1
|
Sharma J, Jangale V, Swain AK, Yadav P. An optimized instrument variable selection approach to improve causality estimation in association studies. Sci Rep 2024; 14:22781. [PMID: 39354059 PMCID: PMC11445377 DOI: 10.1038/s41598-024-73970-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
Mendelian randomization (MR) is an emerging tool for inferring causality in genetic epidemiology. MR studies suffer bias from weak genetic instrument variables (IVs) and horizontal pleiotropy. We introduce a robust integrative framework strictly adhering with STROBE-MR guidelines to improve causality inference through MR studies. We implemented novel t-statistics-based criteria to improve the reliability of selected IVs followed by various MR methods. Further, we include sensitivity analyses to remove horizontal-pleiotropy bias. For functional validation, we perform enrichment analysis of identified causal SNPs. We demonstrate effectiveness of our proposed approach on 5 different MR datasets selected from diverse populations. Our pipeline outperforms its counterpart MR analyses using default parameters on these datasets. Notably, we found a significant association between total cholesterol and coronary artery disease (P = 1.16 × 10-71) in a single-sample dataset using our pipeline. Contrarily, this same association was deemed ambiguous while using default parameters. Moreover, in a two-sample dataset, we uncover 13 new causal SNPs with enhanced statistical significance (P = 1.06 × 10-11) for liver-iron-content and liver-cell-carcinoma. Likewise, these SNPs remained undetected using the default parameters (P = 7.58 × 10-4). Furthermore, our analysis confirmed previously known pathways, such as hyperlipidemia in heart diseases and gene ME1 in liver cancer. In conclusion, we propose a robust and powerful framework to infer causality across diverse populations and easily adaptable to different diseases.
Collapse
Affiliation(s)
- Jyoti Sharma
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Rajasthan, 342030, India
| | - Vaishnavi Jangale
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Rajasthan, 342030, India
| | - Asish Kumar Swain
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Rajasthan, 342030, India
| | - Pankaj Yadav
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Rajasthan, 342030, India.
- School of Artificial Intelligence and Data Science, Indian Institute of Technology Jodhpur, Rajasthan, 342030, India.
| |
Collapse
|
2
|
Tumenbayar BI, Pham K, Biber JC, Drewes R, Bae Y. Transcriptomic and Multi-scale Network Analyses Reveal Key Drivers of Cardiovascular Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612437. [PMID: 39345636 PMCID: PMC11429675 DOI: 10.1101/2024.09.11.612437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cardiovascular diseases (CVDs) and pathologies are often driven by changes in molecular signaling and communication, as well as in cellular and tissue components, particularly those involving the extracellular matrix (ECM), cytoskeleton, and immune response. The fine-wire vascular injury model is commonly used to study neointimal hyperplasia and vessel stiffening, but it is not typically considered a model for CVDs. In this paper, we hypothesize that vascular injury induces changes in gene expression, molecular communication, and biological processes similar to those observed in CVDs at both the transcriptome and protein levels. To investigate this, we analyzed gene expression in microarray datasets from injured and uninjured femoral arteries in mice two weeks post-injury, identifying 1,467 significantly and differentially expressed genes involved in several CVDs such as including vaso-occlusion, arrhythmia, and atherosclerosis. We further constructed a protein-protein interaction network with seven functionally distinct clusters, with notable enrichment in ECM, metabolic processes, actin-based process, and immune response. Significant molecular communications were observed between the clusters, most prominently among those involved in ECM and cytoskeleton organizations, inflammation, and cell cycle. Machine Learning Disease pathway analysis revealed that vascular injury-induced crosstalk between ECM remodeling and immune response clusters contributed to aortic aneurysm, neovascularization of choroid, and kidney failure. Additionally, we found that interactions between ECM and actin cytoskeletal reorganization clusters were linked to cardiac damage, carotid artery occlusion, and cardiac lesions. Overall, through multi-scale bioinformatic analyses, we demonstrated the robustness of the vascular injury model in eliciting transcriptomic and molecular network changes associated with CVDs, highlighting its potential for use in cardiovascular research.
Collapse
Affiliation(s)
- Bat-Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Khanh Pham
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - John C. Biber
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Rhonda Drewes
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, NY 14260, USA
| |
Collapse
|
3
|
Maaniitty E, Sinisilta S, Jalkanen J, Vasankari T, Biancari F, Gunn J, Jalkanen S, Airaksinen KJ, Hollmén M, Kiviniemi T. Distinct circulating cytokine levels in patients with angiography-proven coronary artery disease compared to disease-free controls. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2024; 22:200307. [PMID: 39091640 PMCID: PMC11292512 DOI: 10.1016/j.ijcrp.2024.200307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Background Systemic inflammation has a critical role in the development of symptomatic coronary artery disease (CAD). Identification of inflammatory pathways may provide a platform for novel therapeutic approaches. We sought to determine whether there are differences in circulating cytokine profiles between patients with CAD and disease-free controls as well as according to the severity of the disease. Methods Case-control study's population consisted of 452 patients who underwent diagnostic invasive coronary angiography due to clinical indications. We measured the serum concentrations of 48 circulating cytokines. Extent of CAD was assessed using the SYNTAX Score in 116 patients. Cytokine differences between groups were tested using Mann-Whitney U test and associations with CAD were explored using a logistic regression model. Results Overall, 310 patients had angiographically verified CAD whereas 142 had no angiographically-detected coronary atherosclerosis. In multivariable logistic regression models adjusted for age, sex, hypertension, atrial fibrillation, history of smoking and treatment for diabetes and hyperlipidemia, increased levels of interleukin 9 (OR 1.359, 95%CI 1.046-1.766, p = 0.022), IL-17 (1.491, 95%CI 1.115-1.994, p = 0.007) and tumor necrosis factor alpha (TNF-α) (OR 1.440, 95%CI 1.089-1.904, p = 0.011) were independently associated with CAD. Patients with SYNTAX Score>22 had increased levels of stromal cell-derived factor 1 alfa (SDF-1α), beta-nerve growth factor (β-NGF), IL-3 and decreased level of IL-17 compared to those with score ≤22 when adjusted for smoking and use of beta-blockers. Conclusions Patients with CAD have distinct circulating cytokine profiles compared to disease-free controls. Distinct cytokines may have pivotal roles at different stages of coronary atherosclerosis. ClinicalTrials.gov Identifier: NCT03444259 (https://clinicaltrials.gov/study/NCT03444259).
Collapse
Affiliation(s)
- Eveliina Maaniitty
- Heart Center, Turku University Hospital and University of Turku, POB 52, FI-20521, Turku, Finland
| | - Sami Sinisilta
- Heart Center, Turku University Hospital and University of Turku, POB 52, FI-20521, Turku, Finland
| | - Juho Jalkanen
- Vascular Surgery, Turku University Hospital and University of Turku, POB 52, FI-20521, Turku, Finland
| | - Tuija Vasankari
- Heart Center, Turku University Hospital and University of Turku, POB 52, FI-20521, Turku, Finland
| | - Fausto Biancari
- Heart Center, Turku University Hospital and University of Turku, POB 52, FI-20521, Turku, Finland
- Department of Medicine, South Karelia Central Hospital, University of Helsinki, Valto Käkelän Katu 1, FI-53130, Lappeenranta, Finland
| | - Jarmo Gunn
- Heart Center, Turku University Hospital and University of Turku, POB 52, FI-20521, Turku, Finland
| | - Sirpa Jalkanen
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland
| | - K.E. Juhani Airaksinen
- Heart Center, Turku University Hospital and University of Turku, POB 52, FI-20521, Turku, Finland
| | - Maija Hollmén
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Tuomas Kiviniemi
- Heart Center, Turku University Hospital and University of Turku, POB 52, FI-20521, Turku, Finland
| |
Collapse
|
4
|
Becker SH, Ronayne CE, Bold TD, Jenkins MK. CD4 + T cells recruit, then engage macrophages in cognate interactions to clear Mycobacterium tuberculosis from the lungs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609198. [PMID: 39229103 PMCID: PMC11370583 DOI: 10.1101/2024.08.22.609198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
IFN-γ-producing CD4 + T cells are required for protection against lethal Mycobacterium tuberculosis ( Mtb ) infections. However, the ability of CD4 + T cells to suppress Mtb growth cannot be fully explained by IFN-γ or other known T cell products. In this study, we show that CD4 + T cell-derived IFN-γ promoted the recruitment of monocyte-derived macrophages (MDMs) to the lungs of Mtb -infected mice. Although the recruited MDMs became quickly and preferentially infected with Mtb , CD4 + T cells rapidly disinfected the MDMs. Clearance of Mtb from MDMs was not explained by IFN-γ, but rather by MHCII-mediated cognate interactions with CD4 + T cells. These interactions promoted MDM expression of glycolysis genes essential for Mtb control. Thus, by recruiting MDMs, CD4 + T cells initiate a cycle of bacterial phagocytosis, Mtb antigen presentation and disinfection in an attempt to clear the bacteria from the lungs.
Collapse
|
5
|
Xiang Q, Geng ZX, Yi X, Wei X, Zhu XH, Jiang DS. PANoptosis: a novel target for cardiovascular diseases. Trends Pharmacol Sci 2024; 45:739-756. [PMID: 39003157 DOI: 10.1016/j.tips.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/15/2024]
Abstract
PANoptosis is a unique innate immune inflammatory lytic cell death pathway initiated by an innate immune sensor and driven by caspases and RIPKs. As a distinct pathway, the execution of PANoptosis cannot be hindered by targeting other cell death pathways, such as pyroptosis, apoptosis, or necroptosis. Instead, targeting key PANoptosome components can serve as a strategy to prevent this form of cell death. Given the physiological relevance in several diseases, PANoptosis is a pivotal therapeutic target. Notably, previous research has primarily focused on the role of PANoptosis in cancer and infectious and inflammatory diseases. By contrast, its role in cardiovascular diseases has not been comprehensively discussed. Here, we review the available evidence on PANoptosis in cardiovascular diseases, including cardiomyopathy, atherosclerosis, myocardial infarction, myocarditis, and aortic aneurysm and dissection, and explore a variety of agents that target PANoptosis, with the overarching goal of providing a novel complementary approach to combatting cardiovascular diseases.
Collapse
Affiliation(s)
- Qi Xiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen-Xi Geng
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xue-Hai Zhu
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
6
|
Godbole S, Solomon JL, Johnson M, Srivastava A, Carsons SE, Belilos E, De Leon J, Reiss AB. Treating Cardiovascular Disease in the Inflammatory Setting of Rheumatoid Arthritis: An Ongoing Challenge. Biomedicines 2024; 12:1608. [PMID: 39062180 PMCID: PMC11275112 DOI: 10.3390/biomedicines12071608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/30/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Despite progress in treating rheumatoid arthritis, this autoimmune disorder confers an increased risk of developing cardiovascular disease (CVD). Widely used screening protocols and current clinical guidelines are inadequate for the early detection of CVD in persons with rheumatoid arthritis. Traditional CVD risk factors alone cannot be applied because they underestimate CVD risk in rheumatoid arthritis, missing the window of opportunity for prompt intervention to decrease morbidity and mortality. The lipid profile is insufficient to assess CVD risk. This review delves into the connection between systemic inflammation in rheumatoid arthritis and the premature onset of CVD. The shared inflammatory and immunologic pathways between the two diseases that result in subclinical atherosclerosis and disrupted cholesterol homeostasis are examined. The treatment armamentarium for rheumatoid arthritis is summarized, with a particular focus on each medication's cardiovascular effect, as well as the mechanism of action, risk-benefit profile, safety, and cost. A clinical approach to CVD screening and treatment for rheumatoid arthritis patients is proposed based on the available evidence. The mortality gap between rheumatoid arthritis and non-rheumatoid arthritis populations due to premature CVD represents an urgent research need in the fields of cardiology and rheumatology. Future research areas, including risk assessment tools and novel immunotherapeutic targets, are highlighted.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (J.L.S.); (M.J.); (A.S.); (S.E.C.); (E.B.); (J.D.L.)
| |
Collapse
|
7
|
Kanno Y, Toyama K, Shibata H, Matsuo O, Ozaki KI. α2-Antiplasmin is associated with macrophage activation and fibrin deposition in a macrophage activation syndrome mouse model. Clin Exp Immunol 2024; 216:272-279. [PMID: 38457368 PMCID: PMC11097911 DOI: 10.1093/cei/uxae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/04/2024] [Accepted: 03/07/2024] [Indexed: 03/10/2024] Open
Abstract
Macrophage activation syndrome (MAS) is a life-threatening condition, characterized by cytopenia, multi-organ dysfunction, and coagulopathy associated with excessive activation of macrophages. In this study, we investigated the roles of alpha2-antiplasmin (α2AP) in the progression of MAS using fulminant MAS mouse model induced by toll-like receptor-9 agonist (CpG) and D-(+)-galactosamine hydrochloride (DG). α2AP deficiency attenuated macrophage accumulation, liver injury, and fibrin deposition in the MAS model mice. Interferon-γ (IFN-γ) is associated with macrophage activation, including migration, and plays a pivotal role in MAS progression. α2AP enhanced the IFN-γ-induced migration, and tissue factor production. Additionally, we showed that fibrin-induced macrophage activation and tumor necrosis factor-α production. Moreover, the blockade of α2AP by neutralizing antibodies attenuated macrophage accumulation, liver injury, and fibrin deposition in the MAS model mice. These data suggest that α2AP may regulate IFN-γ-induced responses and be associated with macrophage activation and fibrin deposition in the MAS progression.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Molecular Pathology, Faculty of Pharmaceutical Science, Doshisha Women’s College of Liberal Arts, Kodo Kyo-tanabe, Kyoto, Japan
| | - Kinomi Toyama
- Department of Molecular Pathology, Faculty of Pharmaceutical Science, Doshisha Women’s College of Liberal Arts, Kodo Kyo-tanabe, Kyoto, Japan
| | - Haruna Shibata
- Department of Molecular Pathology, Faculty of Pharmaceutical Science, Doshisha Women’s College of Liberal Arts, Kodo Kyo-tanabe, Kyoto, Japan
| | - Osamu Matsuo
- Faculty of Medicine, Kindai University, Osaka-sayama, Japan
| | - Kei-ichi Ozaki
- Department of Molecular Pathology, Faculty of Pharmaceutical Science, Doshisha Women’s College of Liberal Arts, Kodo Kyo-tanabe, Kyoto, Japan
| |
Collapse
|
8
|
Tran DT, Batchu SN, Advani A. Interferons and interferon-related pathways in heart disease. Front Cardiovasc Med 2024; 11:1357343. [PMID: 38665231 PMCID: PMC11043610 DOI: 10.3389/fcvm.2024.1357343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Interferons (IFNs) and IFN-related pathways play key roles in the defence against microbial infection. However, these processes may also be activated during the pathogenesis of non-infectious diseases, where they may contribute to organ injury, or function in a compensatory manner. In this review, we explore the roles of IFNs and IFN-related pathways in heart disease. We consider the cardiac effects of type I IFNs and IFN-stimulated genes (ISGs); the emerging role of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway; the seemingly paradoxical effects of the type II IFN, IFN-γ; and the varied actions of the interferon regulatory factor (IRF) family of transcription factors. Recombinant IFNs and small molecule inhibitors of mediators of IFN receptor signaling are already employed in the clinic for the treatment of some autoimmune diseases, infections, and cancers. There has also been renewed interest in IFNs and IFN-related pathways because of their involvement in SARS-CoV-2 infection, and because of the relatively recent emergence of cGAS-STING as a pattern recognition receptor-activated pathway. Whether these advances will ultimately result in improvements in the care of those experiencing heart disease remains to be determined.
Collapse
Affiliation(s)
| | | | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
9
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
10
|
Yang X, Deng H, Lv J, Chen X, Zeng L, Weng J, Liang H, Xu W. Comparison of changes in adipokine and inflammatory cytokine levels in patients with newly diagnosed type 2 diabetes treated with exenatide, insulin, or pioglitazone: A post-hoc study of the CONFIDENCE trial. Heliyon 2024; 10:e23309. [PMID: 38169889 PMCID: PMC10758788 DOI: 10.1016/j.heliyon.2023.e23309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Background Adipokines and inflammatory cytokines (ADICs) play important roles in type 2 diabetes mellitus (T2DM). This study aimed to compare the changes of ADIC levels (ΔADICs) in patients with newly diagnosed T2DM treated with different antihyperglycemic agents, and further investigate the impact of these changes on metabolic indices, β-cell function and insulin resistance (IR). Methods Four hundred and sixteen patients with newly diagnosed T2DM from 25 centers in China randomly received 48-week intervention with exenatide, insulin or pioglitazone. Anthropometric and laboratory data, indices of β-cell function and IR, and levels of AIDCs, including interleukin-1 beta (IL-1β), interferon-gamma (IFN-γ), leptin, and fibroblast growth factor 21 (FGF21) were detected at baseline and the end of the study. Results In total, 281 participants (68 % male, age: 50.3 ± 9.4 years) completed the study. After 48- week treatment, IL-1β and IFN-γ were significantly decreased with exenatide treatment (P < 0.001 and P = 0.001, respectively), but increased with insulin (P = 0.009 and P = 0.026, respectively). However, pioglitazone treatment had no impact on ADICs. No significant change in leptin or FGF21 was detected with any of the treatments. After adjustment for baseline values and changes of body weight, waist and HbA1c, the between-group differences were found in ΔIL-1β (exenatide vs. insulin: P = 0.048; and exenatide vs. pioglitazone: P = 0.003, respectively) and ΔIFN-γ (exenatide vs. insulin: P = 0.049; and exenatide vs. pioglitazone: P < 0.001, respectively). Multiple linear regression analysis indicated that Δweight was associated with ΔIL-1β (β = 0.753; 95 % CI, 0.137-1.369; P = 0.017). After adjusting for treatment effects, Δweight was also be correlated with ΔFGF21 (β = 1.097; 95%CI, 0.250-1.944; P = 0.012); furthermore, ΔHOMA-IR was correlated with Δleptin (β = 0.078; 95%CI, 0.008-0.147; P = 0.029) as well. However, ΔHOMA-IR was not significantly associated with ΔIL-1β after adjusting for treatment effects (P = 0.513). Conclusion Exenatide treatment led to significant changes of inflammatory cytokines levels (IL-1β and IFN-γ), but not adipokines (leptin and FGF21), in newly diagnosed T2DM patients. The exenatide-mediated improvement in weight and IR may be associated with a decrease in inflammatory cytokine levels.
Collapse
Affiliation(s)
- Xubin Yang
- Department of Endocrinology and Metabolism, the 3rd Affiliated Hospital of Sun Yat-sen University. NO.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, 510630, China
| | - Hongrong Deng
- Department of Endocrinology and Metabolism, the 3rd Affiliated Hospital of Sun Yat-sen University. NO.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, 510630, China
| | - Jing Lv
- Department of Endocrinology and Metabolism, the 3rd Affiliated Hospital of Sun Yat-sen University. NO.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, 510630, China
| | - Xueyan Chen
- Department of Endocrinology and Metabolism, the 3rd Affiliated Hospital of Sun Yat-sen University. NO.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, 510630, China
| | - Longyi Zeng
- Department of Endocrinology and Metabolism, the 3rd Affiliated Hospital of Sun Yat-sen University. NO.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, 510630, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Hua Liang
- Department of Endocrinology and Metabolism, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Wen Xu
- Department of Endocrinology and Metabolism, the 3rd Affiliated Hospital of Sun Yat-sen University. NO.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, 510630, China
| |
Collapse
|
11
|
Ledoux B, Zanin N, Yang J, Mercier V, Coster C, Dupont-Gillain C, Alsteens D, Morsomme P, Renard HF. Plasma membrane nanodeformations promote actin polymerization through CIP4/CDC42 recruitment and regulate type II IFN signaling. SCIENCE ADVANCES 2023; 9:eade1660. [PMID: 38091386 PMCID: PMC10848735 DOI: 10.1126/sciadv.ade1660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/10/2023] [Indexed: 12/18/2023]
Abstract
In their environment, cells must cope with mechanical stresses constantly. Among these, nanoscale deformations of plasma membrane induced by substrate nanotopography are now largely accepted as a biophysical stimulus influencing cell behavior and function. However, the mechanotransduction cascades involved and their precise molecular effects on cellular physiology are still poorly understood. Here, using homemade fluorescent nanostructured cell culture surfaces, we explored the role of Bin/Amphiphysin/Rvs (BAR) domain proteins as mechanosensors of plasma membrane geometry. Our data reveal that distinct subsets of BAR proteins bind to plasma membrane deformations in a membrane curvature radius-dependent manner. Furthermore, we show that membrane curvature promotes the formation of dynamic actin structures mediated by the Rho GTPase CDC42, the F-BAR protein CIP4, and the presence of PI(4,5)P2. In addition, these actin-enriched nanodomains can serve as platforms to regulate receptor signaling as they appear to contain interferon-γ receptor (IFNγ-R) and to lead to the partial inhibition of IFNγ-induced JAK/STAT signaling.
Collapse
Affiliation(s)
- Benjamin Ledoux
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Group of Molecular Physiology, Croix du Sud 4-5 bte L7.07.14, Louvain-la-Neuve 1348, Belgium
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Croix du Sud 4-5 bte L7.07.07, Louvain-la-Neuve 1348, Belgium
- UNamur, Morph-Im platform, Rue de Bruxelles 61, Namur 5000, Belgium
| | - Natacha Zanin
- UNamur, NAmur Research Institute for LIfe Sciences, Unité de Recherche en Biologie Cellulaire animale, Rue de Bruxelles 61, Namur 5000, Belgium
| | - Jinsung Yang
- Gyeongsang National University, Department of Biochemistry, College of Medicine, Department of Convergence Medical Sciences, Institute of Medical Science, Jinju 52727, South Korea
| | - Vincent Mercier
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Charlotte Coster
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Group of Molecular Physiology, Croix du Sud 4-5 bte L7.07.14, Louvain-la-Neuve 1348, Belgium
| | - Christine Dupont-Gillain
- UCLouvain, Institute of Condensed Matter and Nanosciences, Bio- and Soft Matter, Place Louis Pasteur 1 bte L4.01.10, Louvain-la-Neuve 1348, Belgium
| | - David Alsteens
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, NanoBiophysics lab, Croix du Sud 4-5 bte L7.07.07, Louvain-la-Neuve 1348, Belgium
| | - Pierre Morsomme
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Group of Molecular Physiology, Croix du Sud 4-5 bte L7.07.14, Louvain-la-Neuve 1348, Belgium
| | - Henri-François Renard
- UNamur, Morph-Im platform, Rue de Bruxelles 61, Namur 5000, Belgium
- UNamur, NAmur Research Institute for LIfe Sciences, Unité de Recherche en Biologie Cellulaire animale, Rue de Bruxelles 61, Namur 5000, Belgium
| |
Collapse
|
12
|
Keirns BH, Keirns NG, Tsotsoros CE, Layman HM, Stout ME, Medlin AR, Sciarrillo CM, Teague TK, Emerson SR, Hawkins MAW. Adverse childhood experiences and obesity linked to indicators of gut permeability and inflammation in adult women. Physiol Behav 2023; 271:114319. [PMID: 37562704 PMCID: PMC10592146 DOI: 10.1016/j.physbeh.2023.114319] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Gut permeability may increase cardiovascular disease risk by allowing bacterial components (e.g., lipopolysaccharide or LPS) to enter the bloodstream, leading to low-grade inflammation. People with adverse childhood experiences (ACEs) consistently display evidence of chronic inflammation, but the source of this inflammation, and whether gut permeability may contribute, is unknown. Moreover, whether ACE status may further perturb obesity-associated gut permeability and inflammation is unknown. Women (N = 79, aged 18-84y) free of cardiometabolic diseases and inflammatory conditions and not regularly taking anti-inflammatory medications were included in a 2 × 2 factorial design with low or high ACE status (either 0 ACEs or 3+ ACEs) and body mass index (BMI) (either normal-weight [18.5-24.9 kg/m2; NW] or obesity [>30 kg/m2; OB]) as factors (n = 15-27/group). Serum LPS binding protein (LBP), soluble CD14 (sCD14), fatty-acid binding protein-2 (FABP2), LPS core IgM, and the ratio of LBP:sCD14 were used as indicators of gut permeability. Inflammatory markers C-reactive protein (CRP), tumor necrosis factor (TNF)-α, and interleukin (IL)-6 were also measured. Data were analyzed using 2-way ANCOVA (age-adjusted). LBP, LBP:sCD14 and FABP2 were higher in OB versus NW, regardless of ACE status (PBMI < 0.05). Higher ACE status was associated with increased circulating LBP:sCD14 and LPS core IgM (PACE < 0.05). sCD14 was unrelated to BMI or ACEs. CRP was elevated in OB versus NW (PBMI < 0.001) and tended to be higher with 3+ ACEs compared to 0 ACEs (PACE = 0.06). Moreover, TNF-α was greater in 3+ ACEs relative to 0 ACEs (PACE = 0.03). IL-6 was unrelated to BMI or ACE status. No interaction effects were observed for any marker of gut permeability or inflammation. In sum, ACE status and obesity were independently associated with evidence of gut permeability and systemic inflammation but did not interact in relation to indicators of gut permeability.
Collapse
Affiliation(s)
- Bryant H Keirns
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, United States of America.
| | - Natalie G Keirns
- Lifespan Cardiovascular Institute, The Miriam Hospital, Providence, RI, United States of America
| | - Cindy E Tsotsoros
- Department of Human Development and Family Science, University of Rhode Island, 2 Lower College Road, Kingston, RI 02881, United States of America
| | - Harley M Layman
- Department of Psychology, Oklahoma State University, 116 Psychology Building, Stillwater, OK, US 74078, United States of America
| | - Madison E Stout
- Department of Psychology, Oklahoma State University, 116 Psychology Building, Stillwater, OK, US 74078, United States of America
| | - Austin R Medlin
- Department of Health & Wellness Design, Indiana University School of Public Health, 1025 E. Seventh St., Bloomington, IN 47405, United States of America
| | - Christina M Sciarrillo
- Department of Nutritional Sciences, Oklahoma State University, 301 Nancy Randolph Davis, Stillwater, OK, US 74078, United States of America
| | - T Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK 74135, United States of America; Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK 74135, United States of America; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK 74107
| | - Sam R Emerson
- Department of Nutritional Sciences, Oklahoma State University, 301 Nancy Randolph Davis, Stillwater, OK, US 74078, United States of America
| | - Misty A W Hawkins
- Department of Health & Wellness Design, Indiana University School of Public Health, 1025 E. Seventh St., Bloomington, IN 47405, United States of America
| |
Collapse
|
13
|
Marvanova A, Kasik P, Elsnicova B, Tibenska V, Galatik F, Hornikova D, Zvolska V, Vebr P, Vodicka P, Hejnova L, Matous P, Szeiff Bacova B, Sykora M, Novotny J, Neuzil J, Kolar F, Novakova O, Zurmanova JM. Continuous short-term acclimation to moderate cold elicits cardioprotection in rats, and alters β-adrenergic signaling and immune status. Sci Rep 2023; 13:18287. [PMID: 37880253 PMCID: PMC10600221 DOI: 10.1038/s41598-023-44205-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/04/2023] [Indexed: 10/27/2023] Open
Abstract
Moderate cold acclimation (MCA) is a non-invasive intervention mitigating effects of various pathological conditions including myocardial infarction. We aim to determine the shortest cardioprotective regimen of MCA and the response of β1/2/3-adrenoceptors (β-AR), its downstream signaling, and inflammatory status, which play a role in cell-survival during myocardial infarction. Adult male Wistar rats were acclimated (9 °C, 1-3-10 days). Infarct size, echocardiography, western blotting, ELISA, mitochondrial respirometry, receptor binding assay, and quantitative immunofluorescence microscopy were carried out on left ventricular myocardium and brown adipose tissue (BAT). MultiPlex analysis of cytokines and chemokines in serum was accomplished. We found that short-term MCA reduced myocardial infarction, improved resistance of mitochondria to Ca2+-overload, and downregulated β1-ARs. The β2-ARs/protein kinase B/Akt were attenuated while β3-ARs translocated on the T-tubular system suggesting its activation. Protein kinase G (PKG) translocated to sarcoplasmic reticulum and phosphorylation of AMPKThr172 increased after 10 days. Principal component analysis revealed a significant shift in cytokine/chemokine serum levels on day 10 of acclimation, which corresponds to maturation of BAT. In conclusion, short-term MCA increases heart resilience to ischemia without any negative side effects such as hypertension or hypertrophy. Cold-elicited cardioprotection is accompanied by β1/2-AR desensitization, activation of the β3-AR/PKG/AMPK pathways, and an immunomodulatory effect.
Collapse
Affiliation(s)
- Aneta Marvanova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Petr Kasik
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Barbara Elsnicova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Veronika Tibenska
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - František Galatik
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Daniela Hornikova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Veronika Zvolska
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Pavel Vebr
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Petr Vodicka
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Libechov, Czech Republic
| | - Lucie Hejnova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Petr Matous
- First Faculty of Medicine, Center for Advanced Preclinical Imaging (CAPI), Charles University, Prague, Czech Republic
| | - Barbara Szeiff Bacova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Matus Sykora
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Jiri Novotny
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Jiri Neuzil
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD, Australia
| | - Frantisek Kolar
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Olga Novakova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Jitka M Zurmanova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic.
| |
Collapse
|
14
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Haybar H, Bandar B, Torfi E, Mohebbi A, Saki N. Cytokines and their role in cardiovascular diseases. Cytokine 2023; 169:156261. [PMID: 37413877 DOI: 10.1016/j.cyto.2023.156261] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/28/2023] [Accepted: 06/01/2023] [Indexed: 07/08/2023]
Abstract
The evaluation of diagnostic and prognostic biomarkers has always been a hot topic in various diseases. Considering that cardiovascular diseases (CVDs) have the highest mortality and morbidity rates in the world, various studies have been conducted so far to find CVD associated biomarkers, including cardiac troponin (cTn) and NT-proBNP. Cytokines are components of the immune system that are involved in the pathogenesis of CVD due to their contribution to the inflammation process. The level of cytokines varies in many cardiovascular diseases. For instance, the plasma level of IL-1α, IL-18, IL-33, IL-6 and IL-8 is positively correlated with atherosclerosis and that of some other interleukins such as IL-35 is negatively correlated with acute myocardial infarction or cardiac angina. Due to its pivotal role in the inflammation process, IL-1 super family is involved in many CVDs, including atherosclerosis. IL-20 among the interleukins of IL-10 family has a pro-atherogenic role, while others, such as IL-10 and IL-19, play an anti-atherogenic role. In the present review, we have collected the latest published evidence in this respect to discuss valuable cytokines from the diagnostic and prognostic stand point in CVDs.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bita Bandar
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ekhlas Torfi
- Department of Cardiovascular Disease, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Mohebbi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
16
|
Jiang Z, Jiang X, Chen A, He W. Platelet activation: a promoter for psoriasis and its comorbidity, cardiovascular disease. Front Immunol 2023; 14:1238647. [PMID: 37654493 PMCID: PMC10465348 DOI: 10.3389/fimmu.2023.1238647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease with a prevalence of 0.14% to 1.99%. The underlying pathology is mainly driven by the abnormal immune responses including activation of Th1, Th17, Th22 cells and secretion of cytokines. Patients with psoriasis are more likely to develop cardiovascular disease (CVD) which has been well recognized as a comorbidity of psoriasis. As mediators of hemostasis and thromboinflammation, platelets play an important part in CVD. However, less is known about their pathophysiological contribution to psoriasis and psoriasis-associated CVD. A comprehensive understanding of the role of platelet activation in psoriasis might pave the path for more accurate prediction of cardiovascular (CV) risk and provide new strategies for psoriasis management, which alleviates the increased CV burden associated with psoriasis. Here we review the available evidence about the biomarkers and mechanisms of platelet activation in psoriasis and the role of platelet activation in intriguing the common comorbidity, CVD. We further discussed the implications and efficacy of antiplatelet therapies in the treatment of psoriasis and prevention of psoriasis-associated CVD.
Collapse
Affiliation(s)
- Ziqi Jiang
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoran Jiang
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Aijun Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenyan He
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Dimitroglou Y, Aggeli C, Theofilis P, Tsioufis P, Oikonomou E, Chasikidis C, Tsioufis K, Tousoulis D. Novel Anti-Inflammatory Therapies in Coronary Artery Disease and Acute Coronary Syndromes. Life (Basel) 2023; 13:1669. [PMID: 37629526 PMCID: PMC10455741 DOI: 10.3390/life13081669] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Evidence suggests that inflammation plays an important role in atherosclerosis and the consequent clinical presentation, including stable coronary artery disease (CAD) and acute coronary syndromes (ACS). The most essential elements are cytokines, proteins with hormone-like properties that are produced by the immune cells, endothelial cells, platelets, fibroblasts, and some stromal cells. Interleukins (IL-1β and IL-6), chemokines, interferon-γ (IFN-γ), and tumor necrosis factor-alpha (TNF-α) are the cytokines commonly associated with endothelial dysfunction, vascular inflammation, and atherosclerosis. These molecules can be targeted by commonly used therapeutic substances or selective molecules that exert targeted anti-inflammatory actions. The most significant anti-inflammatory therapies are aspirin, statins, colchicine, IL-1β inhibitors, and IL-6 inhibitors, along with novel therapies such as TNF-α inhibitors and IL-1 receptor antagonists. Aspirin and statins are well-established therapies for atherosclerosis and CAD and their pleiotropic and anti-inflammatory actions contribute to their efficacy and favorable profile. Colchicine may also be considered in high-risk patients if recurrent ACS episodes occur when on optimal medical therapy according to the most recent guidelines. Recent randomized studies have also shown that therapies specifically targeting inflammatory interleukins and inflammation can reduce the risk for cardiovascular events, but these therapies are yet to be fully implemented in clinical practice. Preclinical research is also intense, targeting various inflammatory mediators that are believed to be implicated in CAD, namely repeated transfers of the soluble mutant of IFN-γ receptors, NLRP3 inflammasome inhibitors, IL-10 delivery by nanocarriers, chemokine modulatory treatments, and reacting oxygen species (ROS) targeting nanoparticles. Such approaches, although intriguing and promising, ought to be tested in clinical settings before safe conclusions can be drawn. Although the link between inflammation and atherosclerosis is significant, further studies are needed in order to elucidate this association and improve outcomes in patients with CAD.
Collapse
Affiliation(s)
- Yannis Dimitroglou
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Constantina Aggeli
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Panagiotis Theofilis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Panagiotis Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Evangelos Oikonomou
- Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece;
| | - Christos Chasikidis
- Department of Cardiology, General Hospital of Corinth, 20100 Corinth, Greece;
| | - Konstantinos Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| |
Collapse
|
18
|
Khandker SS, Kotyla PJ, Musa KI, Islam MA. Editorial: Autoimmunity and cardiovascular diseases. Front Cardiovasc Med 2023; 10:1249525. [PMID: 37534278 PMCID: PMC10393120 DOI: 10.3389/fcvm.2023.1249525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023] Open
Affiliation(s)
- Shahad Saif Khandker
- Department of Microbiology, Gonoshasthaya Samaj Vittik Medical College, Savar, Dhaka Division, Bangladesh
| | - Przemysław J. Kotyla
- Department of Internal Medicine, Rheumatology and Clinical Immunology, Faculty in Katowice, Medical University of Silesia, Katowice, Poland
| | - Kamarul Imran Musa
- Department of Community Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Md Asiful Islam
- WHO Collaborating Centre for Global Women’s Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
19
|
Somersalo E, Kuuliala K, Kuuliala A, Wasenius NS, Klemetti MM, Kivimäki AS, Kautiainen H, Eriksson JG, Laine MK. Circulating Cytokine Levels and Cardiovascular Disease Risk Profile in Young Adult Offspring of Women with Type 1 Diabetes. Diabetes Ther 2023:10.1007/s13300-023-01428-y. [PMID: 37286850 PMCID: PMC10299958 DOI: 10.1007/s13300-023-01428-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/22/2023] [Indexed: 06/09/2023] Open
Abstract
INTRODUCTION Cytokines are key players in the development of both type 1 diabetes (T1D) and cardiovascular disease (CVD). Offspring of women with T1D are known to have an increased risk of early-onset CVD. We studied whether an increased risk of CVD can be observed in the cytokine profile among young adult offspring of women with T1D. METHODS This cross-sectional case-control study included 67 offspring of women with T1D (cases) and 79 control participants (controls). At an age of 18-23 years, they participated in a clinical assessment including laboratory tests and questionnaires. Cytokine levels were analyzed from venous blood samples after 10 h fasting using Quansys biosciences Q-Plex™ High Sensitivity Human Cytokine Array. RESULTS Circulating cytokine levels were in general similar between the groups. The circulating levels of interferon-γ (1.78 [IQR 1.20, 2.36] pg/mL versus 2.57 [IQR 1.50, 3.89] pg/mL) (p = 0.006) were lower in cases than controls. CONCLUSION The findings did not support our hypothesis that serum cytokine profile, determined in early adulthood, was associated with a more adverse CVD risk profile in offspring of women with T1D. Further studies are warranted to find out whether cytokines could serve as early biomarkers of CVD development or whether changes in the cytokine levels over years could be used to monitor CVD progression in offspring of women with T1D.
Collapse
Affiliation(s)
- Erik Somersalo
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Folkhälsan Research Center, Helsinki, Finland.
| | - Krista Kuuliala
- Department of Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Kuuliala
- Department of Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Niko S Wasenius
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Miira M Klemetti
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Anne S Kivimäki
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Hannu Kautiainen
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Primary Health Care Unit, Kuopio University Hospital, Kuopio, Finland
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Human Potential Translational Research Programme and Department of Obstetrics and Gynecology, National University Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore
- Agency for Science, Technology and Research (A*STAR), Singapore Institute for Clinical Sciences (SICS), Singapore, Singapore
| | - Merja K Laine
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| |
Collapse
|
20
|
Liu J, Chen L, Zheng X, Guo C. Identification of immune-related genes in acute myocardial infarction based on integrated bioinformatical methods and experimental verification. PeerJ 2023; 11:e15058. [PMID: 37214088 PMCID: PMC10198157 DOI: 10.7717/peerj.15058] [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: 08/23/2022] [Accepted: 02/22/2023] [Indexed: 05/24/2023] Open
Abstract
Background Acute myocardial infarction (AMI) is one of the leading causes of death worldwide. The etiology of AMI is complex and has not been fully defined. In recent years, the role of immune response in the development, progression and prognosis of AMI has received increasing attention. The aim of this study was to identify key genes associated with the immune response in AMI and to analyze their immune infiltration. Methods The study included a total of two GEO databases, containing 83 patients with AMI and 54 healthy individuals. We used the linear model of microarray data (limma) package to find the differentially expressed genes associated with AMI, performing weighted gene co-expression analysis (WGCNA) to further identify the genes associated with inflammatory response to AMI. We found the final hub genes through the protein-protein interaction (PPI) network and least absolute shrinkage and selection operator (LASSO) regression model. To verify the above conclusions, we constructed mice AMI model, extracting myocardial tissue to perform qRT-PCR. Furthermore, the CIBERSORT tool for immune cells infiltration analysis was also carried out. Results A total of 5,425 significant up-regulated and 2,126 down-regulated genes were found in GSE66360 and GSE24519. A total of 116 immune-related genes in close association with AMI were screened by WGCNA analysis. These genes were mostly clustered in the immune response on the basis of GO and KEGG enrichment. With construction of PPI network and LASSO regression analysis, this research found three hub genes (SOCS2, FFAR2, MYO10) among these differentially expressed genes. The immune cell infiltration results revealed that significant differences could be found on T cells CD4 memory activated, Tregs (regulatory T cells), macrophages M2, neutrophils, T cells CD8, T cells CD4 naive, eosinophils between controls and AMI patients.
Collapse
Affiliation(s)
- Jian Liu
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lu Chen
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiang Zheng
- Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Ng CT, Fong LY, Abdullah MNH. Interferon-gamma (IFN-γ): Reviewing its mechanisms and signaling pathways on the regulation of endothelial barrier function. Cytokine 2023; 166:156208. [PMID: 37088004 DOI: 10.1016/j.cyto.2023.156208] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023]
Abstract
Interferon-gamma (IFN-γ) is a pleiotropic cytokine that plays a critical role in mediating an array of immune responses including promotes antiviral activity, facilitates macrophage activation, controls Th1/Th2 balance, and regulates cellular apoptosis and proliferation. A few articles have previously reviewed the effects of IFN-γ in the regulation of barrier permeability, but none of these articles focuses on barrier function of endothelial cells. This review aims to discuss the regulatory mechanisms of IFN-γ on endothelial barrier function and its underlying signaling pathways. Articles were retrieved from electronic databases such as PubMed and Google Scholar using keywords "Interferon-gamma", "endothelial cells", "barrier function", and "signaling pathway". The articles published between 2000 and 2022 that are related to the aforementioned topics were selected. A few journals published beyond this period were also included due to limited information available. The results showed that IFN-γ modulates endothelial barrier function, mainly involves small GTPases, STAT1-dependent pathway, p38 MAPK and nitric oxide. In conclusion, more in depth cellular and molecular studies are needed to elucidate the pathways of IFN-γ in the regulation of endothelial barrier function.
Collapse
Affiliation(s)
- Chin Theng Ng
- Unit of Physiology, Faculty of Medicine, AIMST University, Bedong, 08100 Kedah, Malaysia.
| | - Lai Yen Fong
- Department of Pre-clinical Sciences, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, 43000 Selangor, Malaysia
| | - Muhammad Nazrul Hakim Abdullah
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia
| |
Collapse
|
22
|
Dark Sweet Cherry ( Prunus avium) Supplementation Reduced Blood Pressure and Pro-Inflammatory Interferon Gamma (IFNγ) in Obese Adults without Affecting Lipid Profile, Glucose Levels and Liver Enzymes. Nutrients 2023; 15:nu15030681. [PMID: 36771387 PMCID: PMC9920461 DOI: 10.3390/nu15030681] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Dark sweet cherries (DSC) are rich in fiber and polyphenols that decrease risk factors associated with obesity. This single-blind randomized placebo-controlled study investigated DSC effects on inflammation, cardiometabolic, and liver health biomarkers in obese adults. Participants (>18 years, body mass index (BMI) = 30-40 kg/m2) consumed 200 mL of DSC drink (juice supplemented with DSC powder) (n = 19) or a placebo drink (n = 21) twice/day for 30 days. Anthropometric and physiological biomarkers were monitored at baseline (D1), mid-point (D15), and endpoint (D30) visits. Blood inflammatory biomarkers were assessed at D1, D15, and D30, and blood lipids, glucose, and liver enzymes at D1 and D30. DSC consumption lowered systolic blood pressure (SBP) (p = 0.05) and decreased diastolic blood pressure (DBP) compared to placebo (p = 0.04). Stratification of participants by BMI revealed a greater (p = 0.008) SBP reduction in BMI > 35 participants. DSC lowered pro-inflammatory interferon-gamma (IFNγ) (p = 0.001), which correlated with SBP changes. The interleukin (IL)-1RA and SBP changes were correlated in the placebo group, as well as triglycerides (TG) with DBP. The increased IL-10 levels in the placebo group suggested a compensatory mechanism to counteract elevated IFNγ levels. No significant between-group differences were detected for blood lipids, glucose, and liver enzymes. In conclusion, DSC helped to decrease blood pressure levels and inflammation in obese adults.
Collapse
|
23
|
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
|
24
|
González L, Rivera K, Andia ME, Martínez Rodriguez G. The IL-1 Family and Its Role in Atherosclerosis. Int J Mol Sci 2022; 24:17. [PMID: 36613465 PMCID: PMC9820551 DOI: 10.3390/ijms24010017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
The IL-1 superfamily of cytokines is a central regulator of immunity and inflammation. The family is composed of 11 cytokines (with agonist, antagonist, and anti-inflammatory properties) and 10 receptors, all tightly regulated through decoy receptor, receptor antagonists, and signaling inhibitors. Inflammation not only is an important physiological response against infection and injury but also plays a central role in atherosclerosis development. Several clinical association studies along with experimental studies have implicated the IL-1 superfamily of cytokines and its receptors in the pathogenesis of cardiovascular disease. Here, we summarize the key features of the IL-1 family, its role in immunity and disease, and how it helps shape the development of atherosclerosis.
Collapse
Affiliation(s)
- Leticia González
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Instituto Milenio de Ingeniería e Inteligencia Artificial Para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Katherine Rivera
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Programa de Doctorado en Ciencias Médicas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
| | - Marcelo E. Andia
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Instituto Milenio de Ingeniería e Inteligencia Artificial Para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Gonzalo Martínez Rodriguez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
| |
Collapse
|
25
|
Chiorescu RM, Mocan M, Inceu AI, Buda AP, Blendea D, Vlaicu SI. Vulnerable Atherosclerotic Plaque: Is There a Molecular Signature? Int J Mol Sci 2022; 23:13638. [PMID: 36362423 PMCID: PMC9656166 DOI: 10.3390/ijms232113638] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2023] Open
Abstract
Atherosclerosis and its clinical manifestations, coronary and cerebral artery diseases, are the most common cause of death worldwide. The main pathophysiological mechanism for these complications is the rupture of vulnerable atherosclerotic plaques and subsequent thrombosis. Pathological studies of the vulnerable lesions showed that more frequently, plaques rich in lipids and with a high level of inflammation, responsible for mild or moderate stenosis, are more prone to rupture, leading to acute events. Identifying the vulnerable plaques helps to stratify patients at risk of developing acute vascular events. Traditional imaging methods based on plaque appearance and size are not reliable in prediction the risk of rupture. Intravascular imaging is a novel technique able to identify vulnerable lesions, but it is invasive and an operator-dependent technique. This review aims to summarize the current data from literature regarding the main biomarkers involved in the attempt to diagnose vulnerable atherosclerotic lesions. These biomarkers could be the base for risk stratification and development of the new therapeutic drugs in the treatment of patients with vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Roxana Mihaela Chiorescu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Mihaela Mocan
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Andreea Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine, 400349 Cluj-Napoca, Romania
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Andreea Paula Buda
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Dan Blendea
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
- Department of Cardiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400437 Cluj-Napoca, Romania
| | - Sonia Irina Vlaicu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| |
Collapse
|
26
|
Roach LA, Meyer BJ, Fitton JH, Winberg P. Improved Plasma Lipids, Anti-Inflammatory Activity, and Microbiome Shifts in Overweight Participants: Two Clinical Studies on Oral Supplementation with Algal Sulfated Polysaccharide. Mar Drugs 2022; 20:md20080500. [PMID: 36005503 PMCID: PMC9410082 DOI: 10.3390/md20080500] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 02/06/2023] Open
Abstract
Seaweed polysaccharides in the diet may influence both inflammation and the gut microbiome. Here we describe two clinical studies with an Ulva sp. 84-derived sulfated polysaccharide—“xylorhamnoglucuronan” (SXRG84)—on metabolic markers, inflammation, and gut flora composition. The first study was a double-blind, randomized placebo-controlled trial with placebo, and either 2 g/day or 4 g/day of SXRG84 daily for six weeks in 64 overweight or obese participants (median age 55 years, median body mass index (BMI) 29 kg/m2). The second study was a randomized double-blind placebo-controlled crossover trial with 64 participants (median BMI 29 kg/m2, average age 52) on placebo for six weeks and then 2 g/day of SXRG84 treatment for six weeks, or vice versa. In Study 1, the 2 g/day dose exhibited a significant reduction in non-HDL (high-density lipoprotein) cholesterol (−10% or −0.37 mmol/L, p = 0.02) and in the atherogenic index (−50%, p = 0.05), and two-hour insulin (−12% or −4.83 mU/L) showed trends for reduction in overweight participants. CRP (C-reactive protein) was significantly reduced (−27% or −0.78 mg/L, p = 0.03) with the 4 g/day dose in overweight participants. Significant gut flora shifts included increases in Bifidobacteria, Akkermansia, Pseudobutyrivibrio, and Clostridium and a decrease in Bilophila. In Study 2, no significant differences in lipid measures were observed, but inflammatory cytokines were improved. At twelve weeks after the SXRG84 treatment, plasma cytokine concentrations were significantly lower than at six weeks post placebo for IFN-γ (3.4 vs. 7.3 pg/mL), IL-1β (16.2 vs. 23.2 pg/mL), TNF-α (9.3 vs. 12.6 pg/mL), and IL-10 (1.6 vs. 2.1 pg/mL) (p < 0.05). Gut microbiota abundance and composition did not significantly differ between groups (p > 0.05). Together, the studies illustrate improvements in plasma lipids and an anti-inflammatory effect of dietary SXRG84 that is participant specific.
Collapse
Affiliation(s)
- Lauren A. Roach
- Molecular Horizons, Illawarra Health and Medical Research Institute, School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW 2522, Australia
- Correspondence: (L.A.R.); (B.J.M.); (P.W.)
| | - Barbara J. Meyer
- Molecular Horizons, Illawarra Health and Medical Research Institute, School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW 2522, Australia
- Correspondence: (L.A.R.); (B.J.M.); (P.W.)
| | | | - Pia Winberg
- Venus Shell Systems Pty Ltd., Nowra, NSW 2540, Australia
- Correspondence: (L.A.R.); (B.J.M.); (P.W.)
| |
Collapse
|
27
|
Zhao T, Jiang Q, Li W, Wang Y, Zou Y, Chai X, Yuan Z, Ma L, Yu R, Deng T, Yu C, Wang T. Antigen-Presenting Cell-Like Neutrophils Foster T Cell Response in Hyperlipidemic Patients and Atherosclerotic Mice. Front Immunol 2022; 13:851713. [PMID: 35251050 PMCID: PMC8891125 DOI: 10.3389/fimmu.2022.851713] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/31/2022] [Indexed: 12/30/2022] Open
Abstract
Neutrophils constitute abundant cellular components in atherosclerotic plaques. Most of the current studies are focused on the roles of granular proteins released by neutrophils in atherosclerosis. Here, we revealed a unique subset of neutrophils which exhibit the characteristics of antigen-presenting cell (APC) (which were called APC-like neutrophils afterwards) in atherosclerosis. The roles of APC-like neutrophils and relevant mechanisms were investigated in hyperlipidemic patients and atherosclerotic mice. Higher percentages of neutrophils and APC-like neutrophils were found in peripheral blood of hyperlipidemic patients than that of healthy donors. Meanwhile, we also identified higher infiltration of neutrophils and APC-like neutrophils in atherosclerotic mice. Ox-LDL induced Phorbol-12-myristate-13-acetate (PMA)-activated neutrophils to acquire the APC-like phenotype. Importantly, upon over-expression of APC-like markers, neutrophils acquired APC functions to promote the proliferation and interferon-γ production of CD3+ T cells via HLA-DR/CD80/CD86. In accordance with what found in vitro, positive correlation between neutrophils and CD3+ T cells was observed in hyperlipidemic patients. In conclusion, our work identifies a proinflammatory neutrophil subset in both hyperlipidemic patients and atherosclerotic mice. This unique phenotype of neutrophils could activate the adaptive immune response to promote atherosclerosis progression. Thus, this neutrophil subset may be a new target for immunotherapy of atherosclerosis.
Collapse
Affiliation(s)
- Tingrui Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| | - Qingsong Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Wenming Li
- Department of Clinical Laboratory, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yin Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| | - Yao Zou
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| | - Xinyu Chai
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| | - Limei Ma
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| | - Ruihong Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| | - Tao Deng
- Research Center of Pharmaceutical Preparations and Nanomedicine, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| | - Tingting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, China
| |
Collapse
|
28
|
Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, Qiang L. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci 2022; 79:272. [PMID: 35503385 PMCID: PMC11073100 DOI: 10.1007/s00018-022-04286-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/11/2022] [Accepted: 04/03/2022] [Indexed: 12/12/2022]
Abstract
Characterized by a surplus of whole-body adiposity, obesity is strongly associated with the prognosis of atherosclerosis, a hallmark of coronary artery disease (CAD) and the major contributor to cardiovascular disease (CVD) mortality. Adipose tissue serves a primary role as a lipid-storage organ, secreting cytokines known as adipokines that affect whole-body metabolism, inflammation, and endocrine functions. Emerging evidence suggests that adipokines can play important roles in atherosclerosis development, progression, as well as regression. Here, we review the versatile functions of various adipokines in atherosclerosis and divide these respective functions into three major groups: protective, deteriorative, and undefined. The protective adipokines represented here are adiponectin, fibroblast growth factor 21 (FGF-21), C1q tumor necrosis factor-related protein 9 (CTRP9), and progranulin, while the deteriorative adipokines listed include leptin, chemerin, resistin, Interleukin- 6 (IL-6), and more, with additional adipokines that have unclear roles denoted as undefined adipokines. Comprehensively categorizing adipokines in the context of atherosclerosis can help elucidate the various pathways involved and potentially pave novel therapeutic approaches to treat CVDs.
Collapse
Affiliation(s)
- Longhua Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China.
| | - Zunhan Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiaohui Ji
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Wenqian Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Jinwen Luan
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Tarik Zahr
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Li Qiang
- Department of Pathology and Cellular Biology and Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA.
| |
Collapse
|
29
|
Inflammatory and Oxidative Stress Markers Related to Adherence to the Mediterranean Diet in Patients with Metabolic Syndrome. Antioxidants (Basel) 2022; 11:antiox11050901. [PMID: 35624765 PMCID: PMC9137637 DOI: 10.3390/antiox11050901] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/28/2022] Open
Abstract
Metabolic syndrome (MetS) is characterized by increased pro-oxidative stress and a pro-inflammatory state. Several studies emphasized the protective effect of the Mediterranean dietary pattern (MDP). To assess the oxidative and inflammatory state according to the adherence to MDP using biomarkers in patients with MetS. Antioxidant and pro-inflammatory biomarkers were determined in plasma, peripheral blood mononuclear cells (PBMCs), and neutrophils of adults (aged 55–75 years old; 60% women) with MetS living in Mallorca (Spain). Anthropometrics, dietary intake by a validated semi-quantitative 143-item food frequency questionnaire, and a Dietary Inflammatory Index were measured. Patients with low adherence to MDP showed higher levels of glycated haemoglobin A1c and triglycerides, and lower levels of HDL cholesterol. Plasma levels of interleukin-1β, IL-6, IL-15, tumour necrosis factor α, xanthine oxidase, and ghrelin, and activities of superoxide dismutase, and myeloperoxidase were higher in subjects with low adherence to the MDP. Reactive oxygen species production in PBMCs and neutrophils stimulated with lipopolysaccharide was higher in participants with low adherence to the MDP. Patients with MetS and higher adherence to the MDP showed less altered anthropometric parameters, blood biochemical profile, and better oxidative and inflammatory status.
Collapse
|
30
|
Lorey MB, Öörni K, Kovanen PT. Modified Lipoproteins Induce Arterial Wall Inflammation During Atherogenesis. Front Cardiovasc Med 2022; 9:841545. [PMID: 35310965 PMCID: PMC8927694 DOI: 10.3389/fcvm.2022.841545] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/26/2022] [Indexed: 12/15/2022] Open
Abstract
Circulating apolipoprotein B-containing lipoproteins, notably the low-density lipoproteins, enter the inner layer of the arterial wall, the intima, where a fraction of them is retained and modified by proteases, lipases, and oxidizing agents and enzymes. The modified lipoproteins and various modification products, such as fatty acids, ceramides, lysophospholipids, and oxidized lipids induce inflammatory reactions in the macrophages and the covering endothelial cells, initiating an increased leukocyte diapedesis. Lipolysis of the lipoproteins also induces the formation of cholesterol crystals with strong proinflammatory properties. Modified and aggregated lipoproteins, cholesterol crystals, and lipoproteins isolated from human atherosclerotic lesions, all can activate macrophages and thereby induce the secretion of proinflammatory cytokines, chemokines, and enzymes. The extent of lipoprotein retention, modification, and aggregation have been shown to depend largely on differences in the composition of the circulating lipoprotein particles. These properties can be modified by pharmacological means, and thereby provide opportunities for clinical interventions regarding the prevention and treatment of atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Martina B. Lorey
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- *Correspondence: Katariina Öörni
| | - Petri T. Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| |
Collapse
|
31
|
Verdú E, Homs J, Boadas-Vaello P. Physiological Changes and Pathological Pain Associated with Sedentary Lifestyle-Induced Body Systems Fat Accumulation and Their Modulation by Physical Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:13333. [PMID: 34948944 PMCID: PMC8705491 DOI: 10.3390/ijerph182413333] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
A sedentary lifestyle is associated with overweight/obesity, which involves excessive fat body accumulation, triggering structural and functional changes in tissues, organs, and body systems. Research shows that this fat accumulation is responsible for several comorbidities, including cardiovascular, gastrointestinal, and metabolic dysfunctions, as well as pathological pain behaviors. These health concerns are related to the crosstalk between adipose tissue and body systems, leading to pathophysiological changes to the latter. To deal with these health issues, it has been suggested that physical exercise may reverse part of these obesity-related pathologies by modulating the cross talk between the adipose tissue and body systems. In this context, this review was carried out to provide knowledge about (i) the structural and functional changes in tissues, organs, and body systems from accumulation of fat in obesity, emphasizing the crosstalk between fat and body tissues; (ii) the crosstalk between fat and body tissues triggering pain; and (iii) the effects of physical exercise on body tissues and organs in obese and non-obese subjects, and their impact on pathological pain. This information may help one to better understand this crosstalk and the factors involved, and it could be useful in designing more specific training interventions (according to the nature of the comorbidity).
Collapse
Affiliation(s)
- Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| | - Judit Homs
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
- Department of Physical Therapy, EUSES-University of Girona, 17190 Salt, Spain
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| |
Collapse
|
32
|
Ding T, Li B, Su R, Su R, Wang Y, Gao C, Li X, Wang C. Elevated Th17 cells are associated with cardiovascular complications in ankylosing spondylitis. Rheumatology (Oxford) 2021; 61:3481-3490. [PMID: 34894210 DOI: 10.1093/rheumatology/keab888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/22/2021] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE Patients with ankylosing spondylitis (AS) carry an increased burden of cardiovascular diseases (CVD), but features denoting the development of CVD in AS are unclear. This study aimed to evaluate the percentage and absolute number of lymphocytes and CD4+T cells in AS patients complicated with CVD (AS-CVD) and determine whether circulating Th17 cells are associated with the development of CVD in AS. METHOD A total of 117 AS patients (46 had CVD and 71 had no CVD) were enrolled in this retrospective study. The percentage and absolute number of lymphocytes and CD4+T cells were determined by Flow cytometry. Associations between CVD and clinical markers were analyzed using logistic regression. RESULTS The ratio of Th17/Treg cells (0.30 vs 0.19, p = 0.014) and the absolute number of Th17 cells (7.27 cells/μL vs 4.34 cells/μL, p < 0.001) was significantly elevated in AS-CVD group compared with AS-no-CVD group. Multivariate logistic regression revealed that elevated Th17 cells (OR = 1.20, p = 0.016) were associated with CVD complications in AS. Receiver operating characteristic (ROC) curves showed a contribution of Th17 cell for distinguishing AS patients with CVD, with the areas under the ROC curve (AUCs) of 0.729 (95%CI: 0.632-0.825, p < 0.001). CONCLUSION Our findings provide evidence for the association between Th17 cells and increased cardiovascular risk in AS. Th17 cells may contribute to accelerated atherogenesis and increased cardiovascular burden in AS and be valuable for early assessment and management of AS-CVD.
Collapse
Affiliation(s)
- Tingting Ding
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Baochen Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ronghui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanyan Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
33
|
Talepoor AG, Rastegari B, Kalani M, Doroudchi M. Decrease in the inflammatory cytokines of LPS-stimulated PBMCs of patients with atherosclerosis by a TLR-4 antagonist in the co-culture with HUVECs. Int Immunopharmacol 2021; 101:108295. [PMID: 34735917 DOI: 10.1016/j.intimp.2021.108295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/22/2021] [Accepted: 10/19/2021] [Indexed: 11/29/2022]
Abstract
Toll-like receptors (TLRs) are among the players of inflammation during atherosclerosis. We assessed the effects of Eritoran, a TLR-4 antagonist, on lipopolysaccharide (LPS)-induced cytokines production by Peripheral Blood Mononuclear Cells (PBMCs) of patients with high-stenosis (HS) (n = 6) and healthy controls (HCs) (n = 6) co-cultured with Human Umbilical Vein Endothelial Cells (HUVECs). LPS stimulation significantly increased the levels of IL-6 (P = 0.007 and P = 0.005), TNF-α (P = 0.006 and P = 0.005), IL-2 (P = 0.007 and P = 0.002), IFN-γ (P = 0.006 and P = 0.003), IL-17A (P = 0.004 and P = 0.003), IL-17F (P = 0.005 and P = 0.003), IL-5 (P = 0.007 and P = 0.005), IL-13 (P = 0.006 and P = 0.005), IL-9 (P = 0.005 and P = 0.005) and IL-21 (P = 0.007 and P = 0.005) in HUVECs co-cultured with HC and HS PBMCs as compared with un-stimulated co-culture condition, respectively. Eritoran treatment (50 μg/mL and 100 μg/mL) significantly reduced the levels of LPS-induced IL-6 (P = 0.007 and P = 0.006; P = 0.007 and P = 0.007), TNF-α (P = 0.005 and P = 0.003; P = 0.007 and P = 0.005), IL-2 (P = 0.007 and P = 0.005; P = 0.005 and P = 0.004), IFN-γ (P = 0.007 and P = 0.005; P = 0.005 and P = 0.004), IL-17A (P = 0.005 and P = 0.002; P = 0.005 and P = 0.002), IL-17F (P = 0.006 and P = 0.006; P = 0.005 and P = 0.005), IL-5 (P = 0.007 and P = 0.006; P = 0.007 and P = 0.007), IL-9 (P = 0.005 and P = 0.005; P = 0.005 and P = 0.005) and IL-21 (P = 0.007 and P = 0.007; P = 0.005 and P = 0.005) in stimulated HUVECs co-cultured with HC and HS PBMCs, compared to un-treated condition, respectively. Our results demonstrate that attenuating effect of Eritoran on the inflammatory responses to LPS is higher in PBMCs of patients with high stenosis, suggesting its potential role in ameliorating inflammatory conditions in atherosclerosis.
Collapse
Affiliation(s)
- Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Banafsheh Rastegari
- Diagnostic Laboratory of Sciences and Technology Research Center, Paramedical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Kalani
- Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
34
|
Diakos NA, Taleb I, Kyriakopoulos CP, Shah KS, Javan H, Richins TJ, Yin MY, Yen C, Dranow E, Bonios MJ, Alharethi R, Koliopoulou AG, Taleb M, Fang JC, Selzman CH, Stellos K, Drakos SG. Circulating and Myocardial Cytokines Predict Cardiac Structural and Functional Improvement in Patients With Heart Failure Undergoing Mechanical Circulatory Support. J Am Heart Assoc 2021; 10:e020238. [PMID: 34595931 PMCID: PMC8751895 DOI: 10.1161/jaha.120.020238] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Recent prospective multicenter data from patients with advanced heart failure demonstrated that left ventricular assist device (LVAD) support combined with standard heart failure medications, induced significant cardiac structural and functional improvement, leading to high rates of LVAD weaning in selected patients. We investigated whether preintervention myocardial and systemic inflammatory burden could help identify the subset of patients with advanced heart failure prone to LVAD-mediated cardiac improvement to guide patient selection, treatment, and monitoring. Methods and Results Ninety-three patients requiring durable LVAD were prospectively enrolled. Myocardial tissue and blood were acquired during LVAD implantation, for measurement of inflammatory markers. Cardiac structural and functional improvement was prospectively assessed via serial echocardiography. Eleven percent of the patients showed significant reverse remodeling following LVAD support (ie, responders). Circulating tumor necrosis factor alpha, interleukin (IL)-4, IL-5, IL-6, IL-7, IL-13, and interferon gamma were lower in responders, compared with nonresponders (P<0.05, all comparisons). The myocardial tissue signal transducer and activator of transcription-3, an inflammatory response regulator, was less activated in responders (P=0.037). Guided by our tissue studies and a multivariable dichotomous regression analysis, we identified that low levels of circulating interferon gamma (odds ratio [OR], 0.06; 95% CI, 0.01-0.35) and tumor necrosis factor alpha (OR, 0.05; 95% CI, 0.00-0.43), independently predict cardiac improvement, creating a 2-cytokine model effectively predicting responders (area under the curve, 0.903; P<0.0001). Conclusions Baseline myocardial and systemic inflammatory burden inversely correlates with cardiac improvement following LVAD support. A circulating 2-cytokine model predicting significant reverse remodeling was identified, warranting further investigation as a practical preintervention tool in identifying patients prone to LVAD-mediated cardiac improvement and device weaning.
Collapse
Affiliation(s)
- Nikolaos A. Diakos
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUT,Present address:
Division of CardiologyColumbia University Medical CenterNew YorkNY
| | - Iosif Taleb
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUT,University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Christos P. Kyriakopoulos
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUT,University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Kevin S. Shah
- University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Hadi Javan
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUT,University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Tyler J. Richins
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUT
| | - Michael Y. Yin
- University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Chi‐Gang Yen
- University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Elizabeth Dranow
- University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Michael J. Bonios
- University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT,Present address:
Onassis Cardiac Surgery CenterAthensGreece
| | - Rami Alharethi
- University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Antigone G. Koliopoulou
- University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT,Present address:
Onassis Cardiac Surgery CenterAthensGreece
| | - Mariam Taleb
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUT
| | - James C. Fang
- University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Craig H. Selzman
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUT,University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| | - Konstantinos Stellos
- Cardiovascular Research CentreNewcastle University & Cardiothoracic CentreNewcastle upon Tyne HospitalsNewcastleUK
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUT,University of Utah Health and School of Medicine, Intermountain Medical Center, George E. Wahlen Department of Veterans Affairs Medical CenterU.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant ProgramSalt Lake CityUT
| |
Collapse
|
35
|
Balestra C, Lambrechts K, Mrakic-Sposta S, Vezzoli A, Levenez M, Germonpré P, Virgili F, Bosco G, Lafère P. Hypoxic and Hyperoxic Breathing as a Complement to Low-Intensity Physical Exercise Programs: A Proof-of-Principle Study. Int J Mol Sci 2021; 22:ijms22179600. [PMID: 34502508 PMCID: PMC8431767 DOI: 10.3390/ijms22179600] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammation is an adaptive response to both external and internal stimuli including infection, trauma, surgery, ischemia-reperfusion, or malignancy. A number of studies indicate that physical activity is an effective means of reducing acute systemic and low-level inflammation occurring in different pathological conditions and in the recovery phase after disease. As a proof-of-principle, we hypothesized that low-intensity workout performed under modified oxygen supply would elicit a "metabolic exercise" inducing a hormetic response, increasing the metabolic load and oxidative stress with the same overall effect expected after a higher intensity or charge exercise. Herein, we report the effect of a 5-week low-intensity, non-training, exercise program in a group of young healthy subjects in combination with the exposure to hyperoxia (30% and 100% pO2, respectively) or light hypoxia (15% pO2) during workout sessions on several inflammation and oxidative stress parameters, namely hemoglobin (Hb), redox state, nitric oxide metabolite (NOx), inducible nitric oxide synthase (iNOS), inflammatory cytokine expression (TNF-α, interleukin (IL)-6, IL-10), and renal functional biomarkers (creatinine, neopterin, and urates). We confirmed our previous reports demonstrating that intermittent hyperoxia induces the normobaric oxygen paradox (NOP), a response overlapping the exposure to hypoxia. Our data also suggest that the administration of modified air composition is an expedient complement to a light physical exercise program to achieve a significant modulation of inflammatory and immune parameters, including cytokines expression, iNOS activity, and oxidative stress parameters. This strategy can be of pivotal interest in all those conditions characterized by the inability to achieve a sufficient workload intensity, such as severe cardiovascular alterations and articular injuries failing to effectively gain a significant improvement of physical capacity.
Collapse
Affiliation(s)
- Costantino Balestra
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1180 Brussels, Belgium; (K.L.); (M.L.); (P.G.); (P.L.)
- Physical Activity Teaching Unit, Motor Sciences Faculty, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
- DAN Europe Research Division, 1160 Brussels, Belgium
- Correspondence: (C.B.); (F.V.); (G.B.)
| | - Kate Lambrechts
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1180 Brussels, Belgium; (K.L.); (M.L.); (P.G.); (P.L.)
| | - Simona Mrakic-Sposta
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Piazza dell’Ospedale Maggiore, 3, 20162 Milan, Italy; (S.M.-S.); (A.V.)
| | - Alessandra Vezzoli
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Piazza dell’Ospedale Maggiore, 3, 20162 Milan, Italy; (S.M.-S.); (A.V.)
| | - Morgan Levenez
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1180 Brussels, Belgium; (K.L.); (M.L.); (P.G.); (P.L.)
| | - Peter Germonpré
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1180 Brussels, Belgium; (K.L.); (M.L.); (P.G.); (P.L.)
- DAN Europe Research Division, 1160 Brussels, Belgium
- Centre for Hyperbaric Oxygen Therapy, Queen Astrid Military Hospital, 1120 Brussels, Belgium
| | - Fabio Virgili
- Council for Agricultural Research and Economics—Food and Nutrition Research Centre (C.R.E.A.-AN), 00178 Rome, Italy
- Correspondence: (C.B.); (F.V.); (G.B.)
| | - Gerardo Bosco
- Environmental Physiology & Medicine Lab, Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Correspondence: (C.B.); (F.V.); (G.B.)
| | - Pierre Lafère
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1180 Brussels, Belgium; (K.L.); (M.L.); (P.G.); (P.L.)
- DAN Europe Research Division, 1160 Brussels, Belgium
- Centre for Hyperbaric Oxygen Therapy, Queen Astrid Military Hospital, 1120 Brussels, Belgium
| |
Collapse
|
36
|
López-Díez R, Egaña-Gorroño L, Senatus L, Shekhtman A, Ramasamy R, Schmidt AM. Diabetes and Cardiovascular Complications: The Epidemics Continue. Curr Cardiol Rep 2021; 23:74. [PMID: 34081211 PMCID: PMC8173334 DOI: 10.1007/s11886-021-01504-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW The cardiovascular complications of type 1 and 2 diabetes are major causes of morbidity and mortality. Extensive efforts have been made to maximize glycemic control; this strategy reduces certain manifestations of cardiovascular complications. There are drawbacks, however, as intensive glycemic control does not impart perennial protective benefits, and these efforts are not without potential adverse sequelae, such as hypoglycemic events. RECENT FINDINGS Here, the authors have focused on updates into key areas under study for mechanisms driving these cardiovascular disorders in diabetes, including roles for epigenetics and gene expression, interferon networks, and mitochondrial dysfunction. Updates on the cardioprotective roles of the new classes of hyperglycemia-targeting therapies, the sodium glucose transport protein 2 inhibitors and the agonists of the glucagon-like peptide 1 receptor system, are reviewed. In summary, insights from ongoing research and the cardioprotective benefits of the newer type 2 diabetes therapies are providing novel areas for therapeutic opportunities in diabetes and CVD.
Collapse
Affiliation(s)
- Raquel López-Díez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, Science Building, Room 615, New York, NY 10016 USA
| | - Lander Egaña-Gorroño
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, Science Building, Room 615, New York, NY 10016 USA
| | - Laura Senatus
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, Science Building, Room 615, New York, NY 10016 USA
| | - Alexander Shekhtman
- Department of Chemistry, The State University of New York at Albany, Albany, NY USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, Science Building, Room 615, New York, NY 10016 USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, Science Building, Room 615, New York, NY 10016 USA
| |
Collapse
|
37
|
Huaman MA, De Cecco CN, Bittencourt MS, Ticona E, Kityo C, Ballena I, Nalukwago S, Nazzinda R, Ticona C, Azañero R, Zhang B, Farquhar C, Hawn TR, Sterling TR, Fichtenbaum CJ, Longenecker CT. Latent Tuberculosis Infection and Subclinical Coronary Atherosclerosis in Peru and Uganda. Clin Infect Dis 2021; 73:e3384-e3390. [PMID: 33388766 DOI: 10.1093/cid/ciaa1934] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tuberculosis has been linked to an increased risk of atherosclerotic cardiovascular disease (ASCVD). We assessed whether latent tuberculosis infection (LTBI) is associated with subclinical coronary atherosclerosis in two TB-prevalent areas. METHODS We analyzed cross-sectional data from studies conducted in Lima, Peru, and Kampala, Uganda. Individuals ≥40 years old were included. We excluded persons with known history of ASCVD events or active TB. Participants underwent QuantiFERON®-TB (QFT) testing to define LTBI, and computed tomography angiography to examine coronary atherosclerosis. A Coronary Artery Disease-Reporting Data System (CAD-RADS) score ≥3 defined obstructive CAD (plaque causing ≥50% stenosis). RESULTS 113 persons with LTBI and 91 persons without LTBI were included. There were no significant differences between LTBI and non-LTBI participants in terms of age (median [interquartile range]; 56 [51-62] vs. 55 [49-64], p=0.829), male sex (38% vs. 42%; p=0.519), or 10-year ASCVD risk scores (7.1 [3.2-11.7] vs. 6.1 [2.8-10.8]; p=0.533). CAD prevalence (any plaque) was similar between groups (29% vs. 24%; p=0.421). Obstructive CAD was present in 9% of LTBI and 3% of non-LTBI individuals; p=0.095. LTBI was associated with obstructive CAD after adjusting for ASCVD risk score, HIV status, and study site (adjusted odds ratio, 4.96, 95% CI 1.05-23.44; p=0.043). Quantitative QFT TB antigen minus nil interferon-gamma responses were associated with obstructive CAD (adjusted odds ratio, 1.2, 95% CI 1.03-1.41; p=0.022). CONCLUSIONS LTBI was independently associated with an increased likelihood of subclinical obstructive CAD. Our data indicates that LTBI is a non-traditional correlate of ASCVD risk.
Collapse
Affiliation(s)
- Moises A Huaman
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati College of Medicine, Ohio, United States of America
| | - Carlo N De Cecco
- Division of Cardiothoracic Imaging, Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | | | - Eduardo Ticona
- Hospital Nacional Dos de Mayo, Lima, Peru.,Department of Internal Medicine, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Cissy Kityo
- Joint Clinical Research Centre, Kampala, Uganda
| | | | | | | | | | | | - Bin Zhang
- Division of Biostatistics and Epidemiology, Cincinnati Children's Medical Center, Ohio, United States of America
| | - Carey Farquhar
- Departments of Medicine and Global Health, University of Washington School of Medicine, Seattle, United States of America
| | - Thomas R Hawn
- Departments of Medicine and Global Health, University of Washington School of Medicine, Seattle, United States of America
| | - Timothy R Sterling
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University School of Medicine, United States of America
| | - Carl J Fichtenbaum
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati College of Medicine, Ohio, United States of America
| | - Chris T Longenecker
- Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Ohio, United States of America
| |
Collapse
|