1
|
Lv L, Guo Y, Zheng Z, Li B. Exploring causal correlations between inflammatory cytokines and coronary heart disease: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e39789. [PMID: 39465855 DOI: 10.1097/md.0000000000039789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
Coronary heart disease (CHD) is a global health concern, with inflammation significantly contributing to its pathogenesis. It is crucial to understand the relationship between inflammatory cytokines and CHD. This study investigates the causal correlations between circulating inflammatory cytokines and CHD using Mendelian randomization (MR), assessing both causative and resultant roles of these cytokines in CHD. In this bidirectional MR analysis, we used genetic data from a genome-wide association study (GWAS) of 60,801 CHD cases and 123,504 controls of European ancestry. We derived inflammatory cytokine data from a GWAS summary of 14,824 participants. The primary analytical approach was the inverse variance-weighted (IVW) method, supported by MR-Egger, weighted median, and weighted mode analyses. Heterogeneity was assessed using the Cochrane Q test, and horizontal pleiotropy was evaluated through the MR-Egger intercept and the MR-PRESSO global test, ensuring robustness against potential pleiotropic bias. This study pinpointed several cytokines as key upstream influencers on the risk of CHD, including eotaxin (CCL11) (odds ratio [OR]: 1.10, 95% confidence interval [CI]: 1.03-1.18, P = .003), C-C motif chemokine ligand 20 (CCL20) (OR: 1.15, 95% CI: 1.05-1.25, P = .002), macrophage colony-stimulating factor 1 (CSF1) (OR: 1.09, 95% CI: 1.01-1.17, P = .020), Fibroblast growth factor 21 (FGF21) (OR: 1.14, 95% CI: 1.01-1.29, P = .038), Fms-related tyrosine kinase 3 ligand (FLT3LG) (OR: 1.26, 95% CI: 1.09-1.44, P = .001), neurotrophin-3 (NT-3) (OR: 1.12, 95% CI: 1.01-1.24, P = .026), and leukemia inhibitory factor (LIF) (OR: 0.89, 95% CI: 0.80-0.99, P = .029). Conversely, T-cell surface glycoprotein CD5 (CD5) (beta: -0.15, 95% CI: -0.29 to -0.01, P = .042) were identified as downstream factors impacted by CHD. No evidence of heterogeneity or horizontal pleiotropy was detected across all results, and a leave-one-out analysis substantiated the robustness of these findings. These findings suggest that CCL11, CCL20, CSF1, FGF21, FLT3LG, NT-3, and LIF may play a crucial role in the pathogenesis of CHD. Additionally, CHD may impact the expression of CD5. Additional research is needed to explore the potential of these biomarkers in the prevention and treatment of CHD.
Collapse
Affiliation(s)
- Luo Lv
- Department of Cardiology, The Second Hospital of Shanxi Medical University, School of Medicine, Shanxi Medical University, Taiyuan, China
| | - Yuli Guo
- Department of Cardiology, The Frist Hospital of Shanxi Medical University, School of Medicine, Shanxi Medical University, Taiyuan, China
| | - Zhongyi Zheng
- Department of Urology, The First Hospital of Shanxi Medical University, School of Medicine, Shanxi Medical University, Taiyuan, China
| | - Bao Li
- Department of Cardiology, The Second Hospital of Shanxi Medical University, School of Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Nagar N, Naidu G, Panda SK, Gulati K, Singh RP, Poluri KM. Elucidating the role of chemokines in inflammaging associated atherosclerotic cardiovascular diseases. Mech Ageing Dev 2024; 220:111944. [PMID: 38782074 DOI: 10.1016/j.mad.2024.111944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Age-related inflammation or inflammaging is a critical deciding factor of physiological homeostasis during aging. Cardiovascular diseases (CVDs) are exquisitely associated with aging and inflammation and are one of the leading causes of high mortality in the elderly population. Inflammaging comprises dysregulation of crosstalk between the vascular and cardiac tissues that deteriorates the vasculature network leading to development of atherosclerosis and atherosclerotic-associated CVDs in elderly populations. Leukocyte differentiation, migration and recruitment holds a crucial position in both inflammaging and atherosclerotic CVDs through relaying the activity of an intricate network of inflammation-associated protein-protein interactions. Among these interactions, small immunoproteins such as chemokines play a major role in the progression of inflammaging and atherosclerosis. Chemokines are actively involved in lymphocyte migration and severe inflammatory response at the site of injury. They relay their functions via chemokine-G protein-coupled receptors-glycosaminoglycan signaling axis and is a principal part for the detection of age-related atherosclerosis and related CVDs. This review focuses on highlighting the detailed intricacies of the effects of chemokine-receptor interaction and chemokine oligomerization on lymphocyte recruitment and its evident role in clinical manifestations of atherosclerosis and related CVDs. Further, the role of chemokine mediated signaling for formulating next-generation therapeutics against atherosclerosis has also been discussed.
Collapse
Affiliation(s)
- Nupur Nagar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Goutami Naidu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Santosh Kumar Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Khushboo Gulati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Ravindra Pal Singh
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Gujarat International Finance Tec-City, Gandhinagar, Gujarat 382355, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| |
Collapse
|
3
|
Xia Y, Wei K, Jiang L, Zou D, Yang Y, Wu S, Hu F, Ma Y. Expression levels and clinical significance of serum miR-19a/CCL20 in patients with acute cerebral infarction. Open Med (Wars) 2024; 19:20240977. [PMID: 38961881 PMCID: PMC11221218 DOI: 10.1515/med-2024-0977] [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: 10/10/2023] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 07/05/2024] Open
Abstract
Acute cerebral infarction (ACI) is a lethal disease whose early diagnosis is critical for treatment. microRNA (miR)-19a targets CC chemokine ligand 20 (CCL20) in myocardial infarction. We investigated the expression patterns of serum miR-19a and CCL20 of ACI patients and assessed their clinical values. Serum samples of 50 healthy subjects and110 ACI patients were collected. Serum levels of miR-19a, CCL20 mRNA, and biochemical indexes were assessed. miR-19a downstream target gene and the binding relationship between miR-19a and CCL20 were predicted and verified. miR-19a and CCL20 mRNA were subjected to correlation and diagnostic efficiency analysis. miR-19a was poorly expressed in the serum of ACI patients, especially in patients with unstable plaque and large infarction. tumor necrosis factor-α, low-density lipoprotein, and platelet/lymphocyte ratio negatively correlated with serum miR-19a level and positively correlated with CCL20. Dual-luciferase assay revealed that miR-19a could negatively regulate CCL20 expression. CCL20 was highly expressed in the serum of ACI patients. The area under receiver-operating characteristic curve of miR-19a combined with CCL20 was 0.9741 (98.00% specificity, 90.91% sensitivity), higher than their single diagnosis. Collectively, miR-19a had high diagnostic value for ACI and could target to restrain CCL20. The combination of miR-19a and CCL20 improved diagnostic value for ACI.
Collapse
Affiliation(s)
- Yongli Xia
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
- Clinical Medicine Department, Sichuan College of Traditional Chinese Medicine, Mianyang621000, Sichuan, China
- Department of Neurosurgery, Anzhou District People’s Hospital, Mianyang622650, Sichuan, China
| | - Kun Wei
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
| | - Lingli Jiang
- Department of Neurosurgery, General Hospital of The Western Theater Command, Chengdu610083, Sichuan, China
| | - Dongbo Zou
- Department of Neurosurgery, General Hospital of The Western Theater Command, Chengdu610083, Sichuan, China
| | - Yuting Yang
- Department of Neurosurgery, General Hospital of The Western Theater Command, Chengdu610083, Sichuan, China
| | - Song Wu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
| | - Fei Hu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
| | - Yuan Ma
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou646000, Sichuan, China
- Department of Neurosurgery, General Hospital of The Western Theater Command, Chengdu610083, Sichuan, China
| |
Collapse
|
4
|
Fransén K, Hiyoshi A, Paramel GV, Hurtig-Wennlöf A. Association between C10X polymorphism in the CARD8 gene and inflammatory markers in young healthy individuals in the LBA study. BMC Cardiovasc Disord 2024; 24:103. [PMID: 38350853 PMCID: PMC10863129 DOI: 10.1186/s12872-024-03765-7] [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: 08/15/2023] [Accepted: 02/03/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND The Caspase activation and recruitment domain 8 (CARD8) protein is a component of innate immunity as a negative regulator of NF- ĸB, and has been associated with regulation of proteins involved in inflammation. Expression of CARD8 mRNA and protein has been identified in human atherosclerotic lesions, and the truncated T30A variant (rs2043211) of CARD8 has been associated with lower C-reactive (CRP) and MCP-1 levels in myocardial infarction patients. The present study examines the role of a genetic variation in the CARD8 gene in relation to a selection of markers of inflammation. METHODS In a cross-sectional study of young healthy individuals (18.0-25.9 yrs, n = 744) the association between the rs2043211 variant in the CARD8 gene and protein markers of inflammation was assessed. Genotyping of the CARD8 C10X (rs2043211) polymorphism was performed with TaqMan real time PCR on DNA from blood samples. Protein levels were studied via Olink inflammation panel ( https://olink.com/ ). Using linear models, we analyzed men and two groups of women with and without estrogen containing contraceptives separately, due to previous findings indicating differences between estrogen users and non-estrogen using women. Genotypes were analyzed by additive, recessive and dominant models. RESULTS The minor (A) allele of the rs2043211 polymorphism in the CARD8 gene was associated with lower levels of CCL20 and IL-6 in men (CCL20, Additive model: p = 0.023; Dominant model: p = 0.016. IL-6, Additive model: p = 0.042; Dominant model: p = 0.039). The associations remained significant also after adjustment for age and potential intermediate variables. CONCLUSIONS Our data indicate that CARD8 may be involved in the regulation of CCL20 and IL-6 in men. No such association was observed in women. These findings strengthen and support previous in vitro data on IL-6 and CCL20 and highlight the importance of CARD8 as a factor in the regulation of inflammatory proteins. The reason to the difference between sexes is however not clear, and the influence of estrogen as a possible factor important for the inflammatory response needs to be further explored.
Collapse
Affiliation(s)
- Karin Fransén
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | - Ayako Hiyoshi
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Geena V Paramel
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Anita Hurtig-Wennlöf
- Department of Clinical Diagnostics, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| |
Collapse
|
5
|
Bondi D, Bevere M, Piccirillo R, Sorci G, Di Felice V, Re Cecconi AD, D'Amico D, Pietrangelo T, Fulle S. Integrated procedures for accelerating, deepening, and leading genetic inquiry: A first application on human muscle secretome. Mol Genet Metab 2023; 140:107705. [PMID: 37837864 DOI: 10.1016/j.ymgme.2023.107705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/15/2023] [Accepted: 10/01/2023] [Indexed: 10/16/2023]
Abstract
PURPOSE Beyond classical procedures, bioinformatic-assisted approaches and computational biology offer unprecedented opportunities for scholars. However, these amazing possibilities still need epistemological criticism, as well as standardized procedures. Especially those topics with a huge body of data may benefit from data science (DS)-assisted methods. Therefore, the current study dealt with the combined expert-assisted and DS-assisted approaches to address the broad field of muscle secretome. We aimed to apply DS tools to fix the literature research, suggest investigation targets with a data-driven approach, predict possible scenarios, and define a workflow. METHODS Recognized scholars with expertise on myokines were invited to provide a list of the most important myokines. GeneRecommender, GeneMANIA, HumanNet, and STRING were selected as DS tools. Networks were built on STRING and GeneMANIA. The outcomes of DS tools included the top 5 recommendations. Each expert-led discussion has been then integrated with an DS-led approach to provide further perspectives. RESULTS Among the results, 11 molecules had already been described as bona-fide myokines in literature, and 11 molecules were putative myokines. Most of the myokines and the putative myokines recommended by the DS tools were described as present in the cargo of extracellular vesicles. CONCLUSIONS Including both supervised and unsupervised learning methods, as well as encompassing algorithms focused on both protein interaction and gene represent a comprehensive approach to tackle complex biomedical topics. DS-assisted methods for reviewing existent evidence, recommending targets of interest, and predicting original scenarios are worth exploring as in silico recommendations to be integrated with experts' ideas for optimizing molecular studies.
Collapse
Affiliation(s)
- Danilo Bondi
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Michele Bevere
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.
| | - Rosanna Piccirillo
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy.
| | - Guglielmo Sorci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Valentina Di Felice
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Andrea David Re Cecconi
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy.
| | - Daniela D'Amico
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Tiziana Pietrangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Stefania Fulle
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| |
Collapse
|
6
|
Li L, Zeng Z, Yagublu V, Rahbari N, Reißfelder C, Keese M. Analysis of Inflammation-Related Genes in Patients with Stanford Type A Aortic Dissection. J Pers Med 2023; 13:990. [PMID: 37373979 DOI: 10.3390/jpm13060990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Background: Aortic dissection (AD) is a life-threatening cardiovascular disease. Pathophysiologically, it has been shown that aortic wall inflammation promotes the occurrence and development of aortic dissection. Thus, the aim of the current research was to determine the inflammation-related biomarkers in AD. Methods: In this study, we conducted differentially expressed genes (DEGs) analysis using the GSE153434 dataset containing 10 type A aortic dissection (TAAD) and 10 normal samples downloaded from the Gene Expression Omnibus (GEO) database. The intersection of DEGs and inflammation-related genes was identified as differential expressed inflammation-related genes (DEIRGs). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed for DEIRGs. We then constructed the protein-protein interaction (PPI) network using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database and identified hub genes using the Cytoscape plugin MCODE. Finally, least absolute shrinkage and selection operator (LASSO) logistic regression was used to construct a diagnostic model. Results: A total of 1728 DEGs were identified between the TAAD and normal samples. Thereafter, 61 DEIRGs are obtained by taking the intersection of DEGs and inflammation-related genes. The GO indicated that DEIRGs were mainly enriched in response to lipopolysaccharide, in response to molecules of bacterial origin, secretory granule membrane, external side of plasma, receptor ligand activity, and signaling receptor activator activity. KEGG analysis indicated that DEIRGs were mainly enriched in cytokine-cytokine receptor interaction, TNF signaling pathway, and proteoglycans in cancer. We identified MYC, SELL, HIF1A, EDN1, SERPINE1, CCL20, IL1R1, NOD2, TLR2, CD69, PLAUR, MMP14, and HBEGF as hub genes using the MCODE plug-in. The ROC indicated these genes had a good diagnostic performance for TAAD. Conclusion: In conclusion, our study identified 13 hub genes in the TAAD. This study will be of significance for the future development of a preventive therapy of TAAD.
Collapse
Affiliation(s)
- Lin Li
- Department of Vascular Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- European Center of Angioscience ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ziwei Zeng
- Department of Vascular Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- European Center of Angioscience ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Vugar Yagublu
- Surgical Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Nuh Rahbari
- Surgical Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Christoph Reißfelder
- Surgical Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Michael Keese
- European Center of Angioscience ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Department of Vascular Surgery, Theresienkrankenhaus, 68165 Mannheim, Germany
| |
Collapse
|
7
|
Mylonas KS, Peroulis M, Schizas D, Kapelouzou A. MYD88 and Proinflammatory Chemokines in Aortic Atheromatosis: Exploring Novel Statin Effects. Int J Mol Sci 2023; 24:ijms24119248. [PMID: 37298199 DOI: 10.3390/ijms24119248] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Atherosclerosis is driven by a diverse range of cellular and molecular processes. In the present study, we sought to better understand how statins mitigate proatherogenic inflammation. 48 male New Zealand rabbits were divided into eight groups, each including 6 animals. The control groups received normal chow for 90 and 120 days. Three groups underwent a hypercholesterolemic diet (HCD) for 30, 60, and 90 days. Another three groups underwent HCD for 3 months, followed by normal chow for one month, with or without rosuvastatin or fluvastatin. The cytokine and chemokine expressions were assessed in the samples of thoracic and abdominal aorta. Rosuvastatin significantly reduced MYD88, CCL4, CCL20, CCR2, TNF-α, IFN-β, IL-1b, IL-2, IL-4, IL-8, and IL-10, both in the thoracic and abdominal aorta. Fluvastatin also downregulated MYD88, CCR2, IFN-β, IFN-γ, IL-1b, IL-2, IL-4, and IL-10 in both aortic segments. Rosuvastatin curtailed the expression of CCL4, IFN-β, IL-2, IL-4, and IL-10 more effectively than fluvastatin in both types of tissue. MYD88, TNF-α, IL-1b, and IL-8 showed a stronger downregulation with rosuvastatin compared to fluvastatin only in the thoracic aorta. The CCL20 and CCR2 levels reduced more extensively with rosuvastatin treatment only in abdominal aortic tissue. In conclusion, statin therapy can halt proatherogenic inflammation in hyperlipidemic animals. Rosuvastatin may be more effective in downregulating MYD88 in atherosclerotic thoracic aortas.
Collapse
Affiliation(s)
| | - Michail Peroulis
- Vascular Surgery Unit, Department of Surgery, Faculty of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| | - Dimitrios Schizas
- First Department of Cardiac Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, 176 71 Athens, Greece
| | - Alkistis Kapelouzou
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, 176 71 Athens, Greece
| |
Collapse
|
8
|
Geelen IGP, Gullaksen SE, Ilander MM, Olssen-Strömberg U, Mustjoki S, Richter J, Blijlevens NMA, Smit WM, Gjertsen BT, Gedde-Dahl T, Markevärn B, Koppes MMA, Westerweel PE, Hjorth-Hansen H, Janssen JJWM. Switching from imatinib to nilotinib plus pegylated interferon-α2b in chronic phase CML failing to achieve deep molecular response: clinical and immunological effects. Ann Hematol 2023; 102:1395-1408. [PMID: 37119314 DOI: 10.1007/s00277-023-05199-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 03/23/2023] [Indexed: 05/01/2023]
Abstract
In order to improve molecular response for a discontinuation attempt in chronic myeloid leukemia (CML) patients in chronic phase, who had not achieved at least a molecular response <0.01% BCR-ABL1IS (MR4.0) after at least 2 years of imatinib therapy, we prospectively evaluated whether they could attain MR4.0 after a switch to a combination of nilotinib and 9 months of pegylated interferon-α2b (PegIFN). The primary endpoint of confirmed MR4.0 at month 12 (a BCR-ABL1IS level ≤ 0.01% both at 12 and 15 months) was reached by 44% (7/16 patients, 95% confidence interval (CI): 23- 67%) of patients, with 81% (13/16 patients, 95% CI: 57-93%) of patients achieving an unconfirmed MR4.0. The scheduled combination was completed by 56% of the patients, with premature discontinuations, mainly due to mood disturbances after the introduction of PegIFN, questioning the feasibility of the combination of nilotinib and PegIFN for this patient population and treatment goal. A comprehensive clinical substudy program was implemented to characterize the impact of the treatment changes on the immunological profile. This trial was registered at www.clinicaltrials.gov as #NCT01866553.
Collapse
Affiliation(s)
- Inge G P Geelen
- Department of Internal Medicine / Hematology, Albert Schweitzer Hospital, Dordrecht, The Netherlands.
| | - Stein-Erik Gullaksen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology section, Helse Bergen, Bergen, Norway
| | - Mette M Ilander
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer center, Helsinki, Finland
| | | | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Johan Richter
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | | - Willem M Smit
- Department of Hematology, Medical Spectrum Twente, Enschede, The Netherlands
| | - Bjorn T Gjertsen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology section, Helse Bergen, Bergen, Norway
| | - Tobias Gedde-Dahl
- Department of Hematology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Berit Markevärn
- Department of Hematology, Umeå University Hospital, Umeå, Sweden
| | - Malika M A Koppes
- Department of Hematology, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| | - Peter E Westerweel
- Department of Internal Medicine / Hematology, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Henrik Hjorth-Hansen
- Department of Hematology, St Olavs Hospital, Trondheim, Norway
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jeroen J W M Janssen
- Department of Hematology, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Xu Y, Yang S, Xue G. The role of long non-coding RNA in abdominal aortic aneurysm. Front Genet 2023; 14:1153899. [PMID: 37007957 PMCID: PMC10050724 DOI: 10.3389/fgene.2023.1153899] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
The abdominal aortic aneurysm (AAA) is characterized by segmental expansion of the abdominal aorta and a high mortality rate. The characteristics of AAA suggest that apoptosis of smooth muscle cells, the production of reactive oxygen species, and inflammation are potential pathways for the formation and development of AAA. Long non-coding RNA (lncRNA) is becoming a new and essential regulator of gene expression. Researchers and physicians are focusing on these lncRNAs to use them as clinical biomarkers and new treatment targets for AAAs. LncRNA studies are beginning to emerge, suggesting that they may play a significant but yet unidentified role in vascular physiology and disease. This review examines the role of lncRNA and their target genes in AAA to increase our understanding of the disease’s onset and progression, which is crucial for developing potential AAA therapies.
Collapse
|
10
|
Kennon AM, Stewart JA. Paracrine Signals in Calcified Conditioned Media Elicited Differential Responses in Primary Aortic Vascular Smooth Muscle Cells and in Adventitial Fibroblasts. Int J Mol Sci 2023; 24:ijms24043599. [PMID: 36835011 PMCID: PMC9961433 DOI: 10.3390/ijms24043599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Our goal was to determine if paracrine signals from different aortic layers can impact other cell types in the diabetic microenvironment, specifically medial vascular smooth muscle cells (VSMCs) and adventitial fibroblasts (AFBs). The diabetic hyperglycemic aorta undergoes mineral dysregulation, causing cells to be more responsive to chemical messengers eliciting vascular calcification. Advanced glycation end-products (AGEs)/AGE receptors (RAGEs) signaling has been implicated in diabetes-mediated vascular calcification. To elucidate responses shared between cell types, pre-conditioned calcified media from diabetic and non-diabetic VSMCs and AFBs were collected to treat cultured murine diabetic, non-diabetic, diabetic RAGE knockout (RKO), and non-diabetic RKO VSMCs and AFBs. Calcium assays, western blots, and semi-quantitative cytokine/chemokine profile kits were used to determine signaling responses. VSMCs responded to non-diabetic more than diabetic AFB calcified pre-conditioned media. AFB calcification was not significantly altered when VSMC pre-conditioned media was used. No significant changes in VSMCs signaling markers due to treatments were reported; however, genotypic differences existed. Losses in AFB α-smooth muscle actin were observed with diabetic pre-conditioned VSMC media treatment. Superoxide dismutase-2 (SOD-2) increased with non-diabetic calcified + AGE pre-conditioned VSMC media, while same treatment decreased diabetic AFBs levels. Overall, non-diabetic and diabetic pre-conditioned media elicited different responses from VSMCs and AFBs.
Collapse
Affiliation(s)
- Amber M. Kennon
- Department of Investigational Cancer, Division of Cancer Medicine, U.T.M.D Anderson Cancer Center, Houston, TX 77030, USA
| | - James A. Stewart
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
- Correspondence: ; Tel.: +1-(662)-915-2309
| |
Collapse
|
11
|
Ebadi N, Arefizadeh R, Nasrollahzadeh Sabet M, Goodarzi N. Identification of Key Genes and Biological Pathways Related to Myocardial Infarction through Integrated Bioinformatics Analysis. IRANIAN JOURNAL OF MEDICAL SCIENCES 2023; 48:35-42. [PMID: 36688193 PMCID: PMC9843455 DOI: 10.30476/ijms.2022.92656.2395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/10/2022] [Accepted: 01/31/2022] [Indexed: 01/24/2023]
Abstract
Background Coronary heart disease is the leading cause of death worldwide. Myocardial infarction (MI) is a fatal manifestation of coronary heart disease, which can present as sudden death. Although the molecular mechanisms of coronary heart disease are still unknown, global gene expression profiling is regarded as a useful approach for deciphering the pathophysiology of this disease and subsequent diseases. This study used a bioinformatics analysis approach to better understand the molecular mechanisms underlying coronary heart disease. Methods This experimental study was conducted in the department of cardiology, Aja University of Medical Sciences (2021-2022), Tehran, Iran. To identify the key deregulated genes and pathways in coronary heart disease, an integrative approach was used by merging three gene expression datasets, including GSE19339, GSE66360, and GSE29111, into a single matrix. The t test was used for the statistical analysis, with a significance level of P<0.05. Results The limma package in R was used to identify a total of 133 DEGs, consisting of 124 upregulated and nine downregulated genes. KDM5D, EIF1AY, and CCL20 are among the top upregulated genes. Moreover, the interleukin 17 (IL-17) signaling pathway and four other signaling pathways were identified as the potent underlying pathogenesis of both coronary artery disease (CAD) and MI using a systems biology approach. Accordingly, these findings can provide expression signatures and potential biomarkers in CAD and MI pathophysiology, which can contribute to both diagnosis and therapeutic purposes. Conclusion Five signaling pathways were introduced in MI and CAD that were primarily involved in inflammation, including the IL-17 signaling pathway, TNF signaling pathway, toll-like receptor signaling pathway, C-type lectin receptor signaling pathway, and rheumatoid arthritis signaling pathway.
Collapse
Affiliation(s)
- Nader Ebadi
- Department of Cardiology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran,
Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Arefizadeh
- Department of Cardiology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | | | - Naser Goodarzi
- Department of Clinical Psychology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Van Nynatten LR, Slessarev M, Martin CM, Leligdowicz A, Miller MR, Patel MA, Daley M, Patterson EK, Cepinskas G, Fraser DD. Novel plasma protein biomarkers from critically ill sepsis patients. Clin Proteomics 2022; 19:50. [PMID: 36572854 PMCID: PMC9792322 DOI: 10.1186/s12014-022-09389-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Despite the high morbidity and mortality associated with sepsis, the relationship between the plasma proteome and clinical outcome is poorly understood. In this study, we used targeted plasma proteomics to identify novel biomarkers of sepsis in critically ill patients. METHODS Blood was obtained from 15 critically ill patients with suspected/confirmed sepsis (Sepsis-3.0 criteria) on intensive care unit (ICU) Day-1 and Day-3, as well as age- and sex-matched 15 healthy control subjects. A total of 1161 plasma proteins were measured with proximal extension assays. Promising sepsis biomarkers were narrowed with machine learning and then correlated with relevant clinical and laboratory variables. RESULTS The median age for critically ill sepsis patients was 56 (IQR 51-61) years. The median MODS and SOFA values were 7 (IQR 5.0-8.0) and 7 (IQR 5.0-9.0) on ICU Day-1, and 4 (IQR 3.5-7.0) and 6 (IQR 3.5-7.0) on ICU Day-3, respectively. Targeted proteomics, together with feature selection, identified the leading proteins that distinguished sepsis patients from healthy control subjects with ≥ 90% classification accuracy; 25 proteins on ICU Day-1 and 26 proteins on ICU Day-3 (6 proteins overlapped both ICU days; PRTN3, UPAR, GDF8, NTRK3, WFDC2 and CXCL13). Only 7 of the leading proteins changed significantly between ICU Day-1 and Day-3 (IL10, CCL23, TGFα1, ST2, VSIG4, CNTN5, and ITGAV; P < 0.01). Significant correlations were observed between a variety of patient clinical/laboratory variables and the expression of 15 proteins on ICU Day-1 and 14 proteins on ICU Day-3 (P < 0.05). CONCLUSIONS Targeted proteomics with feature selection identified proteins altered in critically ill sepsis patients relative to healthy control subjects. Correlations between protein expression and clinical/laboratory variables were identified, each providing pathophysiological insight. Our exploratory data provide a rationale for further hypothesis-driven sepsis research.
Collapse
Affiliation(s)
| | - Marat Slessarev
- grid.39381.300000 0004 1936 8884Medicine, Western University, London, ON Canada ,grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, London, ON Canada
| | - Claudio M. Martin
- grid.39381.300000 0004 1936 8884Medicine, Western University, London, ON Canada ,grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, London, ON Canada
| | - Aleks Leligdowicz
- grid.39381.300000 0004 1936 8884Medicine, Western University, London, ON Canada ,grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, London, ON Canada
| | - Michael R. Miller
- grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, London, ON Canada ,grid.39381.300000 0004 1936 8884Pediatrics, Western University, London, ON Canada
| | - Maitray A. Patel
- grid.39381.300000 0004 1936 8884Computer Science, Western University, London, ON N6A 3K7 Canada
| | - Mark Daley
- grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, London, ON Canada ,grid.39381.300000 0004 1936 8884Computer Science, Western University, London, ON N6A 3K7 Canada ,grid.494618.6The Vector Institute for Artificial Intelligence, Toronto, ON M5G 1M1 Canada
| | - Eric K. Patterson
- grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, London, ON Canada
| | - Gediminas Cepinskas
- grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, London, ON Canada ,grid.39381.300000 0004 1936 8884Medical Biophysics, Western University, London, ON N6A 3K7 Canada
| | - Douglas D. Fraser
- grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, London, ON Canada ,grid.39381.300000 0004 1936 8884Pediatrics, Western University, London, ON Canada ,grid.39381.300000 0004 1936 8884Clinical Neurological Sciences, Western University, London, ON Canada ,grid.39381.300000 0004 1936 8884Physiology and Pharmacology, Western University, London, ON Canada ,grid.412745.10000 0000 9132 1600London Health Sciences Centre, Room C2-C82, 800 Commissioners Road East, London, ON N6A 5W9 Canada
| |
Collapse
|
13
|
Li W, Jin K, Luo J, Xu W, Wu Y, Zhou J, Wang Y, Xu R, Jiao L, Wang T, Yang G. NF-κB and its crosstalk with endoplasmic reticulum stress in atherosclerosis. Front Cardiovasc Med 2022; 9:988266. [PMID: 36204587 PMCID: PMC9530249 DOI: 10.3389/fcvm.2022.988266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis (AS) is a common cardiovascular disease with complex pathogenesis, in which multiple pathways and their interweaving regulatory mechanism remain unclear. The primary transcription factor NF-κB plays a critical role in AS via modulating the expression of a series of inflammatory mediators under various stimuli such as cytokines, microbial antigens, and intracellular stresses. Endoplasmic reticulum (ER) stress, caused by the disrupted synthesis and secretion of protein, links inflammation, metabolic signals, and other cellular processes via the unfolded protein response (UPR). Both NF-κB and ER stress share the intersection regarding their molecular regulation and function and are regarded as critical individual contributors to AS. In this review, we summarize the multiple interactions between NF-κB and ER stress activation, including the UPR, NLRP3 inflammasome, and reactive oxygen species (ROS) generation, which have been ignored in the pathogenesis of AS. Given the multiple links between NF-κB and ER stress, we speculate that the integrated network contributes to the understanding of molecular mechanisms of AS. This review aims to provide an insight into these interactions and their underlying roles in the progression of AS, highlighting potential pharmacological targets against the atherosclerotic inflammatory process.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jia Zhou
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Liqun Jiao,
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Tao Wang,
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- Tao Wang,
| |
Collapse
|
14
|
Jiménez-Fernández M, Rodríguez-Sinovas C, Cañes L, Ballester-Servera C, Vara A, Requena S, de la Fuente H, Martínez-González J, Sánchez-Madrid F. CD69-oxLDL ligand engagement induces Programmed Cell Death 1 (PD-1) expression in human CD4 + T lymphocytes. Cell Mol Life Sci 2022; 79:468. [PMID: 35930205 PMCID: PMC9355928 DOI: 10.1007/s00018-022-04481-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/28/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022]
Abstract
The mechanisms that control the inflammatory–immune response play a key role in tissue remodelling in cardiovascular diseases. T cell activation receptor CD69 binds to oxidized low-density lipoprotein (oxLDL), inducing the expression of anti-inflammatory NR4A nuclear receptors and modulating inflammation in atherosclerosis. To understand the downstream T cell responses triggered by the CD69-oxLDL binding, we incubated CD69-expressing Jurkat T cells with oxLDL. RNA sequencing revealed a differential gene expression profile dependent on the presence of CD69 and the degree of LDL oxidation. CD69-oxLDL binding induced the expression of NR4A receptors (NR4A1 and NR4A3), but also of PD-1. These results were confirmed using oxLDL and a monoclonal antibody against CD69 in CD69-expressing Jurkat and primary CD4 + lymphocytes. CD69-mediated induction of PD-1 and NR4A3 was dependent on NFAT activation. Silencing NR4A3 slightly increased PD-1 levels, suggesting a potential regulation of PD-1 by this receptor. Moreover, expression of PD-1, CD69 and NR4A3 was increased in human arteries with chronic inflammation compared to healthy controls, with a strong correlation between PD-1 and CD69 mRNA expression (r = 0.655 P < 0.0001). Moreover, PD-1 was expressed in areas enriched in CD3 infiltrating T cells. Our results underscore a novel mechanism of PD-1 induction independent of TCR signalling that might contribute to the role of CD69 in the modulation of inflammation and vascular remodelling in cardiovascular diseases.
Collapse
Affiliation(s)
- María Jiménez-Fernández
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cristina Rodríguez-Sinovas
- Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), IIB-Sant Pau, Barcelona, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Laia Cañes
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Instituto de Investigaciones Biomédicas de Barcelona - Consejo Superior de Investigaciones Científicas (IIBB-CSIC), IIB-Sant Pau, C/ Rosselló, 161, 08036, Barcelona, Spain
| | - Carme Ballester-Servera
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Instituto de Investigaciones Biomédicas de Barcelona - Consejo Superior de Investigaciones Científicas (IIBB-CSIC), IIB-Sant Pau, C/ Rosselló, 161, 08036, Barcelona, Spain
| | - Alicia Vara
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain
| | - Silvia Requena
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain
| | - Hortensia de la Fuente
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - José Martínez-González
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain. .,Instituto de Investigaciones Biomédicas de Barcelona - Consejo Superior de Investigaciones Científicas (IIBB-CSIC), IIB-Sant Pau, C/ Rosselló, 161, 08036, Barcelona, Spain.
| | - Francisco Sánchez-Madrid
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), c/ Diego de León, 62, 28006, Madrid, Spain. .,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain. .,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
15
|
Endothelial Dysfunction in the Pathogenesis of Abdominal Aortic Aneurysm. Biomolecules 2022; 12:biom12040509. [PMID: 35454098 PMCID: PMC9030795 DOI: 10.3390/biom12040509] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/18/2022] [Accepted: 03/27/2022] [Indexed: 12/25/2022] Open
Abstract
Abdominal aortic aneurysm (AAA), defined as a focal dilation of the abdominal aorta beyond 50% of its normal diameter, is a common and potentially life-threatening vascular disease. The molecular and cellular mechanisms underlying AAA pathogenesis remain unclear. Healthy endothelial cells (ECs) play a critical role in maintaining vascular homeostasis by regulating vascular tone and maintaining an anti-inflammatory, anti-thrombotic local environment. Increasing evidence indicates that endothelial dysfunction is an early pathologic event in AAA formation, contributing to both oxidative stress and inflammation in the degenerating arterial wall. Recent studies utilizing single-cell RNA sequencing revealed heterogeneous EC sub-populations, as determined by their transcriptional profiles, in aortic aneurysm tissue. This review summarizes recent findings, including clinical evidence of endothelial dysfunction in AAA, the impact of biomechanical stress on EC in AAA, the role of endothelial nitric oxide synthase (eNOS) uncoupling in AAA, and EC heterogeneity in AAA. These studies help to improve our understanding of AAA pathogenesis and ultimately may lead to the generation of EC-targeted therapeutics to treat or prevent this deadly disease.
Collapse
|
16
|
Wu PH, Glerup RI, Svensson MHS, Eriksson N, Christensen JH, de Laval P, Soveri I, Westerlund M, Linde T, Ljunggren Ö, Fellström B. Novel Biomarkers Detected by Proteomics Predict Death and Cardiovascular Events in Hemodialysis Patients. Biomedicines 2022; 10:biomedicines10040740. [PMID: 35453489 PMCID: PMC9026983 DOI: 10.3390/biomedicines10040740] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/06/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
End-stage kidney disease increases mortality and the risk of cardiovascular (CV) disease. It is crucial to explore novel biomarkers to predict CV disease in the complex setting of patients receiving hemodialysis (HD). This study investigated the association between 92 targeted proteins with all-cause death, CV death, and composite vascular events (CVEs) in HD patients. From December 2010 to March 2011, 331 HD patients were included and followed prospectively for 5 years. Serum was analyzed for 92 CV-related proteins using Proseek Multiplex Cardiovascular I panel, a high-sensitivity assay based on proximity extension assay (PEA) technology. The association between biomarkers and all-cause death, CV death, and CVEs was evaluated using Cox-regression analyses. Of the PEA-based proteins, we identified 20 proteins associated with risk of all-cause death, 7 proteins associated with risk of CV death, and 17 proteins associated with risk of CVEs, independent of established risk factors. Interleukin-8 (IL-8), T-cell immunoglobulin and mucin domain 1 (TIM-1), and C-C motif chemokine 20 (CCL20) were associated with increased risk of all-cause death, CV death, and CVE in multivariable-adjusted models. Stem cell factor (SCF) and Galanin peptides (GAL) were associated with both decreased risk of all-cause death and CV death. In conclusion, IL-8, TIM-1, and CCL20 predicted death and CV outcomes in HD patients. Novel findings were that SCF and GAL were associated with a lower risk of all-cause death and CV death. The SCF warrants further study with regard to its possible biological effect in HD patients.
Collapse
Affiliation(s)
- Ping-Hsun Wu
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Big Data Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Rie Io Glerup
- Department of Nephrology, Aalborg University Hospital, 9000 Aalborg, Denmark; (R.I.G.); (J.H.C.)
| | - My Hanna Sofia Svensson
- Division of Medicine, Department of Nephrology, Akershus University Hospital, 1478 Oslo, Norway;
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, 75185 Uppsala, Sweden;
| | | | - Philip de Laval
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Inga Soveri
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Magnus Westerlund
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Torbjörn Linde
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Östen Ljunggren
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Bengt Fellström
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
- Correspondence: ; Tel.: +46-18-6114348
| |
Collapse
|
17
|
Sobolev VV, Soboleva AG, Denisova EV, Pechatnikova EA, Dvoryankova E, Korsunskaya IM, Mezentsev A. Proteomic Studies of Psoriasis. Biomedicines 2022; 10:biomedicines10030619. [PMID: 35327421 PMCID: PMC8945259 DOI: 10.3390/biomedicines10030619] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
In this review paper, we discuss the contribution of proteomic studies to the discovery of disease-specific biomarkers to monitor the disease and evaluate available treatment options for psoriasis. Psoriasis is one of the most prevalent skin disorders driven by a Th17-specific immune response. Although potential patients have a genetic predisposition to psoriasis, the etiology of the disease remains unknown. During the last two decades, proteomics became deeply integrated with psoriatic research. The data obtained in proteomic studies facilitated the discovery of novel mechanisms and the verification of many experimental hypotheses of the disease pathogenesis. The detailed data analysis revealed multiple differentially expressed proteins and significant changes in proteome associated with the disease and drug efficacy. In this respect, there is a need for proteomic studies to characterize the role of the disease-specific biomarkers in the pathogenesis of psoriasis, develop clinical applications to choose the most efficient treatment options and monitor the therapeutic response.
Collapse
Affiliation(s)
- Vladimir V. Sobolev
- Centre of Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.G.S.); (E.V.D.); (E.D.); (I.M.K.)
- Correspondence: (V.V.S.); (A.M.)
| | - Anna G. Soboleva
- Centre of Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.G.S.); (E.V.D.); (E.D.); (I.M.K.)
- Scientific Research Institute of Human Morphology, 117418 Moscow, Russia
| | - Elena V. Denisova
- Centre of Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.G.S.); (E.V.D.); (E.D.); (I.M.K.)
- Moscow Scientific and Practical Center of Dermatovenereology and Cosmetology, 119071 Moscow, Russia
| | - Eva A. Pechatnikova
- Department of Dermatology and Cosmetology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Eugenia Dvoryankova
- Centre of Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.G.S.); (E.V.D.); (E.D.); (I.M.K.)
| | - Irina M. Korsunskaya
- Centre of Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.G.S.); (E.V.D.); (E.D.); (I.M.K.)
| | - Alexandre Mezentsev
- Centre of Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.G.S.); (E.V.D.); (E.D.); (I.M.K.)
- Correspondence: (V.V.S.); (A.M.)
| |
Collapse
|
18
|
Quercetin Relieves the Excised Great Saphenous Vein Oxidative Damage and Inflammatory Reaction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2021:6251559. [PMID: 35003306 PMCID: PMC8741364 DOI: 10.1155/2021/6251559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/13/2021] [Accepted: 11/19/2021] [Indexed: 12/03/2022]
Abstract
Objective The patency and quality of transplanted great saphenous vein (GSV) can seriously influence the physical state and life quality of patients who accepted the coronary artery bypass grafting (CABG). Quercetin is known for antioxidant, antithrombotic, anti-inflammatory, and antitumor properties. In this study, we examined the protection of quercetin to the great saphenous vein from oxidative and inflammatory damage. Methods The GSVs were collected from 15 patients undergoing CABG and cultured. Treated the veins by H2O2 and detected the NO, SOD, and MDA content by the relevant kits to explore the quercetin protection against oxidative damage. Then, for another group of GSVs, sheared them and detected the inflammatory cytokines, such as IL-6, TNFα, CCL20, PCNA, and VEGF. Collect the veins for H&E staining and PCNA and VEGF immunofluorescent staining. Results Pretreatment by quercetin reduced the production of NO and MDA induced by H2O2, and increased SOD activity. Quercetin also supressed the mRNA expressions of IL-6, TNFα after mechanical damage and had no influence on CCL20 and VEGF. Consistent with the lower expression of PCNA treated by quercetin, the vein intima was thinner. Conclusion These results demonstrated that quercetin protects GSVs by reducing the oxidative damage and inflammatory response and also suppresses the abnormal thickening of venous endothelium by inhibiting cell proliferation. It reminded that, to some extent, quercetin has the potential to release the great saphenous vein graft damage.
Collapse
|
19
|
Srikakulapu P, Upadhye A, Drago F, Perry HM, Bontha SV, McSkimming C, Marshall MA, Taylor AM, McNamara CA. Chemokine Receptor-6 Promotes B-1 Cell Trafficking to Perivascular Adipose Tissue, Local IgM Production and Atheroprotection. Front Immunol 2021; 12:636013. [PMID: 33679793 PMCID: PMC7933012 DOI: 10.3389/fimmu.2021.636013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Chemokine receptor-6 (CCR6) mediates immune cell recruitment to inflammatory sites and has cell type-specific effects on diet-induced atherosclerosis in mice. Previously we showed that loss of CCR6 in B cells resulted in loss of B cell-mediated atheroprotection, although the B cell subtype mediating this effect was unknown. Perivascular adipose tissue (PVAT) harbors high numbers of B cells including atheroprotective IgM secreting B-1 cells. Production of IgM antibodies is a major mechanism whereby B-1 cells limit atherosclerosis development. Yet whether CCR6 regulates B-1 cell number and production of IgM in the PVAT is unknown. In this present study, flow cytometry experiments demonstrated that both B-1 and B-2 cells express CCR6, albeit at a higher frequency in B-2 cells in both humans and mice. Nevertheless, B-2 cell numbers in peritoneal cavity (PerC), spleen, bone marrow and PVAT were no different in ApoE -/- CCR6 -/- compared to ApoE -/- CCR6 +/+ mice. In contrast, the numbers of atheroprotective IgM secreting B-1 cells were significantly lower in the PVAT of ApoE -/- CCR6 -/- compared to ApoE -/- CCR6 +/+ mice. Surprisingly, adoptive transfer (AT) of CD43- splenic B cells into B cell-deficient μMT -/- ApoE -/- mice repopulated the PerC with B-1 and B-2 cells and reduced atherosclerosis when transferred into ApoE -/- CCR6 +/+ sIgM -/- mice only when those cells expressed both CCR6 and sIgM. CCR6 expression on circulating human B cells in subjects with a high level of atherosclerosis in their coronary arteries was lower only in the putative human B-1 cells. These results provide evidence that B-1 cell CCR6 expression enhances B-1 cell number and IgM secretion in PVAT to provide atheroprotection in mice and suggest potential human relevance to our murine findings.
Collapse
Affiliation(s)
- Prasad Srikakulapu
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Aditi Upadhye
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Fabrizio Drago
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Heather M Perry
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Sai Vineela Bontha
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Chantel McSkimming
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Melissa A Marshall
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Angela M Taylor
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Coleen A McNamara
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
20
|
Elnabawi YA, Garshick MS, Tawil M, Barrett TJ, Fisher EA, Lo Sicco K, Neimann AL, Scher JU, Krueger J, Berger JS. CCL20 in psoriasis: A potential biomarker of disease severity, inflammation, and impaired vascular health. J Am Acad Dermatol 2020; 84:913-920. [PMID: 33259876 DOI: 10.1016/j.jaad.2020.10.094] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/29/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Psoriasis is associated with increased cardiovascular risk that is not captured by traditional proinflammatory biomarkers. OBJECTIVE To investigate the relationship between Psoriasis Area and Severity Index, circulating proinflammatory biomarkers, and vascular health in psoriasis. METHODS In patients with psoriasis and in age and sex-matched controls, 273 proteins were analyzed with the Proseek Multiplex Cardiovascular disease reagents kit and Inflammatory reagents kit (Olink Bioscience), whereas vascular endothelial inflammation and health were measured via direct transcriptomic analysis of brachial vein endothelial cells. RESULTS In psoriasis, chemokine ligand 20 (CCL20), interleukin (IL) 6, and IL-17A were the top 3 circulating proinflammatory cytokines. Vascular endothelial inflammation correlated with CCL20 (r = 0.55; P < .001) and less so with IL-6 (r = 0.36; P = .04) and IL-17A (r = 0.29; P = .12). After adjustment for potential confounders, the association between CCL20 and vascular endothelial inflammation remained significant (β = 1.71; P = .02). In nested models, CCL20 added value (χ2 = 79.22; P < .001) to a model already incorporating the Psoriasis Area and Severity Index, Framingham risk, high-sensitivity C-reactive protein, Il-17A, and IL-6 (χ2 = 48.18; P < .001) in predicting vascular endothelial inflammation. LIMITATIONS Our study was observational and did not allow for causal inference in the relationship between CCL20 and cardiovascular risk. CONCLUSION We demonstrate that CCL20 expression has a strong association with vascular endothelial inflammation, reflects systemic inflammation, and may serve as a potential biomarker of impaired vascular health in psoriasis.
Collapse
Affiliation(s)
- Youssef A Elnabawi
- Department of Internal Medicine, New York University School of Medicine, New York, New York
| | - Michael S Garshick
- Center for the Prevention of Cardiovascular Disease, New York University School of Medicine, New York, New York; Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, New York.
| | - Michael Tawil
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, New York
| | - Tessa J Barrett
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, New York
| | - Edward A Fisher
- Center for the Prevention of Cardiovascular Disease, New York University School of Medicine, New York, New York; Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, New York
| | - Kristen Lo Sicco
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York
| | - Andrea L Neimann
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York
| | - Jose U Scher
- Psoriatic Arthritis Center, Division of Rheumatology, New York University School of Medicine, New York, New York
| | - James Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, New York
| | - Jeffrey S Berger
- Center for the Prevention of Cardiovascular Disease, New York University School of Medicine, New York, New York; Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, New York; Division of Hematology, New York University School of Medicine, New York, New York; Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, New York, New York
| |
Collapse
|
21
|
Munjal A, Khandia R. Atherosclerosis: orchestrating cells and biomolecules involved in its activation and inhibition. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 120:85-122. [PMID: 32085889 DOI: 10.1016/bs.apcsb.2019.11.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The term atherosclerosis refers to the condition of deposition of lipids and other substances in and on the artery walls, called as plaque that restricts the normal blood flow. The plaque may be stable or unstable in nature. Unstable plaque can burst and trigger clot formation adding further adversities. The process of plaque formation involves various stages including fatty streak, intermediate or fibro-fatty lesion and advanced lesion. The cells participating in the formation of atherosclerotic plaque include endothelial cells, vascular smooth muscle cells (VSMC), monocytes, monocytes derived macrophages, macrophages and dendritic cells and regulatory T cells (TREG). The role of a variety of cytokines and chemokines have been studied which either help in progression of atherosclerotic plaque or vice versa. The cytokines involved in atherosclerotic plaque formation include IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12, IL-13, IL-15, IL-17, IL-18, IL-20, IL-25, IL-27, IL-33, IL-37, TNF-α, TGF-β and IFN-γ; whereas amongst the chemokines (family of small cytokines) are CCL2, CCL3, CXCL4, CCL5, CXCL1, CX3CL1, CCL17, CXCL8, CXCL10, CCL20, CCL19 and CCL21 and macrophage migration-inhibitory factor. These are involved in the atherosclerosis advancements, whereas the chemokine CXCL12 is play atheroprotective roles. Apart this, contradictory functions have been documented for few other chemokines such as CXCL16. Since the cytokines and chemokines are amongst the key molecules involved in orchestrating the atherosclerosis advancements, targeting them might be an effective strategy to encumber the atherosclerotic progression. Blockage of cytokines and chemokines via the means of broad-spectrum inhibitors, neutralizing antibodies, usage of decoy receptors or RNA interference have been proved to be useful intervention against atherosclerosis.
Collapse
Affiliation(s)
- Ashok Munjal
- Department of Genetics, Barkatullah University, Bhopal, MP, India
| | - Rekha Khandia
- Department of Genetics, Barkatullah University, Bhopal, MP, India
| |
Collapse
|
22
|
Bhattacharya S, Kawamura A. Using evasins to target the chemokine network in inflammation. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 119:1-38. [PMID: 31997766 DOI: 10.1016/bs.apcsb.2019.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Inflammation, is driven by a network comprising cytokines, chemokines, their target receptors and leukocytes, and is a major pathologic mechanism that adversely affects organ function in diverse human diseases. Despite being supported by substantial target validation, no successful anti-chemokine therapeutic to treat inflammatory disease has yet been developed. This is in part because of the robustness of the chemokine network, which emerges from a large total chemokine load in disease, promiscuous expression of receptors on leukocytes, promiscuous and synergistic interactions between chemokines and receptors, and feedforward loops created by secretion of chemokines by leukocytes themselves. Many parasites, including viruses, helminths and ticks, evade the chemokine network by producing proteins that bind promiscuously to chemokines or their receptors. Evasins - three small glycoproteins identified in the saliva of the brown dog tick - bind multiple chemokines, and are active in several animal models of inflammatory disease. Over 50 evasin homologs have recently been identified from diverse tick species. Characterization of the chemokine binding patterns of evasins show that several have anti-chemokine activities that extend substantially beyond those previously described. These studies indicate that evasins function at the site of the tick bite by reducing total chemokine load. This not only reduces chemokine signaling to receptors, but also interrupts feedforward loops, thus disabling the chemokine network. Taking the lead from nature, a goal for the development of new anti-chemokine therapeutics would be to reduce the total chemokine load in disease. This could be achieved by administering appropriate evasin combinations or by smaller peptides that mimic evasin action.
Collapse
Affiliation(s)
- Shoumo Bhattacharya
- RDM Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Akane Kawamura
- RDM Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
23
|
Ulleryd MA, Mjörnstedt F, Panagaki D, Yang LJ, Engevall K, Gutiérrez S, Wang Y, Gan LM, Nilsson H, Michaëlsson E, Johansson ME. Stimulation of alpha 7 nicotinic acetylcholine receptor (α7nAChR) inhibits atherosclerosis via immunomodulatory effects on myeloid cells. Atherosclerosis 2019; 287:122-133. [PMID: 31260875 DOI: 10.1016/j.atherosclerosis.2019.06.903] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/16/2019] [Accepted: 06/13/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIMS Alpha 7 nicotinic acetylcholine receptor (α7nAChR) stimulation can regulate acute inflammation, and lack of α7nAChR accelerates atherosclerosis in mice. In this study, we aimed to investigate the effects of the novel α7nAChR agonist, AZ6983, on atherosclerosis and assess its possible immunomodulating effects. METHODS AZ6983 was tested in vitro in LPS-challenged mouse and human blood and in vivo using the acute inflammatory air pouch model. Thereafter, long-term effects of AZ6983 treatment on atherosclerosis and immune responses were assessed in apoE-/- mice after 8 and 12 weeks. Atherosclerosis was investigated in the aortic root and thoracic aorta, serum levels of cytokines were analysed and RNAseq was used to study aortic gene expression. Further, bone-marrow-derived macrophages were used to assess phagocytosis in vitro. RESULTS α7nAChR activation by AZ6983 decreased pro-inflammatory cytokines in acute stimulations of human and mouse blood in vitro, as well as in vivo using the air pouch model. Treating apoE-/- mice with AZ6983 decreased atherosclerosis by 37-49% and decreased serum cytokine levels. RNAseq analysis of aortae suggested the involvement of several specific myeloid cell functions, including phagocytosis. In line with this, AZ6983 significantly increased phagocytosis in bone marrow-derived macrophages. CONCLUSIONS This study demonstrates that activation of α7nAChR with AZ6983 inhibits atherosclerosis in apoE-/-mice and that immunomodulating effects on myeloid cells, such as enhanced phagocytosis and suppression of inflammatory cytokines, could be part of the athero-protective mechanisms. The observed anti-inflammatory effect in human blood supports the idea that AZ6983 may decrease disease also in humans.
Collapse
Affiliation(s)
- Marcus A Ulleryd
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Filip Mjörnstedt
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dimitra Panagaki
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Li Jin Yang
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kajsa Engevall
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Saray Gutiérrez
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Li-Ming Gan
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Early Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Holger Nilsson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erik Michaëlsson
- Early Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Maria E Johansson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
24
|
Almén MS, Björk J, Nyman U, Lindström V, Jonsson M, Abrahamson M, Vestergren AS, Lindhe Ö, Franklin G, Christensson A, Grubb A. Shrunken Pore Syndrome Is Associated With Increased Levels of Atherosclerosis-Promoting Proteins. Kidney Int Rep 2019; 4:67-79. [PMID: 30596170 PMCID: PMC6308389 DOI: 10.1016/j.ekir.2018.09.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Shrunken pore syndrome (SPS), originally defined by cystatin C-based estimated glomerular filtration rate (eGFRcystatin C) being less than 60% of creatinine-based estimated glomerular filtration rate (eGFRcreatinine) in the absence of extrarenal influences on the plasma levels of cystatin C or creatinine, is associated with a high increase in mortality, even in the absence of reduced glomerular filtration rate (GFR). The objective of the present study was to determine whether the proteome of patients with SPS shows differences from that of patients with normal or reduced measured GFR (mGFR) without SPS. METHODS Four patient cohorts were included: 1 cohort with normal mGFR without SPS, 1 with normal mGFR with SPS, 1 with reduced mGFR without SPS, and 1 with reduced mGFR with SPS. The plasma levels of 177 selected proteins were analyzed. RESULTS Differences in the levels of 30 proteins were specific for SPS; 31 differences were specific for patients with both SPS and reduced mGFR; and 27 were specific for reduced mGFR. Eighteen of the differences specific for SPS concerned proteins described as promoting, or being associated with, atherosclerosis. Twelve of the differences specific for patients with both SPS and reduced mGFR and 10 of the differences specific for reduced mGFR also concerned proteins described as promoting, or being associated with, atherosclerosis. Almost all (82 of 88) of the concentration differences represented increased levels. For SPS, but not for reduced mGFR, a correlation between protein size and increase in level was observed, with smaller proteins being associated with higher levels. CONCLUSION The high mortality in shrunken pore syndrome might be caused by the accumulation of atherosclerosis-promoting proteins in this condition.
Collapse
Affiliation(s)
| | - Jonas Björk
- Department of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Ulf Nyman
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Veronica Lindström
- Department of Clinical Chemistry, Skåne University Hospital, Lund, Lund University, Sweden
| | - Magnus Jonsson
- Department of Clinical Chemistry, Skåne University Hospital, Malmö, Sweden
| | | | | | | | | | - Anders Christensson
- Department of Nephrology, Skåne University Hospital, Malmö, Lund University, Sweden
| | - Anders Grubb
- Department of Clinical Chemistry, Skåne University Hospital, Lund, Lund University, Sweden
| |
Collapse
|
25
|
Extracellular volume by cardiac magnetic resonance is associated with biomarkers of inflammation in hypertensive heart disease. J Hypertens 2019; 37:65-72. [DOI: 10.1097/hjh.0000000000001875] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
The Potential of Fluocinolone Acetonide to Mitigate Inflammation and Lipid Accumulation in 2D and 3D Foam Cell Cultures. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3739251. [PMID: 30596089 PMCID: PMC6282138 DOI: 10.1155/2018/3739251] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/29/2018] [Accepted: 11/07/2018] [Indexed: 12/27/2022]
Abstract
Inflammation plays an important role in all stages of atherosclerosis development. Therefore, the use of anti-inflammatory drugs could reduce the risk of major adverse cardiovascular events due to atherosclerosis. Herein, we explored the capacity of fluocinolone acetonide (FA), a glucocorticoid (GC), in modulating foam cell formation and response. Human THP-1 derived foam cells were produced using 100 μg/mL oxidized low-density lipoproteins (OxLDL) and fetal bovine serum (1 and 10%). 2D cultures of these cells were treated with FA (0.1, 1, 10, and 50 μg/mL) in comparison with dexamethasone (Dex). Results showed that treatment with 0.1 and 1 μg/mL FA and Dex improved foam cell survival. FA and Dex also inhibited inflammatory cytokine (CD14, M-CSF, MIP-3α, and TNF-α) secretion. Notably, at the concentration of 1 μg/mL, both FA and Dex reduced cholesteryl ester accumulation. Compared to Dex, FA was significantly better in reducing lipid accumulation at the therapeutic concentrations of 1 and 10 μg/mL. In a novel 3D foam cell spheroid model, FA was shown to be more effective than Dex in diminishing lipid accumulation, at the concentration of 0.1 μg/mL. Taken together, FA was demonstrated to be effective in preventing both lipid accumulation and inflammation in foam cells.
Collapse
|
27
|
Varona S, Orriols M, Galán M, Guadall A, Cañes L, Aguiló S, Sirvent M, Martínez-González J, Rodríguez C. Lysyl oxidase (LOX) limits VSMC proliferation and neointimal thickening through its extracellular enzymatic activity. Sci Rep 2018; 8:13258. [PMID: 30185869 PMCID: PMC6125287 DOI: 10.1038/s41598-018-31312-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/14/2018] [Indexed: 02/06/2023] Open
Abstract
Lysyl oxidase (LOX) plays a critical role in extracellular matrix maturation and limits VSMC proliferation and vascular remodeling. We have investigated whether this anti-proliferative effect relies on the extracellular catalytically active LOX or on its biologically active propeptide (LOX-PP). High expression levels of both LOX and LOX-PP were detected in the vascular wall from transgenic mice over-expressing the full-length human LOX cDNA under the control of SM22α promoter (TgLOX), which targets the transgene to VSMC without affecting the expression of mouse LOX isoenzymes. TgLOX VSMC also secrete high amounts of both mature LOX and LOX-PP. Wild-type (WT) mouse VSMC exposed to VSMC supernatants from transgenic animals showed reduced proliferative rates (low [3H]-thymidine uptake and expression of PCNA) than those incubated with conditioned media from WT cells, effect that was abrogated by β-aminopropionitrile (BAPN), an inhibitor of LOX activity. Lentiviral over-expression of LOX, but not LOX-PP, decreased human VSMC proliferation, effect that was also prevented by BAPN. LOX transgenesis neither impacted local nor systemic inflammatory response induced by carotid artery ligation. Interestingly, in this model, BAPN normalized the reduced neointimal thickening observed in TgLOX mice. Therefore, extracellular enzymatically active LOX is required to limit both VSMC proliferation and vascular remodeling.
Collapse
Affiliation(s)
- Saray Varona
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain.,Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Mar Orriols
- CIBER de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain.,Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - María Galán
- CIBER de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain.,Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain.,Institut de Recerca del Hospital de la Santa Creu i Sant Pau-Programa ICCC, Barcelona, Spain
| | - Anna Guadall
- Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain.,Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Silvia Aguiló
- Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain.,Institut de Recerca del Hospital de la Santa Creu i Sant Pau-Programa ICCC, Barcelona, Spain
| | - Marc Sirvent
- Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain. .,CIBER de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain. .,Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain.
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), ISCIII, Madrid, Spain. .,Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain. .,Institut de Recerca del Hospital de la Santa Creu i Sant Pau-Programa ICCC, Barcelona, Spain.
| |
Collapse
|
28
|
Martí-Pàmies I, Cañes L, Alonso J, Rodríguez C, Martínez-González J. The nuclear receptor NOR-1/NR4A3 regulates the multifunctional glycoprotein vitronectin in human vascular smooth muscle cells. FASEB J 2017; 31:4588-4599. [PMID: 28666984 DOI: 10.1096/fj.201700136rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/19/2017] [Indexed: 01/04/2023]
Abstract
The nuclear receptor NOR-1 (NR4A3) has recently been involved in the regulation of extracellular matrix (ECM) proteins associated with neointimal thickening and the vascular control of hemostasis. We sought to find as-yet unidentified NOR-1 target genes in human vascular smooth muscle cells (VSMCs). An in silico analysis identified putative NOR-1 response elements in the proximal promoter region of several genes encoding for ECM proteins, including vitronectin (VTN). Lentiviral overexpression of NOR-1 strongly increased VTN mRNA and protein levels, whereas NOR-1 silencing significantly reduced VTN expression. Deletion and site-directed mutagenesis studies, as well as EMSA and chromatin immunoprecipitation, identified the NBRE(-202/-195) site in the VTN promoter as an essential element for NOR-1 responsiveness. Furthermore, NOR-1 and VTN colocalized in VSMCs in human atherosclerotic lesions. VTN levels were increased in cell supernatants from VSMCs that overexpress NOR-1. Cell supernatants from VSMCs overexpressing NOR-1 induced cell migration to a greater extent than supernatants from control cells, and this effect was attenuated when cell supernatants were preincubated with anti-VTN blocking antibodies or VTN was silenced in supernatant-generating cells. These results indicate that VTN is a target of NOR-1 and suggest that this multifunctional glycoprotein may participate in vascular responses mediated by this nuclear receptor.-Martí-Pàmies, I., Cañes, L., Alonso, J., Rodríguez, C., Martínez-González, J. The nuclear receptor NOR-1/NR4A3 regulates the multifunctional glycoprotein vitronectin in human vascular smooth muscle cells.
Collapse
Affiliation(s)
- Ingrid Martí-Pàmies
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Sant Pau Biomedical Research Institute, Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Sant Pau Biomedical Research Institute, Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Sant Pau Biomedical Research Institute, Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain
| | - Cristina Rodríguez
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain.,Institut Català de Ciències Cardiovasculars (ICCC), Sant Pau Biomedical Research Institute, Barcelona, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Sant Pau Biomedical Research Institute, Barcelona, Spain; .,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain
| |
Collapse
|
29
|
Plotkin JD, Elias MG, Dellinger AL, Kepley CL. NF-κB inhibitors that prevent foam cell formation and atherosclerotic plaque accumulation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2037-2048. [PMID: 28457935 DOI: 10.1016/j.nano.2017.04.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 03/30/2017] [Accepted: 04/16/2017] [Indexed: 11/30/2022]
Abstract
The transformation of monocyte-derived macrophages into lipid-laden foam cells is one inflammatory process underlying atherosclerotic disease. Previous studies have demonstrated that fullerene derivatives (FDs) have inflammation-blunting properties. Thus, it was hypothesized that FD could inhibit the transformation process underlying foam cell formation. Fullerene derivatives inhibited the phorbol myristic acid/oxidized low-density lipoprotein-induced differentiation of macrophages into foam cells as determined by lipid staining and morphology.Lipoprotein-induced generation of TNF-α, C5a-induced MC activation, ICAM-1 driven adhesion, and CD36 expression were significantly inhibited in FD treated cells compared to non-treated cells. Inhibition appeared to be mediated through the NF-κB pathway as FD reduced expression of NF-κB and atherosclerosis-associated genes. Compared to controls, FD dramatically inhibited plaque formation in arteries of apolipoprotein E null mice. Thus, FD may be an unrecognized therapy to prevent atherosclerotic lesions via inhibition of foam cell formation and MC stabilization.
Collapse
Affiliation(s)
- Jesse D Plotkin
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, United States
| | - Michael G Elias
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, United States
| | - Anthony L Dellinger
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, United States
| | - Christopher L Kepley
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, United States.
| |
Collapse
|
30
|
McEvoy C, de Gaetano M, Giffney HE, Bahar B, Cummins EP, Brennan EP, Barry M, Belton O, Godson CG, Murphy EP, Crean D. NR4A Receptors Differentially Regulate NF-κB Signaling in Myeloid Cells. Front Immunol 2017; 8:7. [PMID: 28167941 PMCID: PMC5256039 DOI: 10.3389/fimmu.2017.00007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/03/2017] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of inflammatory responses is a hallmark of multiple diseases such as atherosclerosis and rheumatoid arthritis. As constitutively active transcription factors, NR4A nuclear receptors function to control the magnitude of inflammatory responses and in chronic inflammatory disease can be protective or pathogenic. Within this study, we demonstrate that TLR4 stimulation using the endotoxin lipopolysaccharide (LPS) rapidly enhances NR4A1–3 expression in human and murine, primary and immortalized myeloid cells with concomitant gene transcription and protein secretion of MIP-3α, a central chemokine implicated in numerous pathologies. Deficiency of NR4A2 and NR4A3 in human and murine myeloid cells reveals that both receptors function as positive regulators of enhanced MIP-3α expression. In contrast, within the same cell types and conditions, altered NR4A activity leads to suppression of LPS-induced MCP-1 gene and protein expression. An equivalent pattern of inflammatory gene regulation is replicated in TNFα-treated myeloid cells. We show that NF-κB is the critical regulator of NR4A1–3, MIP-3α, and MCP-1 during TLR4 stimulation in myeloid cells and highlight a parallel mechanism whereby NR4A activity can repress or enhance NF-κB target gene expression simultaneously. Mechanistic insight reveals that NR4A2 does not require DNA-binding capacity in order to enhance or repress NF-κB target gene expression simultaneously and establishes a role for NF-κB family member Relb as a novel NR4A target gene involved in the positive regulation of MIP-3α. Thus, our data reveal a dynamic role for NR4A receptors concurrently enhancing and repressing NF-κB activity in myeloid cells leading to altered transcription of key inflammatory mediators.
Collapse
Affiliation(s)
- Caitriona McEvoy
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Monica de Gaetano
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Hugh E Giffney
- Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Bojlul Bahar
- International Institute of Nutritional Sciences and Applied Food Safety Studies, University of Central Lancashire , Preston , UK
| | - Eoin P Cummins
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Eoin P Brennan
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Mary Barry
- St. Vincent's University Hospital , Dublin , Ireland
| | - Orina Belton
- Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin , Dublin , Ireland
| | - Catherine G Godson
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Evelyn P Murphy
- Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Daniel Crean
- Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
Qun L, Wenda X, Weihong S, Jianyang M, Wei C, Fangzhou L, Zhenyao X, Pingjin G. miRNA-27b modulates endothelial cell angiogenesis by directly targeting Naa15 in atherogenesis. Atherosclerosis 2016; 254:184-192. [PMID: 27755984 DOI: 10.1016/j.atherosclerosis.2016.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/06/2016] [Accepted: 10/04/2016] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND AIMS The CCL20/CCR6 axis has been shown to play a vital role in the pathogenesis of atherosclerosis (AS). However, the regulatory mechanism remains unclear. Here, we studied the miRNA-mediated epigenetic regulation of the CCL20/CCR6 axis in atherogenesis. METHODS CCR6+/+ApoE-/- and CCR6-/-ApoE-/- mice were fed a high-fat diet for 24 weeks. Plaque size was evaluated via ultrasound biomicroscope and hematoxylin and eosin. Protein expression were measured by Western blotting or immunofluorescence/immunohistochemistry or ELISA, and gene mRNA levels were detected by RT-PCR. Seven hundred and sixty miRNAs were screened via miRNA profiling. miRNA-27b target genes were predicted using software and verified with a dual luciferase reporter assay. The tube formation of mouse aortic endothelial cells (MAECs) was performed on Matrigel. RESULTS In contrast to wild-type ApoE-/- mice, CCR6 deficiency led to a significantly decreased plaque size, CD31, CCR6, CCL20 expression and number of CCL20+ macrophages in atherosclerotic plaques. Stimulation of mouse primary peritoneal macrophages (MPPMs) resulted in increased IL-23 release. miRNA-27b was the most highly expressed (5.19-fold increase) miRNA among the 760 miRNAs screened in the vessel. Naa15 was verified as miRNA-27b target gene, which was diminished in the plaques. Transfection of siRNA Naa15 or miRNA-27b mimic into MAECs caused an increase tube formation. CONCLUSIONS CCR6 deletion effectively ameliorates atherosclerosis progression by reducing macrophage accumulation, resulting in reduced secretion of CCL20 and IL-23. Mechanistically, the decreased miRNA-27b regulates the activity of the CCL20/CCR6 axis by targeting Naa15, and promotes plaque stability in atherosclerosis.
Collapse
Affiliation(s)
- Li Qun
- The State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Key Laboratory of Stem Cell Biology and Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xi Wenda
- The State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Sun Weihong
- Key Laboratory of Stem Cell Biology and Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ma Jianyang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 20001, China
| | - Cai Wei
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lou Fangzhou
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xu Zhenyao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Gao Pingjin
- The State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Key Laboratory of Stem Cell Biology and Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
32
|
Orriols M, Varona S, Aguiló S, Galán M, Martínez González J, Rodríguez C. [Inflammation inhibits vascular fibulin-5 expression: Involvement of transcription factor SOX9]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2016; 28:271-280. [PMID: 27692634 DOI: 10.1016/j.arteri.2016.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 05/27/2016] [Accepted: 06/03/2016] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Fibulin-5 (FBLN5) is an elastogenic protein critically involved in extracellular matrix (ECM) remodelling, a key process in abdominal aortic aneurysm (AAA). However, the possible contribution of FBLN5 to AAA development has not been addressed. METHODS Expression levels were determined by real-time PCR and Western blot in human abdominal aorta from patients with AAA or healthy donors, as well as in human aortic vascular smooth muscle cells (VSMC). Lentiviral transduction, transient transfections, and chromatin immunoprecipitation (ChIP) assays were also performed. RESULTS The expression of FBLN5 in human AAA was significantly lower than in healthy donors. FBLN5 mRNA and protein levels and their secretion to the extracellular environment were down-regulated in VSMC exposed to inflammatory stimuli. Interestingly, FBLN5 transcriptional activity was inhibited by TNFα and lipopolysaccharide (LPS), and depends on a SOX response element. In fact, SOX9 expression was reduced in VMSC induced by inflammatory mediators and in human AAA, and correlated with that of FBLN5. Furthermore, SOX9 over-expression limited the reduction of FBLN5 expression induced by cytokines in VSMC. Finally, it was observed that SOX9 interacts with FBLN5 promoter, and that this binding was reduced upon TNFα exposure. CONCLUSIONS FBLN5 downregulation in human AAA could contribute to extracellular matrix remodelling induced by the inflammatory component of the disease.
Collapse
Affiliation(s)
- Mar Orriols
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, España
| | - Saray Varona
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, España
| | - Silvia Aguiló
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, España
| | - María Galán
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, España
| | | | - Cristina Rodríguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, España.
| |
Collapse
|
33
|
Orriols M, Varona S, Martí-Pàmies I, Galán M, Guadall A, Escudero JR, Martín-Ventura JL, Camacho M, Vila L, Martínez-González J, Rodríguez C. Down-regulation of Fibulin-5 is associated with aortic dilation: role of inflammation and epigenetics. Cardiovasc Res 2016; 110:431-42. [DOI: 10.1093/cvr/cvw082] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 04/14/2016] [Indexed: 01/04/2023] Open
|
34
|
Pozzobon T, Facchinello N, Bossi F, Capitani N, Benagiano M, Di Benedetto G, Zennaro C, West N, Codolo G, Bernardini M, Baldari CT, D'Elios MM, Pellegrini L, Argenton F, de Bernard M. Treponema pallidum (syphilis) antigen TpF1 induces angiogenesis through the activation of the IL-8 pathway. Sci Rep 2016; 6:18785. [PMID: 26728351 PMCID: PMC4700457 DOI: 10.1038/srep18785] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/26/2015] [Indexed: 12/25/2022] Open
Abstract
Over 10 million people every year become infected by Treponema pallidum and develop syphilis, a disease with broad symptomatology that, due to the difficulty to eradicate the pathogen from the highly vascularized secondary sites of infection, is still treated with injections of penicillin. Unlike most other bacterial pathogens, T. pallidum infection produces indeed a strong angiogenic response whose mechanism of activation, however, remains unknown. Here, we report that one of the major antigen of T. pallidum, the TpF1 protein, has growth factor-like activity on primary cultures of human endothelial cells and activates specific T cells able to promote tissue factor production. The growth factor-like activity is mediated by the secretion of IL-8 but not of VEGF, two known angiogenic factors. The pathogen’s factor signals IL-8 secretion through the activation of the CREB/NF-κB signalling pathway. These findings are recapitulated in an animal model, zebrafish, where we observed that TpF1 injection stimulates angiogenesis and IL-8, but not VEGF, secretion. This study suggests that the angiogenic response observed during secondary syphilis is triggered by TpF1 and that pharmacological therapies directed to inhibit IL-8 response in patients should be explored to treat this disease.
Collapse
Affiliation(s)
| | | | - Fleur Bossi
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy.,Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giulietta Di Benedetto
- Institute of Neuroscience, Italian National Research Council (CNR), Padua, Italy.,Venetian Institute of Molecular Medicine, Padua, Italy
| | - Cristina Zennaro
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Nicole West
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Gaia Codolo
- Department of Biology, University of Padua, Padua, Italy
| | - Marialina Bernardini
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | | | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luca Pellegrini
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec, Canada
| | | | | |
Collapse
|
35
|
Circulating CD105 shows significant impact in patients of oral cancer and promotes malignancy of cancer cells via CCL20. Tumour Biol 2015; 37:1995-2005. [PMID: 26334621 DOI: 10.1007/s13277-015-3991-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/25/2015] [Indexed: 10/23/2022] Open
Abstract
CD105 is rich in endothelium cells and is involved in angiogenesis. Higher microvascular density of tumor is also related to the prognosis in a variety of cancers. In this present study, patients with positive N classification, advanced T classification, advanced TNM stage, extracapsular spread of lymph nodes (ECS), and perineural invasion had significantly higher levels of peripheral vein (pCD105) and venous return from tumor (tCD105) in 71 patients with OSCC compared to 13 healthy volunteers. Those with higher pCD105 or tCD105 levels had significantly poorer 5-year disease-specific survival rate (DDS) and overall survival rate (OS). The tCD105 and pCD105 levels and ECS were the independent prognostic factors by the multivariate analysis according to the Cox regression model in 5-year DDS and OS rate. SAS and SCC4 cells treated with CD105 showed the increase in migration, invasion, and proliferation in vitro and in vivo. Furthermore, CCL20 expression participated in CD105-elicited cell motility in oral cancer cells. In conclusion, higher level of circulating CD105 is related to adverse pathological features among patients with OSCC. It is also a useful marker for evaluating the prognosis and targeting therapeutics of OSCC.
Collapse
|
36
|
Rodríguez-Calvo R, Ferrán B, Alonso J, Martí-Pàmies I, Aguiló S, Calvayrac O, Rodríguez C, Martínez-González J. NR4A receptors up-regulate the antiproteinase alpha-2 macroglobulin (A2M) and modulate MMP-2 and MMP-9 in vascular smooth muscle cells. Thromb Haemost 2015; 113:1323-34. [PMID: 25809189 DOI: 10.1160/th14-07-0645] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/20/2015] [Indexed: 11/05/2022]
Abstract
Matrix metalloproteinases (MMPs) are associated with tissue remodelling and repair. In non-vascular tissues, NR4A receptors have been involved in the regulation of MMPs by transcriptional repression mechanisms. Here, we analyse alternative mechanisms involving NR4A receptors in the modulation of MMP activity in vascular smooth muscle cells (VSMC). Lentiviral overexpression of NR4A receptors (NOR-1, Nurr1 and Nur77) in human VSMC strongly decreased MMP-2 and MMP-9 activities (analysed by zymography and DQ-gelatin assays) and protein levels. NR4A receptors also down-regulated MMP-2 mRNA levels. Real-time PCR analysis evidenced that alpha-2-macroglobulin (A2M), but not other MMP inhibitors (TIMP-1 and TIMP-2) were up-regulated in NR4A-transduced cells. Interestingly, A2M was expressed in human vascular tissues including the smooth muscle media layer. While NR4A receptors increased A2M expression and secretion in VSMC, NR4A knockdown significantly reduced basal A2M expression in these cells. The direct transcriptional regulation of the human A2M promoter by NR4A receptors was characterised in luciferase reporter assays, electrophoretic mobility shift assays and by chromatin immunoprecipitation, identifying a NGFI-B response element (NBRE-71/-64) essential for the NR4A-mediated induction. The blockade of A2M partially prevented the reduction of MMPs activity observed in NR4A-transduced cells. Although mouse A2M promoter was unresponsive to NR4A receptors, vascular MMP expression was attenuated in transgenic mice over-expressing human NOR-1 in VSMC challenged with lipopolysaccharide. Our results show that the pan-proteinase inhibitor A2M is expressed in the vasculature and that NR4A receptors modulate VSMC MMP activity by several mechanisms including the up-regulation of A2M.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - José Martínez-González
- José Martínez-González, Centro de Investigación Cardiovascular (CSIC-ICCC), Hospital de la Santa Creu i Sant Pau, Avda. Sant Antoni Maria Claret 167, 08025 Barcelona, Spain, Tel.: +34 935565896, Fax: +34 935565559, E-mail:
| |
Collapse
|
37
|
Atherosclerosis regulation via media lipid-driven VSMC cholesterol efflux switch. Med Hypotheses 2015; 84:141-4. [DOI: 10.1016/j.mehy.2014.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 11/27/2014] [Accepted: 12/04/2014] [Indexed: 02/02/2023]
|
38
|
Calvayrac O, Rodríguez-Calvo R, Martí-Pamies I, Alonso J, Ferrán B, Aguiló S, Crespo J, Rodríguez-Sinovas A, Rodríguez C, Martínez-González J. NOR-1 modulates the inflammatory response of vascular smooth muscle cells by preventing NFκB activation. J Mol Cell Cardiol 2014; 80:34-44. [PMID: 25536180 DOI: 10.1016/j.yjmcc.2014.12.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 11/19/2014] [Accepted: 12/07/2014] [Indexed: 11/25/2022]
Abstract
Recent work has highlighted the role of NR4A receptors in atherosclerosis and inflammation. In vascular smooth muscle cell (VSMC) proliferation, however, NOR-1 (neuron-derived orphan receptor-1) exerts antagonistic effects to Nur77 and Nurr1. The aim of this study was to analyse the effect of NOR-1 in VSMC inflammatory response. We assessed the consequence of a gain-of-function of this receptor on the response of VSMC to inflammatory stimuli. In human VSMC, lentiviral over-expression of NOR-1 reduced lipopolysaccharide (LPS)-induced up-regulation of cytokines (IL-1β, IL-6 and IL-8) and chemokines (MCP-1 and CCL20). Similar effects were obtained in cells stimulated with TNFα or oxLDL. Conversely, siRNA-mediated NOR-1 inhibition significantly increased the expression of pro-inflammatory mediators. Interestingly, in the aortas from transgenic mice that over-express human NOR-1 in VSMC (TgNOR-1), the up-regulation of cytokine/chemokine by LPS was lower compared to wild-type littermates. Similar results were obtained in VSMC from transgenic animals. NOR-1 reduced the transcriptional activity of NFκB sensitive promoters (in transient transfections), and the binding of NFκB to its responsive element (in electrophoretic mobility shift assays). Furthermore, NOR-1 prevented the activation of NFκB pathway by decreasing IκBα phosphorylation/degradation and inhibiting the phosphorylation and subsequent translocation of p65 to the nucleus (assessed by Western blot and immunocytochemistry). These effects were associated with an attenuated phosphorylation of ERK1/2, p38 MAPK and Jun N-terminal kinase, pathways involved in the activation of NFκB. In mouse challenged with LPS, the activation of the NFκB signalling was also attenuated in the aorta from TgNOR-1. Our data support a role for NOR-1 as a negative modulator of the acute response elicited by pro-inflammatory stimuli in the vasculature.
Collapse
Affiliation(s)
- Olivier Calvayrac
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | | | - Ingrid Martí-Pamies
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Judith Alonso
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Beatriz Ferrán
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Silvia Aguiló
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Javier Crespo
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | | | - Cristina Rodríguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain.
| | | |
Collapse
|
39
|
Sligh J, Janda J, Jandova J. Mutations in BALB mitochondrial DNA induce CCL20 up-regulation promoting tumorigenic phenotypes. Mutat Res 2014; 769:49-58. [PMID: 25177208 PMCID: PMC4144272 DOI: 10.1016/j.mrfmmm.2014.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
mtDNA mutations are common in human cancers and are thought to contribute to the process of neoplasia. We examined the role of mtDNA mutations in skin cancer by generating fibroblast cybrids harboring a mutation in the gene encoding the mitochondrial tRNA for arginine. This somatic mutation (9821insA) was previously reported in UV-induced hyperkeratotic skin tumors in hairless mice and confers specific tumorigenic phenotypes to mutant cybrids. Microarray analysis revealed and RT-PCR along with Western blot analysis confirmed the up-regulation of CCL20 and its receptor CCR6 in mtBALB haplotype containing the mt-Tr 9821insA allele compared to wild type mtB6 haplotype. Based on reported role of CCL20 in cancer progression we examined whether the hyper-proliferation and enhanced motility of mtBALB haplotype would be associated with CCL20 levels. Treatment of both genotypes with recombinant CCL20 (rmCCL20) resulted in enhanced growth and motility of mtB6 cybrids. Furthermore, the acquired somatic alteration increased the in vivo tumor growth of mtBALB cybrids through the up-regulation of CCL20 since neutralizing antibody significantly decreased in vivo tumor growth of these cells; and tumors from anti-CCL20 treated mice injected with mtBALB cybrids showed significantly decreased CCL20 levels. When rmCCL20 or mtBALB cybrids were used as chemotactic stimuli, mtB6 cybrids showed increased motility while anti-CCL20 antibody decreased the migration and in vivo tumor growth of mtBALB cybrids. Moreover, the inhibitors of MAPK signaling and NF-κB activation inhibited CCL20 expression in mtBALB cybrids and decreased their migratory capabilities. Thus, acquired mtDNA mutations may promote tumorigenic phenotypes through up-regulation of chemokine CCL20.
Collapse
Affiliation(s)
- James Sligh
- Department of Medicine – Dermatology Division, University of Arizona, Tucson, AZ, 857 24, USA
- University of Arizona Cancer Center, Tucson, AZ, 85724, USA
| | - Jaroslav Janda
- University of Arizona Cancer Center, Tucson, AZ, 85724, USA
| | - Jana Jandova
- Department of Medicine – Dermatology Division, University of Arizona, Tucson, AZ, 857 24, USA
- University of Arizona Cancer Center, Tucson, AZ, 85724, USA
| |
Collapse
|
40
|
Zhao L, Xia J, Wang X, Xu F. Transcriptional regulation of CCL20 expression. Microbes Infect 2014; 16:864-70. [DOI: 10.1016/j.micinf.2014.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/03/2014] [Accepted: 08/07/2014] [Indexed: 12/19/2022]
|
41
|
Pateras I, Giaginis C, Tsigris C, Patsouris E, Theocharis S. NF-κB signaling at the crossroads of inflammation and atherogenesis: searching for new therapeutic links. Expert Opin Ther Targets 2014; 18:1089-101. [DOI: 10.1517/14728222.2014.938051] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
42
|
Orriols M, Guadall A, Galán M, Martí-Pàmies I, Varona S, Rodríguez-Calvo R, Briones AM, Navarro MA, de Diego A, Osada J, Martínez-González J, Rodríguez C. Lysyl oxidase (LOX) in vascular remodelling. Insight from a new animal model. Thromb Haemost 2014; 112:812-24. [PMID: 24990180 DOI: 10.1160/th14-01-0024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 05/06/2014] [Indexed: 12/18/2022]
Abstract
Lysyl oxidase (LOX) is an extracellular matrix-modifying enzyme that seems to play a critical role in vascular remodelling. However, the lack of viable LOX-deficient animal models has been an obstacle to deep in LOX biology. In this study we have developed a transgenic mouse model that over-expresses LOX in vascular smooth muscle cells (VSMC) to clarify whether LOX could regulate VSMC phenotype and vascular remodelling. The SM22α proximal promoter drove the expression of a transgene containing the human LOX cDNA. Two stable transgenic lines, phenotypically indistinguishable, were generated by conventional methods (TgLOX). Transgene expression followed the expected SMC-specific pattern. In TgLOX mice, real-time PCR and immunohistochemistry evidenced a strong expression of LOX in the media from aorta and carotid arteries, coincident with a higher proportion of mature collagen. VSMC isolated from TgLOX mice expressed high levels of LOX pro-enzyme, which was properly secreted and processed into mature and bioactive LOX. Interestingly, cell proliferation was significantly reduced in cells from TgLOX mice. Transgenic VSMC also exhibited low levels of Myh10 (marker of SMC phenotypic switching), PCNA (marker of cell proliferation) and MCP-1, and a weak activation of Akt and ERK1/2 in response to mitogenic stimuli. Accordingly, neointimal thickening induced by carotid artery ligation was attenuated in TgLOX mice that also displayed a reduction in PCNA and MCP-1 immunostaining. Our results give evidence that LOX plays a critical role in vascular remodelling. We have developed a new animal model to study the role of LOX in vascular biology.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Cristina Rodríguez
- José Martínez-González or Cristina Rodríguez, Centro de Investigación Cardiovascular (CSIC-ICCC), Hospital de la Santa Creu i Sant Pau (pabellón Nº 11), Avda. Sant Antoni Maria Claret 167, 08025 Barcelona, Spain, Tel.: +34 93 5565897, Fax: +34 93 5565559, E-mail: ;
| |
Collapse
|
43
|
Roles of the chemokine system in development of obesity, insulin resistance, and cardiovascular disease. J Immunol Res 2014; 2014:181450. [PMID: 24741577 PMCID: PMC3987870 DOI: 10.1155/2014/181450] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 02/04/2014] [Indexed: 12/13/2022] Open
Abstract
The escalating epidemic of obesity has increased the incidence of obesity-induced complications to historically high levels. Adipose tissue is a dynamic energy depot, which stores energy and mobilizes it during nutrient deficiency. Excess nutrient intake resulting in adipose tissue expansion triggers lipid release and aberrant adipokine, cytokine and chemokine production, and signaling that ultimately lead to adipose tissue inflammation, a hallmark of obesity. This low-grade chronic inflammation is thought to link obesity to insulin resistance and the associated comorbidities of metabolic syndrome such as dyslipidemia and hypertension, which increase risk of type 2 diabetes and cardiovascular disease. In this review, we focus on and discuss members of the chemokine system for which there is clear evidence of participation in the development of obesity and obesity-induced pathologies.
Collapse
|
44
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
45
|
Manthey HD, Cochain C, Barnsteiner S, Karshovska E, Pelisek J, Koch M, Chaudhari SM, Busch M, Eckstein HH, Weber C, Koenen RR, Zernecke A. CCR6 selectively promotes monocyte mediated inflammation and atherogenesis in mice. Thromb Haemost 2013; 110:1267-77. [PMID: 24114205 DOI: 10.1160/th13-01-0017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 08/19/2013] [Indexed: 12/22/2022]
Abstract
The chemokine receptor CCR6 is expressed by various cell subsets implicated in atherogenesis, such as monocytes, Th17 and regulatory T cells. In order to further define the role of CCR6 in atherosclerosis, CCR6-deficient (Ccr6-/-) mice were crossed with low-density lipoprotein receptor-deficient (Ldlr-/-) mice to generate atherosclerosis-prone mice deficient in CCR6. Compared to Ldlr-/- controls, atherosclerotic burden in the aortic sinus and aorta were reduced in Ccr6-/-Ldlr-/- mice fed a high fat diet, associated with a profound depression in lesional macrophage accumulation. Local and systemic distributions of T cells, including frequencies of Th1, Th17 and regulatory T cells were unaltered. In contrast, circulating counts of both Gr-1(high) and Gr1(low) monocytes were reduced in Ccr6-/-Ldlr-/- mice. Moreover, CCR6 was revealed to promote monocyte adhesion to inflamed endothelium in vitro and leukocyte adhesion to carotid arteries in vivo. Finally, CCR6 selectively recruited monocytes but not T cells in an acute inflammatory air pouch model. We here show that CCR6 functions on multiple levels and regulates the mobilisation, adhesion and recruitment of monocytes/macrophages to the inflamed vessel, thereby promoting atherosclerosis, but is dispensable for hypercholesterolaemia-associated adaptive immune priming. Targeting CCR6 or its ligand CCL20 may therefore be a promising therapeutic strategy to alleviate atherosclerosis.
Collapse
Affiliation(s)
- Helga D Manthey
- Univ.-Prof. Dr. Alma Zernecke, Klinikum rechts der Isar, Klinik für Gefäßchirurgie, Technische Universität München, Ismaninger Str. 22, 81675 München, Germany, Tel: +49 89 4140 2167, Fax: +49 89 4140 4861, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Son DS, Kabir SM, Dong Y, Lee E, Adunyah SE. Characteristics of chemokine signatures elicited by EGF and TNF in ovarian cancer cells. JOURNAL OF INFLAMMATION-LONDON 2013; 10:25. [PMID: 23800251 PMCID: PMC3694479 DOI: 10.1186/1476-9255-10-25] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 06/19/2013] [Indexed: 01/26/2023]
Abstract
Background Ovarian cancer, an inflammation-associated cancer, is the fifth leading cause of cancer deaths in women. The malignancy produces a large amount of tumor necrosis factor (TNF) which promotes a proinflammatory tumor microenvironment. In addition, the epidermal growth factor receptor (EGFR) is frequently overexpressed in high-grade ovarian cancer, which likely aggravates cancer progression. Since ovarian cancer progression is closely associated with chemokine networks driven by inflammation or EGFR activation, we investigated the chemokine signatures elicited by EGF and TNF in ovarian cancer cells to determine their individual profiles and if there was in fact some kind of synergy between their actions on the chemokine network. Methods We used a PCR array for the chemokine network to examine the signature of chemokines and their receptors elicited by EGF and TNF in four ovarian cancer cell lines (OVCAR-3, SKOV-3, CaOV-3 and TOV-21G). Results The chemokine network revealed that ovarian cancer cells commonly expressed high levels of proinflammatory chemokines such as CCL20, CXCL1-3 and CXCL8 in response to EGF or TNF. However, the responsiveness to EGF or TNF displayed a cell line specific pattern. Although OVCAR-3 and SKOV-3 cells were responsive to either EGF or TNF, their TNF responsiveness was dominant. On the other hand, CaOV-3 and TOV-21G cells were responsive to EGF but less to TNF, probably due to the high levels of non-canonical nuclear factor (NF)-κB components such as IKKα and p52 in these cell lines compared to OVCAR-3 and SKOV-3 cells. Among chemokine receptors, only CXCR5 was responsive to EGF or TNF in CaOV-3 cells. Finally, CCL20 and CXCL8 responded synergistically in response to EGF and TNF in OVCAR-3 and SKOV-3 cells. Conclusion Our results indicate that CCL20, CXCL1-3 and CXCL8 are the primary chemokines induced by EGF or TNF and are elicited in these ovarian cancer cells via NF-κB, Akt and Erk signaling pathways. Of interest, there was a syngergistic response in terms of CCL20 and CXCL8 levels, when OVCAR-3 and SKOV-3 cells were exposed to EGF plus TNF. Targeting these proinflammatory chemokines may be a promising therapeutic strategy for ovarian cancer with abundant TNF and EGFR activation patterns.
Collapse
Affiliation(s)
- Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN, USA.
| | | | | | | | | |
Collapse
|
47
|
Abstract
Lysophosphatidic acid (LPA) is a potent bioactive phospholipid. As many other biological active lipids, LPA is an autacoid: it is formed locally on demand, and it acts locally near its site of synthesis. LPA has a plethora of biological activities on blood cells (platelets, monocytes) and cells of the vessel wall (endothelial cells, smooth muscle cells, macrophages) that are all key players in atherosclerotic and atherothrombotic processes. The specific cellular actions of LPA are determined by its multifaceted molecular structures, the expression of multiple G-protein coupled LPA receptors at the cell surface and their diverse coupling to intracellular signalling pathways. Numerous studies have now shown that LPA has thrombogenic and atherogenic actions. Here, we aim to provide a comprehensive, yet concise, thoughtful and critical review of this exciting research area and to pinpoint potential pharmacological targets for inhibiting thrombogenic and atherogenic activities of LPA. We hope that the review will serve to accelerate knowledge of basic and clinical science, and to foster drug development in the field of LPA and atherosclerotic/atherothrombotic diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
48
|
Regulation of atherogenesis by chemokines and chemokine receptors. Arch Immunol Ther Exp (Warsz) 2012; 61:1-14. [PMID: 23224338 DOI: 10.1007/s00005-012-0202-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 11/18/2012] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is a chronic inflammatory and metabolic disorder affecting large- and medium-sized arteries, and the leading cause of mortality worldwide. The pathogenesis of atherosclerosis involves accumulation of lipids and leukocytes in the intima of blood vessel walls creating plaque. How leukocytes accumulate in plaque remains poorly understood; however, chemokines acting at specific G protein-coupled receptors appear to be important. Studies using knockout mice suggest that chemokine receptor signaling may either promote or inhibit atherogenesis, depending on the receptor. These proof of concept studies have spurred efforts to develop drugs targeting the chemokine system in atherosclerosis, and several have shown beneficial effects in animal models. This study will review key discoveries in basic and translational research in this area.
Collapse
|
49
|
Lipinski MJ, Campbell KA, Duong SQ, Welch TJ, Garmey JC, Doran AC, Skaflen MD, Oldham SN, Kelly KA, McNamara CA. Loss of Id3 increases VCAM-1 expression, macrophage accumulation, and atherogenesis in Ldlr-/- mice. Arterioscler Thromb Vasc Biol 2012; 32:2855-61. [PMID: 23042815 DOI: 10.1161/atvbaha.112.300352] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Inhibitor of differention-3 (Id3) promotes B cells homing to the aorta and atheroprotection in Apoe(-/-) mice. We sought to determine the impact of loss of Id3 in the Ldlr((-/-)) mouse model of diet-induced atherosclerosis and identify novel Id3 targets in the vessel wall. METHODS AND RESULTS Ex vivo optical imaging confirmed that Id3((-/-)) Ldlr((-/-)) mice have significantly fewer aortic B cells than Id3((+/+)) Ldlr(-/-) mice. After 8 and 16 weeks of Western diet, Id3((-/-)) Ldlr((-/-)) mice developed significantly more atherosclerosis than Id3((+/+)) Ldlr((-/-)) mice, with Id3(+/-) Ldlr(-/-) mice demonstrating an intermediate phenotype. There were no differences in serum lipid levels between genotypes. Immunostaining demonstrated that aortas from Id3((-/-)) Ldlr((-/-)) mice had greater intimal macrophage density and C-C chemokine ligand 20 and vascular cell adhesion molecule 1 (VCAM-1) expression compared with Id3((+/+)) Ldlr(-/-) mice. Real-time polymerase chain reaction demonstrated increased VCAM-1 mRNA levels in the aortas of Id3(-/-) Ldlr(-/-) mice. Primary vascular smooth muscle cells from Id3((-/-)) mice expressed greater amounts of VCAM-1 protein compared with control. Gain and loss of function studies in primary vascular smooth muscle cells identified a role for Id3 in repressing VCAM-1 promoter activation. Chromatin immunoprecipitation demonstrated interaction of E12 with the VCAM-1 promoter, which is inhibited by Id3. CONCLUSIONS Id3 is an atheroprotective transcription regulator with targets in both B cells and vessel wall cells leading to reduced macrophage accumulation and reduced atherosclerosis formation.
Collapse
Affiliation(s)
- Michael J Lipinski
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Alexander MR, Murgai M, Moehle CW, Owens GK. Interleukin-1β modulates smooth muscle cell phenotype to a distinct inflammatory state relative to PDGF-DD via NF-κB-dependent mechanisms. Physiol Genomics 2012; 44:417-29. [PMID: 22318995 PMCID: PMC3339851 DOI: 10.1152/physiolgenomics.00160.2011] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 01/17/2012] [Indexed: 12/14/2022] Open
Abstract
Smooth muscle cell (SMC) phenotypic modulation in atherosclerosis and in response to PDGF in vitro involves repression of differentiation marker genes and increases in SMC proliferation, migration, and matrix synthesis. However, SMCs within atherosclerotic plaques can also express a number of proinflammatory genes, and in cultured SMCs the inflammatory cytokine IL-1β represses SMC marker gene expression and induces inflammatory gene expression. Studies herein tested the hypothesis that IL-1β modulates SMC phenotype to a distinct inflammatory state relative to PDGF-DD. Genome-wide gene expression analysis of IL-1β- or PDGF-DD-treated SMCs revealed that although both stimuli repressed SMC differentiation marker gene expression, IL-1β distinctly induced expression of proinflammatory genes, while PDGF-DD primarily induced genes involved in cell proliferation. Promoters of inflammatory genes distinctly induced by IL-1β exhibited over-representation of NF-κB binding sites, and NF-κB inhibition in SMCs reduced IL-1β-induced upregulation of proinflammatory genes as well as repression of SMC differentiation marker genes. Interestingly, PDGF-DD-induced SMC marker gene repression was not NF-κB dependent. Finally, immunofluorescent staining of mouse atherosclerotic lesions revealed the presence of cells positive for the marker of an IL-1β-stimulated inflammatory SMC, chemokine (C-C motif) ligand 20 (CCL20), but not the PDGF-DD-induced gene, regulator of G protein signaling 17 (RGS17). Results demonstrate that IL-1β- but not PDGF-DD-induced phenotypic modulation of SMC is characterized by NF-κB-dependent activation of proinflammatory genes, suggesting the existence of a distinct inflammatory SMC phenotype. In addition, studies provide evidence for the possible utility of CCL20 and RGS17 as markers of inflammatory and proliferative state SMCs within atherosclerotic plaques in vivo.
Collapse
Affiliation(s)
- Matthew R Alexander
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | | | | | | |
Collapse
|