SM-originated fibroblasts.
Collapse
Affiliation(s)
- Isabell Matz
- Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Kathleen Pappritz
- Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Jochen Springer
- Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- *Correspondence: Sophie Van Linthout,
| |
Collapse
|
239
|
Li T, Peng J, Li Q, Shu Y, Zhu P, Hao L. The Mechanism and Role of ADAMTS Protein Family in Osteoarthritis. Biomolecules 2022; 12:biom12070959. [PMID: 35883515 PMCID: PMC9313267 DOI: 10.3390/biom12070959] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 01/27/2023] Open
Abstract
Osteoarthritis (OA) is a principal cause of aches and disability worldwide. It is characterized by the inflammation of the bone leading to degeneration and loss of cartilage function. Factors, including diet, age, and obesity, impact and/or lead to osteoarthritis. In the past few years, OA has received considerable scholarly attention owing to its increasing prevalence, resulting in a cumbersome burden. At present, most of the interventions only relieve short-term symptoms, and some treatments and drugs can aggravate the disease in the long run. There is a pressing need to address the safety problems due to osteoarthritis. A disintegrin-like and metalloprotease domain with thrombospondin type 1 repeats (ADAMTS) metalloproteinase is a kind of secretory zinc endopeptidase, comprising 19 kinds of zinc endopeptidases. ADAMTS has been implicated in several human diseases, including OA. For example, aggrecanases, ADAMTS-4 and ADAMTS-5, participate in the cleavage of aggrecan in the extracellular matrix (ECM); ADAMTS-7 and ADAMTS-12 participate in the fission of Cartilage Oligomeric Matrix Protein (COMP) into COMP lyase, and ADAMTS-2, ADAMTS-3, and ADAMTS-14 promote the formation of collagen fibers. In this article, we principally review the role of ADAMTS metalloproteinases in osteoarthritis. From three different dimensions, we explain how ADAMTS participates in all the following aspects of osteoarthritis: ECM, cartilage degeneration, and synovial inflammation. Thus, ADAMTS may be a potential therapeutic target in osteoarthritis, and this article may render a theoretical basis for the study of new therapeutic methods for osteoarthritis.
Collapse
Affiliation(s)
- Ting Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330000, China; (T.L.); (J.P.); (Q.L.); (Y.S.); (P.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330000, China
| | - Jie Peng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330000, China; (T.L.); (J.P.); (Q.L.); (Y.S.); (P.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330000, China
| | - Qingqing Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330000, China; (T.L.); (J.P.); (Q.L.); (Y.S.); (P.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330000, China
| | - Yuan Shu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330000, China; (T.L.); (J.P.); (Q.L.); (Y.S.); (P.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330000, China
| | - Peijun Zhu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330000, China; (T.L.); (J.P.); (Q.L.); (Y.S.); (P.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330000, China
| | - Liang Hao
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330000, China; (T.L.); (J.P.); (Q.L.); (Y.S.); (P.Z.)
- Correspondence: ; Tel.: +86-13607008562; Fax: +86-86415785
| |
Collapse
|
240
|
Lee EK, Koh EM, Kim YN, Song J, Song CH, Jung KJ. Immunomodulatory Effect of Hispolon on LPS-Induced RAW264.7 Cells and Mitogen/Alloantigen-Stimulated Spleen Lymphocytes of Mice. Pharmaceutics 2022; 14:pharmaceutics14071423. [PMID: 35890318 PMCID: PMC9322787 DOI: 10.3390/pharmaceutics14071423] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
Hispolon is a potent anticancer, anti-inflammatory, antioxidant, and antidiabetic agent isolated from Phellinus linteus, an oriental medicinal mushroom. However, the immunomodulatory mechanisms by which hispolon affects macrophages and lymphocytes remain poorly characterized. We investigated the immunomodulatory effects of hispolon on oxidative stress, inflammatory responses, and lymphocyte proliferation using lipopolysaccharide (LPS)-treated RAW264.7 macrophages or mitogen/alloantigen-treated mouse splenocytes. Hispolon inhibited LPS-induced reactive oxygen and nitrogen species (ROS/RNS) generation and decreased total sulfhydryl (SH) levels in a cell-free system and RAW264.7 cells. Hispolon exerted significant anti-inflammatory effects by inhibiting production of the proinflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) and activation of nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) in LPS-treated RAW264.7 cells. Hispolon also modulated NF-κB and STAT3 activation by suppressing the NF-κB p65 interaction with phospho-IκBα and the STAT3 interaction with JAK1, as determined via coimmunoprecipitation analysis. Additionally, hispolon significantly decreased lymphocyte proliferation, T cell responses and T helper type 1 (Th1)/type 2 (Th2) cytokines production in mitogen/alloantigen-treated splenocytes. We conclude that hispolon exerts immunomodulatory effects on LPS-treated macrophages or mitogen/alloantigen-treated splenocytes through antioxidant, anti-inflammatory, and antiproliferative activities. Thus, hispolon may be a therapeutic agent for treating immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Eun Kyeong Lee
- Immunotoxicology Research Group, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea; (E.K.L.); (E.M.K.); (Y.N.K.); (C.H.S.)
| | - Eun Mi Koh
- Immunotoxicology Research Group, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea; (E.K.L.); (E.M.K.); (Y.N.K.); (C.H.S.)
| | - Yu Na Kim
- Immunotoxicology Research Group, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea; (E.K.L.); (E.M.K.); (Y.N.K.); (C.H.S.)
| | - Jeongah Song
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup 56212, Korea;
| | - Chi Hun Song
- Immunotoxicology Research Group, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea; (E.K.L.); (E.M.K.); (Y.N.K.); (C.H.S.)
| | - Kyung Jin Jung
- Immunotoxicology Research Group, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea; (E.K.L.); (E.M.K.); (Y.N.K.); (C.H.S.)
- Correspondence: ; Tel.: +82-42-610-8279; Fax: +82-42-610-8099
| |
Collapse
|
241
|
He L, Zhang CL, Chen Q, Wang L, Huang Y. Endothelial shear stress signal transduction and atherogenesis: From mechanisms to therapeutics. Pharmacol Ther 2022; 235:108152. [PMID: 35122834 DOI: 10.1016/j.pharmthera.2022.108152] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/13/2022] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
Atherosclerotic vascular disease and its complications are among the top causes of mortality worldwide. In the vascular lumen, atherosclerotic plaques are not randomly distributed. Instead, they are preferentially localized at the curvature and bifurcations along the arterial tree, where shear stress is low or disturbed. Numerous studies demonstrate that endothelial cell phenotypic change (e.g., inflammation, oxidative stress, endoplasmic reticulum stress, apoptosis, autophagy, endothelial-mesenchymal transition, endothelial permeability, epigenetic regulation, and endothelial metabolic adaptation) induced by oscillatory shear force play a fundamental role in the initiation and progression of atherosclerosis. Mechano-sensors, adaptor proteins, kinases, and transcriptional factors work closely at different layers to transduce the shear stress force from the plasma membrane to the nucleus in endothelial cells, thereby controlling the expression of genes that determine cell fate and phenotype. An in-depth understanding of these mechano-sensitive signaling cascades shall provide new translational strategies for therapeutic intervention of atherosclerotic vascular disease. This review updates the recent advances in endothelial mechano-transduction and its role in the pathogenesis of atherosclerosis, and highlights the perspective of new anti-atherosclerosis therapies through targeting these mechano-regulated signaling molecules.
Collapse
Affiliation(s)
- Lei He
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Cheng-Lin Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Qinghua Chen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
242
|
Schwartz DM, Parel P, Li H, Sorokin AV, Berg AR, Chen M, Dey A, Hong CG, Playford M, Sylvester M, Teague H, Siegel E, Mehta NN. PET/CT-Based Characterization of 18F-FDG Uptake in Various Tissues Reveals Novel Potential Contributions to Coronary Artery Disease in Psoriatic Arthritis. Front Immunol 2022; 13:909760. [PMID: 35720288 PMCID: PMC9201918 DOI: 10.3389/fimmu.2022.909760] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 11/30/2022] Open
Abstract
Background and Objectives Psoriasis is a heterogeneous inflammatory disease that involves the skin, joints, liver, heart, and other organs. Psoriatic arthritis (PsA) is associated with cardiovascular disease (CVD), but the relative contributions of inflammatory and metabolic dysregulation to CVD are incompletely understood. We set out to discover novel potential contributors to CVD in PsA patients by comprehensively phenotyping a cohort of PsA patients using these advanced technologies. Methods In this cross-sectional analysis of a cohort study, we investigated associations of systemic inflammation and metabolic dysregulation with Coronary CT angiography (CCTA)-proven coronary artery disease (CAD) in 39 subjects with PsA. We measured traditional CVD risk factors [blood pressure, Body Mass Index (BMI), diabetes, age, sex, smoking], serum markers of systemic inflammation (hsCRP, GlycA) and metabolic dysfunction (cholesterol efflux capacity), and inflammatory cytokines (IL-1β, IL-6, IL-12/IL-23, IL-17A, TNF-α, IFN-γ). We also incorporated radiographic measures of metabolic dysfunction (visceral and subcutaneous adipose volume) and tissue-specific inflammation (positron emission tomography-computed tomography, PET-CT). To quantify relative contributions of FDG (fluorodeoxyglucose) uptake and adiposity to coronary plaque, we performed multiple linear regression, controlling for Framingham risk score (FRS) and FRS + visceral adiposity. Results Compared with non-psoriatic volunteers, subjects with PsA had elevated markers of metabolic and inflammatory disease, which was more pronounced in subjects with moderate-to-severe skin disease. This included visceral (p = 0.005) and subcutaneous (p = 0.004) adiposity, BMI (p = 0.001), hemoglobin A1C (p = 0.037), high sensitivity C-reactive protein (p = 0.005), IL-6 (p = 0.003), IFN-γ (p = 0.006), and liver FDG uptake (p = 0.03). In subjects with PsA, visceral adiposity correlated significantly with subclinical CAD (standardized β = 0.681, p = 0.002), as did FDG uptake in bone marrow (standardized β = 0.488, p = 0.008), liver (standardized β = 0.619, p < 0.001), spleen (standardized β = 0.523, p = 0.004), and subcutaneous adipose (standardized β = 0.524, p = 0.003). Interpretation Together, these findings reveal inflammatory and metabolic potential contributors to subclinical CAD in PsA, including adipose inflammation, and suggesting novel targets for CVD prevention and treatment in PsA.
Collapse
Affiliation(s)
- Daniella M Schwartz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Philip Parel
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Haiou Li
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Alexander V Sorokin
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Alexander R Berg
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Marcus Chen
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Amit Dey
- Department of Internal Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Christin G Hong
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Martin Playford
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - McKella Sylvester
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Heather Teague
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Evan Siegel
- Arthritis and Rheumatism Associates, Wheaton, MD, United States
| | - Nehal N Mehta
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
243
|
Kamtchum-Tatuene J, Saba L, Heldner MR, Poorthuis MHF, Borst GJD, Rundek T, Kakkos SK, Chaturvedi S, Topakian R, Polak JF, Jickling GC. Interleukin-6 Predicts Carotid Plaque Severity, Vulnerability, and Progression. Circ Res 2022; 131:e22-e33. [PMID: 35713008 DOI: 10.1161/circresaha.122.320877] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND IL-6 (interleukin-6) has important roles in atherosclerosis pathophysiology. To determine if anti-IL-6 therapy warrants evaluation as an adjuvant stroke prevention strategy in patients with carotid atherosclerosis, we tested whether circulating IL-6 levels predict carotid plaque severity, vulnerability, and progression in the prospective population-based CHS (Cardiovascular Health Study). METHODS Duplex carotid ultrasound was performed at baseline and 5 years. Baseline plaque severity was scored 0 to 5 based on North American Symptomatic Carotid Endarterectomy Trial grade of stenosis. Plaque vulnerability at baseline was the presence of markedly irregular, ulcerated, or echolucent plaques. Plaque progression at 5 years was a ≥1 point increase in stenosis severity. The relationship of baseline plasma IL-6 levels with plaque characteristics was modeled using multivariable linear (severity) or logistic (vulnerability and progression) regression. Risk factors of atherosclerosis were included as independent variables. Stepwise backward elimination was used with P>0.05 for variable removal. To assess model stability, we computed the E-value or minimum strength of association (odds ratio scale) that unmeasured confounders must have with log IL-6 and the outcome to suppress the association. We performed internal validation with 100 bootstrap samples. RESULTS There were 4334 participants with complete data (58.9% women, mean age: 72.7±5.1 years), including 1267 (29.2%) with vulnerable plaque and 1474 (34.0%) with plaque progression. Log IL-6 predicted plaque severity (β=0.09, P=1.3×10-3), vulnerability (OR, 1.21 [95% CI, 1.05-1.40]; P=7.4×10-3, E-value=1.71), and progression (OR, 1.44 [95% CI, 1.23-1.69], P=9.1×10-6, E-value 2.24). In participants with >50% predicted probability of progression, mean log IL-6 was 0.54 corresponding to 2.0 pg/mL. Dichotomizing IL-6 levels did not affect the performance of prediction models. CONCLUSIONS Circulating IL-6 predicts carotid plaque severity, vulnerability, and progression. The 2.0 pg/mL cutoff could facilitate the selection of individuals that would benefit from anti-IL-6 drugs for stroke prevention.
Collapse
Affiliation(s)
- Joseph Kamtchum-Tatuene
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada. (J.K.-T.)
| | - Luca Saba
- Department of Radiology, University of Cagliari, Italy (L.S.)
| | - Mirjam R Heldner
- Department of Neurology, University Hospital Bern, Switzerland (M.R.H.)
| | - Michiel H F Poorthuis
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, the Netherlands. (M.H.F.P.)
| | - Gert J de Borst
- Department of Vascular Surgery, University Medical Center Utrecht, the Netherlands. (G.J.d.B.)
| | | | - Stavros K Kakkos
- Department of Vascular Surgery, University of Patras Medical School, Greece (S.K.K.)
| | | | - Raffi Topakian
- Department of Neurology, University of Miami Miller School of Medicine (T.R.).,Academic Teaching Hospital Wels-Grieskirchen, Austria (R.T.)
| | - Joseph F Polak
- Department of Radiology, Tufts University School of Medicine and Boston University School of Medicine (J.F.P.)
| | - Glen C Jickling
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada. (G.C.J.)
| | | |
Collapse
|
244
|
Kataoka Y, Funabashi S, Doi T, Harada-Shiba M. How Can We Identify Very High-Risk Heterozygous Familial Hypercholesterolemia? J Atheroscler Thromb 2022; 29:795-807. [PMID: 35022364 PMCID: PMC9174089 DOI: 10.5551/jat.rv17063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 11/11/2022] Open
Abstract
Heterozygous familial hypercholesterolemia (HeFH) is a genetic disorder that elevates low-density lipoprotein cholesterol and increases the risk of premature atherosclerotic cardiovascular disease (ASCVD). However, despite their atherogenic lipid profiles, the cardiovascular risk of HeFH varies in each individual. Their variety of phenotypic features suggests the need for better risk stratification to optimize their therapeutic management. The current review summarizes three potential approaches, including (1) definition of familial hypercholesterolemia (FH)-related risk scores, (2) genetic analysis, and (3) biomarkers. The International Atherosclerosis Society has recently proposed a definition of severe FH to identify very high-risk HeFH subjects according to their clinical characteristics. Furthermore, published studies have shown the association of FH-related genetic phenotypes with ASCVD, which indicates the genetic analysis's potential to evaluate individual cardiovascular risks. Biomarkers reflecting disease activity have been considered to predict the formation of atherosclerosis and the occurrence of ASCVD in HeFH subjects. Incorporating these risk stratifications will be expected to allocate adequate intensity of lipid-lowering therapies in HeFH subjects, which ultimately improves cardiovascular outcomes.
Collapse
Affiliation(s)
- Yu Kataoka
- Department of Cardiovascular Medicine, National Cerebral & Cardiovascular Centre, Osaka, Japan
| | - Sayaka Funabashi
- Department of Cardiology, Kyorin University School of Medicine, Tokyo, Japan
| | - Takahito Doi
- Department of Cardiovascular Medicine, National Cerebral & Cardiovascular Centre, Osaka, Japan
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral & Cardiovascular Centre, Osaka, Japan
| |
Collapse
|
245
|
Rade JJ, Barton BA, Vasan RS, Kronsberg SS, Xanthakis V, Keaney JF, Hamburg NM, Kakouros N, Kickler TA. Association of Thromboxane Generation With Survival in Aspirin Users and Nonusers. J Am Coll Cardiol 2022; 80:233-250. [PMID: 35660296 DOI: 10.1016/j.jacc.2022.04.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Persistent systemic thromboxane generation, predominantly from nonplatelet sources, in aspirin (ASA) users with cardiovascular disease (CVD) is a mortality risk factor. OBJECTIVES This study sought to determine the mortality risk associated with systemic thromboxane generation in an unselected population irrespective of ASA use. METHODS Stable thromboxane B2 metabolites (TXB2-M) were measured by enzyme-linked immunosorbent assay in banked urine from 3,044 participants (mean age 66 ± 9 years, 53.8% women) in the Framingham Heart Study. The association of TXB2-M to survival over a median observation period of 11.9 years (IQR: 10.6-12.7 years) was determined by multivariable modeling. RESULTS In 1,363 (44.8%) participants taking ASA at the index examination, median TXB2-M were lower than in ASA nonusers (1,147 pg/mg creatinine vs 4,179 pg/mg creatinine; P < 0.0001). TXB2-M were significantly associated with all-cause and cardiovascular mortality irrespective of ASA use (HR: 1.96 and 2.41, respectively; P < 0.0001 for both) for TXB2-M in the highest quartile based on ASA use compared with lower quartiles, and remained significant after adjustment for mortality risk factors for similarly aged individuals (HR: 1.49 and 1.82, respectively; P ≤ 0.005 for both). In 2,353 participants without CVD, TXB2-M were associated with cardiovascular mortality in ASA nonusers (adjusted HR: 3.04; 95% CI: 1.29-7.16) but not in ASA users, while ASA use was associated with all-cause mortality in those with low (adjusted HR: 1.46; 95% CI: 1.14-1.87) but not elevated TXB2-M. CONCLUSIONS Systemic thromboxane generation is an independent risk factor for all-cause and cardiovascular mortality irrespective of ASA use, and its measurement may be useful for therapy modification, particularly in those without CVD.
Collapse
Affiliation(s)
- Jeffrey J Rade
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
| | - Bruce A Barton
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | - Shari S Kronsberg
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | - John F Keaney
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Boston University School of Medicine, Boston, Massachusetts, USA
| | - Naomi M Hamburg
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Nikolaos Kakouros
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
246
|
Hengel FE, Benitah JP, Wenzel UO. Mosaic theory revised: inflammation and salt play central roles in arterial hypertension. Cell Mol Immunol 2022; 19:561-576. [PMID: 35354938 PMCID: PMC9061754 DOI: 10.1038/s41423-022-00851-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
The mosaic theory of hypertension was advocated by Irvine Page ~80 years ago and suggested that hypertension resulted from the close interactions of different causes. Increasing evidence indicates that hypertension and hypertensive end-organ damage are not only mediated by the proposed mechanisms that result in hemodynamic injury. Inflammation plays an important role in the pathophysiology and contributes to the deleterious consequences of arterial hypertension. Sodium intake is indispensable for normal body function but can be detrimental when it exceeds dietary requirements. Recent data show that sodium levels also modulate the function of monocytes/macrophages, dendritic cells, and different T-cell subsets. Some of these effects are mediated by changes in the microbiome and metabolome due to high-salt intake. The purpose of this review is to propose a revised and extended version of the mosaic theory by summarizing and integrating recent advances in salt, immunity, and hypertension research. Salt and inflammation are placed in the middle of the mosaic because both factors influence each of the remaining pieces.
Collapse
|
247
|
Guo J, Zhang Y, Liu T, Levy BD, Libby P, Shi GP. Allergic asthma is a risk factor for human cardiovascular diseases. NATURE CARDIOVASCULAR RESEARCH 2022; 1:417-430. [PMID: 39195946 DOI: 10.1038/s44161-022-00067-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 04/08/2022] [Indexed: 08/29/2024]
Abstract
Asthma is an allergic airway disease in which type 2-mediated inflammation has a pathogenic role. Cardiovascular diseases (CVDs) are type 1-dominant inflammatory diseases in which type 2 cytokines often have a protective role. However, clinical studies demonstrate that allergic asthma and associated allergies are essential risk factors for CVD, including coronary heart diseases, aortic diseases, peripheral arterial diseases, pulmonary embolism, right ventricular dysfunction, atrial fibrillation, cardiac hypertrophy and even hypertension. Mast cells, eosinophils, inflammatory cytokines and immunoglobulin (Ig)E accumulate in asthmatic lungs and in the injured heart and vasculature of patients with CVD. Clinical studies show that many anti-asthmatic therapies affect the risk of CVD. As such, allergic asthma and CVD may share common pathogenic mechanisms. Preclinical investigations indicate that anti-asthmatic drugs have therapeutic potential in certain CVDs. In this Review, we discuss how asthma and allied allergic conditions may contribute to the prevalence, incidence and progression of CVD and vice versa.
Collapse
Affiliation(s)
- Junli Guo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province & Key Laboratory of Emergency and Trauma of Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yuanyuan Zhang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province & Key Laboratory of Emergency and Trauma of Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Tianxiao Liu
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bruce D Levy
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
248
|
Adam CA, Șalaru DL, Prisacariu C, Marcu DTM, Sascău RA, Stătescu C. Novel Biomarkers of Atherosclerotic Vascular Disease-Latest Insights in the Research Field. Int J Mol Sci 2022; 23:ijms23094998. [PMID: 35563387 PMCID: PMC9103799 DOI: 10.3390/ijms23094998] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 02/06/2023] Open
Abstract
The atherosclerotic vascular disease is a cardiovascular continuum in which the main role is attributed to atherosclerosis, from its appearance to its associated complications. The increasing prevalence of cardiovascular risk factors, population ageing, and burden on both the economy and the healthcare system have led to the development of new diagnostic and therapeutic strategies in the field. The better understanding or discovery of new pathophysiological mechanisms and molecules modulating various signaling pathways involved in atherosclerosis have led to the development of potential new biomarkers, with key role in early, subclinical diagnosis. The evolution of technological processes in medicine has shifted the attention of researchers from the profiling of classical risk factors to the identification of new biomarkers such as midregional pro-adrenomedullin, midkine, stromelysin-2, pentraxin 3, inflammasomes, or endothelial cell-derived extracellular vesicles. These molecules are seen as future therapeutic targets associated with decreased morbidity and mortality through early diagnosis of atherosclerotic lesions and future research directions.
Collapse
Affiliation(s)
- Cristina Andreea Adam
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iași, Romania; (C.A.A.); (C.P.); (R.A.S.); (C.S.)
| | - Delia Lidia Șalaru
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iași, Romania; (C.A.A.); (C.P.); (R.A.S.); (C.S.)
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania;
- Correspondence:
| | - Cristina Prisacariu
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iași, Romania; (C.A.A.); (C.P.); (R.A.S.); (C.S.)
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania;
| | - Dragoș Traian Marius Marcu
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania;
| | - Radu Andy Sascău
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iași, Romania; (C.A.A.); (C.P.); (R.A.S.); (C.S.)
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania;
| | - Cristian Stătescu
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iași, Romania; (C.A.A.); (C.P.); (R.A.S.); (C.S.)
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania;
| |
Collapse
|
249
|
Wang P, Yuan D, Zhang C, Zhu P, Jia S, Song Y, Tang X, Xu J, Li T, Zeng G, Zhao X, Yang Y, Xu B, Gao R, Yuan J. High fibrinogen-to-albumin ratio with type 2 diabetes mellitus is associated with poor prognosis in patients undergoing percutaneous coronary intervention: 5-year findings from a large cohort. Cardiovasc Diabetol 2022; 21:46. [PMID: 35313877 PMCID: PMC8939137 DOI: 10.1186/s12933-022-01477-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Inflammation plays a crucial role in coronary atherosclerosis progression, and growing evidence has demonstrated that the fibrinogen-to-albumin ratio (FAR), as a novel inflammation biomarker, is associated with the severity of coronary artery disease (CAD). However, the long-term risk of cardiovascular events remains indistinct in patients with different level of FAR and different glycemic metabolism status. This study was to assess 5-year clinical outcomes of diabetic and non-diabetic patients who underwent percutaneous coronary intervention (PCI) with different level of FAR. METHODS We consecutively enrolled 10,724 patients with CAD hospitalized for PCI and followed up for the major adverse cardiac and cerebrovascular events (MACCE) covering all-cause mortality, cardiac mortality, non-fatal myocardial infarction, non-fatal ischemic stroke, and unplanned coronary revascularization. FAR was computed using the following formula: Fibrinogen (g/L)/Albumin (g/L). According to the optimal cut-off value of FAR for MACCE prediction, patients were divided into higher level of FAR (FAR-H) and lower level of FAR (FAR-L) subgroups, and were further categorized into four groups as FAR-H with DM and non-DM, and FAR-L with DM and non-DM. RESULTS 5298 patients (58.36 ± 10.36 years, 77.7% male) were ultimately enrolled in the present study. A total of 1099 (20.7%) MACCEs were documented during the 5-year follow-up. The optimal cut-off value of FAR was 0.0783 by the surv_cutpoint function. Compared to ones with FAR-H and DM, patients with FAR-L and non-DM, FAR-H and non-DM, FAR-L and DM had decreased risk of MACCEs [adjusted hazard ratio (HR): 0.75, 95% confidence interval (CI) 0.64-0.89, P = 0.001; HR: 0.78, 95% CI 0.66-0.93, P = 0.006; HR: 0.81, 95% CI 0.68-0.97, P = 0.019; respectively]. Notably, non-diabetic patients with lower level of FAR also had lower all-cause mortality and cardiac mortality risk than those in the FAR-H/DM group (HR: 0.41, 95% CI 0.27-0.63, P < 0.001; HR: 0.30, 95% CI 0.17-0.53, P < 0.001; respectively). Multivariate Cox proportional hazards regression analysis also indicated the highest risk of MACCEs in patients with FAR-H and DM than others (P for trend = 0.005). In addition, post-hoc analysis revealed consistent effects on 5-year MACCE across various subgroups. CONCLUSION In this real-world cohort study, higher level of FAR combined with DM was associated with worse 5-year outcomes among patients with CAD undergoing PCI. The level of FAR may help to identify high-risk individuals in this specific population, where more precise risk assessment should be performed.
Collapse
Affiliation(s)
- Peizhi Wang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Deshan Yuan
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Ce Zhang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Pei Zhu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Sida Jia
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Ying Song
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Xiaofang Tang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jingjing Xu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Tianyu Li
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Guyu Zeng
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Xueyan Zhao
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yuejin Yang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Bo Xu
- Catheterization Laboratories, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runlin Gao
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jinqing Yuan
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China. .,National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
250
|
Papadopoulos A, Palaiopanos K, Björkbacka H, Peters A, de Lemos JA, Seshadri S, Dichgans M, Georgakis MK. Circulating Interleukin-6 Levels and Incident Ischemic Stroke: A Systematic Review and Meta-analysis of Prospective Studies. Neurology 2022; 98:e1002-e1012. [PMID: 34969940 PMCID: PMC8967391 DOI: 10.1212/wnl.0000000000013274] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 12/21/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Human genetic studies support a key role of interleukin-6 (IL-6) in the pathogenesis of ischemic stroke. However, there are only limited data from observational studies exploring circulating IL-6 levels as a risk factor for ischemic stroke. We set out to perform a systematic review and meta-analysis of aggregate data on cohort studies to determine the magnitude and shape of the association between circulating IL-6 levels and risk of incident ischemic stroke in the general population. METHODS Following the PRISMA guidelines, we systematically screened the PubMed search engine from inception to March 2021 for population-based prospective cohort studies exploring the association between circulating IL-6 levels and risk of incident ischemic stroke. We pooled association estimates for ischemic stroke risk with random-effects models and explored nonlinear effects in dose-response meta-analyses. Risk of bias was assessed with the Newcastle-Ottawa Scale (NOS). We used funnel plots and trim-to-fill analyses to assess publication bias. RESULTS We identified 11 studies (n = 27,411 individuals; 2,669 stroke events) meeting our eligibility criteria. Mean age of all included participants was 60.5 years and 54.8% were female. Overall, quality of the included studies was high (median 8 out of 9 NOS points, interquartile range 7-9). In meta-analyses, 1 SD increment in circulating log-transformed IL-6 levels was associated with a 19% increase in risk of incident ischemic stroke over a mean follow-up of 12.4 years (relative risk 1.19; 95% confidence interval 1.10 to 1.28). A dose-response meta-analysis showed a linear association between circulating IL-6 levels and ischemic stroke risk. There was only moderate heterogeneity and the results were consistent in sensitivity analyses restricted to studies of low risk of bias and studies fully adjusting for demographic and vascular risk factors. The results also remained stable following adjustment for publication bias. DISCUSSION Higher circulating IL-6 levels in community-dwelling individuals are associated with higher long-term risk of incident ischemic stroke in a linear pattern and independently of conventional vascular risk factors. Along with findings from genetic studies and clinical trials, these results provide additional support for a key role of IL-6 signaling in ischemic stroke.
Collapse
Affiliation(s)
- Andreas Papadopoulos
- From the Department of Radiology (A.P.), 401 General Military Hospital of Athens; National Public Health Organization (K.P.), Athens, Greece; Department of Clinical Sciences Malmö (H.B.), Lund University, Sweden; Institute of Epidemiology (A.P.), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg; German Center for Diabetes Research (DZD) (A.P.), München-Neuherberg, Neuherberg; German Research Center for Cardiovascular Disease (DZHK) (A.P.), Partner Site Munich Heart Alliance; Institute of Medical Information Sciences, Biometry and Epidemiology (A.P.), and Institute for Stroke and Dementia Research, University Hospital (M.D., M.K.G.), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology (J.A.d.L.), University of Texas Southwestern Medical Center, Dallas; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study (S.S.), Framingham; Department of Medicine (S.S.), Boston University School of Medicine, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio; Munich Cluster for Systems Neurology (SyNergy) (M.D.); and German Centre for Neurodegenerative Diseases (DZNE) (M.D.), Munich, Germany. M.K.G. is currently at the Center for Genomic Medicine, Massachusetts General Hospital, Boston and the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Konstantinos Palaiopanos
- From the Department of Radiology (A.P.), 401 General Military Hospital of Athens; National Public Health Organization (K.P.), Athens, Greece; Department of Clinical Sciences Malmö (H.B.), Lund University, Sweden; Institute of Epidemiology (A.P.), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg; German Center for Diabetes Research (DZD) (A.P.), München-Neuherberg, Neuherberg; German Research Center for Cardiovascular Disease (DZHK) (A.P.), Partner Site Munich Heart Alliance; Institute of Medical Information Sciences, Biometry and Epidemiology (A.P.), and Institute for Stroke and Dementia Research, University Hospital (M.D., M.K.G.), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology (J.A.d.L.), University of Texas Southwestern Medical Center, Dallas; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study (S.S.), Framingham; Department of Medicine (S.S.), Boston University School of Medicine, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio; Munich Cluster for Systems Neurology (SyNergy) (M.D.); and German Centre for Neurodegenerative Diseases (DZNE) (M.D.), Munich, Germany. M.K.G. is currently at the Center for Genomic Medicine, Massachusetts General Hospital, Boston and the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Harry Björkbacka
- From the Department of Radiology (A.P.), 401 General Military Hospital of Athens; National Public Health Organization (K.P.), Athens, Greece; Department of Clinical Sciences Malmö (H.B.), Lund University, Sweden; Institute of Epidemiology (A.P.), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg; German Center for Diabetes Research (DZD) (A.P.), München-Neuherberg, Neuherberg; German Research Center for Cardiovascular Disease (DZHK) (A.P.), Partner Site Munich Heart Alliance; Institute of Medical Information Sciences, Biometry and Epidemiology (A.P.), and Institute for Stroke and Dementia Research, University Hospital (M.D., M.K.G.), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology (J.A.d.L.), University of Texas Southwestern Medical Center, Dallas; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study (S.S.), Framingham; Department of Medicine (S.S.), Boston University School of Medicine, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio; Munich Cluster for Systems Neurology (SyNergy) (M.D.); and German Centre for Neurodegenerative Diseases (DZNE) (M.D.), Munich, Germany. M.K.G. is currently at the Center for Genomic Medicine, Massachusetts General Hospital, Boston and the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Annette Peters
- From the Department of Radiology (A.P.), 401 General Military Hospital of Athens; National Public Health Organization (K.P.), Athens, Greece; Department of Clinical Sciences Malmö (H.B.), Lund University, Sweden; Institute of Epidemiology (A.P.), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg; German Center for Diabetes Research (DZD) (A.P.), München-Neuherberg, Neuherberg; German Research Center for Cardiovascular Disease (DZHK) (A.P.), Partner Site Munich Heart Alliance; Institute of Medical Information Sciences, Biometry and Epidemiology (A.P.), and Institute for Stroke and Dementia Research, University Hospital (M.D., M.K.G.), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology (J.A.d.L.), University of Texas Southwestern Medical Center, Dallas; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study (S.S.), Framingham; Department of Medicine (S.S.), Boston University School of Medicine, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio; Munich Cluster for Systems Neurology (SyNergy) (M.D.); and German Centre for Neurodegenerative Diseases (DZNE) (M.D.), Munich, Germany. M.K.G. is currently at the Center for Genomic Medicine, Massachusetts General Hospital, Boston and the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - James A de Lemos
- From the Department of Radiology (A.P.), 401 General Military Hospital of Athens; National Public Health Organization (K.P.), Athens, Greece; Department of Clinical Sciences Malmö (H.B.), Lund University, Sweden; Institute of Epidemiology (A.P.), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg; German Center for Diabetes Research (DZD) (A.P.), München-Neuherberg, Neuherberg; German Research Center for Cardiovascular Disease (DZHK) (A.P.), Partner Site Munich Heart Alliance; Institute of Medical Information Sciences, Biometry and Epidemiology (A.P.), and Institute for Stroke and Dementia Research, University Hospital (M.D., M.K.G.), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology (J.A.d.L.), University of Texas Southwestern Medical Center, Dallas; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study (S.S.), Framingham; Department of Medicine (S.S.), Boston University School of Medicine, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio; Munich Cluster for Systems Neurology (SyNergy) (M.D.); and German Centre for Neurodegenerative Diseases (DZNE) (M.D.), Munich, Germany. M.K.G. is currently at the Center for Genomic Medicine, Massachusetts General Hospital, Boston and the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Sudha Seshadri
- From the Department of Radiology (A.P.), 401 General Military Hospital of Athens; National Public Health Organization (K.P.), Athens, Greece; Department of Clinical Sciences Malmö (H.B.), Lund University, Sweden; Institute of Epidemiology (A.P.), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg; German Center for Diabetes Research (DZD) (A.P.), München-Neuherberg, Neuherberg; German Research Center for Cardiovascular Disease (DZHK) (A.P.), Partner Site Munich Heart Alliance; Institute of Medical Information Sciences, Biometry and Epidemiology (A.P.), and Institute for Stroke and Dementia Research, University Hospital (M.D., M.K.G.), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology (J.A.d.L.), University of Texas Southwestern Medical Center, Dallas; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study (S.S.), Framingham; Department of Medicine (S.S.), Boston University School of Medicine, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio; Munich Cluster for Systems Neurology (SyNergy) (M.D.); and German Centre for Neurodegenerative Diseases (DZNE) (M.D.), Munich, Germany. M.K.G. is currently at the Center for Genomic Medicine, Massachusetts General Hospital, Boston and the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Martin Dichgans
- From the Department of Radiology (A.P.), 401 General Military Hospital of Athens; National Public Health Organization (K.P.), Athens, Greece; Department of Clinical Sciences Malmö (H.B.), Lund University, Sweden; Institute of Epidemiology (A.P.), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg; German Center for Diabetes Research (DZD) (A.P.), München-Neuherberg, Neuherberg; German Research Center for Cardiovascular Disease (DZHK) (A.P.), Partner Site Munich Heart Alliance; Institute of Medical Information Sciences, Biometry and Epidemiology (A.P.), and Institute for Stroke and Dementia Research, University Hospital (M.D., M.K.G.), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology (J.A.d.L.), University of Texas Southwestern Medical Center, Dallas; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study (S.S.), Framingham; Department of Medicine (S.S.), Boston University School of Medicine, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio; Munich Cluster for Systems Neurology (SyNergy) (M.D.); and German Centre for Neurodegenerative Diseases (DZNE) (M.D.), Munich, Germany. M.K.G. is currently at the Center for Genomic Medicine, Massachusetts General Hospital, Boston and the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Marios K Georgakis
- From the Department of Radiology (A.P.), 401 General Military Hospital of Athens; National Public Health Organization (K.P.), Athens, Greece; Department of Clinical Sciences Malmö (H.B.), Lund University, Sweden; Institute of Epidemiology (A.P.), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg; German Center for Diabetes Research (DZD) (A.P.), München-Neuherberg, Neuherberg; German Research Center for Cardiovascular Disease (DZHK) (A.P.), Partner Site Munich Heart Alliance; Institute of Medical Information Sciences, Biometry and Epidemiology (A.P.), and Institute for Stroke and Dementia Research, University Hospital (M.D., M.K.G.), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology (J.A.d.L.), University of Texas Southwestern Medical Center, Dallas; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study (S.S.), Framingham; Department of Medicine (S.S.), Boston University School of Medicine, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio; Munich Cluster for Systems Neurology (SyNergy) (M.D.); and German Centre for Neurodegenerative Diseases (DZNE) (M.D.), Munich, Germany. M.K.G. is currently at the Center for Genomic Medicine, Massachusetts General Hospital, Boston and the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA.
| |
Collapse
|