1
|
Li M, Han J, Muegge C, Zollinger T, Zhou LY, Monahan P, Wessel J, Kleinschmidt V, Moffatt S, Nan H. Age, inflammation, alkaline phosphatase, and coronary artery calcification in firefighters. BMC Cardiovasc Disord 2025; 25:309. [PMID: 40269684 DOI: 10.1186/s12872-025-04750-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Firefighting involves exposure to hazardous conditions that may contribute to adverse long term health outcomes, including cardiovascular disease. While coronary artery disease (CAD) is a leading cause of morbidity among firefighters, the specific occupational contributions to Coronary Artery Calcification (CAC), a reliable predictor of CAD, are not well understood. METHODS We conducted a cross-sectional study involving 410 firefighters, aged 35-68, who underwent comprehensive health assessments, including CAC measurement using computed tomography. Multiple logistic regression models were built to examine the associations of demographic, lifestyle, and clinical variables with CAC score. RESULTS Our analysis revealed statistically significant associations between several clinical indicators and CAC score. Age (odds ratio (OR): 1.12; 95% confidence interval (95% CI): [1.05, 1.19]) and percentage of monocytes (OR: 1.29; 95% CI: [1.06, 1.58]) were positively correlated with higher CAC score, highlighting the role of inflammation in CAD among firefighters. Moreover, the enzyme alkaline phosphatase emerged as an independent predictor of CAC score (OR: 1.02; 95% CI: [1.01, 1.04]), suggesting a novel biomarker of cardiovascular risk in this population. CONCLUSION Our study identified several risk factors associated with increased CAC score in firefighters, including age, inflammation, and alkaline phosphatase. These findings underscore the importance of tailored health monitoring and interventions to mitigate CAD risk in firefighters, considering both general and occupation-specific risk factors. This study contributes to a better understanding of the occupational health challenges faced by firefighters and provides a foundation for future research and preventive strategies in this high-risk group.
Collapse
Affiliation(s)
- Mingyue Li
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Carolyn Muegge
- National Institute for Public Safety Health, Indianapolis, IN, USA
- College of Health Science, University of Indianapolis, Indianapolis, IN, USA
| | - Terrell Zollinger
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
| | - Laura Y Zhou
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Patrick Monahan
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jennifer Wessel
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
| | - Vanessa Kleinschmidt
- National Institute for Public Safety Health, Indianapolis, IN, USA
- Center for Global Health Equity, Indiana University, Indianapolis, IN, USA
| | - Steven Moffatt
- National Institute for Public Safety Health, Indianapolis, IN, USA
| | - Hongmei Nan
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
2
|
Long Z, Du J, Hu J, Xiao Y, Hou C. The prognostic value of the systemic immunity-inflammation index for cardiovascular and all-cause mortality in cardiovascular disease patients with diabetes or prediabetes. J Diabetes Investig 2025; 16:510-520. [PMID: 39718039 PMCID: PMC11871382 DOI: 10.1111/jdi.14383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND The relationship between the systemic immune-inflammatory index (SII) and the prognosis of cardiovascular disease (CVD) patients with diabetes or prediabetes remains uncertain. This study investigated the association between baseline SII and all-cause and cardiovascular mortality in American adults with CVD and diabetes or prediabetes. METHODS Our survey included 4,060 adults with cardiovascular disease and diabetes or prediabetes from the National Health and Nutrition Examination Survey (1998-2020). Using restricted cubic splines (RCS) based on Cox regression models and a two-piecewise Cox proportional hazards model for both sides of the inflection point, we elucidated the nonlinear relationship between baseline SII and mortality. Mediation analysis was used to explore the indirect impact of SII on mortality through eGFR. RESULTS In the median 129 months of follow-up, 620 people died from cardiovascular causes and 1,800 from all causes. In the CVD population with diabetes or prediabetes, SII showed a U-shaped relationship with all-cause mortality. The association between SII and CVD mortality was nonlinear and J-shaped. Stratified and interaction analysis confirmed the stability of the core results. Notably, eGFR partially mediated the association between SII and both all-cause and cardiovascular mortality by 9.4% and 6.9%, respectively. CONCLUSIONS SII revealed a U-shaped relationship with all-cause mortality (inflection point: lnSII = 6) and a J-shaped association with CVD mortality (inflection point: lnSII = 5.73) in CVD patients with diabetes or prediabetes among American patients. Thus, assessing the SII index may offer valuable insights into risk assessment and prognosis in patients with CVD who are diabetic or prediabetic.
Collapse
Affiliation(s)
- Zhengyi Long
- Key Laboratory Diseases Immunology, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Ministry of EducationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Jingyun Du
- Key Laboratory Diseases Immunology, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Ministry of EducationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Jie Hu
- Key Laboratory Diseases Immunology, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Ministry of EducationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Yang Xiao
- Key Laboratory Diseases Immunology, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Ministry of EducationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Can Hou
- Key Laboratory Diseases Immunology, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Ministry of EducationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| |
Collapse
|
3
|
Wu G, Wang J, Zhu W, Liu H, Ding W, Liu Y, Wang Z, Tao L. A causal link between circulating immune cells and erectile dysfunction: Two-sample bidirectional Mendelian randomization study. Medicine (Baltimore) 2024; 103:e41124. [PMID: 39969333 PMCID: PMC11688085 DOI: 10.1097/md.0000000000041124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/11/2024] [Indexed: 02/20/2025] Open
Abstract
Emerging observational studies have highlighted a robust association between circulating immune cells and erectile dysfunction (ED); however, these associations may be confounded by underlying factors. To elucidate this relationship, we conducted a comprehensive two-sample Mendelian randomization (MR) analysis to evaluate the potential causal links between circulating immune cell and ED. Utilizing large-scale genetic datasets from publicly available sources, we systematically investigated causal associations across 731 distinct immunophenotypes and ED risk, supported by rigorous sensitivity analyses to ensure robustness, evaluate heterogeneity, and detect horizontal pleiotropy. In forward MR analyses, 28 immune phenotypes were identified as having a causal association with ED. Notably, 3 specific phenotypes: CD25 + resting Treg cells, CD33dim HLA-DR + CD11b + antigen-presenting cells, and CD3- lymphocyte antigen-presenting cells, exhibited consistent significance across multiple MR methods, including inverse-variance weighted, MR-Egger, and weighted median analyses. Additionally, reverse MR analyses revealed that ED could causally influence 41 immune phenotypes, with 11 phenotypes showing consistent significance across the same analytical approaches. These findings underscore the complex bidirectional relationship between circulating immune cells and ED, providing critical insights into ED pathogenesis and potential targets for novel therapeutic interventions.
Collapse
Affiliation(s)
- Guanwei Wu
- Department of Urology, Affiliated Wuhu Hospital of East China Normal University, Wuhu, China
| | - Jiawei Wang
- Department of Urology, Affiliated Wuhu Hospital of East China Normal University, Wuhu, China
| | - Weiyu Zhu
- Department of Ultrasound, Affiliated Wuhu Hospital of East China Normal University, Wuhu, China
| | - Heqian Liu
- Department of Urology, Affiliated Wuhu Hospital of East China Normal University, Wuhu, China
| | - Wei Ding
- Department of Urology, Affiliated Wuhu Hospital of East China Normal University, Wuhu, China
| | - Yingqing Liu
- Department of Urology, Affiliated Wuhu Hospital of East China Normal University, Wuhu, China
| | - Zhonglang Wang
- Department of Urology, Affiliated Wuhu Hospital of East China Normal University, Wuhu, China
| | - Lingsong Tao
- Department of Urology, Affiliated Wuhu Hospital of East China Normal University, Wuhu, China
| |
Collapse
|
4
|
Zhao Z, Qin Y, Wu R, Li W, Dong Y. Single-cell analysis identified key macrophage subpopulations associated with atherosclerosis. Open Med (Wars) 2024; 19:20241088. [PMID: 39726810 PMCID: PMC11669903 DOI: 10.1515/med-2024-1088] [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: 07/17/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 12/28/2024] Open
Abstract
Background Atherosclerosis is a lipid-driven inflammatory disease characterized by plaque formation in major arteries. These plaques contain lipid-rich macrophages that accumulate through monocyte recruitment, local macrophage differentiation, and proliferation. Objective We identify the macrophage subsets that are closely related to atherosclerosis and reveal the key pathways in the progression of atherosclerotic disease. Materials and methods In this study, we characterize the single-cell landscape of atherosclerosis, identifying macrophage subsets closely related to the disease and revealing key pathways in its progression. Using analytical methods like CytoTRACE, Monocle2, Slingshot, and CellChat, we study macrophage differentiation and infer cell trajectory. Results The 8,417 macrophages were divided into six subtypes, macrophages: C0 C1QC+ macrophages, C1 SPP1+ macrophages, C2 FCN1+ macrophages, C3 IGKC+ macrophages, C4 FCER1A+ macrophages, C5CALD1+ macrophages. The results of gene set enrichment analysis, Monocle2, and Slingshot suggest that C2 FCN1+ macrophages may play an important role in the progression of atherosclerosis. C2 FCN1+ macrophages interact with endothelial cells via CCL, CXCL, APP, and other pathways to regulate the progression of atherosclerosis. Conclusion We identify a key macrophage subgroup (C2 FCN1+ macrophages) associated with atherosclerosis, which interacts with endothelial cells via CCL, CXCL, APP, and other pathways to regulate disease progression.
Collapse
Affiliation(s)
- Zhenzhen Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
- Department of Cardiovascular Disease, The Second Affiliated Hospital of Shandong University of Chinese Medicine, Jinan, 250001, China
| | - Yuelong Qin
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Rui Wu
- Pingyi County Hospital of Traditional Chinese Medicine Cardiology Department, Linyi, 273300, China
| | - Wenwu Li
- Department of Burn Plastic and Wound Repair Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Laboratory of the Atherosclerosis and Ischemic Cardiovascular Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yujiang Dong
- Department of Cardiovascular Disease, The Second Affiliated Hospital of Shandong University of Chinese Medicine, Jinan, 250001, China
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| |
Collapse
|
5
|
Panduga S, Vasishta S, Subramani R, Vincent S, Mutalik S, Joshi MB. Epidrugs in the clinical management of atherosclerosis: Mechanisms, challenges and promises. Eur J Pharmacol 2024; 980:176827. [PMID: 39038635 DOI: 10.1016/j.ejphar.2024.176827] [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: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Atherosclerosis is a complex and multigenic pathology associated with significant epigenetic reprogramming. Traditional factors (age, sex, obesity, hyperglycaemia, dyslipidaemia, hypertension) and non-traditional factors (foetal indices, microbiome alteration, clonal hematopoiesis, air pollution, sleep disorders) induce endothelial dysfunction, resulting in reduced vascular tone and increased vascular permeability, inflammation and shear stress. These factors induce paracrine and autocrine interactions between several cell types, including vascular smooth muscle cells, endothelial cells, monocytes/macrophages, dendritic cells and T cells. Such cellular interactions lead to tissue-specific epigenetic reprogramming regulated by DNA methylation, histone modifications and microRNAs, which manifests in atherosclerosis. Our review outlines epigenetic signatures during atherosclerosis, which are viewed as potential clinical biomarkers that may be adopted as new therapeutic targets. Additionally, we emphasize epigenetic modifiers referred to as 'epidrugs' as potential therapeutic molecules to correct gene expression patterns and restore vascular homeostasis during atherosclerosis. Further, we suggest nanomedicine-based strategies involving the use of epidrugs, which may selectively target cells in the atherosclerotic microenvironment and reduce off-target effects.
Collapse
Affiliation(s)
- Sushma Panduga
- Department of Biochemistry, Palamur Biosciences Private Limited, Hyderabad, 500026, Telangana, India; PhD Program, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Sampara Vasishta
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Ramamoorthy Subramani
- Department of Biochemistry, Palamur Biosciences Private Limited, Hyderabad, 500026, Telangana, India
| | - Sthevaan Vincent
- Department of Pathology, Palamur Biosciences Private Limited, Hyderabad, 500026, Telangana, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
6
|
Gunasena M, Alles M, Wijewantha Y, Mulhern W, Bowman E, Gabriel J, Kettelhut A, Kumar A, Weragalaarachchi K, Kasturiratna D, Horowitz JC, Scrape S, Pannu SR, Liu SL, Vilgelm A, Wijeratne S, Bednash JS, Demberg T, Funderburg NT, Liyanage NP. Synergy Between NK Cells and Monocytes in Potentiating Cardiovascular Disease Risk in Severe COVID-19. Arterioscler Thromb Vasc Biol 2024; 44:e243-e261. [PMID: 38989579 PMCID: PMC11448863 DOI: 10.1161/atvbaha.124.321085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Evidence suggests that COVID-19 predisposes to cardiovascular diseases (CVDs). While monocytes/macrophages play a central role in the immunopathogenesis of atherosclerosis, less is known about their immunopathogenic mechanisms that lead to CVDs during COVID-19. Natural killer (NK) cells, which play an intermediary role during pathologies like atherosclerosis, are dysregulated during COVID-19. Here, we sought to investigate altered immune cells and their associations with CVD risk during severe COVID-19. METHODS We measured plasma biomarkers of CVDs and determined phenotypes of circulating immune subsets using spectral flow cytometry. We compared these between patients with severe COVID-19 (severe, n=31), those who recovered from severe COVID-19 (recovered, n=29), and SARS-CoV-2-uninfected controls (controls, n=17). In vivo observations were supported using in vitro assays to highlight possible mechanistic links between dysregulated immune subsets and biomarkers during and after COVID-19. We performed multidimensional analyses of published single-cell transcriptome data of monocytes and NK cells during severe COVID-19 to substantiate in vivo findings. RESULTS During severe COVID-19, we observed alterations in cardiometabolic biomarkers including oxidized-low-density lipoprotein, which showed decreased levels in severe and recovered groups. Severe patients exhibited dysregulated monocyte subsets, including increased frequencies of proinflammatory intermediate monocytes (also observed in the recovered) and decreased nonclassical monocytes. All identified NK-cell subsets in the severe COVID-19 group displayed increased expression of activation and tissue-resident markers, such as CD69 (cluster of differentiation 69). We observed significant correlations between altered immune subsets and plasma oxidized-low-density lipoprotein levels. In vitro assays revealed increased uptake of oxidized-low-density lipoprotein into monocyte-derived macrophages in the presence of NK cells activated by plasma of patients with severe COVID-19. Transcriptome analyses confirmed enriched proinflammatory responses and lipid dysregulation associated with epigenetic modifications in monocytes and NK cells during severe COVID-19. CONCLUSIONS Our study provides new insights into the involvement of monocytes and NK cells in the increased CVD risk observed during and after COVID-19.
Collapse
Affiliation(s)
- Manuja Gunasena
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Mario Alles
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Yasasvi Wijewantha
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Will Mulhern
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Emily Bowman
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Janelle Gabriel
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Aaren Kettelhut
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Amrendra Kumar
- Department of pathology, College of Medicine, The Ohio State University
| | | | - Dhanuja Kasturiratna
- Department of Mathematics and Statistics, Northern Kentucky University, KY, Highland Heights, KY, USA
| | - Jeffrey C Horowitz
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Scott Scrape
- Department of pathology, College of Medicine, The Ohio State University
| | - Sonal R Pannu
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Shan-Lu Liu
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Anna Vilgelm
- Department of pathology, College of Medicine, The Ohio State University
| | - Saranga Wijeratne
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Joseph S Bednash
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Thorsten Demberg
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Nicholas T Funderburg
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Namal P.M. Liyanage
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| |
Collapse
|
7
|
Henni Mansour AS, Ragues M, Brevier J, Borowczyk C, Grevelinger J, Laroche-Traineau J, Garaude J, Marais S, Jacobin-Valat MJ, Gerbaud E, Clofent-Sanchez G, Ottones F. Phenotypic, Metabolic, and Functional Characterization of Experimental Models of Foamy Macrophages: Toward Therapeutic Research in Atherosclerosis. Int J Mol Sci 2024; 25:10146. [PMID: 39337629 PMCID: PMC11432604 DOI: 10.3390/ijms251810146] [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: 08/15/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Different types of macrophages (Mφ) are involved in atherogenesis, including inflammatory Mφ and foamy Mφ (FM). Our previous study demonstrated that two-photon excited fluorescence (TPEF) imaging of NADH and FAD autofluorescence (AF) could distinguish experimental models that mimic the different atherosclerotic Mφ types. The present study assessed whether optical differences correlated with phenotypic and functional differences, potentially guiding diagnostic and therapeutic strategies. Phenotypic differences were investigated using three-dimensional principal component analysis and multi-color flow cytometry. Functional analyses focused on cytokine production, metabolic profiles, and cellular oxidative stress, in LDL dose-dependent assays, to understand the origin of AF in the FAD spectrum and assess FM ability to transition toward an immunoregulatory phenotype and function. Phenotypic studies revealed that FM models generated with acetylated LDL (Mac) were closer to immunoregulatory Mφ, while those generated with oxidized LDL (Mox) more closely resembled inflammatory Mφ. The metabolic analysis confirmed that inflammatory Mφ primarily used glycolysis, while immunoregulatory Mφ mainly depended on mitochondrial respiration. FM models employed both pathways; however, FM models generated with high doses of modified LDL showed reduced mitochondrial respiration, particularly Mox FM. Thus, the high AF in the FAD spectrum in Mox was not linked to increased mitochondrial respiration, but correlated with the dose of oxidized LDL, leading to increased production of reactive oxygen species (ROS) and lysosomal ceroid accumulation. High FAD-like AF, ROS, and ceroid accumulation were reduced by incubation with α-tocopherol. The cytokine profiles supported the phenotypic analysis, indicating that Mox FM exhibited greater inflammatory activity than Mac FM, although both could be redirected toward immunoregulatory functions, albeit to different degrees. In conclusion, in the context of immunoregulatory therapies for atherosclerosis, it is crucial to consider FM, given their prevalence in plaques and our results, as potential targets, regardless of their inflammatory status, alongside non-foamy inflammatory Mφ.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sébastien Marais
- Bordeaux Imaging Center, BIC, UAR 3420, US 4, 33000 Bordeaux, France
| | | | - Edouard Gerbaud
- Centre de Recherche Cardio Thoracique, INSERM U 1045, 33000 Bordeaux, France
| | | | | |
Collapse
|
8
|
Chen Y, Lu S, Ren Y, Fan J, Bao CP, Zhang X, Shi YK, Wang Y, Yang LX. Integrating genomic profiling to clinical data: assessing the impact of CD147 expression on plaque stability. Front Cardiovasc Med 2024; 11:1425817. [PMID: 39355350 PMCID: PMC11444025 DOI: 10.3389/fcvm.2024.1425817] [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: 04/30/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024] Open
Abstract
Background Acute Coronary Syndrome (ACS) continues to be a leading cause of death and illness worldwide. Differentiating stable from unstable coronary plaques is essential for enhancing patient outcomes. This research investigates the role of CD147 as a biomarker for plaque stability among coronary artery disease patients. Methods The study began with high-throughput sequencing of blood samples from six patients, divided equally between those with Stable Angina (SA) and Unstable Angina (UA), followed by bioinformatics analysis. Expanding upon these findings, the study included 31 SA patients and 30 patients with ACS, using flow cytometry to examine CD147 expression on platelets and monocytes. Additionally, logistic regression was utilized to integrate traditional risk factors and evaluate the predictive value of CD147 expression for plaque stability. Results Initial sequencing displayed a notable difference in CD147 expression between SA and UA groups, with a significant increase in UA patients. Further analysis confirmed that elevated platelet CD147 expression was strongly associated with unstable plaques (OR = 277.81, P < .001), after adjusting for conventional risk factors, whereas monocyte CD147 levels did not show a significant difference. Conclusion Elevated CD147 expression on platelets is a crucial biomarker for identifying unstable coronary artery plaques, offering insights into patient risk stratification and the development of targeted treatment strategies. This underscores the pivotal role of molecular research in understanding and managing coronary artery disease, paving the way for improved clinical outcomes.
Collapse
Affiliation(s)
- Yu Chen
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, China
| | - Si Lu
- Department of Clinical Medical College, Dali University, Dali, China
| | - Yong Ren
- Department of Clinical Medical College, Dali University, Dali, China
| | - Jun Fan
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, China
| | - Chun-Ping Bao
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, China
| | - Xin Zhang
- Department of Pulmonary and Critical Care Medicine, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, China
| | - Yan-Kun Shi
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, China
| | - Yan Wang
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li-Xia Yang
- Department of Cardiology, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, China
| |
Collapse
|
9
|
Krishnaraj A, Dennis F, Teoh H, Verma S, Hess DA. Vascular regenerative cell content in South Asians: the key learnings. Curr Opin Cardiol 2024; 39:444-450. [PMID: 38847610 DOI: 10.1097/hco.0000000000001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
PURPOSE OF REVIEW We aim to provide a comprehensive examination of the literature linking elevated rates of cardiovascular disease (CVD) in individuals of South Asian ethnicity with the severity of circulating vascular regenerative cell exhaustion. RECENT FINDINGS Recent findings have demonstrated reduced bioavailability of pro-vascular progenitor cell subsets in individuals with T2D and obesity. Depletion of vascular regenerative cells in the bone marrow - coupled with decreased mobilization into circulation - can negatively impact the capacity for vascular repair and exacerbate CVD risk. Several recent studies have established that although South Asian individuals possess similar inflammatory cell burden compared with other ethnicities, they exhibit marked decreases in vessel regenerative hematopoietic progenitor cells and monocyte subsets. Validation of these findings and investigation the functional capacity of vascular regenerative cell subsets to mediate vessel repair is highly warranted. SUMMARY Vascular regenerative cells play a key role coordinating angiogenic and arteriogenic vessel remodelling. Recent studies have demonstrated that South Asian individuals with T2D show severe depletion in circulating vascular regenerative cell subsets. Because the reversal of vascular regenerative cell exhaustion by current glucose-lowering pharmaceutical agents has recently been documented, early intervention to bolster vascular regenerative cell content may prevent CVD co-morbidities in South Asian individuals with cardiometabolic disease.
Collapse
Affiliation(s)
- Aishwarya Krishnaraj
- Division of Cardiac Surgery
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital
- Department of Pharmacology and Toxicology, University of Toronto
| | - Fallon Dennis
- Division of Cardiac Surgery
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital
- Department of Pharmacology and Toxicology, University of Toronto
| | - Hwee Teoh
- Division of Cardiac Surgery
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital
- Division of Endocrinology and Metabolism, St. Michael's Hospital
| | - Subodh Verma
- Division of Cardiac Surgery
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital
- Department of Pharmacology and Toxicology, University of Toronto
- Department of Surgery, University of Toronto, Toronto
| | - David A Hess
- Division of Cardiac Surgery
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital
- Department of Pharmacology and Toxicology, University of Toronto
- Department of Physiology and Pharmacology, Western University
- Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| |
Collapse
|
10
|
Ge X, Slütter B, Lambooij JM, Zhou E, Ying Z, Agirman C, Heijink M, Rimbert A, Guigas B, Kuiper J, Müller C, Bracher F, Giera M, Kooijman S, Rensen PC, Wang Y, Schönke M. DHCR24 inhibitor SH42 increases desmosterol without preventing atherosclerosis development in mice. iScience 2024; 27:109830. [PMID: 38770137 PMCID: PMC11103367 DOI: 10.1016/j.isci.2024.109830] [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: 10/18/2023] [Revised: 02/29/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
The liver X receptor (LXR) is considered a therapeutic target for atherosclerosis treatment, but synthetic LXR agonists generally also cause hepatic steatosis and hypertriglyceridemia. Desmosterol, a final intermediate in cholesterol biosynthesis, has been identified as a selective LXR ligand that suppresses inflammation without inducing lipogenesis. Δ24-Dehydrocholesterol reductase (DHCR24) converts desmosterol into cholesterol, and we previously showed that the DHCR24 inhibitor SH42 increases desmosterol to activate LXR and attenuate experimental peritonitis and metabolic dysfunction-associated steatotic liver disease. Here, we aimed to evaluate the effect of SH42 on atherosclerosis development in APOE∗3-Leiden.CETP mice and low-density lipoproteins (LDL) receptor knockout mice, models for lipid- and inflammation-driven atherosclerosis, respectively. In both models, SH42 increased desmosterol without affecting plasma lipids. While reducing liver lipids in APOE∗3-Leiden.CETP mice, and regulating populations of circulating monocytes in LDL receptor knockout mice, SH42 did not attenuate atherosclerosis in either model.
Collapse
Affiliation(s)
- Xiaoke Ge
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Bram Slütter
- Div. of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Leiden 2333 AL, the Netherlands
| | - Joost M. Lambooij
- Department of Parasitology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Enchen Zhou
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Zhixiong Ying
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Ceren Agirman
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Marieke Heijink
- The Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, France
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Johan Kuiper
- Div. of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Leiden 2333 AL, the Netherlands
| | - Christoph Müller
- Department of Pharmacy, Center for Drug Research, Ludwig Maximilians Universität München, 80539 Munich, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Ludwig Maximilians Universität München, 80539 Munich, Germany
| | - Martin Giera
- The Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Patrick C.N. Rensen
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Yanan Wang
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
- Med-X institute, Center for Immunological and Metabolic Diseases, and Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an 710061, China
| | - Milena Schönke
- Department of Medicine, Div. of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| |
Collapse
|
11
|
Liu S, Zhang B, Zhou J, Lv J, Zhang J, Li X, Yang W, Guo Y. Inhibition of differentiation of monocyte-derived macrophages toward an M2-Like phenotype May Be a neglected mechanism of β-AR receptor blocker therapy for atherosclerosis. Front Pharmacol 2024; 15:1378787. [PMID: 38903990 PMCID: PMC11188457 DOI: 10.3389/fphar.2024.1378787] [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: 01/30/2024] [Accepted: 05/13/2024] [Indexed: 06/22/2024] Open
Abstract
The clinical efficacy of adrenergic β-receptor (β-AR) blockers in significantly stabilizing atherosclerotic plaques has been extensively supported by evidence-based medical research; however, the underlying mechanism remains unclear. Recent findings have highlighted the impact of lipid-induced aberrant polarization of macrophages during normal inflammatory-repair and regenerative processes on atherosclerosis formation and progression. In this review, we explore the relationship between macrophage polarization and atherosclerosis, as well as the influence of β-AR blockers on macrophage polarization. Based on the robust evidence supporting the use of β-AR blockers for treating atherosclerosis, we propose that their main mechanism involves inhibiting monocyte-derived macrophage differentiation towards an M2-like phenotype.
Collapse
Affiliation(s)
| | | | - Jingqun Zhou
- Affiliated Renhe Hospital, China Three Gorges University, Yichang, China
| | | | | | | | | | | |
Collapse
|
12
|
Wei J, Shen S, Tian Y, Kang P, Sun G. Correlation Analysis of Macrophage Distribution and Pathological Features of Carotid Atherosclerotic Plaque. Ann Vasc Surg 2024; 98:355-364. [PMID: 37852365 DOI: 10.1016/j.avsg.2023.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/24/2023] [Accepted: 08/22/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Macrophages play an important role in maintaining the chronic inflammatory of atherosclerosis (AS) and are hallmark of atherosclerotic plaques. They differentiate into different subpopulations under the influence of oxidized lipids and cytokines and play different roles in the formation and development of plaque. To explore the differences in the amount and distribution of different macrophage subpopulations around different carotid plaque pathological features in human AS, and based on these results, to explore the correlation between some macrophage subpopulations and AS pathological features. METHODS First, we analyzed the single cells RNA-sequence data from the Gene Expression Omnibus DataSets (GSE159677). Second, we investigated the distribution difference of macrophage subpopulations in 61 surgically resected AS plaques by markers staining include CD68, inducible nitric oxide synthase, Arg-1, CD163 and HO-1. RESULTS The result of single cells RNA-Sequence analysis showed that there were a large number of macrophages infiltrated in AS and they can be categorized into different subpopulations with different transcriptional features and functions; moreover in different part of AS (calcified AS core versus proximal adjacent), the total number and subpopulation ratios were all different. The result of staining analysis showed that macrophages mainly distributed in some pathological lesions such as necrosis, fibrous tissue degeneration, cholesterol crystallization etc., and different subpopulations were distributed differently in these lesions. CONCLUSIONS This study confirmed that macrophages were heavily infiltrated in atherosclerotic plaques, and there existed subtype variability in different pathological lesions; meanwhile, these results suggested that different macrophage subpopulations may contribute differently in different pathological lesions.
Collapse
Affiliation(s)
- Jianhui Wei
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China; Department of Neurosurgery, Harrison International Peace Hospital, Hengshiu, People's Republic of China
| | - Shaoping Shen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yanan Tian
- Department of Neurosurgery, Harrison International Peace Hospital, Hengshiu, People's Republic of China
| | - Pengpeng Kang
- Department of Pathology, Harrison International Peace Hospital, Hengshiu, People's Republic of China
| | - Guozhu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China.
| |
Collapse
|
13
|
Jung I, Cho YJ, Park M, Park K, Lee SH, Kim WH, Jeong H, Lee JE, Kim GY. Proteomic analysis reveals activation of platelet- and fibrosis-related pathways in hearts of ApoE -/- mice exposed to diesel exhaust particles. Sci Rep 2023; 13:22636. [PMID: 38114606 PMCID: PMC10730529 DOI: 10.1038/s41598-023-49790-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023] Open
Abstract
Air pollution is an environmental risk factor linked to multiple human diseases including cardiovascular diseases (CVDs). While particulate matter (PM) emitted by diesel exhaust damages multiple organ systems, heart disease is one of the most severe pathologies affected by PM. However, the in vivo effects of diesel exhaust particles (DEP) on the heart and the molecular mechanisms of DEP-induced heart dysfunction have not been investigated. In the current study, we attempted to identify the proteomic signatures of heart fibrosis caused by diesel exhaust particles (DEP) in CVDs-prone apolipoprotein E knockout (ApoE-/-) mice model using tandem mass tag (TMT)-based quantitative proteomic analysis. DEP exposure induced mild heart fibrosis in ApoE-/- mice compared with severe heart fibrosis in ApoE-/- mice that were treated with CVDs-inducing peptide, angiotensin II. TMT-based quantitative proteomic analysis of heart tissues between PBS- and DEP-treated ApoE-/- mice revealed significant upregulation of proteins associated with platelet activation and TGFβ-dependent pathways. Our data suggest that DEP exposure could induce heart fibrosis, potentially via platelet-related pathways and TGFβ induction, causing cardiac fibrosis and dysfunction.
Collapse
Affiliation(s)
- Inkyo Jung
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeng2-ro, Osong-eub, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28159, Republic of Korea
| | - Yoon Jin Cho
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Department of Chemistry, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-gu, Seoul, 04310, Republic of Korea
| | - Minhan Park
- School of Earth Science and Environmental Engineering, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Kihong Park
- School of Earth Science and Environmental Engineering, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Seung Hee Lee
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeng2-ro, Osong-eub, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28159, Republic of Korea
| | - Won-Ho Kim
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeng2-ro, Osong-eub, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28159, Republic of Korea
| | - Hyuk Jeong
- Department of Chemistry, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-gu, Seoul, 04310, Republic of Korea
| | - Ji Eun Lee
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| | - Geun-Young Kim
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeng2-ro, Osong-eub, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28159, Republic of Korea.
| |
Collapse
|
14
|
Song J, Zhang Y, Frieler RA, Andren A, Wood S, Tyrrell DJ, Sajjakulnukit P, Deng JC, Lyssiotis CA, Mortensen RM, Salmon M, Goldstein DR. Itaconate suppresses atherosclerosis by activating a Nrf2-dependent antiinflammatory response in macrophages in mice. J Clin Invest 2023; 134:e173034. [PMID: 38085578 PMCID: PMC10849764 DOI: 10.1172/jci173034] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024] Open
Abstract
Itaconate has emerged as a critical immunoregulatory metabolite. Here, we examined the therapeutic potential of itaconate in atherosclerosis. We found that both itaconate and the enzyme that synthesizes it, aconitate decarboxylase 1 (Acod1, also known as immune-responsive gene 1 [IRG1]), are upregulated during atherogenesis in mice. Deletion of Acod1 in myeloid cells exacerbated inflammation and atherosclerosis in vivo and resulted in an elevated frequency of a specific subset of M1-polarized proinflammatory macrophages in the atherosclerotic aorta. Importantly, Acod1 levels were inversely correlated with clinical occlusion in atherosclerotic human aorta specimens. Treating mice with the itaconate derivative 4-octyl itaconate attenuated inflammation and atherosclerosis induced by high cholesterol. Mechanistically, we found that the antioxidant transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2), was required for itaconate to suppress macrophage activation induced by oxidized lipids in vitro and to decrease atherosclerotic lesion areas in vivo. Overall, our work shows that itaconate suppresses atherogenesis by inducing Nrf2-dependent inhibition of proinflammatory responses in macrophages. Activation of the itaconate pathway may represent an important approach to treat atherosclerosis.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yanling Zhang
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Ryan A. Frieler
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Anthony Andren
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sherri Wood
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel J. Tyrrell
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- University of Michigan Rogel Cancer Center
| | - Jane C. Deng
- Graduate Program in Immunology, and
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Richard M. Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes
| | | | - Daniel R. Goldstein
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Graduate Program in Immunology, and
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
15
|
Gunasena M, Alles M, Wijewantha Y, Mulhern W, Bowman E, Gabriel J, Kettelhut A, Kumar A, Weragalaarachchi K, Kasturiratna D, Horowitz JC, Scrape S, Pannu SR, Liu SL, Vilgelm A, Wijeratne S, Bednash JS, Demberg T, Funderburg NT, Liyanage NPM. Synergistic Role of NK Cells and Monocytes in Promoting Atherogenesis in Severe COVID-19 Patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.10.23298322. [PMID: 37986806 PMCID: PMC10659469 DOI: 10.1101/2023.11.10.23298322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Clinical data demonstrate an increased predisposition to cardiovascular disease (CVD) following severe COVID-19 infection. This may be driven by a dysregulated immune response associated with severe disease. Monocytes and vascular tissue resident macrophages play a critical role in atherosclerosis, the main pathology leading to ischemic CVD. Natural killer (NK) cells are a heterogenous group of cells that are critical during viral pathogenesis and are known to be dysregulated during severe COVID-19 infection. Their role in atherosclerotic cardiovascular disease has recently been described. However, the contribution of their altered phenotypes to atherogenesis following severe COVID-19 infection is unknown. We demonstrate for the first time that during and after severe COVID-19, circulating proinflammatory monocytes and activated NK cells act synergistically to increase uptake of oxidized low-density lipoprotein (Ox-LDL) into vascular tissue with subsequent foam cell generation leading to atherogenesis despite recovery from acute infection. Our data provide new insights, revealing the roles of monocytes/macrophages, and NK cells in COVID-19-related atherogenesis.
Collapse
|
16
|
Mirza Z, Al-Saedi DA, Saddeek S, Almowallad S, AlMassabi RF, Huwait E. Atheroprotective Effect of Fucoidan in THP-1 Macrophages by Potential Upregulation of ABCA1. Biomedicines 2023; 11:2929. [PMID: 38001931 PMCID: PMC10669811 DOI: 10.3390/biomedicines11112929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
Targeting foam cells reduces the risk and pathophysiology of atherosclerosis, of which they are one of its early hallmarks. The precise mechanism of action of fucoidan, a potential anti-atherogenic drug, is still unknown. Our objective was to assess the ability of fucoidan to regulate expression of ATP-binding cassette transporter A1 (ABCA1) in ox-LDL-induced THP-1 macrophages. Molecular docking was used to predict how fucoidan interacts with anti-foam cell markers, and further in vitro experiments were performed to evaluate the protective effect of fucoidan on modulating uptake and efflux of lipids. THP-1 macrophages were protected by 50 µg/mL of fucoidan and were then induced to form foam cells with 25 µg/mL of ox-LDL. Expression levels were assessed using RT-qPCR, and an Oil Red O stain was used to observe lipid accumulation in THP-1 macrophages. In addition, ABCA1 protein was examined by Western blot, and cellular cholesterol efflux was determined using fluorescently labeled cholesterol. Under a light microscope, decreased lipid accumulation in ox-LDL-induced-THP-1 macrophages pre-treated with fucoidan showed a significant effect, although it did not affect the expression of scavenger receptors (SR-AI and CD36). It is interesting to note that fucoidan dramatically increased the gene and protein expression of ABCA1, perhaps via the liver X receptor-α (LXR-α). Moreover, fucoidan's ability to increase and control the efflux of cholesterol from ox-LDL-induced THP-1 macrophages revealed how it may alter ABCA1's conformation and have a major effect on how it interacts with apolipoprotein A (ApoA1). In vitro results support a rationale for predicting fucoidan and its interaction with its receptor targets' predicted data, hence validating its anti-atherogenic properties and suggesting that fucoidan could be promising as an atheroprotective.
Collapse
Affiliation(s)
- Zeenat Mirza
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Dalal A. Al-Saedi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Cell Culture Lab, Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Salma Saddeek
- Department of Chemistry, Faculty of Sciences, University of Hafr Al Batin, Hafr Al Batin 39511, Saudi Arabia;
| | - Sanaa Almowallad
- Department of Biochemistry, Faculty of Sciences, University of Tabuk, Tabuk 48322, Saudi Arabia (R.F.A.)
| | - Rehab F. AlMassabi
- Department of Biochemistry, Faculty of Sciences, University of Tabuk, Tabuk 48322, Saudi Arabia (R.F.A.)
| | - Etimad Huwait
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Cell Culture Lab, Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
17
|
Kotlyarov SN, Suchkov IA, Uryas'yev OM, Yakusheva EN, Shchulkin AV, Kotlyarova AA. Analysis of Influence of Cigarette Smoke on Signaling Pathways of Innate Immune System in Monocytes of Peripheral Blood. I.P. PAVLOV RUSSIAN MEDICAL BIOLOGICAL HERALD 2023; 31:391-404. [DOI: 10.17816/pavlovj306495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
INTRODUCTION: Tobacco smoking is an important medical problem since it has a significant impact on the development and progression of chronic obstructive pulmonary disease (COPD). The components of tobacco smoke can initiate and support local and systemic inflammation with participation of monocytes and macrophages.
AIM: To study molecular mechanisms associated with the impact of cigarette smoke on signaling pathways of the innate immune system in monocytes of peripheral blood.
MATERIALS AND METHODS: The methods of in silico analysis was used to identify genes associated with the impact of tobacco smoke. On the basis of the data obtained, a cellular model of inflammation was created in vitro using tobacco smoke extract and monocytes of peripheral blood isolated by immunomagnetic separation. An enzyme-linked immunoassay (ELISA) kit was used to measure the concentration of tumor necrosis factor- (TNF-), interleukin-1 (IL-1) in cell supernatants, and of Toll-like receptor 4 (TLR4), ATP-binding cassette A1 (ABCA1) in homogenates of cell membranes of native monocytes and monocytes exposed to 4% tobacco smoke extract. These data were compared with the levels of TNF-, IL-1, TLR4 and ABCA1 in monocytes of peripheral blood of patients with COPD with frequent exacerbation phenotype and with obliterating atherosclerosis of lower limb arteries (OALLA). For statistical processing and visualization of the data, MedCalc 20.1.4 and R (version 4.2.2) software was used.
RESULTS: Tobacco smoke influences TLR4, TNF- signaling pathways and lipid metabolism. Cigarette smoke extract enhanced the expression of proinflammatory cytokines TNF- and IL-1 in cell supernatants, increased the level of TLR4 and decreased that of ABCA1 in plasmolemma of monocytes of peripheral blood. In patients with COPD with frequent exacerbation phenotype and with OALLA, there were shown increase in the levels of proinflammatory TNF- and IL-1 cytokines in cell supernatants, increase in the level of TLR4 and reduction of the level of ABCA1 in plasmolemma of monocytes of peripheral blood compared to native monocytes of healthy individuals.
CONCLUSION: Cigarette smoke enhances the production of proinflammatory TNF- and IL-1 cytokines, increases the levels of TLR4 protein and reduces the amount of ABCA1 transporter in membranes of monocytes of peripheral blood. This may partially explain the cause of the influence of cigarette smoke on development of the pulmonary and cardiovascular diseases. COPD with frequent exacerbation phenotype and OALLA are characterized by enhancement of inflammation with participation of monocytes.
Collapse
|
18
|
Afzoon S, Amiri MA, Mohebbi M, Hamedani S, Farshidfar N. A systematic review of the impact of Porphyromonas gingivalis on foam cell formation: Implications for the role of periodontitis in atherosclerosis. BMC Oral Health 2023; 23:481. [PMID: 37442956 PMCID: PMC10347812 DOI: 10.1186/s12903-023-03183-9] [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: 04/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND The current literature suggests the significant role of foam cells in the initiation of atherosclerosis through the formation of a necrotic core in atherosclerotic plaques. Moreover, an important periodontal pathogen called Porphyromonas gingivalis (P. gingivalis) is indicated to play a significant role in this regard. Thus, the aim of this systematic review was to comprehensively study the pathways by which P. gingivalis as a prominent bacterial species in periodontal disease, can induce foam cells that would initiate the process of atherosclerosis formation. METHODS An electronic search was undertaken in three databases (Pubmed, Scopus, and Web of Science) to identify the studies published from January 2000 until March 2023. The risk of bias in each study was also assessed using the QUIN risk of bias assessment tool. RESULTS After the completion of the screening process, 11 in-vitro studies met the inclusion criteria and were included for further assessments. Nine of these studies represented a medium risk of bias, while the other two had a high risk of bias. All of the studies have reported that P. gingivalis can significantly induce foam cell formation by infecting the macrophages and induction of oxidized low-density lipoprotein (oxLDL) uptake. This process is activated through various mediators and pathways. The most important factors in this regard are the lipopolysaccharide of P. gingivalis and its outer membrane vesicles, as well as the changes in the expression rate of transmembrane lipid transportation channels, including transient receptor potential channel of the vanilloid subfamily 4 (TRPV4), lysosomal integral protein 2 (LIMP2), CD36, etc. The identified molecular pathways involved in this process include but are not limited to NF-κB, ERK1/2, p65. CONCLUSION Based on the results of this study, it can be concluded that P. gingivalis can effectively promote foam cell formation through various pathogenic elements and this bacterial species can affect the expression rate of various genes and the function of specific receptors in the cellular and lysosomal membranes. However, due to the moderate to high level of risk of bias among the studies, further studies are required in this regard.
Collapse
Affiliation(s)
- Saeed Afzoon
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Amin Amiri
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mostafa Mohebbi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahram Hamedani
- Oral and Dental Disease Research Center, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nima Farshidfar
- Orthodontic Research Center, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
19
|
In Het Panhuis W, Schönke M, Modder M, Tom HE, Lalai RA, Pronk ACM, Streefland TCM, van Kerkhof LWM, Dollé MET, Depuydt MAC, Bot I, Vos WG, Bosmans LA, van Os BW, Lutgens E, Rensen PCN, Kooijman S. Time-restricted feeding attenuates hypercholesterolaemia and atherosclerosis development during circadian disturbance in APOE∗3-Leiden.CETP mice. EBioMedicine 2023; 93:104680. [PMID: 37356205 DOI: 10.1016/j.ebiom.2023.104680] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Circadian disturbance (CD) is the consequence of a mismatch between endogenous circadian rhythms, behaviour, and/or environmental cycles, and frequently occurs during shift work. Shift work has been associated with elevated risk for atherosclerotic cardiovascular disease (asCVD) in humans, but evidence for the effectiveness of prevention strategies is lacking. METHODS Here, we applied time-restricted feeding (TRF) as a strategy to counteract atherosclerosis development during CD in female APOE∗3-Leiden.CETP mice, a well-established model for humanized lipoprotein metabolism. Control groups were subjected to a fixed 12:12 h light-dark cycle, while CD groups were subjected to 6-h phase advancement every 3 days. Groups had either ad libitum (AL) access to food or were subjected to TRF with restricted food access to the dark phase. FINDINGS TRF did not prevent the increase in the relative abundance of circulating inflammatory monocytes and elevation of (postprandial) plasma triglycerides during CD. Nonetheless, TRF reduced atherosclerotic lesion size and prevented an elevation in macrophage content of atherosclerotic lesions during CD, while it increased the relative abundance of anti-inflammatory monocytes, prevented activation of T cells, and lowered plasma total cholesterol levels and markers of hepatic cholesterol synthesis. These effects were independent of total food intake. INTERPRETATION We propose that time restricted eating could be a promising strategy for the primary prevention of asCVD risk in shift workers, which warrants future study in humans. FUNDING This work was funded by the Novo Nordisk Foundation, the Netherlands Ministry of Social Affairs and Employment, Amsterdam Cardiovascular Sciences, and the Dutch Heart Foundation.
Collapse
Affiliation(s)
- Wietse In Het Panhuis
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Melanie Modder
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Hannah E Tom
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Reshma A Lalai
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Trea C M Streefland
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Linda W M van Kerkhof
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Martijn E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Marie A C Depuydt
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, the Netherlands
| | - Ilze Bot
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, the Netherlands
| | - Winnie G Vos
- Department of Medical Biochemistry, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Immunity and Infection, Amsterdam, the Netherlands
| | - Laura A Bosmans
- Department of Medical Biochemistry, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Immunity and Infection, Amsterdam, the Netherlands
| | - Bram W van Os
- Department of Medical Biochemistry, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Immunity and Infection, Amsterdam, the Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Immunity and Infection, Amsterdam, the Netherlands; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
20
|
Alalawi S, Albalawi F, Ramji DP. The Role of Punicalagin and Its Metabolites in Atherosclerosis and Risk Factors Associated with the Disease. Int J Mol Sci 2023; 24:ijms24108476. [PMID: 37239823 DOI: 10.3390/ijms24108476] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ACVD) is the leading cause of death worldwide. Although current therapies, such as statins, have led to a marked reduction in morbidity and mortality from ACVD, they are associated with considerable residual risk for the disease together with various adverse side effects. Natural compounds are generally well-tolerated; a major recent goal has been to harness their full potential in the prevention and treatment of ACVD, either alone or together with existing pharmacotherapies. Punicalagin (PC) is the main polyphenol present in pomegranates and pomegranate juice and demonstrates many beneficial actions, including anti-inflammatory, antioxidant, and anti-atherogenic properties. The objective of this review is to inform on our current understanding of the pathogenesis of ACVD and the potential mechanisms underlying the beneficial actions of PC and its metabolites in the disease, including the attenuation of dyslipidemia, oxidative stress, endothelial cell dysfunction, foam cell formation, and inflammation mediated by cytokines and immune cells together with the regulation of proliferation and migration of vascular smooth muscle cells. Some of the anti-inflammatory and antioxidant properties of PC and its metabolites are due to their strong radical-scavenging activities. PC and its metabolites also inhibit the risk factors of atherosclerosis, including hyperlipidemia, diabetes mellitus, inflammation, hypertension, obesity, and non-alcoholic fatty liver disease. Despite the promising findings that have emerged from numerous in vitro, in vivo, and clinical studies, deeper mechanistic insights and large clinical trials are required to harness the full potential of PC and its metabolites in the prevention and treatment of ACVD.
Collapse
Affiliation(s)
- Sulaiman Alalawi
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Faizah Albalawi
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| |
Collapse
|
21
|
Xie L, Chen J, Wang Y, Jin C, Xie Y, Ma H, Xiang M. Emerging roles of macrophages in heart failure and associated treatment approaches. Ther Adv Chronic Dis 2023; 14:20406223231168755. [PMID: 37152348 PMCID: PMC10155014 DOI: 10.1177/20406223231168755] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/23/2023] [Indexed: 05/09/2023] Open
Abstract
Heart failure is typically caused by different cardiovascular conditions and has a poor prognosis. Despite the advances in treatment in recent decades, heart failure has remained a major cause of morbidity and mortality worldwide. As revealed by in vivo and in vitro experiments, inflammation plays a crucial role in adverse cardiac remodeling, ultimately leading to heart failure. Macrophages are central to the innate immune system, and they are the most indispensable cell type for all cardiac injuries and remodeling stages. The immediate microenvironment regulates their polarization and secretion. In this review, we summarize the phenotypic heterogeneity and governing roles of macrophages in the infarcted, inflamed, and aging heart and assess their significance as potential therapeutic targets in heart failure. We also highlight the current missing links and major challenges in the field that remain to be addressed before macrophages can be exploited for therapeutic applications.
Collapse
Affiliation(s)
- Lan Xie
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinyong Chen
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yidong Wang
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengjiang Jin
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Ma
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou 310009,
China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou 310009,
China
| |
Collapse
|
22
|
Yu L, Zhang Y, Liu C, Wu X, Wang S, Sui W, Zhang Y, Zhang C, Zhang M. Heterogeneity of macrophages in atherosclerosis revealed by single-cell RNA sequencing. FASEB J 2023; 37:e22810. [PMID: 36786718 DOI: 10.1096/fj.202201932rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/15/2023]
Abstract
Technology at the single-cell level has advanced dramatically in characterizing molecular heterogeneity. These technologies have enabled cell subtype diversity to be seen in all tissues, including atherosclerotic plaques. Critical in atherosclerosis pathogenesis and progression are macrophages. Previous studies have only determined macrophage phenotypes within the plaque, mainly by bulk analysis. However, recent progress in single-cell technologies now enables the comprehensive mapping of macrophage subsets and phenotypes present in plaques. In this review, we have updated and discussed the definition and classification of macrophage subsets in mice and humans using single-cell RNA sequencing. We summarized the different classification methods and perspectives: traditional classification with an updated scoring system, inflammatory macrophages, foamy macrophages, and atherosclerotic-resident macrophages. In addition, some special types of macrophages were identified by specific markers, including IFN-inducible and cavity macrophages. Furthermore, we discussed macrophage subset-specific markers and their functions. In the future, these novel insights into the characteristics and phenotypes of these macrophage subsets within atherosclerotic plaques can provide additional therapeutic targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Liwen Yu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yujie Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changhao Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao Wu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shasha Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenhai Sui
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
23
|
O’Morain VL, Chen J, Plummer SF, Michael DR, Ramji DP. Anti-Atherogenic Actions of the Lab4b Consortium of Probiotics In Vitro. Int J Mol Sci 2023; 24:ijms24043639. [PMID: 36835055 PMCID: PMC9964490 DOI: 10.3390/ijms24043639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Probiotic bacteria have many protective effects against inflammatory disorders, though the mechanisms underlying their actions are poorly understood. The Lab4b consortium of probiotics contains four strains of lactic acid bacteria and bifidobacteria that are reflective of the gut of newborn babies and infants. The effect of Lab4b on atherosclerosis, an inflammatory disorder of the vasculature, has not yet been determined and was investigated on key processes associated with this disease in human monocytes/macrophages and vascular smooth muscle cells in vitro. The Lab4b conditioned medium (CM) attenuated chemokine-driven monocytic migration, monocyte/macrophage proliferation, uptake of modified LDL and macropinocytosis in macrophages together with the proliferation and platelet-derived growth factor-induced migration of vascular smooth muscle cells. The Lab4b CM also induced phagocytosis in macrophages and cholesterol efflux from macrophage-derived foam cells. The effect of Lab4b CM on macrophage foam cell formation was associated with a decrease in the expression of several key genes implicated in the uptake of modified LDL and induced expression of those involved in cholesterol efflux. These studies reveal, for the first time, several anti-atherogenic actions of Lab4b and strongly implicate further studies in mouse models of the disease in vivo and in clinical trials.
Collapse
Affiliation(s)
- Victoria L. O’Morain
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Jing Chen
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Sue F. Plummer
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, UK
| | - Daryn R. Michael
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, UK
| | - Dipak P. Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
- Correspondence: ; Tel.: +44-(0)29-20876753
| |
Collapse
|
24
|
Dorenkamp M, Nasiry M, Semo D, Koch S, Löffler I, Wolf G, Reinecke H, Godfrey R. Pharmacological Targeting of the RAGE-NFκB Signalling Axis Impedes Monocyte Activation under Diabetic Conditions through the Repression of SHP-2 Tyrosine Phosphatase Function. Cells 2023; 12:cells12030513. [PMID: 36766855 PMCID: PMC9914555 DOI: 10.3390/cells12030513] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 02/09/2023] Open
Abstract
Monocytes play a vital role in the development of cardiovascular diseases. Type 2 diabetes mellitus (T2DM) is a major CVD risk factor, and T2DM-induced aberrant activation and enhanced migration of monocytes is a vital pathomechanism that leads to atherogenesis. We recently reported the upregulation of SHP-2 phosphatase expression in mediating the VEGF resistance of T2DM patient-derived monocytes or methylglyoxal- (MG, a glucose metabolite and advanced glycation end product (AGE) precursor) treated monocytes. However, the exact mechanisms leading to SHP-2 upregulation in hyperglycemic monocytes are unknown. Since inflammation and accumulation of AGEs is a hallmark of T2DM, we hypothesise that inflammation and AGE-RAGE (Receptor-for-AGEs) signalling drive SHP-2 expression in monocytes and blockade of these pathways will repress SHP-2 function. Indeed, monocytes from T2DM patients revealed an elevated SHP-2 expression. Under normoglycemic conditions, the serum from T2DM patients strongly induced SHP-2 expression, indicating that the T2DM serum contains critical factors that directly regulate SHP-2 expression. Activation of pro-inflammatory TNFα signalling cascade drove SHP-2 expression in monocytes. In line with this, linear regression analysis revealed a significant positive correlation between TNFα expression and SHP-2 transcript levels in T2DM monocytes. Monocytes exposed to MG or AGE mimetic AGE-BSA, revealed an elevated SHP-2 expression and co-treatment with an NFκB inhibitor or genetic inhibition of p65 reversed it. The pharmacological inhibition of RAGE was sufficient to block MG- or AGE-BSA-induced SHP-2 expression and activity. Confirming the importance of RAGE-NFκB signalling in regulating SHP-2 expression, the elevated binding of NFκB to the SHP-2 promoter-induced by MG or AGE-BSA-was reversed by RAGE and NFκB inhibition. Besides, we detected elevated RAGE levels in human and murine T2DM monocytes and monocytes exposed to MG or AGE-BSA. Importantly, MG and AGE-BSA treatment of non-T2DM monocytes phenocopied the aberrant pro-migratory phenotype of T2DM monocytes, which was reversed entirely by either SHP-2- or RAGE inhibition. In conclusion, these findings suggest a new therapeutic approach to prevent accelerated atherosclerosis in T2DM patients since inhibiting the RAGE-NFκB-SHP-2 axis impeded the T2DM-driven, SHP-2-dependent monocyte activation.
Collapse
Affiliation(s)
- Marc Dorenkamp
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Madina Nasiry
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Dilvin Semo
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Sybille Koch
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Ivonne Löffler
- Department of Internal Medicine III, University Hospital Jena, 07743 Jena, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, 07743 Jena, Germany
| | - Holger Reinecke
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Rinesh Godfrey
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
- Correspondence: ; Tel.: +49-251-83-57089; Fax: +49-251-83-55747
| |
Collapse
|
25
|
Shin U, You H, Lee GY, Son Y, Han SN. The effects of 1,25(OH) 2D 3 treatment on metabolic reprogramming and maturation in bone marrow-derived dendritic cells from control and diabetic mice. J Steroid Biochem Mol Biol 2023; 225:106197. [PMID: 36183994 DOI: 10.1016/j.jsbmb.2022.106197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/17/2022] [Accepted: 09/26/2022] [Indexed: 02/01/2023]
Abstract
Activated dendritic cells (DCs) undergo significant metabolic reprogramming, which is characterized by an increase in aerobic glycolysis and a concurrent progressive loss of oxidative phosphorylation. The modulation of metabolic reprogramming is believed to be closely related to the function of DCs. Vitamin D has been reported to inhibit the maturation of DCs. DC dysfunction has been reported in diabetic patients, and hyperglycemia is associated with impaired glycolytic metabolism in immune cells. Therefore, vitamin D and diabetes may affect intracellular metabolism, thereby regulating the activity of DCs. We investigated the effect of in vitro treatment of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) on metabolic reprogramming and maturation of bone marrow-derived dendritic cells (BMDCs) from diabetic mouse. Six-week-old male C57BLKS/J-m+/m+ mice (CON) and C57BLKS/J-db/db mice (db/db) were fed with a 10% kcal fat diet for seven weeks. BMDCs were generated by culturing bone marrow cells from the mice with rmGM-CSF (20 ng/mL) in the absence or presence of 10 nM 1,25(OH)2D3. The maturation of BMDCs was induced via lipopolysaccharide (LPS, 50 ng/mL) stimulation for 24 h. LPS stimulation induced iNOS protein expression and decreased the mitochondrial respiration, while increased lactate production and the expression of glycolytic pathway-related genes (Glut1 and Pfkfb3) in BMDCs from both CON and db/db groups. In LPS-stimulated mature BMDCs, 1,25(OH)2D3 treatment decreased the expression of surface markers related to immunostimulatory functions (MHC class II, CD80, CD86, and CD40) and production of IL-12p70 in both CON and db/db groups. While the mRNA level of the gene related to glucose uptake (Glut1) was increased in both groups, lactate production was decreased by 1,25(OH)2D3 treatment. mTORC1 activity was suppressed following 1,25(OH)2D3 treatment. Collectively, our findings confirmed that metabolic reprogramming occurred in BMDCs following LPS stimulation. In vitro 1,25(OH)2D3 treatment induced tolerogenic phenotypes by reducing the expression of surface markers, as well as cytokine production. However, no significant difference was observed regarding the effects of 1,25(OH)2D3 treatment on metabolic conversion and maturation of BMDCs between the control and diabetic mice. Additionally, the decreased aerobic glycolysis induced by the 1,25(OH)2D3 treatment appeared to be associated with the diminished maturation of BMDCs, and mTORC1 appears to play a key role in the 1,25(OH)2D3-mediated regulation of glycolysis.
Collapse
Affiliation(s)
- Ungue Shin
- Department of Food and Nutrition, Seoul National University, Seoul, the Republic of Korea.
| | - Hyeyoung You
- Department of Food and Nutrition, Seoul National University, Seoul, the Republic of Korea.
| | - Ga Young Lee
- Department of Food and Nutrition, Seoul National University, Seoul, the Republic of Korea.
| | - YeKyoung Son
- Department of Food and Nutrition, Seoul National University, Seoul, the Republic of Korea.
| | - Sung Nim Han
- Department of Food and Nutrition, Seoul National University, Seoul, the Republic of Korea; Research Institute of Human Ecology, Seoul National University, Seoul, the Republic of Korea.
| |
Collapse
|
26
|
Han JL, Song YX, Yao WJ, Zhou J, Du Y, Xu T. Follicle-Stimulating Hormone Provokes Macrophages to Secrete IL-1β Contributing to Atherosclerosis Progression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:25-32. [PMID: 36368721 DOI: 10.4049/jimmunol.2200475] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/06/2022] [Indexed: 02/17/2024]
Abstract
Abnormally high follicle-stimulating hormone (FSH) has been reported to associate with cardiovascular diseases in prostate cancer patients with specific androgen deprivation therapy and in menopausal women. All of the cardiovascular diseases were involved in atherosclerosis. However, the pathogenic mechanism of FSH-associated atherosclerosis remains uncertain. Apolipoprotein E-deficient mice were chosen to develop atherosclerosis, of which the plaques were analyzed with administration of short- and long-term FSH imitating androgen deprivation therapy-induced and menopausal FSH elevation. The study showed that short- and long-term exposure of FSH significantly accelerated atherosclerosis progression in apolipoprotein E-deficient mice, manifested as strikingly increased plaques in the aorta and its roots, increased macrophage content, reduced fibrin, and an enlarged necrotic core, suggesting a decrease in plaque stability. Furthermore, expression profiles from the Gene Expression Omnibus GSE21545 dataset revealed that macrophage inflammation was tightly associated with FSH-induced atherosclerotic progression. The human monocyte cell line THP-1 was induced by PMA and worked as a macrophage model to detect inflammatory factors and cellular functions. FSH remarkably promoted the expression of IL-1β in macrophages and strikingly increased the chemotactic migratory capacity of macrophages toward MCP-1, but the promigratory capacity of FSH was attenuated in foam cells. Overall, we revealed that FSH significantly promoted the inflammatory response and migration of macrophages, thereby provoking atherosclerosis development.
Collapse
Affiliation(s)
- Jing-Li Han
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Yu-Xuan Song
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Wei-Juan Yao
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; and
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Jing Zhou
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; and
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yiqing Du
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing, China;
| |
Collapse
|
27
|
Chumakova SP, Urazova OI, Denisenko OA, Vins MV, Shipulin VM, Pryakhin AS, Nevskaya KV, Gladkovskaya MV, Churina EG. Cytokines in the mechanisms of regulation of monocytopoiesis in ischemic heart disease. RUSSIAN JOURNAL OF HEMATOLOGY AND TRANSFUSIOLOGY 2022. [DOI: 10.35754/0234-5730-2022-67-4-511-524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction. The relationship of the violation of the subpopulation composition of blood monocytes in ischemic cardiomyopathy (ICMP) with changes in monocytopoiesis, as well as the effect of colony-stimulating factor of macrophages (M-CSF) and cytokines on the differentiation of monocytes of various immunophenotypes in the bone marrow is of great relevance.Aim – to study the role of cytokines in the mechanisms of local and distant regulation of differentiation of classical, intermediate, non-classical and transitional bone marrow monocytes in combination with the content of VEGFR2+-monocytes and hypoxia-induced factor-1a (HIF-1a) in the blood of patients with ischemic heart disease (IHD), suffering and not suffering from ischemic cardiomyopathy.Materials and methods. Seventy-four patients with IHD, suffering and not suffering from ICMP (30 and 44 people, respectively) were examined. The number of subpopulations of classical (CD14++CD16–), intermediate (CD14++CD16+), nonclassical (CD14+CD16++) and transitional (CD14+CD16–) monocytes (in bone marrow samples) and CD14+VEGFR2+-monocytes (in blood and bone marrow) was determined by flow cytofluorimetry; the concentration of cytokines IL-10, IL-13, TNF-α, IFN-γ, M-CSF in bone marrow and blood, as well as HIF-1a in the blood, was determined by ELISA.Results. Content of hematopoietins IL-10, IL-13, TNF-α, M-CSF in the bone marrow, as well as the ability of M-CSF to activate and IL-13 to inhibit the differentiation of classical monocytes from transitional cell forms were comparable between groups of patients with IHD. In the blood of patients with ICMP the concentration of IL-10 was higher, and the content of HIF-1α and CD14+VEGFR2+-cells was lower than in patients with IHD without ICMP (IL-10 – 30.00 (26.25–34.50) pg/ mL vs. 0 (23.0–28.0) pg/mL, p < 0.05; HIF-1α – 0.040 (0.029–0.053) ng/mL vs. 0.063 (0.054–0.122) ng/mL, p < 0.05; CD14+VEGFR2+ – 7.00 (5.67–7.15) % vs. 7.80 (7.23–8.17) %, p < 0.05). A feature of monocytopoiesis in ICMP compared with patients with IHD without ICMP is a high concentration of IFN-γ in the BM and a low ratio of M-CSF/IL-13 (10.00 (0.65–18.23) and 0.02 [0–0.15) pg/mL, p < 0.001; 1.02 (0.41–2.00) and 9.00 (2.13–22.09), p < 0.05, respectively), in association with a decrease in the number of classical, intermediate monocytes and an increase in the number of transitional cells in the BM in patients with ICMP relative to patients without cardiomyopathy (21.0 (19.5–23.0) and 47 (41–61.5) %, p < 0.001; 0.3 (0.0–1.2) and 18.5 (6.5–28.0) %, p < 0.01; 76.2 (73.0–78.5) and 30.5 (13.0–41.5) %, p < 0.001, respectively). At the same time, regardless of the clinical form of IHD, IL-10 and IL-13 are distant hematopoietins, TNF-α is local hematopoietin.Conclusion. An increase in the concentration of IFN-γ and a low ratio of M-CSF/IL-13 in the bone marrow, as well as an excess of IL-10 and a lack of HIF-1a and CD14+VEGFR2+-cells in the blood of IHD patients, are associated with inhibition of differentiation of mature forms of monocytes and the development of ICMP.
Collapse
Affiliation(s)
| | - O. I. Urazova
- Siberian State Medical University; Tomsk State University of Control Systems and Radioelectronics
| | | | | | - V. M. Shipulin
- Siberian State Medical University; Tomsk National Research Medical Center of the Russian Academy of Sciences, Cardiology Research Institute
| | - A. S. Pryakhin
- Tomsk National Research Medical Center of the Russian Academy of Sciences, Cardiology Research Institute
| | | | | | - E. G. Churina
- Siberian State Medical University; National Research Tomsk State University
| |
Collapse
|
28
|
Shin E, Kim D, Choi YY, Youn H, Seong KM, Youn B. LDR-adapted liver-derived cytokines have potential to induce atherosclerosis. Int J Radiat Biol 2022; 99:791-806. [PMID: 36383216 DOI: 10.1080/09553002.2023.2145028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE Atherosclerosis is a lipid-driven chronic inflammatory disease that causes cardiovascular diseases (CVD). The association between radiation and atherosclerosis has already been demonstrated; however, the effects of low-dose radiation (LDR) exposure on atherosclerosis have not been reported. Our study aims to propose that LDR may cause atherosclerosis phenotypes by the upregulation of plasminogen activator inhibitor-1 (PAI-1) and downregulation of androgen receptor (AR), which are cytokines secreted from the liver. METHODS Low-density lipoprotein (LDL) receptor deficient (Ldlr-/-) mice were irradiated at 50 mGy, 100 mGy, and 1000 mGy. LDR irradiated Ldlr-/- mice serum was analyzed by cytokine array and proteomics with silver staining. Oil Red O staining and BODIPY staining were performed to determine lipid accumulation in Human umbilical vein endothelial cells (HUVECs). Foam cell formation and monocyte recruitment were assessed through co-culture system with HUVECs and THP-1 cells. RESULTS After irradiation with LDR (100 mGy) the mice showed atherosclerotic phenotypes and through analysis results, we selected regulated cytokines, PAI-1 and AR, and found that these were changed in the liver. LDR-regulated cytokines have the potential to be transported to endothelial cells and induce lipid accumulation, inflammation of monocytes, increased oxidized low-density lipoprotein (oxLDL) and foam cells formation, that were series of phenotypes lead to plaque formation in endothelial cells and induces atherosclerosis. As a further aspect of this study, testosterone undecanoate (TU) was found to pharmacologically inhibit a series of atherosclerotic phenotypes exhibited by LDR. This study suggests a role for PAI-1 and AR in regulating the development of atherosclerosis after LDR exposure. Targeting PAI-1 and AR could serve as an attractive strategy for the management of atherosclerosis following LDR exposure.
Collapse
Affiliation(s)
- Eunguk Shin
- Department of Integrated Biological Science, Pusan National University, Busan, Korea
| | - Dahye Kim
- Department of Integrated Biological Science, Pusan National University, Busan, Korea
| | - You Yeon Choi
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center (NREMC), Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, Korea
| | - Ki Moon Seong
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center (NREMC), Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan, Korea
- Department of Biological Sciences, Pusan National University, Busan, Korea
| |
Collapse
|
29
|
Palshikar MG, Palli R, Tyrell A, Maggirwar S, Schifitto G, Singh MV, Thakar J. Executable models of immune signaling pathways in HIV-associated atherosclerosis. NPJ Syst Biol Appl 2022; 8:35. [PMID: 36131068 PMCID: PMC9492768 DOI: 10.1038/s41540-022-00246-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
Atherosclerosis (AS)-associated cardiovascular disease is an important cause of mortality in an aging population of people living with HIV (PLWH). This elevated risk has been attributed to viral infection, anti-retroviral therapy, chronic inflammation, and lifestyle factors. However, the rates at which PLWH develop AS vary even after controlling for length of infection, treatment duration, and for lifestyle factors. To investigate the molecular signaling underlying this variation, we sequenced 9368 peripheral blood mononuclear cells (PBMCs) from eight PLWH, four of whom have atherosclerosis (AS+). Additionally, a publicly available dataset of PBMCs from persons before and after HIV infection was used to investigate the effect of acute HIV infection. To characterize dysregulation of pathways rather than just measuring enrichment, we developed the single-cell Boolean Omics Network Invariant Time Analysis (scBONITA) algorithm. scBONITA infers executable dynamic pathway models and performs a perturbation analysis to identify high impact genes. These dynamic models are used for pathway analysis and to map sequenced cells to characteristic signaling states (attractor analysis). scBONITA revealed that lipid signaling regulates cell migration into the vascular endothelium in AS+ PLWH. Pathways implicated included AGE-RAGE and PI3K-AKT signaling in CD8+ T cells, and glucagon and cAMP signaling pathways in monocytes. Attractor analysis with scBONITA facilitated the pathway-based characterization of cellular states in CD8+ T cells and monocytes. In this manner, we identify critical cell-type specific molecular mechanisms underlying HIV-associated atherosclerosis using a novel computational method.
Collapse
Affiliation(s)
- Mukta G Palshikar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Rohith Palli
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Alicia Tyrell
- University of Rochester Clinical & Translational Science Institute, Rochester, USA
| | - Sanjay Maggirwar
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Giovanni Schifitto
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Imaging Sciences, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Meera V Singh
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Juilee Thakar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, USA.
| |
Collapse
|
30
|
Seidel F, Kleemann R, van Duyvenvoorde W, van Trigt N, Keijzer N, van der Kooij S, van Kooten C, Verschuren L, Menke A, Kiliaan AJ, Winter J, Hughes TR, Morgan BP, Baas F, Fluiter K, Morrison MC. Therapeutic Intervention with Anti-Complement Component 5 Antibody Does Not Reduce NASH but Does Attenuate Atherosclerosis and MIF Concentrations in Ldlr-/-.Leiden Mice. Int J Mol Sci 2022; 23:ijms231810736. [PMID: 36142647 PMCID: PMC9506266 DOI: 10.3390/ijms231810736] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/07/2022] [Accepted: 09/10/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Chronic inflammation is an important driver in the progression of non-alcoholic steatohepatitis (NASH) and atherosclerosis. The complement system, one of the first lines of defense in innate immunity, has been implicated in both diseases. However, the potential therapeutic value of complement inhibition in the ongoing disease remains unclear. Methods: After 20 weeks of high-fat diet (HFD) feeding, obese Ldlr-/-.Leiden mice were treated twice a week with an established anti-C5 antibody (BB5.1) or vehicle control. A separate group of mice was kept on a chow diet as a healthy reference. After 12 weeks of treatment, NASH was analyzed histopathologically, and genome-wide hepatic gene expression was analyzed by next-generation sequencing and pathway analysis. Atherosclerotic lesion area and severity were quantified histopathologically in the aortic roots. Results: Anti-C5 treatment considerably reduced complement system activity in plasma and MAC deposition in the liver but did not affect NASH. Anti-C5 did, however, reduce the development of atherosclerosis, limiting the total lesion size and severity independently of an effect on plasma cholesterol but with reductions in oxidized LDL (oxLDL) and macrophage migration inhibitory factor (MIF). Conclusion: We show, for the first time, that treatment with an anti-C5 antibody in advanced stages of NASH is not sufficient to reduce the disease, while therapeutic intervention against established atherosclerosis is beneficial to limit further progression.
Collapse
Affiliation(s)
- Florine Seidel
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
- Department Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, 6525 EZ Nijmegen, The Netherlands
- Correspondence:
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Nikki van Trigt
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Nanda Keijzer
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Sandra van der Kooij
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands
| | - Aswin Menke
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| | - Amanda J. Kiliaan
- Department Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, 6525 EZ Nijmegen, The Netherlands
| | - Johnathan Winter
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- UK Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Timothy R. Hughes
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- UK Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - B. Paul Morgan
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- UK Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Kees Fluiter
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Martine C. Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands
| |
Collapse
|
31
|
Kelley WJ, Wragg KM, Chen J, Murthy T, Xu Q, Boyne MT, Podojil JR, Elhofy A, Goldstein DR. Nanoparticles reduce monocytes within the lungs to improve outcomes after influenza virus infection in aged mice. JCI Insight 2022; 7:156320. [PMID: 35737459 PMCID: PMC9462478 DOI: 10.1172/jci.insight.156320] [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: 11/02/2021] [Accepted: 06/21/2022] [Indexed: 01/26/2023] Open
Abstract
Older people exhibit dysregulated innate immunity to respiratory viral infections, including influenza and SARS-CoV-2, and show an increase in morbidity and mortality. Nanoparticles are a potential practical therapeutic that could reduce exaggerated innate immune responses within the lungs during viral infection. However, such therapeutics have not been examined for effectiveness during respiratory viral infection, particular in aged hosts. Here, we employed a lethal model of influenza viral infection in vulnerable aged mice to examine the ability of biodegradable, cargo-free nanoparticles, designated ONP-302, to resolve innate immune dysfunction and improve outcomes during infection. We administered ONP-302 via i.v. injection to aged mice at day 3 after infection, when the hyperinflammatory innate immune response was already established. During infection, we found that ONP-302 treatment reduced the numbers of inflammatory monocytes within the lungs and increased their number in both the liver and spleen, without impacting viral clearance. Importantly, cargo-free nanoparticles reduced lung damage, reduced histological lung inflammation, and improved gas exchange and, ultimately, the clinical outcomes in influenza-infected aged mice. In conclusion, ONP-302 improves outcomes in influenza-infected aged mice. Thus, our study provides information concerning a practical therapeutic, which, if translated clinically, could improve disease outcomes for vulnerable older patients suffering from respiratory viral infections.
Collapse
Affiliation(s)
| | | | - Judy Chen
- Department of Internal Medicine and,Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Tushar Murthy
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Qichen Xu
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Michael T. Boyne
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Joseph R. Podojil
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Adam Elhofy
- Research and Development, COUR Pharmaceuticals Development Company Inc., Northbrook, Illinois, USA
| | - Daniel R. Goldstein
- Department of Internal Medicine and,Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Microbiology and Immunology, University of Michigan, Michigan, USA
| |
Collapse
|
32
|
Biological Sensing of Nitric Oxide in Macrophages and Atherosclerosis Using a Ruthenium-Based Sensor. Biomedicines 2022; 10:biomedicines10081807. [PMID: 36009353 PMCID: PMC9405170 DOI: 10.3390/biomedicines10081807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 12/04/2022] Open
Abstract
Macrophage-derived nitric oxide (NO) plays a critical role in atherosclerosis and presents as a potential biomarker. We assessed the uptake, distribution, and NO detection capacity of an irreversible, ruthenium-based, fluorescent NO sensor (Ru-NO) in macrophages, plasma, and atherosclerotic plaques. In vitro, incubation of Ru-NO with human THP1 monocytes and THP1-PMA macrophages caused robust uptake, detected by Ru-NO fluorescence using mass-cytometry, confocal microscopy, and flow cytometry. THP1-PMA macrophages had higher Ru-NO uptake (+13%, p < 0.05) than THP1 monocytes with increased Ru-NO fluorescence following lipopolysaccharide stimulation (+14%, p < 0.05). In mice, intraperitoneal infusion of Ru-NO found Ru-NO uptake was greater in peritoneal CD11b+F4/80+ macrophages (+61%, p < 0.01) than CD11b+F4/80− monocytes. Infusion of Ru-NO into Apoe−/− mice fed high-cholesterol diet (HCD) revealed Ru-NO fluorescence co-localised with atherosclerotic plaque macrophages. When Ru-NO was added ex vivo to aortic cell suspensions from Apoe−/− mice, macrophage-specific uptake of Ru-NO was demonstrated. Ru-NO was added ex vivo to tail-vein blood samples collected monthly from Apoe−/− mice on HCD or chow. The plasma Ru-NO fluorescence signal was higher in HCD than chow-fed mice after 12 weeks (37.9%, p < 0.05). Finally, Ru-NO was added to plasma from patients (N = 50) following clinically-indicated angiograms. There was lower Ru-NO fluorescence from plasma from patients with myocardial infarction (−30.7%, p < 0.01) than those with stable coronary atherosclerosis. In conclusion, Ru-NO is internalised by macrophages in vitro, ex vivo, and in vivo, can be detected in atherosclerotic plaques, and generates measurable changes in fluorescence in murine and human plasma. Ru-NO displays promising utility as a sensor of atherosclerosis.
Collapse
|
33
|
Li J, Duan H, Liu Y, Wang L, Zhou X. Biomaterial-Based Therapeutic Strategies for Obesity and Its Comorbidities. Pharmaceutics 2022; 14:1445. [PMID: 35890340 PMCID: PMC9320151 DOI: 10.3390/pharmaceutics14071445] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a global public health issue that results in many health complications or comorbidities, including type 2 diabetes mellitus, cardiovascular disease, and fatty liver. Pharmacotherapy alone or combined with either lifestyle alteration or surgery represents the main modality to combat obesity and its complications. However, most anti-obesity drugs are limited by their bioavailability, target specificity, and potential toxic effects. Only a handful of drugs, including orlistat, liraglutide, and semaglutide, are currently approved for clinical obesity treatment. Thus, there is an urgent need for alternative treatment strategies. Based on the new revelation of the pathogenesis of obesity and the efforts toward the multi-disciplinary integration of materials, chemistry, biotechnology, and pharmacy, some emerging obesity treatment strategies are gradually entering the field of preclinical and clinical research. Herein, by analyzing the current situation and challenges of various new obesity treatment strategies such as small-molecule drugs, natural drugs, and biotechnology drugs, the advanced functions and prospects of biomaterials in obesity-targeted delivery, as well as their biological activities and applications in obesity treatment, are systematically summarized. Finally, based on the systematic analysis of biomaterial-based obesity therapeutic strategies, the future prospects and challenges in this field are proposed.
Collapse
Affiliation(s)
- Jing Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
| | - Hongli Duan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
| | - Yan Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
| | - Lu Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
| | - Xing Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
- Institute of Materia Medica and Center of Translational Medicine, College of Pharmacy, Army Medical University, Chongqing 400038, China
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| |
Collapse
|
34
|
Chen J, Li S, Ma D, Li L, Zhuang W, Chen M. A lipid droplet-specific fluorescence probe for atherosclerotic plaque imaging. Analyst 2022; 147:3081-3086. [PMID: 35678714 DOI: 10.1039/d1an01937f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The dysregulation of lipid droplets (LDs) is closely related to certain metabolic diseases, while the role of LDs during pathological processes remains mysterious. It would be of great value to monitor the dynamic changes of LDs in a visible way so as to study their biological functions. In this study, we report a LD-specific fluorescence probe TBI for precise LD-targeting imaging in cells and atherosclerotic tissues. TBI exhibited great biocompatibility, remarkable oil-enhanced fluorescence emission, good photostability and impressive intracellular and tissular LD-specific imaging performance. Importantly, TBI could efficiently stain the LDs at a low concentration of 50 nM, and the motion tracking of LDs could be observed via fluorescence imaging. Moreover, TBI could efficiently light up the LD distribution in mouse atherosclerotic plaques with high resolution, which revealed the ultra-structure of atherosclerotic plaques. In conclusion, these results imply that TBI could be a potential tool for investigating the physiological and pathological role of LDs.
Collapse
Affiliation(s)
- Jingruo Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China.
| | - Shufen Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China.
| | - Di Ma
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lilan Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China.
| | - Weihua Zhuang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China.
| | - Mao Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China. .,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.,Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China
| |
Collapse
|
35
|
Anti-Inflammatory Potential of Fucoidan for Atherosclerosis: In Silico and In Vitro Studies in THP-1 Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103197. [PMID: 35630678 PMCID: PMC9146328 DOI: 10.3390/molecules27103197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 01/13/2023]
Abstract
Several diseases, including atherosclerosis, are characterized by inflammation, which is initiated by leukocyte migration to the inflamed lesion. Hence, genes implicated in the early stages of inflammation are potential therapeutic targets to effectively reduce atherogenesis. Algal-derived polysaccharides are one of the most promising sources for pharmaceutical application, although their mechanism of action is still poorly understood. The present study uses a computational method to anticipate the effect of fucoidan and alginate on interactions with adhesion molecules and chemokine, followed by an assessment of the cytotoxicity of the best-predicted bioactive compound for human monocytic THP-1 macrophages by lactate dehydrogenase and crystal violet assay. Moreover, an in vitro pharmacodynamics evaluation was performed. Molecular docking results indicate that fucoidan has a greater affinity for L-and E-selectin, monocyte chemoattractant protein 1 (MCP-1), and intercellular adhesion molecule-1 (ICAM-1) as compared to alginate. Interestingly, there was no fucoidan cytotoxicity on THP-1 macrophages, even at 200 µg/mL for 24 h. The strong interaction between fucoidan and L-selectin in silico explained its ability to inhibit the THP-1 monocytes migration in vitro. MCP-1 and ICAM-1 expression levels in THP-1 macrophages treated with 50 µg/mL fucoidan for 24 h, followed by induction by IFN-γ, were shown to be significantly suppressed as eight- and four-fold changes, respectively, relative to cells treated only with IFN-γ. These results indicate that the electrostatic interaction of fucoidan improves its binding affinity to inflammatory markers in silico and reduces their expression in THP-1 cells in vitro, thus making fucoidan a good candidate to prevent inflammation.
Collapse
|
36
|
Increased frequency of proangiogenic tunica intima endothelial kinase 2 (Tie2) expressing monocytes in individuals with type 2 diabetes mellitus. Cardiovasc Diabetol 2022; 21:72. [PMID: 35549955 PMCID: PMC9102255 DOI: 10.1186/s12933-022-01497-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/18/2022] [Indexed: 12/02/2022] Open
Abstract
Background Individuals with type 2 diabetes mellitus (T2DM) have an increased risk for developing macrovascular disease (MVD) manifested by atherosclerosis. Phenotypically and functionally different monocyte subsets (classical; CD14++CD16−, non-classical; CD14+CD16++, and intermediate; CD14++CD16+) including pro-angiogenic monocytes expressing Tie2 (TEMs) can be identified. Here we investigated monocyte heterogeneity and its association with T2DM and MVD. Methods Individuals with (N = 51) and without (N = 56) T2DM were recruited and allocated to "non-MVD" or "with MVD" (i.e., peripheral or coronary artery disease) subgroups. Blood monocyte subsets were quantified based on CD14, CD16 and Tie2 expression levels. Plasma levels of Tie2-ligands angiopoietin-1 and angiopoietin-2 were determined using ELISA. Carotid endarterectomy samples from individuals with (N = 24) and without (N = 22) T2DM were stained for intraplaque CD68+ macrophages (inflammation) and CD34+ (angiogenesis), as plaque vulnerability markers. Results Monocyte counts were similar between individuals with T2DM and healthy controls (non-diabetic, non-MVD). Non-classical monocytes were reduced (p < 0.05) in T2DM, whereas the percentage of TEMs within the intermediate subset was increased (p < 0.05). T2DM was associated with increased angiopoietin-1 (p < 0.05) and angiopoietin-2 (p = 0.0001) levels. Angiopoietin-2 levels were higher in T2DM individuals with MVD compared with non-MVD (p < 0.01). Endarterectomized plaques showed no differences in macrophage influx and microvessel number between individuals with and without T2DM. Conclusions Monocyte subset distribution is altered in T2DM with reduced non-classical monocytes and increased TEM percentage in the intermediate monocyte subset. Increased angiopoietin-2 levels together with increased frequency of TEMs might promote plaque vulnerability in T2DM which could however not be confirmed at tissue level in advanced atherosclerotic lesions.
Collapse
|
37
|
Shayo SC, Ogiso K, Kawade S, Hashiguchi H, Deguchi T, Nishio Y. Dietary obesity and glycemic excursions cause a parallel increase in STEAP4 and pro-inflammatory gene expression in murine PBMCs. Diabetol Int 2022; 13:358-371. [PMID: 35463853 PMCID: PMC8980188 DOI: 10.1007/s13340-021-00542-1] [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: 06/15/2021] [Accepted: 09/05/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND The balance between pro-atherogenic and anti-atherogenic factors is very crucial in the development of atherosclerotic lesions. Although the expression of the six-transmembrane epithelial antigen of the prostate 4 (STEAP4) in myeloid cells is known to be atheroprotective, there is not a single study reporting on the status of STEAP4 expression in circulating monocytes in the early stages of diet-induced obesity or in events of glycemic excursions. METHODS We induced glycemic spikes twice daily for a 1-week duration to rats fed on regular chow and western diet, and analyzed gene expression changes in the peripheral blood mononuclear cells (PBMCs). We also conducted experiments on RAW 264.7 cells to gain insight into some of our in vivo findings. RESULTS Diet-induced obesity and glycemic excursions independently caused a significant increase in STEAP4 mRNA expression in PBMCs. This was also accompanied by an induction of a substantial number of pro-inflammatory cytokines, chemokines, and chemokine receptors. However, the combined effect of western diet and hyperglycemic spikes was subtle and non-additive. In the in vitro setting, either glucose spikes, persistent hyperglycemia, or a combination of palmitic acid and insulin resulted in a parallel increase in expression of STEAP4 and pro-inflammatory genes. This was, however, significantly abrogated with 4-octyl itaconate or attenuated by inhibitors of p38MAPK and NF-kB. CONCLUSIONS STEAP4 expression in mononuclear cells is induced by increasing inflammation or oxidative stress. The observed increase in STEAP4 expression in circulating monocytes due to visceral obesity or glycemic excursions is a compensatory response. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13340-021-00542-1.
Collapse
Affiliation(s)
- Sigfrid Casmir Shayo
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
- Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Kazuma Ogiso
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| | - Shigeru Kawade
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| | - Hiroshi Hashiguchi
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| | - Takahisa Deguchi
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| | - Yoshihiko Nishio
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| |
Collapse
|
38
|
Secreted frizzled-related protein 4 exerts anti-atherosclerotic effects by reducing inflammation and oxidative stress. Eur J Pharmacol 2022; 923:174901. [PMID: 35364070 DOI: 10.1016/j.ejphar.2022.174901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/04/2022] [Accepted: 03/17/2022] [Indexed: 11/20/2022]
Abstract
Atherosclerosis and its sequelae, such as coronary artery disease (CAD), are the most common diseases worldwide and the leading causes of morbidity and mortality in most countries. Our previous studies have shown that circulating secreted frizzled-related protein 4 (SFRP4) levels are increased in patients with CAD. However, the role of SFRP4 in the development of atherosclerosis remains unclear; thus, the purpose of this study was to determine the effect of SFRP4 on high-fat diet (HFD)-induced atherosclerosis and explore the possible mechanisms. In this study, we found for the first time that administration of recombinant SFRP4 alleviates atherosclerosis in ApoE-/- mice by reducing inflammation and oxidative stress. In addition, the anti-atherosclerotic effect of SFRP4 was associated with inhibition of the Wnt/β-catenin signaling pathway, and Wnt1 overexpression abolished the anti-atherosclerotic effects of SFRP4. Taken together, our results highlight the potential beneficial effect of SFRP4 as a therapeutic agent for atherosclerosis and CAD.
Collapse
|
39
|
Ramji DP, Ismail A, Chen J, Alradi F, Al Alawi S. Survey of In Vitro Model Systems for Investigation of Key Cellular Processes Associated with Atherosclerosis. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:39-56. [PMID: 35237957 DOI: 10.1007/978-1-0716-1924-7_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Atherosclerosis progression is associated with a complex array of cellular processes in the arterial wall, including endothelial cell activation/dysfunction, chemokine-driven recruitment of immune cells, differentiation of monocytes to macrophages and their subsequent transformation into lipid laden foam cells, activation of inflammasome and pro-inflammatory signaling, and migration of smooth muscle cells from the media to the intima. The use of in vitro model systems has considerably advanced our understanding of these atherosclerosis-associated processes and they are also often used in drug discovery and other screening platforms. This chapter will describe key in vitro model systems employed frequently in atherosclerosis research.
Collapse
Affiliation(s)
- Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK.
| | - Alaa Ismail
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Jing Chen
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Fahad Alradi
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | | |
Collapse
|
40
|
Liu H, Pietersz G, Peter K, Wang X. Nanobiotechnology approaches for cardiovascular diseases: site-specific targeting of drugs and nanoparticles for atherothrombosis. J Nanobiotechnology 2022; 20:75. [PMID: 35135581 PMCID: PMC8822797 DOI: 10.1186/s12951-022-01279-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/21/2022] [Indexed: 02/18/2023] Open
Abstract
Atherosclerosis and atherothrombosis, the major contributors to cardiovascular diseases (CVDs), represent the leading cause of death worldwide. Current pharmacological therapies have been associated with side effects or are insufficient at halting atherosclerotic progression effectively. Pioneering work harnessing the passive diffusion or endocytosis properties of nanoparticles and advanced biotechnologies in creating recombinant proteins for site-specific delivery have been utilized to overcome these limitations. Since CVDs are complex diseases, the most challenging aspect of developing site-specific therapies is the identification of an individual and unique antigenic epitope that is only expressed in lesions or diseased areas. This review focuses on the pathological mechanism of atherothrombosis and discusses the unique targets that are important during disease progression. We review recent advances in site-specific therapy using novel targeted drug-delivery and nanoparticle-carrier systems. Furthermore, we explore the limitations and future perspectives of site-specific therapy for CVDs.
Collapse
Affiliation(s)
- Haikun Liu
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Geoffrey Pietersz
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Burnet Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia.,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia. .,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia. .,Department of Medicine, Monash University, Melbourne, VIC, Australia. .,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
41
|
Snarski P, Sukhanov S, Yoshida T, Higashi Y, Danchuk S, Chandrasekar B, Tian D, Rivera-Lopez V, Delafontaine P. Macrophage-Specific IGF-1 Overexpression Reduces CXCL12 Chemokine Levels and Suppresses Atherosclerotic Burden in Apoe-Deficient Mice. Arterioscler Thromb Vasc Biol 2022; 42:113-126. [PMID: 34852642 PMCID: PMC8792341 DOI: 10.1161/atvbaha.121.316090] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE IGF-1 (insulin-like growth factor 1) exerts pleiotropic effects including promotion of cellular growth, differentiation, survival, and anabolism. We have shown that systemic IGF-1 administration reduced atherosclerosis in Apoe-/- (apolipoprotein E deficient) mice, and this effect was associated with a reduction in lesional macrophages and a decreased number of foam cells in the plaque. Almost all cell types secrete IGF-1, but the effect of macrophage-derived IGF-1 on the pathogenesis of atherosclerosis is poorly understood. We hypothesized that macrophage-derived IGF-1 will reduce atherosclerosis. Approach and Results: We created macrophage-specific IGF-1 overexpressing mice on an Apoe-/- background. Macrophage-specific IGF-1 overexpression reduced plaque macrophages, foam cells, and atherosclerotic burden and promoted features of stable atherosclerotic plaque. Macrophage-specific IGF1 mice had a reduction in monocyte infiltration into plaque, decreased expression of CXCL12 (CXC chemokine ligand 12), and upregulation of ABCA1 (ATP-binding cassette transporter 1), a cholesterol efflux regulator, in atherosclerotic plaque and in peritoneal macrophages. IGF-1 prevented oxidized lipid-induced CXCL12 upregulation and foam cell formation in cultured THP-1 macrophages and increased lipid efflux. We also found an increase in cholesterol efflux in macrophage-specific IGF1-derived peritoneal macrophages. CONCLUSIONS Macrophage IGF-1 overexpression reduced atherosclerotic burden and increased features of plaque stability, likely via a reduction in CXCL12-mediated monocyte recruitment and an increase in ABCA1-dependent macrophage lipid efflux.
Collapse
Affiliation(s)
- Patricia Snarski
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Sergiy Sukhanov
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Tadashi Yoshida
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Yusuke Higashi
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Svitlana Danchuk
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Bysani Chandrasekar
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO
| | - Di Tian
- Department of Pathology, Tulane University School of Medicine, New Orleans, LA
| | | | - Patrick Delafontaine
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
42
|
Yang W, Su G, Liu Y. Silencing p62 reduces ox-LDL-induced M1 polarization and inflammation in macrophages by inhibiting mTOR/NF-κB signaling pathways. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221110348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Macrophages can change their phenotypes according to the changes in the microenvironment, and thus have various functions, that is, macrophages polarization. Macrophage phenotype is associated with the progression of atherosclerotic plaques. Studies have shown a large accumulation of p62 protein in atherosclerotic plaques. Whether the accumulation of p62 protein affects the level of macrophage polarization and inflammation and its mechanism is not clear. The p62 levels of macrophages treated with ox-LDL were detected by western blotting and qRT-PCR. Several polarizing markers and cytokines associated with atherosclerosis were detected by western blotting, ELISA, qRT-PCR, and flow cytometry to assess macrophage phenotype. The effect of p62 on the treatment of macrophage polarization by ox-LDL was studied by silencing p62 by gene silencing technique. The activity of mTOR and NF-κB signaling pathways was evaluated by detecting p-mTOR and intranuclear p65 levels in western blotting to explore the mechanism of p62. Rapamycin inhibited mTOR to demonstrate its role in activating the NF-κB signaling pathway and in ox-LDL therapy of p62 induced M1 polarization in macrophages. ox-LDL induced a significant increase in p62 and an increase in M1 markers and inflammatory cytokines. After p62 silencing, M1 markers and inflammatory cytokines decreased significantly, while M2 markers and anti-inflammatory cytokines increased significantly. Silencing p62 inhibited p-mTOR and p65 nuclear translocation. Rapamycin inhibited ox-LDL-induced p65 nuclear translocation and M1 markers, and increased M2 markers. p62 protein accumulation in ox-LDL treatment macrophages induces M1 polarization and inflammatory cytokines through the mTOR/NF-κB pathway.
Collapse
Affiliation(s)
- Wei Yang
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Laboratory Diagnosis, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangming Su
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yanhong Liu
- Department of Laboratory Diagnosis, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
43
|
Zhao Q, Wang Z, Meyers AK, Madenspacher J, Zabalawi M, Zhang Q, Boudyguina E, Hsu FC, McCall CE, Furdui CM, Parks JS, Fessler MB, Zhu X. Hematopoietic Cell-Specific SLC37A2 Deficiency Accelerates Atherosclerosis in LDL Receptor-Deficient Mice. Front Cardiovasc Med 2021; 8:777098. [PMID: 34957260 PMCID: PMC8702732 DOI: 10.3389/fcvm.2021.777098] [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: 09/14/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022] Open
Abstract
Macrophages play a central role in the pathogenesis of atherosclerosis. Our previous study demonstrated that solute carrier family 37 member 2 (SLC37A2), an endoplasmic reticulum-anchored phosphate-linked glucose-6-phosphate transporter, negatively regulates macrophage Toll-like receptor activation by fine-tuning glycolytic reprogramming in vitro. Whether macrophage SLC37A2 impacts in vivo macrophage inflammation and atherosclerosis under hyperlipidemic conditions is unknown. We generated hematopoietic cell-specific SLC37A2 knockout and control mice in C57Bl/6 Ldlr−/− background by bone marrow transplantation. Hematopoietic cell-specific SLC37A2 deletion in Ldlr−/− mice increased plasma lipid concentrations after 12-16 wks of Western diet induction, attenuated macrophage anti-inflammatory responses, and resulted in more atherosclerosis compared to Ldlr−/− mice transplanted with wild type bone marrow. Aortic root intimal area was inversely correlated with plasma IL-10 levels, but not total cholesterol concentrations, suggesting inflammation but not plasma cholesterol was responsible for increased atherosclerosis in bone marrow SLC37A2-deficient mice. Our in vitro study demonstrated that SLC37A2 deficiency impaired IL-4-induced macrophage activation, independently of glycolysis or mitochondrial respiration. Importantly, SLC37A2 deficiency impaired apoptotic cell-induced glycolysis, subsequently attenuating IL-10 production. Our study suggests that SLC37A2 expression is required to support alternative macrophage activation in vitro and in vivo. In vivo disruption of hematopoietic SLC37A2 accelerates atherosclerosis under hyperlipidemic pro-atherogenic conditions.
Collapse
Affiliation(s)
- Qingxia Zhao
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Zhan Wang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Allison K Meyers
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jennifer Madenspacher
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, United States
| | - Manal Zabalawi
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Qianyi Zhang
- Department of Biology, Wake Forest University, Winston-Salem, NC, United States
| | - Elena Boudyguina
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Charles E McCall
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States.,Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - John S Parks
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC, United States
| | - Xuewei Zhu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States.,Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
44
|
Irmscher S, Zipfel SLH, Halder LD, Ivanov L, Gonzalez-Delgado A, Waldeyer C, Seiffert M, Brunner FJ, von der Heide M, Löschmann I, Wulf S, Czamara D, Papac-Milicevic N, Strauß O, Lorkowski S, Reichenspurner H, Holers MV, Banda NK, Zeller T, Binder EB, Binder CJ, Wiech T, Zipfel PF, Skerka C. Factor H-related protein 1 (FHR-1) is associated with atherosclerotic cardiovascular disease. Sci Rep 2021; 11:22511. [PMID: 34795372 PMCID: PMC8602345 DOI: 10.1038/s41598-021-02011-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 10/29/2021] [Indexed: 02/08/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ACVD) is a lipid-driven inflammatory disease and one of the leading causes of death worldwide. Lipid deposits in the arterial wall lead to the formation of plaques that involve lipid oxidation, cellular necrosis, and complement activation, resulting in inflammation and thrombosis. The present study found that homozygous deletion of the CFHR1 gene, which encodes the plasma complement protein factor H-related protein 1 (FHR-1), was protective in two cohorts of patients with ACVD, suggesting that FHR-1 accelerates inflammation and exacerbates the disease. To test this hypothesis, FHR-1 was isolated from human plasma and was found to circulate on extracellular vesicles and to be deposited in atherosclerotic plaques. Surface-bound FHR-1 induced the expression of pro-inflammatory cytokines and tissue factor in both monocytes and neutrophils. Notably, plasma concentrations of FHR-1, but not of factor H, were significantly (p < 0.001) elevated in patients with ACVD, and correlated with the expression of the inflammation markers C-reactive protein, apolipoprotein serum amyloid protein A, and neopterin. FHR-1 expression also significantly correlated with plasma concentrations of low-density lipoprotein (LDL) (p < 0.0001) but not high-density lipoprotein (HDL). Taken together, these findings suggest that FHR-1 is associated with ACVD.
Collapse
Affiliation(s)
- Sarah Irmscher
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany.,Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Svante L H Zipfel
- Clinic for Heart and Visceral Surgery, University Heart and Vascular Center Hamburg, Medical University Hamburg-Eppendorf, Hamburg, Germany
| | - Luke D Halder
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Lia Ivanov
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Andres Gonzalez-Delgado
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Christoph Waldeyer
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Medical University Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg/Lübeck/Kiel, Hamburg, Germany
| | - Moritz Seiffert
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Medical University Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg/Lübeck/Kiel, Hamburg, Germany
| | - Fabian J Brunner
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Medical University Hamburg-Eppendorf, Hamburg, Germany
| | - Monika von der Heide
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Ina Löschmann
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Sonia Wulf
- Institute of Pathology, Medical University Hamburg-Eppendorf, Hamburg, Germany
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Olaf Strauß
- Department of Ophthalmology, Charité -University Medicine Berlin, a Corporate Member of Free University, Humboldt-University and the Berlin Institute of Health, Berlin, Germany
| | - Stefan Lorkowski
- Institute for Nutritional Sciences, Friedrich Schiller University, Jena, Germany
| | - Hermann Reichenspurner
- Clinic for Heart and Visceral Surgery, University Heart and Vascular Center Hamburg, Medical University Hamburg-Eppendorf, Hamburg, Germany
| | - Michael V Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States of America
| | - Nirmal K Banda
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States of America
| | - Tania Zeller
- Department of General and Interventional Cardiology, University Heart and Vascular Center Hamburg, Medical University Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg/Lübeck/Kiel, Hamburg, Germany
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Thorsten Wiech
- Institute of Pathology, Medical University Hamburg-Eppendorf, Hamburg, Germany
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany.,Faculty of Biosciences, Friedrich Schiller University, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany.
| |
Collapse
|
45
|
Shang C, Lin H, Fang X, Wang Y, Jiang Z, Qu Y, Xiang M, Shen Z, Xin L, Lu Y, Gao J, Cui X. Beneficial effects of cinnamon and its extracts in the management of cardiovascular diseases and diabetes. Food Funct 2021; 12:12194-12220. [PMID: 34752593 DOI: 10.1039/d1fo01935j] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cardiovascular diseases (CVDs) and diabetes are the leading causes of death worldwide, which underlines the urgent necessity to develop new pharmacotherapies. Cinnamon has been an eminent component of spice and traditional Chinese medicine for thousands of years. Numerous lines of findings have elucidated that cinnamon has beneficial effects against CVDs in various ways, including endothelium protection, regulation of immune response, lowering blood lipids, antioxidative properties, anti-inflammatory properties, suppression of vascular smooth muscle cell (VSMC) growth and mobilization, repression of platelet activity and thrombosis and inhibition of angiogenesis. Furthermore, emerging evidence has established that cinnamon improves diabetes, a crucial risk factor for CVDs, by enhancing insulin sensitivity and insulin secretion; regulating the enzyme activity involved in glucose; regulating glucose metabolism in the liver, adipose tissue and muscle; ameliorating oxidative stress and inflammation to protect islet cells; and improving diabetes complications. In this review, we summarized the mechanisms by which cinnamon regulates CVDs and diabetes in order to provide a theoretical basis for the further clinical application of cinnamon.
Collapse
Affiliation(s)
- Chang Shang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hongchen Lin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xuqin Fang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yuling Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhilin Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yi Qu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mi Xiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Zihuan Shen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Laiyun Xin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,First Clinical Medical School, Shandong University of Chinese Medicine, Shandong, 250355, China
| | - Yingdong Lu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Jialiang Gao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Xiangning Cui
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
46
|
Greene JT, Brian BF, Senevirathne SE, Freedman TS. Regulation of myeloid-cell activation. Curr Opin Immunol 2021; 73:34-42. [PMID: 34601225 DOI: 10.1016/j.coi.2021.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022]
Abstract
Myeloid cells (macrophages, monocytes, dendritic cells, and granulocytes) survey the body for signs of infection and damage and regulate tissue homeostasis, organogenesis, and immunity. They express receptors that initiate the inflammatory response, send signals that alter the vascular and cytokine milieu, and oversee the recruitment, differentiation, and activation of other myeloid and adaptive immune cells. Their activation must therefore be tightly regulated, optimized for maximal innate-immune protection with a minimum of collateral tissue damage or disorganization. In this review we discuss what it means for myeloid cells to become activated, with emphasis on the receptors and signaling molecules important for the recognition of pathogen-associated and damage-associated molecular patterns. We also outline how these signals are regulated by the steric properties of proteins, by adhesive and cytoskeletal interactions, and by negative feedback to keep inflammation in check and support healthy tissue development and homeostasis. Throughout the text we highlight recent publications and reviews and direct readers therein for a comprehensive bibliography.
Collapse
Affiliation(s)
- Joseph T Greene
- Department of Pharmacology, Center for Immunology, Masonic Cancer Center, and Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, United States
| | - Ben F Brian
- Department of Pharmacology, Center for Immunology, Masonic Cancer Center, and Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, United States
| | - S Erandika Senevirathne
- Department of Pharmacology, Center for Immunology, Masonic Cancer Center, and Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, United States
| | - Tanya S Freedman
- Department of Pharmacology, Center for Immunology, Masonic Cancer Center, and Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, United States.
| |
Collapse
|
47
|
Jin L, Deng Z, Bai Y, Ye P. Functions of Monocytes and Macrophages and the Associated Effective Molecules and Mechanisms at the Early Stage of Atherosclerosis. ACTA CARDIOLOGICA SINICA 2021; 37:522-533. [PMID: 34584385 DOI: 10.6515/acs.202109_37(5).20210323c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Objective This study aimed to explore the functions and possible underlying regulatory molecules and mechanisms of monocytes and macrophages under early atherosclerotic conditions. Methods THP-1-derived monocytes or macrophages were induced by 50 μg/ml oxidized low density lipoprotein (ox-LDL) for 24 hours, and the degree of lipid metabolism and inflammation were determined. In addition, we identified differentially expressed genes, noncoding ribonucleic acids (RNAs), pathways and mechanisms by RNA sequencing, and performed further correlation analysis and molecular expression verification. Results Monocytes could not form foam cells with oil red O staining directly and had low levels of lipids as determined by total cholesterol and triglycerides assays, cholesterol uptake molecules CD36, the class A macrophage scavenger receptor and lectin-like oxidized low-density lipoprotein receptor-1 and cholesterol efflux molecules ATP binding cassette transporter A1, ATP binding cassette transporter G1 and liver X receptor α, and inflammatory factors, which were markedly different from those in macrophages. Additionally, sequencing data showed obviously differentially expressed genes, microRNAs and long noncoding RNAs in the atherosclerotic group. We identified 15 upregulated and downregulated genes, and 10 biological processes and pathways involved in atherosclerosis. Specifically, fatty acid desaturase 2 and apolipoprotein A1 in the peroxisome proliferator-activated receptor signaling pathway were differentially expressed in stimulated macrophages, whereas no changes were observed in the monocyte groups. Furthermore, correlation analysis showed differential expressed lncRNAs targeting miRNAs and mRNAs, and 24 competing endogenous RNA (ceRNA) networks of long noncoding RNA-microRNA-messenger RNA in early oxidative macrophages. Conclusions Monocytes did not directly participate in lipid metabolism before differentiation into macrophages at the early stage in vitro. Furthermore, noncoding RNAs and ceRNA networks might play important roles in regulating the lipid metabolism of macrophages at the early stage of atherosclerosis.
Collapse
Affiliation(s)
- Liyuan Jin
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases
| | - Zihui Deng
- Biochemistry Department of Graduate School, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Yongyi Bai
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases
| | - Ping Ye
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases
| |
Collapse
|
48
|
Wu H, Wang M, Li X, Shao Y. The Metaflammatory and Immunometabolic Role of Macrophages and Microglia in Diabetic Retinopathy. Hum Cell 2021; 34:1617-1628. [PMID: 34324139 DOI: 10.1007/s13577-021-00580-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/17/2021] [Indexed: 12/17/2022]
Abstract
Emergent studies reveal the roles of inflammatory cells and cytokines in the development of diabetic retinopathy (DR), which is gradually portrayed as a chronic inflammatory disease accompanied by metabolic disorder. Through the pathogenesis of DR, macrophages or microglia play a critical role in the inflammation, neovascularization, and neurodegeneration of the retina. Conventionally, macrophages are generally divided into M1 and M2 phenotypes which mainly rely on glycolysis and oxidative phosphorylation, respectively. Recently, studies have found that nutrients (including glucose and lipids) and metabolites (such as lactate), can not only provide energy for cells, but also act as signaling molecules to regulate the function and fate of cells. In this review, we discussed the intrinsic correlations among the metabolic status, polarization, and function of macrophage/microglia in DR. Hyperglycemia and hyperlipidemia could induce M1-like and M2-like macrophages polarization in different phases of DR. Targeting the regulation of microglial metabolic profile might be a promising therapeutic strategy to modulate the polarization and function of macrophages/microglia, thus attenuating the progression of DR.
Collapse
Affiliation(s)
- Honglian Wu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Mengqi Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Yan Shao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China. .,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China. .,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.
| |
Collapse
|
49
|
Genkel VV, Kuznetsova AS, Lebedev EV, Shaposhnik II. Factors associated with atherosclerotic plaque echogenicity in patients aged 40-64 with carotid atherosclerosis. ACTA ACUST UNITED AC 2021; 61:35-40. [PMID: 34311686 DOI: 10.18087/cardio.2021.6.n1536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/27/2021] [Accepted: 03/29/2021] [Indexed: 11/18/2022]
Abstract
Aim To identify clinical and laboratory indexes related with the atherosclerotic plaque (ASP) echogenicity based on results of the analysis of grey-scale median (GSM) in patients aged 40-64 years.Material and methods The study included patients aged 40-64 years with carotid atherosclerosis. The carotid duplex scanning was performed for all patients. The GSM analysis of obtained images was performed with the Adobe Photoshop CS6 software.Results Atherosclerotic cardiovascular diseases were found in 31 (21.4 %) patients. Correlation analysis determined inverse interrelationships between GSM and the body weight index (BWI) (r=-0.359; p<0.0001), waist circumference (r=-0.357; p<0.0001), and levels of uric acid (r=-0.244; p=0.021) and glucose (r=-0.205; p=0.032). According to the regression, statistically significant correlations remained between GSM and BWI as well as the waist circumference after the adjustment for sex and age.Conclusion In patients with carotid atherosclerosis aged 40-64 years, the decrease in ASP GSM was associated with increases in BWI and waist circumference.
Collapse
Affiliation(s)
- V V Genkel
- South Ural State Medical University of the Ministry of Health of the Russian Federation, Chelyabinsk, Russia
| | - A S Kuznetsova
- South Ural State Medical University of the Ministry of Health of the Russian Federation, Chelyabinsk, Russia
| | - E V Lebedev
- South Ural State Medical University of the Ministry of Health of the Russian Federation, Chelyabinsk, Russia
| | - I I Shaposhnik
- South Ural State Medical University of the Ministry of Health of the Russian Federation, Chelyabinsk, Russia
| |
Collapse
|
50
|
Poznyak AV, Bharadwaj D, Prasad G, Grechko AV, Sazonova MA, Orekhov AN. Anti-Inflammatory Therapy for Atherosclerosis: Focusing on Cytokines. Int J Mol Sci 2021; 22:ijms22137061. [PMID: 34209109 PMCID: PMC8269273 DOI: 10.3390/ijms22137061] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 01/03/2023] Open
Abstract
Atherosclerosis is a well-known global health problem. Despite the high prevalence of the disease, numerous aspects of pathogenesis remain unclear. Subsequently, there are still no cure or adequate preventive measures available. Atherogenesis is now considered a complex interplay between lipid metabolism alterations, oxidative stress, and inflammation. Inflammation in atherogenesis involves cellular elements of both innate (such as macrophages and monocytes) and adaptive immunity (such as B-cells and T-cells), as well as various cytokines cascades. Because inflammation is, in general, a well-investigated therapeutic target, and strategies for controlling inflammation have been successfully used to combat a number of other diseases, inflammation seems to be the preferred target for the treatment of atherosclerosis as well. In this review, we summarized data on targeting the most studied inflammatory molecular targets, CRP, IL-1β, IL-6, IFN-γ, and TNF-α. Studies in animal models have shown the efficacy of anti-inflammatory therapy, while clinical studies revealed the incompetence of existing data, which blocks the development of an effective atheroprotective drug. However, all data on cytokine targeting give evidence that anti-inflammatory therapy can be a part of a complex treatment.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| | - Dwaipayan Bharadwaj
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi 110025, India;
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Gauri Prasad
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Margarita A. Sazonova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
- Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|