1
|
Antonello J, Roy P. Damage-Associated Molecular Patterns (DAMPs) In Vascular Diseases. J Biol Chem 2025:110241. [PMID: 40381697 DOI: 10.1016/j.jbc.2025.110241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Research into the role of chronic sterile inflammation (i.e. a prolonged inflammatory state not caused by an infectious agent), in vascular disease progression has continued to grow over the last few decades. DAMPs have a critical role in this research due to their ability to link stress-causing cardiovascular risk factors to inflammatory phenotypes seen in vascular disease. In this mini-review, we will briefly summarize the DAMPs and receptor signaling pathways that have been extensively studied in the context of vascular disease, including TLRs, RAGE, cGAS-STING, and the NLRP3 inflammasome. In particular, we will discuss how these pathways can promote the release of pro-inflammatory cytokines and chemokines as well as vascular remodeling. Next, we will summarize the results of studies which have linked the various pro-inflammatory effects of DAMPs with the phenotypes in the context of vascular diseases including atherosclerosis, fibrosis, aneurysm, ischemia, and hypertension. Finally, we will discuss some pre-clinical and clinical trials that have targeted DAMPs, their receptors, or the products of their signaling pathways, and discuss the outlook and future directions for the field at large.
Collapse
Affiliation(s)
| | - Partha Roy
- Bioengineering, University of Pittsburgh; Pathology, University of Pittsburgh.
| |
Collapse
|
2
|
Desouky DA, Nosair NA, Salama MK, El-Magd MA, Desouky MA, Sherif DE. PCSK9 and its relationship with HMGB1, TLR4, and TNFα in non-statin and statin-treated coronary artery disease patients. Mol Cell Biochem 2025; 480:2935-2949. [PMID: 39541017 DOI: 10.1007/s11010-024-05154-2] [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: 07/09/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Despite statin use in coronary artery disease (CAD), significant risk remains, potentially due to increased proprotein convertase subtilisin/kexin-type 9 (PCSK9) production, which raises LDL-C levels and induces inflammation. The exact relationship between PCSK9, inflammatory markers like TNFα, TLR4, CRP, and HMGB1, and monocyte subsets is poorly understood. This study aimed to explore these relationships in non-statin and statin-taking CAD patients. This case-control study included 91 controls and 91 stable CAD patients, divided into no-statin (NS, n = 25), low-dose statin (LDS, n = 25), and high-dose statin (HDS, n = 41) groups. Serum levels of LDL-C, CRP, PCSK9, TLR4, HMGB1, and TNFα were measured. Monocyte subsets were classified using flow cytometry into classical monocytes (CM), intermediate monocytes (IM), and non-classical monocytes (NCM). CAD patients showed elevated PCSK9, LDL-C, and inflammatory markers compared to controls. Statin groups (LDS, HDS) had lower LDL-C and inflammatory markers but higher PCSK9 than the NS group, with the HDS group showing the lowest LDL-C and inflammatory markers but the highest PCSK9. In the NS group, PCSK9 positively correlated with inflammatory markers (HMGB1, TNFα, TLR4, CRP) and monocyte subsets (IM%, NCM%). In the total statin group (LDS + HDS), PCSK9 negatively correlated with HMGB1, TLR4, and NCM%, for each, respectively, and positively with CM%. Multivariable linear regression showed significant associations between PCSK9 and HMGB1, NCM%, and IM% in the NS group, and HMGB1, NCM%, and TLR4 in the total statin group. In conclusion, we recommend combining PCSK9 inhibitors with statins in high-risk CAD patients. This may enhance statin efficacy, reduce LDL-C, and inhibit the TLR4/NF-кB inflammatory pathway, decreasing atherosclerotic inflammation.
Collapse
Affiliation(s)
- Dina A Desouky
- Department of Clinical Pathology, Faculty of Medicine, Kafrelshiekh University, Kafrelsheikh, Egypt.
| | - Nahla A Nosair
- Department of Clinical Pathology, Faculty of Medicine, Kafrelshiekh University, Kafrelsheikh, Egypt
| | - Mohamed K Salama
- Department of Cardiovascular, Faculty of Medicine, Kafrelshiekh University, Kafrelsheikh, Egypt
| | - Mohammed A El-Magd
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Kafrelshiekh University, Kafrelsheikh, Egypt
| | | | - Dalia E Sherif
- Department of Clinical Pathology, Faculty of Medicine, Kafrelshiekh University, Kafrelsheikh, Egypt
| |
Collapse
|
3
|
Wasim R, Singh A, Islam A, Mohammed S, Anwar A, Mahmood T. High Mobility Group Box 1 and Cardiovascular Diseases: Study of Act and Connect. Cardiovasc Toxicol 2024; 24:1268-1286. [PMID: 39242448 DOI: 10.1007/s12012-024-09919-5] [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: 10/16/2023] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Cardiovascular disease is the deadly disease that can result in sudden death, and inflammation plays an important role in its onset and progression. High mobility group box 1 (HMGB1) is a nuclear protein that regulates transcription, DNA replication, repair, and nucleosome assembly. HMGB1 is released passively by necrotic tissues and actively secreted by stressed cells. Extracellular HMGB1 functions as a damage associated molecular patterns molecule, producing numerous redox forms that induce a range of cellular responses by binding to distinct receptors and interactors, including tissue inflammation and regeneration. Extracellular HMGB1 inhibition reduces inflammation and is protective in experimental models of myocardial ischemia/reperfusion damage, myocarditis, cardiomyopathies caused by mechanical stress, diabetes, bacterial infection, or chemotherapeutic drugs. HMGB1 administration following a myocardial infarction followed by permanent coronary artery ligation improves cardiac function by stimulating tissue regeneration. HMGB1 inhibits contractility and produces hypertrophy and death in cardiomyocytes, while also stimulating cardiac fibroblast activity and promoting cardiac stem cell proliferation and differentiation. Maintaining normal nuclear HMGB1 levels, interestingly, protects cardiomyocytes from apoptosis by limiting DNA oxidative stress, and mice with HMGB1cardiomyocyte-specific overexpression are partially protected from cardiac injury. Finally, elevated levels of circulating HMGB1 have been linked to human heart disease. As a result, following cardiac damage, HMGB1 elicits both detrimental and helpful responses, which may be due to the formation and stability of the various redox forms, the particular activities of which in this context are mostly unknown. This review covers recent findings in HMGB1 biology and cardiac dysfunction.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacy, Integral University, Lucknow, 226026, India.
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Aditya Singh
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Anas Islam
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Saad Mohammed
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Aamir Anwar
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Tarique Mahmood
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
4
|
Kim JW, Kim JY, Bae HE, Kim CD. Biophysically stressed vascular smooth muscle cells express MCP-1 via a PDGFR-β-HMGB1 signaling pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:449-456. [PMID: 39198225 PMCID: PMC11361998 DOI: 10.4196/kjpp.2024.28.5.449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 09/01/2024]
Abstract
Vascular smooth muscle cells (VSMCs) under biophysical stress play an active role in the progression of vascular inflammation, but the precise mechanisms are unclear. This study examined the cellular expression of monocyte chemoattractant protein 1 (MCP-1) and its related mechanisms using cultured rat aortic VSMCs stimulated with mechanical stretch (MS, equibiaxial cyclic stretch, 60 cycles/ min). When the cells were stimulated with 10% MS, MCP-1 expression was markedly increased compared to those in the cells stimulated with low MS intensity (3% or 5%). An enzyme-linked immunosorbent assay revealed an increase in HMGB1 released into culture media from the cells stimulated with 10% MS compared to those stimulated with 3% MS. A pretreatment with glycyrrhizin, a HMGB1 inhibitor, resulted in the marked attenuation of MCP-1 expression in the cells stimulated with 10% MS, suggesting a key role of HMGB1 on MCP-1 expression. Western blot analysis revealed higher PDGFR-α and PDGFR-β expression in the cells stimulated with 10% MS than 3% MS-stimulated cells. In the cells deficient of PDGFR-β using siRNA, but not PDGFR-α, HMGB1 released into culture media was significantly attenuated in the 10% MS-stimulated cells. Similarly, MCP-1 expression induced in 10% MS-stimulated cells was also attenuated in cells deficient of PDGFR-β. Overall, the PDGFR-β signaling plays a pivotal role in the increased expression of MCP-1 in VSMCs stressed with 10% MS. Therefore, targeting PDGFR-β signaling in VSMCs might be a promising therapeutic strategy for vascular complications in the vasculatures under excessive biophysical stress.
Collapse
Affiliation(s)
- Ji Won Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Ju Yeon Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Hee Eun Bae
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| |
Collapse
|
5
|
Kiełbowski K, Skórka P, Plewa P, Bakinowska E, Pawlik A. The Role of Alarmins in the Pathogenesis of Atherosclerosis and Myocardial Infarction. Curr Issues Mol Biol 2024; 46:8995-9015. [PMID: 39194749 DOI: 10.3390/cimb46080532] [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: 06/29/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Atherosclerosis is a condition that is associated with lipid accumulation in the arterial intima. Consequently, the enlarging lesion, which is also known as an atherosclerotic plaque, may close the blood vessel lumen, thus leading to organ ischaemia. Furthermore, the plaque may rupture and initiate the formation of a thrombus, which can cause acute ischaemia. Atherosclerosis is a background pathological condition that can eventually lead to major cardiovascular diseases such as acute coronary syndrome or ischaemic stroke. The disorder is associated with an altered profile of alarmins, stress response molecules that are secreted due to cell injury or death and that induce inflammatory responses. High-mobility group box 1 (HMGB1), S100 proteins, interleukin-33, and heat shock proteins (HSPs) also affect the behaviour of endothelial cells and vascular smooth muscle cells (VSMCs). Thus, alarmins control the inflammatory responses of endothelial cells and proliferation of VSMCs, two important processes implicated in the pathogenesis of atherosclerosis. In this review, we will discuss the role of alarmins in the pathophysiology of atherosclerosis and myocardial infarction.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Patryk Skórka
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Paulina Plewa
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
6
|
Otunla AA, Shanmugarajah K, Davies AH, Lucia Madariaga M, Shalhoub J. The Biological Parallels Between Atherosclerosis and Cardiac Allograft Vasculopathy: Implications for Solid Organ Chronic Rejection. Cardiol Rev 2024; 32:2-11. [PMID: 38051983 DOI: 10.1097/crd.0000000000000437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Atherosclerosis and solid organ chronic rejection are pervasive chronic disease states that account for significant morbidity and mortality in developed countries. Recently, a series of shared molecular pathways have emerged, revealing biological parallels from early stages of development up to the advanced forms of pathology. These shared mechanistic processes are inflammatory in nature, reflecting the importance of inflammation in both disorders. Vascular inflammation triggers endothelial dysfunction and disease initiation through aberrant vasomotor control and shared patterns of endothelial activation. Endothelial dysfunction leads to the recruitment of immune cells and the perpetuation of the inflammatory response. This drives lesion formation through the release of key cytokines such as IFN-y, TNF-alpha, and IL-2. Continued interplay between the adaptive and innate immune response (represented by T lymphocytes and macrophages, respectively) promotes lesion instability and thrombotic complications; hallmarks of advanced disease in both atherosclerosis and solid organ chronic rejection. The aim of this study is to identify areas of overlap between atherosclerosis and chronic rejection. We then discuss new approaches to improve current understanding of the pathophysiology of both disorders, and eventually design novel therapeutics.
Collapse
Affiliation(s)
- Afolarin A Otunla
- From the Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | | | - Alun H Davies
- Section of Vascular Surgery, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Imperial Vascular Unit, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Joseph Shalhoub
- Section of Vascular Surgery, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Imperial Vascular Unit, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
7
|
Yu H, Cao H, Yu H. MicroRNA-98 inhibition accelerates the development of atherosclerosis via regulation of dysfunction of endothelial cell. Clin Exp Hypertens 2023; 45:2206068. [PMID: 37183710 DOI: 10.1080/10641963.2023.2206068] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/01/2023] [Accepted: 04/18/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Atherosclerosis has been recognized as a chronic inflammation initiated by dysfunction of endothelial cell that contributes to the increased morbidity and mortality of severe cardiovascular events. The reported important role of microRNA-98 (miR-98) in regulation of endothelial cell behaviors prompt us to hypothesize that miR-98 could be involved in the process of atherosclerosis. METHODS AND RESULTS The current research showed the miR-98 expression was gradually down-regulated in atherosclerotic mouse arteries isolated from ApoE ablation mice subjected to high fat diet. Additionally, a dramatically reduced miR-98 expression in endothelial cells administrated to oxidized low-density lipoprotein (Ox-LDL) but a slight down-regulated level was found in macrophages. Functionally, attenuated miR-98 expression promoted secretion of chemokines and adhesion molecules in human umbilical vein endothelial cells (HUVECs) induced by Ox-LDL, which subsequently increased infiltration and pro-inflammatory genes expression of macrophages, as well as the foam cell formation. Mechanistically, in vitro experiments indicated that the endothelial cell dysfunction regulated by miR-98 knockdown was partially contributed by upregulated expression of HMGB1. Furthermore, the animal experiment with ApoE-/- mice administrated with miR-98 inhibitor demonstrated that miR-98 silencing enhanced the atherosclerotic lesions in aorta and aortic sinus that were accompanied with increased adhesion molecules, chemokines, and pro-inflammatory markers expression. CONCLUSION MicroRNA-98 knockdown promoted endothelial cell dysfunction to affect the inflammatory state of macrophage and the development of atherosclerosis, at least partially, through direct targeting HMGB1. Collected, these data suggested that miR-98 could be a novel drug target for atherogenesis management.
Collapse
Affiliation(s)
- Huai Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Hui Cao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Hang Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| |
Collapse
|
8
|
Giesen A, Mouselimis D, Weichsel L, Giannopoulos AA, Schmermund A, Nunninger M, Schuetz M, André F, Frey N, Korosoglou G. Pericoronary adipose tissue attenuation is associated with non-calcified plaque burden in patients with chronic coronary syndromes. J Cardiovasc Comput Tomogr 2023; 17:384-392. [PMID: 37659885 DOI: 10.1016/j.jcct.2023.08.008] [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: 04/30/2023] [Revised: 07/28/2023] [Accepted: 08/18/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Pericoronary adipose tissue attenuation (PCAT) is a marker of inflammation of the pericoronary fat tissue, which can be assessed by coronary computed tomography angiography (CCTA). Its prognostic value was reported in previous studies. Nevertheless, the relationship between PCAT, plaque burden and coronary artery disease (CAD) severity, are not well defined. AIM We sought to evaluate the relationship between PCAT, CAD severity based on the CAD-RADS 2.0 score and plaque burden in patients with chronic coronary syndrome (CCS). METHODS Consecutive patients with a clinical indication for CCTA due to suspected or known CCS were included in our study. PCAT was measured in the proximal 4 cm of each of the right coronary artery (RCA), left anterior descending artery (LAD), and the left circumflex artery (LCX). The CAD-RADS 2.0 score was assessed in all patients and total, calcified, and non-calcified plaque burden was quantitatively measured. RESULTS 868 patients (median age of 67.0 (IQR = 58.0-75.0)yrs., 400 (46.1%) female) underwent CCTA between September 2020 and August 2022 due to CCS. Weak correlations were found between PCAT and the total plaque burden, as well as with the Agatston score, whereas no correlations were found between PCAT and CAD-RADS 2.0 score. Associations were also observed between the PCAT of the LAD, RCA and LCX with non-calcified plaque burden (Odds ratios of 1.22 (95%CI = 1.15-1.29), 1.11 (95%CI = 1.07-1.17) and 1.14 (95%CI = 1.08-1.14), respectively, p < 0.001 for all) which were independent of age, the Agatston score, and the CAD-RADS 2.0 score). In addition, higher PCAT were noticed with increasing number of plaques, exhibiting high-risk features per patient (p < 0.05 by ANOVA for all). CONCLUSION PCAT exhibits significant associations with non-calcified plaque burden and plaques with high-risk features in patients undergoing CCTA for CCS. Thus, PCAT may identify high-risk patients who could benefit from more aggressive preventive therapy, which merits further investigation in future studies.
Collapse
Affiliation(s)
- Alexander Giesen
- GRN Hospital Weinheim, Cardiology, Vascular Medicine & Pneumology, Weinheim, Germany; Cardiac Imaging Center Weinheim, Hector Foundations, Weinheim, Germany
| | - Dimitrios Mouselimis
- GRN Hospital Weinheim, Cardiology, Vascular Medicine & Pneumology, Weinheim, Germany; Cardiac Imaging Center Weinheim, Hector Foundations, Weinheim, Germany
| | - Loris Weichsel
- GRN Hospital Weinheim, Cardiology, Vascular Medicine & Pneumology, Weinheim, Germany; Cardiac Imaging Center Weinheim, Hector Foundations, Weinheim, Germany
| | - Andreas A Giannopoulos
- Department of Nuclear Medicine, Cardiac Imaging, University and University Hospital Zurich, Zurich, Switzerland
| | - Axel Schmermund
- CCB Hospital, Department of Cardiology and Vascular Medicine, Frankfurt, Germany
| | - Max Nunninger
- Radiology Practice, GRN Hospital Weinheim, Weinheim, Germany
| | - Moritz Schuetz
- GRN Hospital Weinheim, Cardiology, Vascular Medicine & Pneumology, Weinheim, Germany; Cardiac Imaging Center Weinheim, Hector Foundations, Weinheim, Germany
| | - Florian André
- University Hospital Heidelberg, Cardiology, Angiology & Pneumology, Heidelberg, Germany
| | - Norbert Frey
- University Hospital Heidelberg, Cardiology, Angiology & Pneumology, Heidelberg, Germany
| | - Grigorios Korosoglou
- GRN Hospital Weinheim, Cardiology, Vascular Medicine & Pneumology, Weinheim, Germany; Cardiac Imaging Center Weinheim, Hector Foundations, Weinheim, Germany.
| |
Collapse
|
9
|
Ignatz RM, Zirkenbach VA, Kaya M, Stroikova V, Öttl R, Frey N, Kaya Z. No Evidence for Myocarditis or Other Organ Affection by Induction of an Immune Response against Critical SARS-CoV-2 Protein Epitopes in a Mouse Model Susceptible for Autoimmunity. Int J Mol Sci 2023; 24:9873. [PMID: 37373021 DOI: 10.3390/ijms24129873] [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: 05/15/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
After Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) developed into a global pandemic, not only the infection itself but also several immune-mediated side effects led to additional consequences. Immune reactions such as epitope spreading and cross-reactivity may also play a role in the development of long-COVID, although the exact pathomechanisms have not yet been elucidated. Infection with SARS-CoV-2 can not only cause direct damage to the lungs but can also lead to secondary indirect organ damage (e.g., myocardial involvement), which is often associated with high mortality. To investigate whether an immune reaction against the viral peptides can lead to organ affection, a mouse strain known to be susceptible to the development of autoimmune diseases, such as experimental autoimmune myocarditis (EAM), was used. First, the mice were immunized with single or pooled peptide sequences of the virus's spike (SP), membrane (MP), nucleocapsid (NP), and envelope protein (EP), then the heart and other organs such as the liver, kidney, lung, intestine, and muscle were examined for signs of inflammation or other damage. Our results showed no significant inflammation or signs of pathology in any of these organs as a result of the immunization with these different viral protein sequences. In summary, immunization with different SARS-CoV-2 spike-, membrane-, nucleocapsid-, and envelope-protein peptides does not significantly affect the heart or other organ systems adversely, even when using a highly susceptible mouse strain for experimental autoimmune diseases. This suggests that inducing an immune reaction against these peptides of the SARS-CoV-2 virus alone is not sufficient to cause inflammation and/or dysfunction of the myocardium or other studied organs.
Collapse
Affiliation(s)
| | | | - Mansur Kaya
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Vera Stroikova
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Renate Öttl
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ziya Kaya
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
DeWulf B, Minsart L, Verdonk F, Kruys V, Piagnerelli M, Maze M, Saxena S. High Mobility Group Box 1 (HMGB1): Potential Target in Sepsis-Associated Encephalopathy. Cells 2023; 12:cells12071088. [PMID: 37048161 PMCID: PMC10093266 DOI: 10.3390/cells12071088] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) remains a challenge for intensivists that is exacerbated by lack of an effective diagnostic tool and an unambiguous definition to properly identify SAE patients. Risk factors for SAE development include age, genetic factors as well as pre-existing neuropsychiatric conditions. Sepsis due to certain infection sites/origins might be more prone to encephalopathy development than other cases. Currently, ICU management of SAE is mainly based on non-pharmacological support. Pre-clinical studies have described the role of the alarmin high mobility group box 1 (HMGB1) in the complex pathogenesis of SAE. Although there are limited data available about the role of HMGB1 in neuroinflammation following sepsis, it has been implicated in other neurologic disorders, where its translocation from the nucleus to the extracellular space has been found to trigger neuroinflammatory reactions and disrupt the blood–brain barrier. Negating the inflammatory cascade, by targeting HMGB1, may be a strategy to complement non-pharmacologic interventions directed against encephalopathy. This review describes inflammatory cascades implicating HMGB1 and strategies for its use to mitigate sepsis-induced encephalopathy.
Collapse
Affiliation(s)
- Bram DeWulf
- Department of Anesthesia—Critical Care, AZ Sint-Jan Brugge Oostende AV, 8000 Bruges, Belgium
| | - Laurens Minsart
- Department of Anesthesia, Antwerp University Hospital (UZA), 2650 Edegem, Belgium
| | - Franck Verdonk
- Department of Anesthesiology and Intensive Care, GRC 29, DMU DREAM, Hôpital Saint-Antoine and Sorbonne University, Assistance Publique-Hôpitaux de Paris, 75012 Paris, France
| | - Véronique Kruys
- Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, Free University of Brussels (ULB), 6041 Gosselies, Belgium
| | - Michael Piagnerelli
- Department of Intensive Care, CHU-Charleroi, Université Libre de Bruxelles, 6042 Charleroi, Belgium
- Experimental Medicine Laboratory (ULB Unit 222), CHU-Charleroi, Université Libre de Bruxelles, 6110 Montigny-le-Tilleul, Belgium
| | - Mervyn Maze
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sarah Saxena
- Department of Anesthesia—Critical Care, AZ Sint-Jan Brugge Oostende AV, 8000 Bruges, Belgium
- Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, Free University of Brussels (ULB), 6041 Gosselies, Belgium
| |
Collapse
|
11
|
Schoenfeld J, Roeh A, Holdenrieder S, von Korn P, Haller B, Krueger K, Falkai P, Halle M, Hasan A, Scherr J. High-mobility group box 1 protein, receptor for advanced glycation end products and nucleosomes increases after marathon. Front Physiol 2023; 14:1118127. [PMID: 36866178 PMCID: PMC9971726 DOI: 10.3389/fphys.2023.1118127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
Background: Prolonged and strenuous exercise has been linked to potential exercise-induced myocardial damages. One potential key to unmask the discussed underlying mechanisms of this subclinical cardiac damage could be markers of immunogenic cell damage (ICD). We investigated the kinetics of high-mobility group box 1 protein (HMGB1), soluble receptor for advanced glycation end products (sRAGE), nucleosomes, high sensitive troponin T (hs-TnT) and high sensitive C-reactive protein (hs-CRP) before and up to 12 weeks post-race and described associations with routine laboratory markers and physiological covariates. Methods: In our prospective longitudinal study, 51 adults (82% males; 43 ± 9 years) were included. All participants underwent a cardiopulmonary evaluation 10-12 weeks pre-race. HMGB1, sRAGE, nucleosomes, hs-TnT and, hs-CRP were analysed 10-12 weeks prior, 1-2 weeks before, immediately, 24 h, 72 h, and 12 weeks post-race. Results: HMGB1, sRAGE, nucleosomes and hs-TnT increased significantly from pre- to immediately post-race (0.82-2.79 ng/mL; 1132-1388 pg/mL; 9.24-56.65 ng/mL; 6-27 ng/L; p < 0.001) and returned to baseline within 24-72 h. Hs-CRP increased significantly 24 h post-race (0.88-11.5 mg/L; p < 0.001). Change in sRAGE was positively associated with change in hs-TnT (rs = 0.352, p = 0.011). Longer marathon finishing time was significantly associated with decreased levels of sRAGE [-9.2 pg/mL (β = -9.2, SE = 2.2, p < 0.001)]. Conclusion: Prolonged and strenuous exercise increases markers of ICD immediately post-race, followed by a decrease within 72 h. An acute marathon event results in transient alterations of ICD, we assume that this is not solely driven by myocyte damages.
Collapse
Affiliation(s)
- Julia Schoenfeld
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Astrid Roeh
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Bezirkskrankenhaus Augsburg, Augsburg, Germany,Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Centre Munich, Technical University Munich, Munich, Germany
| | - Pia von Korn
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and Epidemiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kimberly Krueger
- Institute of Laboratory Medicine, German Heart Centre Munich, Technical University Munich, Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Bezirkskrankenhaus Augsburg, Augsburg, Germany,Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Johannes Scherr
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,University Center for Preventive and Sports Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland,*Correspondence: Johannes Scherr,
| |
Collapse
|
12
|
Azizidoost S, Nasrolahi A, Sheykhi-Sabzehpoush M, Akiash N, Assareh AR, Anbiyaee O, Antosik P, Dzięgiel P, Farzaneh M, Kempisty B. Potential roles of endothelial cells-related non-coding RNAs in cardiovascular diseases. Pathol Res Pract 2023; 242:154330. [PMID: 36696805 DOI: 10.1016/j.prp.2023.154330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Endothelial dysfunction is identified by a conversion of the endothelium toward decreased vasodilation and prothrombic features and is known as a primary pathogenic incident in cardiovascular diseases. An insight based on particular and promising biomarkers of endothelial dysfunction may possess vital clinical significances. Currently, non-coding RNAs due to their participation in critical cardiovascular processes like initiation and progression have gained much attention as possible diagnostic as well as prognostic biomarkers in cardiovascular diseases. Emerging line of proof has demonstrated that abnormal expression of non-coding RNAs is nearly correlated with the pathogenesis of cardiovascular diseases. In the present review, we focus on the expression and functional effects of various kinds of non-coding RNAs in cardiovascular diseases and negotiate their possible clinical implications as diagnostic or prognostic biomarkers and curative targets.
Collapse
Affiliation(s)
- Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Nehzat Akiash
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Reza Assareh
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Omid Anbiyaee
- Cardiovascular Research Center, Nemazi Hospital, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paweł Antosik
- Institute of Veterinary Medicine, Department of Veterinary Surgery, Nicolaus Copernicus University, Torun, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Bartosz Kempisty
- Institute of Veterinary Medicine, Department of Veterinary Surgery, Nicolaus Copernicus University, Torun, Poland; Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wrocław, Poland; North Carolina State University College of Agriculture and Life Sciences, Raleigh, NC 27695, USA.
| |
Collapse
|
13
|
Műzes G, Bohusné Barta B, Szabó O, Horgas V, Sipos F. Cell-Free DNA in the Pathogenesis and Therapy of Non-Infectious Inflammations and Tumors. Biomedicines 2022; 10:2853. [PMID: 36359370 PMCID: PMC9687442 DOI: 10.3390/biomedicines10112853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
The basic function of the immune system is the protection of the host against infections, along with the preservation of the individual antigenic identity. The process of self-tolerance covers the discrimination between self and foreign antigens, including proteins, nucleic acids, and larger molecules. Consequently, a broken immunological self-tolerance results in the development of autoimmune or autoinflammatory disorders. Immunocompetent cells express pattern-recognition receptors on their cell membrane and cytoplasm. The majority of endogenous DNA is located intracellularly within nuclei and mitochondria. However, extracellular, cell-free DNA (cfDNA) can also be detected in a variety of diseases, such as autoimmune disorders and malignancies, which has sparked interest in using cfDNA as a possible biomarker. In recent years, the widespread use of liquid biopsies and the increasing demand for screening, as well as monitoring disease activity and therapy response, have enabled the revival of cfDNA research. The majority of studies have mainly focused on the function of cfDNA as a biomarker. However, research regarding the immunological consequences of cfDNA, such as its potential immunomodulatory or therapeutic benefits, is still in its infancy. This article discusses the involvement of various DNA-sensing receptors (e.g., absent in melanoma-2; Toll-like receptor 9; cyclic GMP-AMP synthase/activator of interferon genes) in identifying host cfDNA as a potent danger-associated molecular pattern. Furthermore, we aim to summarize the results of the experimental studies that we recently performed and highlight the immunomodulatory capacity of cfDNA, and thus, the potential for possible therapeutic consideration.
Collapse
Affiliation(s)
| | | | | | | | - Ferenc Sipos
- Department of Internal Medicine and Hematology, Semmelweis University, Szentkirályi Street 46, 1088 Budapest, Hungary
| |
Collapse
|
14
|
Rando MM, Biscetti F, Cecchini AL, Nardella E, Nicolazzi MA, Angelini F, Iezzi R, Eraso LH, Dimuzio PJ, Pitocco D, Gasbarrini A, Massetti M, Flex A. Serum high mobility group box-1 levels associated with cardiovascular events after lower extremity revascularization: a prospective study of a diabetic population. Cardiovasc Diabetol 2022; 21:214. [PMID: 36244983 PMCID: PMC9571458 DOI: 10.1186/s12933-022-01650-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/01/2022] [Indexed: 12/24/2022] Open
Abstract
Background Peripheral arterial disease (PAD) is one of the most disabling cardiovascular complications of type 2 diabetes mellitus and is indeed associated with a high risk of cardiovascular and limb adverse events. High mobility group box-1 (HMGB-1) is a nuclear protein involved in the inflammatory response that acts as a pro-inflammatory cytokine when released into the extracellular space. HMBG-1 is associated with PAD in diabetic patients. The aim of this study was to evaluate the association between serum HMGB-1 levels and major adverse cardiovascular events (MACE) and major adverse limb events (MALE) after lower-extremity endovascular revascularization (LER) in a group of diabetic patients with chronic limb-threatening ischemia (CLTI). Methods We conducted a prospective observational study of 201 diabetic patients with PAD and CLTI requiring LER. Baseline serum HMGB-1 levels were determined before endovascular procedure. Data on cardiovascular and limb outcomes were collected in a 12-month follow-up. Results During the follow-up period, 81 cases of MACE and 93 cases of MALE occurred. Patients who subsequently developed MACE and MALE had higher serum HMGB-1 levels. Specifically, 7.5 ng/mL vs 4.9 ng/mL (p < 0.01) for MACE and 7.2 ng/mL vs 4.8 ng/mL (p < 0.01) for MALE. After adjusting for traditional cardiovascular risk factors, the association between serum HMGB-1 levels and cardiovascular outcomes remained significant in multivariable analysis. In our receiver operating characteristic (ROC) curve analysis, serum HMGB-1 levels were a good predictor of MACE incidence (area under the curve [AUC] = 0.78) and MALE incidence (AUC = 0.75). Conclusions This study demonstrates that serum HMGB-1 levels are associated with the incidence of MACE and MALE after LER in diabetic populations with PAD and CLTI.
Collapse
|
15
|
Aftermath of AGE-RAGE Cascade in the pathophysiology of cardiovascular ailments. Life Sci 2022; 307:120860. [PMID: 35940220 DOI: 10.1016/j.lfs.2022.120860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/21/2022]
|
16
|
Genkel VV, Dolgushin II, Baturina IL, Savochkina AY, Nikushkina KV, Minasova AA, Pykhova LR, Kuznetcova AS, Shaposhnik II. Relationships between serum HMGB1 concentration and subpopulation composition of circulating monocytes in patients with subclinical atherosclerosis. MEDICAL IMMUNOLOGY (RUSSIA) 2022; 24:807-820. [DOI: 10.15789/1563-0625-rbs-2508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Chronic non-infectious inflammation of low intensity is the most important mechanism of development and progression in atherosclerosis. Under the conditions of persistent non-resolving inflammation observed in the vascular wall and atherosclerotic plaque (ASB), permanent tissue damage occurs, thus leading to increased formation of endogenous danger-associated molecular patterns (DAMPs). The non-histone chromosomal protein HMGB1 may be regarded as a prototypical DAMPs. HMGB1 acts as a DAMP if entering the extracellular space, causing inflammation by its binding to pattern-recognizing receptors (TLR2, TLR4, RAGE, CD36, etc.). A number of clinical studies have revealed higher HMGB1 levels in the blood of patients with coronary heart disease and atherosclerotic disease of the lower limb arteries, as well as its interrelations with the burden of coronary artery atherosclerosis. Currently, the mechanisms of HMGB1-mediated atherosclerosis progression are studied only fragmentary. The aim of our study was to investigate relationships between the serum HMGB1 level and subsets of circulating monocyte subpopulations in patients with subclinical atherosclerosis.The study enrolled patients aged 40-64 years with subclinical atherosclerosis of peripheral arteries. Serum HMGB1 concentration was determined using enzyme immunoassay kits (Human HMGB1/HMG-1 ELISA Kit, NBP2-62766, Novus Biologicals, USA). The serum HMGB1 threshold was 18.75 pg/ml, whereas the measurement range was 31.25 to 2000 pg/ml. Phenotyping of the blood monocyte subpopulations was performed by flow cytometry using Navios 6/2 device (Beckman Coulter, USA).An increase in serum HMGB1 concentration was associated with decreased number of classical M2 monocytes, and an increase in intermediate and M1 monocytes. Moreover, an increase in HMGB1 concentration was associated with higher numbers of classical, intermediate, and non-classical monocytes expressing CD36 and TLR2. Increased HMGB1 concentration (from Q1 to Q4) correlated with higher numbers of classical (p = 0.001) and intermediate monocytes (p = 0.006) but not with non-classical phenotypes (p = 0.147). Upon increase of HMGB1 concentration (Q1 to Q4), we have found an increase in the number of classical (p < 0.0001), intermediate (p < 0.0001), and non-classical (p < 0.0001), CD36-expressing monocytes. An increased number of intermediate (p = 0.022; p1, 4 = 0.034) and non-classical, TLR2-expressing monocytes was also revealed (p = 0.002; p1, 4 = 0.035). By mean of correlation analysis, IL-1β concentrations showed direct correlation with the number of M1 monocytes (r = 0.268; p = 0.035) and inverse relation with the number of M2 monocytes (r = -0.376; p = 0.003).Increased serum HMGB1 concentration in patients with subclinical atherosclerosis was associated with decreased numbers of classical and M2 monocytes, as well as higher numbers of intermediate and M1 monocytes, like as with increased contents of intermediate and non-classical monocytes expressing CD36 and TLR2. IL-1β levels directly correlated with HMGB1 concentration and the number of Mi-monocytes.
Collapse
|
17
|
Elevated platelet-leukocyte complexes are associated with, but dispensable for myocardial ischemia-reperfusion injury. Basic Res Cardiol 2022; 117:61. [PMID: 36383299 PMCID: PMC9668925 DOI: 10.1007/s00395-022-00970-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/18/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022]
Abstract
AIMS P-selectin is an activatable adhesion molecule on platelets promoting platelet aggregation, and platelet-leukocyte complex (PLC) formation. Increased numbers of PLC are circulating in the blood of patients shortly after acute myocardial infarction and predict adverse outcomes. These correlations led to speculations about whether PLC may represent novel therapeutic targets. We therefore set out to elucidate the pathomechanistic relevance of PLC in myocardial ischemia and reperfusion injury. METHODS AND RESULTS By generating P-selectin deficient bone marrow chimeric mice, the post-myocardial infarction surge in PLC numbers in blood was prevented. Yet, intravital microscopy, flow cytometry and immunohistochemical staining, echocardiography, and gene expression profiling showed unequivocally that leukocyte adhesion to the vessel wall, leukocyte infiltration, and myocardial damage post-infarction were not altered in response to the lack in PLC. CONCLUSION We conclude that myocardial infarction associated sterile inflammation triggers PLC formation, reminiscent of conserved immunothrombotic responses, but without PLC influencing myocardial ischemia and reperfusion injury in return. Our experimental data do not support a therapeutic concept of selectively targeting PLC formation in myocardial infarction.
Collapse
|
18
|
Handayani W, Suharjono, Yogiarto M. Analysis of HMGB-1 level before and after providing atorvastatin standard therapy in coronary artery disease patients with type-2 diabetes mellitus compared to without type-2 diabetes mellitus. J Basic Clin Physiol Pharmacol 2021; 32:439-446. [PMID: 34214372 DOI: 10.1515/jbcpp-2020-0442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/08/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Coronary artery disease (CAD) is one of the main causes of death from cardiovascular disease, because heart attacks result in atherosclerosis which causes narrowing of the arteries. Atorvastatin has a pleiotropic effect as anti-inflammatory through one of the target levels of High Mobility Group Box-1 (HMGB-1). This prospective observational study aimed to analyze the effect of atorvastatin on serum HMGB-1 levels in CAD. METHODS Samples were collected from prospective observation pre-post study in May-July 2018 with consecutive sampling method. Serum HMGB-1 levels were measured in patients with CAD who were given atorvastatin for CAD with type-2 diabetes mellitus compared without type-2 diabetes mellitus in a patient ward. Blood was collected on admission day and before the patient left the hospital. After centrifugation, serum samples were stored at -80 °C before measurement. We used an ELISA kit (IBL International) to determine HMGB-1 concentrations. This research protocol has been approved by the Ethical Committee of Dr. Soetomo General Hospital, Surabaya. RESULTS We enrolled 38 patients and divided them into two groups which 19 patients on CAD with type-2 diabetes mellitus and 19 patients without diabetes mellitus. Serum HMGB-1 levels in CAD with type-2 diabetes mellitus were increased significantly (p = 0.049) and not significantly decreased in CAD without type-2 diabetes mellitus (p = 0.480). The HMGB-1 level was not significantly different between the two groups (p = 0.210). CONCLUSIONS HMGB-1 levels after providing atorvastatin in CAD with type-2 diabetes mellitus increased significantly, meanwhile, in CAD without type-2 diabetes mellitus did not decrease significantly. The HMGB-1 level was not significantly different between the two groups. Longer time and more point for the collected sample needed for further research.
Collapse
Affiliation(s)
| | - Suharjono
- Department of Clinical Pharmacy, Faculty of Pharmacy,Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Mohammad Yogiarto
- Cardiovascular, Department of Cardiology, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| |
Collapse
|
19
|
Biscetti F, Tinelli G, Rando MM, Nardella E, Cecchini AL, Angelini F, Straface G, Filipponi M, Arena V, Pitocco D, Gasbarrini A, Massetti M, Flex A. Association between carotid plaque vulnerability and high mobility group box-1 serum levels in a diabetic population. Cardiovasc Diabetol 2021; 20:114. [PMID: 34044825 PMCID: PMC8161555 DOI: 10.1186/s12933-021-01304-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/19/2021] [Indexed: 12/21/2022] Open
Abstract
Background Carotid atherosclerosis represents one of the complications of diabetes mellitus. In particular, plaque instability contributes to disease progression and stroke incidence. High mobility group box-1 (HMGB1) is a nuclear protein involved in promotion and progression of atherosclerosis and cardiovascular diseases. The aim of this study was to analyze the relationship between HMGB1 serum levels, main inflammatory cytokines, the presence of internal carotid stenosis and unstable plaque in a diabetic population. Research design and methods We studied 873 diabetic patients, including 347 patients with internal carotid artery stenosis (ICAS) who underwent carotid endarterectomy and 526 diabetic patients without internal carotid artery stenosis (WICAS). At baseline, HMGB1 and the main inflammatory cytokines serum levels were evaluated. For ICAS patients, the histological features of carotid plaque were also collected to differentiate them in patients with stable or unstable atherosclerotic lesions. Results We found that HMGB1 serum levels, osteoprotegerin, high-sensitivity C-reactive protein, tumor necrosis factor-alpha and interleukin-6, were significantly higher in diabetic ICAS patients compared to diabetic WICAS patients. Among ICAS patients, individuals with unstable plaque had higher levels of these cytokines, compared to patients with stable plaque. A multivariable stepwise logistic regression analysis showed that HMGB1 and osteoprotegerin remained independently associated with unstable plaque in ICAS patients. Conclusions The present study demonstrated that HMGB1 is an independent risk factor for carotid plaque vulnerability in an Italian population with diabetes mellitus, representing a promising biomarker of carotid plaque instability and a possible molecular target to treat unstable carotid plaques and to prevent stroke.
Collapse
Affiliation(s)
- Federico Biscetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy. .,Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University School of Medicine, Largo Francesco Vito, 1, 00168, Roma, Italy. .,Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, Roma, Italy.
| | - Giovanni Tinelli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Vascular Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Università Cattolica del Sacro Cuore, Roma, Italy
| | - Maria Margherita Rando
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University School of Medicine, Largo Francesco Vito, 1, 00168, Roma, Italy
| | - Elisabetta Nardella
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, Roma, Italy
| | | | - Flavia Angelini
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Giuseppe Straface
- Department of Internal Medicine, St. M. Goretti Hospital, Roma, Italy
| | | | - Vincenzo Arena
- Department of Pathology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Dario Pitocco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Università Cattolica del Sacro Cuore, Roma, Italy.,Diabetology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Antonio Gasbarrini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Università Cattolica del Sacro Cuore, Roma, Italy.,Department of Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Massimo Massetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Università Cattolica del Sacro Cuore, Roma, Italy.,Cardiovascular Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Andrea Flex
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University School of Medicine, Largo Francesco Vito, 1, 00168, Roma, Italy.,Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, Roma, Italy.,Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
20
|
High-mobility group box 1 serves as an inflammation driver of cardiovascular disease. Biomed Pharmacother 2021; 139:111555. [PMID: 33865014 DOI: 10.1016/j.biopha.2021.111555] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 03/15/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is the most deadly disease, which can cause sudden death, in which inflammation is a key factor in its occurrence and development. High-mobility group box 1 (HMGB1) is a novel nuclear DNA-binding protein that activates innate immunity to induce inflammation in CVD. HMGB1 exists in the cytoplasm and nucleus of different cell types, including those in the heart. By binding to its receptors, HMGB1 triggers a variety of signaling cascades, leading to inflammation and CVD. To help develop HMGB1-targeted therapies, here we discuss HMGB1 and its biological functions, receptors, signaling pathways, and pathophysiology related to inflammation and CVD, including cardiac remodeling, cardiac hypertrophy, myocardial infarction, heart failure, pulmonary hypertension, atherosclerosis, and cardiomyopathy.
Collapse
|
21
|
Kumar V. The Trinity of cGAS, TLR9, and ALRs Guardians of the Cellular Galaxy Against Host-Derived Self-DNA. Front Immunol 2021; 11:624597. [PMID: 33643304 PMCID: PMC7905024 DOI: 10.3389/fimmu.2020.624597] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
The immune system has evolved to protect the host from the pathogens and allergens surrounding their environment. The immune system develops in such a way to recognize self and non-self and develops self-tolerance against self-proteins, nucleic acids, and other larger molecules. However, the broken immunological self-tolerance leads to the development of autoimmune or autoinflammatory diseases. Pattern-recognition receptors (PRRs) are expressed by immunological cells on their cell membrane and in the cytosol. Different Toll-like receptors (TLRs), Nod-like receptors (NLRs) and absent in melanoma-2 (AIM-2)-like receptors (ALRs) forming inflammasomes in the cytosol, RIG (retinoic acid-inducible gene)-1-like receptors (RLRs), and C-type lectin receptors (CLRs) are some of the PRRs. The DNA-sensing receptor cyclic GMP–AMP synthase (cGAS) is another PRR present in the cytosol and the nucleus. The present review describes the role of ALRs (AIM2), TLR9, and cGAS in recognizing the host cell DNA as a potent damage/danger-associated molecular pattern (DAMP), which moves out to the cytosol from its housing organelles (nucleus and mitochondria). The introduction opens with the concept that the immune system has evolved to recognize pathogens, the idea of horror autotoxicus, and its failure due to the emergence of autoimmune diseases (ADs), and the discovery of PRRs revolutionizing immunology. The second section describes the cGAS-STING signaling pathway mediated cytosolic self-DNA recognition, its evolution, characteristics of self-DNAs activating it, and its role in different inflammatory conditions. The third section describes the role of TLR9 in recognizing self-DNA in the endolysosomes during infections depending on the self-DNA characteristics and various inflammatory diseases. The fourth section discusses about AIM2 (an ALR), which also binds cytosolic self-DNA (with 80–300 base pairs or bp) that inhibits cGAS-STING-dependent type 1 IFN generation but induces inflammation and pyroptosis during different inflammatory conditions. Hence, this trinity of PRRs has evolved to recognize self-DNA as a potential DAMP and comes into action to guard the cellular galaxy. However, their dysregulation proves dangerous to the host and leads to several inflammatory conditions, including sterile-inflammatory conditions autoinflammatory and ADs.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, St. Lucia, Brisbane, QLD, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Brisbane, QLD, Australia
| |
Collapse
|
22
|
Testa C, DI Lorenzo A, Parlato A, D'Ambrosio G, Merolla A, Pacileo M, Iannuzzo G, Gentile M, Nugara C, Sarullo FM, DE Gregorio C, D'Andrea A, Vigorito C, Venturini E, Giallauria F. Exercise for slowing the progression of atherosclerotic process: effects on inflammatory markers. Panminerva Med 2021; 63:122-132. [PMID: 33565757 DOI: 10.23736/s0031-0808.21.04266-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Atherosclerosis is a dynamic process driven by all cardiovascular risk factors that can be briefly divided into an early and a late phase. Inflammation is one of the fundamental substrates that initiates the atherosclerotic process in the early stages and promotes and maintains it in the final stages. In the last decades, clinical and experimental data have shown that inflammation is supported by mediators that respond to physical activity. The present review aimed at investigating the effect of physical exercise on inflammatory mediators, both the positive ones that have a proinflammatory effect (interleukin 6, c-reactive protein and tumor necrosis factor α, interferon γ, high-mobility group box-1), and the negative ones which have an anti-inflammatory effect (interleukin 10). Pooled data support the evidence that physical exercise can directly modulate the activity of inflammatory cytokines slowing down or preventing the formation of the atherosclerotic stage.
Collapse
Affiliation(s)
- Crescenzo Testa
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Anna DI Lorenzo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Alessandro Parlato
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Giuseppe D'Ambrosio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Aurora Merolla
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Mario Pacileo
- Unit of Cardiology and Intensive Care, "Umberto I" Hospital, Nocera Inferiore, Salerno, Italy
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Marco Gentile
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Cinzia Nugara
- Unit of Cardiovascular Rehabilitation, Buccheri La Ferla Fatebenefratelli Hospital, Palermo, Italy
| | - Filippo M Sarullo
- Unit of Cardiovascular Rehabilitation, Buccheri La Ferla Fatebenefratelli Hospital, Palermo, Italy
| | - Cesare DE Gregorio
- Unit of Cardiology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy.,Post-graduate Residency School in Cardiovascular Diseases, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Antonello D'Andrea
- Unit of Cardiology and Intensive Care, "Umberto I" Hospital, Nocera Inferiore, Salerno, Italy
| | - Carlo Vigorito
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Elio Venturini
- Cardiac Rehabilitation Unit, AUSL Toscana Nord-Ovest, Cecina Civil Hospital, Cecina, Livorno, Italy
| | - Francesco Giallauria
- Department of Translational Medical Sciences, Federico II University, Naples, Italy - .,Faculty of Sciences and Technology, University of New England, Armidale, Australia
| |
Collapse
|
23
|
Yuan X, Bhat OM, Lohner H, Zhang Y, Li PL. Downregulation of Lysosomal Acid Ceramidase Mediates HMGB1-Induced Migration and Proliferation of Mouse Coronary Arterial Myocytes. Front Cell Dev Biol 2020; 8:111. [PMID: 32211403 PMCID: PMC7076051 DOI: 10.3389/fcell.2020.00111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/10/2020] [Indexed: 01/07/2023] Open
Abstract
High-mobility group box 1 protein (HMGB1) has been reported to trigger lysosome destabilization causing a wide of inflammatory diseases. The present study tested whether a lysosomal enzyme, acid ceramidase (AC), plays a critical role in HMGB1-induced alteration in ceramide metabolism and whether such HMGB1-AC interaction is associated with abnormal migration and proliferation of vascular smooth muscle cells (SMCs). We first observed that the expression of AC in the medial layer of mouse coronary arterial wall and colocalization of AC with a lysosome marker Lamp-1. In primary cultured coronary arterial myocytes (CAMs), AC expression and colocalization with Lamp-1 were significantly up-regulated by AC inducer, genistein, but down-regulated by AC inhibitor, N-oleoylethanolamine (NOE). HMGB1 dose-dependently decreased the colocalization of AC with Lamp-1 and reduced mRNA and protein expressions of AC in CAMs, but reversed by genistein. Consistently, HMGB1 significantly induced increases in the levels of long-chain ceramides in CAMs, which were not further enhanced by NOE but blocked by genistein. More importantly, HMGB1 promoted migration and proliferation of CAMs, which were not further increased by NOE but reduced by genistein. Lastly, CAMs isolated from smooth muscle-specific AC knockout mice (AC gene Asah1) exhibited increased ceramide levels and enhanced the migration and proliferation, which resembles the effects of HMGB1 on wild-type CAMs. Together, these results suggest that HMGB1 promotes SMC migration and proliferation via inhibition of AC expression and ceramide accumulation.
Collapse
Affiliation(s)
- Xinxu Yuan
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Owais M. Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Hannah Lohner
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
24
|
Biscetti F, Rando MM, Nardella E, Cecchini AL, Pecorini G, Landolfi R, Flex A. High Mobility Group Box-1 and Diabetes Mellitus Complications: State of the Art and Future Perspectives. Int J Mol Sci 2019; 20:ijms20246258. [PMID: 31835864 PMCID: PMC6940913 DOI: 10.3390/ijms20246258] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 12/17/2022] Open
Abstract
Diabetes mellitus (DM) is an endemic disease, with growing health and social costs. The complications of diabetes can affect potentially all parts of the human body, from the heart to the kidneys, peripheral and central nervous system, and the vascular bed. Although many mechanisms have been studied, not all players responsible for these complications have been defined yet. High Mobility Group Box-1 (HMGB1) is a non-histone nuclear protein that has been implicated in many pathological processes, from sepsis to ischemia. The purpose of this review is to take stock of all the most recent data available on the role of HMGB1 in the complications of DM.
Collapse
Affiliation(s)
- Federico Biscetti
- U.O.C. Clinica Medica e Malattie Vascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (G.P.); (R.L.); (A.F.)
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Correspondence: ; Tel.: +39-06-3015-4335; Fax: +39-06-3550-7232
| | | | - Elisabetta Nardella
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.M.R.); (E.N.); (A.L.C.)
| | | | - Giovanni Pecorini
- U.O.C. Clinica Medica e Malattie Vascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (G.P.); (R.L.); (A.F.)
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.M.R.); (E.N.); (A.L.C.)
| | - Raffaele Landolfi
- U.O.C. Clinica Medica e Malattie Vascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (G.P.); (R.L.); (A.F.)
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.M.R.); (E.N.); (A.L.C.)
| | - Andrea Flex
- U.O.C. Clinica Medica e Malattie Vascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (G.P.); (R.L.); (A.F.)
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.M.R.); (E.N.); (A.L.C.)
| |
Collapse
|
25
|
Gao W, Cui H, Li Q, Zhong H, Yu J, Li P, He X. Upregulation of microRNA-218 reduces cardiac microvascular endothelial cells injury induced by coronary artery disease through the inhibition of HMGB1. J Cell Physiol 2019; 235:3079-3095. [PMID: 31566720 DOI: 10.1002/jcp.29214] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 08/23/2019] [Indexed: 12/19/2022]
Abstract
This study is performed to examine the impacts of microRNA-218 (miR-218) on cardiac microvascular endothelial cells (CMECs) injury induced by coronary artery disease (CAD). Reverse-transcription quantitative polymerase chain reaction (RT-qPCR) was applied for detecting miR-218 expression in serum of patients with CAD and healthy controls, and the correlation between miR-218 expression and the clinical indexes such as creatine kinase, creatine kinase-myocardial band, cardiac troponin I, and coronary Gensini score was analyzed. CMECs were coincubated with homocysteine for 24 hr for CMECs injury, and the cells were transfected with miR-218 mimics or miR-218 inhibitors. Besides, we used oxidized low density lipoprotein as an inducer to incubate with CMECs for 24 hr, and the model of CMECs injury was established to be transfected with miR-218 mimics. RT-qPCR and western blot analysis were used to detect miR-218 and HMGB1 expression in CMECs. A series of experiments were used to determine cell proliferation, apoptosis, migration, and angiogenesis ability of CMECs. Vascular endothelial growth factor expression and inflammatory factor contents were measured. The obtained results suggested that miR-218 expression in peripheral blood of patients with CAD descended substantially versus that of healthy controls. Low miR-218 expression was found in CAD-induced CMECs injury. Overexpressed miR-218 promoted the proliferation, migration, angiogenesis ability, induced apoptosis, and alleviated the inflammatory injury of CAD-induced CMECs. miR-218 may negatively regulate the expression of HMGB1 in CAD. This study demonstrates that upregulation of miR-218 reduces CMECs injury induced by CAD through the inhibition of HMGB1.
Collapse
Affiliation(s)
- Wenhui Gao
- Department of Cardiovascular, Hangzhouwan Hospital, Ningbo, Zhejiang Province, China
| | - Hanbin Cui
- Department of Cardiovascular, No. 1 Hospital, Ningbo, Zhejiang Province, China
| | - Qianjun Li
- Department of Respiratory, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| | - Hai Zhong
- Department of Thoracic Surgery, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| | - Jingjing Yu
- Department of Pathology, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| | - Ping Li
- Department of Anesthesiology, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| | - Xijie He
- Department of Cardiology, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| |
Collapse
|
26
|
Chen Z, Pan X, Sheng Z, Yan G, Chen L, Ma G. Baicalin Suppresses the Proliferation and Migration of Ox-LDL-VSMCs in Atherosclerosis through Upregulating miR-126-5p. Biol Pharm Bull 2019; 42:1517-1523. [DOI: 10.1248/bpb.b19-00196] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Zhongpu Chen
- The Department of Cardiology, Zhongda Hospital, Southeast University
| | - Xiaodong Pan
- The Department of Cardiology, Zhongda Hospital, Southeast University
| | - Zulong Sheng
- The Department of Cardiology, Zhongda Hospital, Southeast University
| | - Gaoliang Yan
- The Department of Cardiology, Zhongda Hospital, Southeast University
| | - Long Chen
- The Department of Cardiology, Zhongda Hospital, Southeast University
| | - Genshan Ma
- The Department of Cardiology, Zhongda Hospital, Southeast University
| |
Collapse
|
27
|
Increased plasma levels of high mobility group box 1 protein in patients with bipolar disorder: A pilot study. J Neuroimmunol 2019; 334:576993. [DOI: 10.1016/j.jneuroim.2019.576993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/21/2022]
|
28
|
High Mobility Group Box 1 Mediates TMAO-Induced Endothelial Dysfunction. Int J Mol Sci 2019; 20:ijms20143570. [PMID: 31336567 PMCID: PMC6678463 DOI: 10.3390/ijms20143570] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/22/2022] Open
Abstract
The intestinal microbe-derived metabolite trimethylamine N-oxide (TMAO) is implicated in the pathogenesis of cardiovascular diseases (CVDs). The molecular mechanisms of how TMAO induces atherosclerosis and CVDs’ progression are still unclear. In this regard, high-mobility group box protein 1 (HMGB1), an inflammatory mediator, has been reported to disrupt cell–cell junctions, resulting in vascular endothelial hyper permeability leading to endothelial dysfunction. The present study tested whether TMAO associated endothelial dysfunction results via HMGB1 activation. Biochemical and RT-PCR analysis showed that TMAO increased the HMGB1 expression in a dose-dependent manner in endothelial cells. However, prior treatment with glycyrrhizin, an HMGB1 binder, abolished the TMAO-induced HMGB1 production in endothelial cells. Furthermore, Western blot and immunofluorescent analysis showed significant decrease in the expression of cell–cell junction proteins ZO-2, Occludin, and VE-cadherin in TMAO treated endothelial cells compared with control cells. However, prior treatment with glycyrrhizin attenuated the TMAO-induced cell–cell junction proteins’ disruption. TMAO increased toll-like receptor 4 (TLR4) expression in endothelial cells. Inhibition of TLR4 expression by TLR4 siRNA protected the endothelial cells from TMAO associated tight junction protein disruption via HMGB1. In conclusion, our results demonstrate that HMGB1 is one of the important mediators of TMAO-induced endothelial dysfunction.
Collapse
|
29
|
Fischer A, Bockstahler M, Müller AM, Stroikova V, Leib C, Pfitzer G, Katus HA, Kaya Z. FN14 Signaling Plays a Pathogenic Role in a Mouse Model of Experimental Autoimmune Myocarditis. J Card Fail 2019; 25:674-685. [PMID: 31212034 DOI: 10.1016/j.cardfail.2019.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/26/2019] [Accepted: 06/11/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND The pathogenesis of inflammatory cardiomyopathy is affected by the activation of autoimmune-mediated cascades. To study these cascades, we developed an experimental model of troponin I (TnI)-induced autoimmune myocarditis (EAM). One factor playing a pivotal role in the context of autoimmune disorders is the receptor fibroblast growth factor-inducible 14 (FN14). Thus, the impact of FN14 in the development of autoimmune myocarditis was investigated. METHODS AND RESULTS TnI-immunization led to a significantly increased myocardial FN14 mRNA and protein expression in wild-type (wt) mice. To investigate the precise role of FN14 in EAM, FN14 knockout (ko) and wt littermates were immunized with TnI or control buffer. The animals were evaluated for cardiac parameters and indicators of myocardial injury. FN14 deficiency resulted in better cardiac performance, less myocardial inflammation, fibrosis, and cardiac damage. A lower myocardial mRNA expression of inflammatory cytokines and chemokines as well as their receptors could be demonstrated in TnI-immunized FN14ko compared to wt mice also immunized with TnI. Western blot analysis revealed a contribution of nuclear factor kappa-light-chain-enhancer of activated B cells to FN14-induced signaling cascades. CONCLUSIONS In the pathogenesis of autoimmune myocarditis, the inflammatory response to cardiac injury is attenuated in FN14ko mice. Thus, inhibition of FN14 in patients might represent a novel therapeutic strategy in the treatment of inflammatory cardiomyopathy.
Collapse
Affiliation(s)
- Andrea Fischer
- Department of Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Mariella Bockstahler
- Department of Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; Cardiology, St. Claraspital, 4058 Basel, Switzerland
| | - Anna-Maria Müller
- Department of Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Vera Stroikova
- Department of Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Christoph Leib
- Department of Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; Cardiology, St. Claraspital, 4058 Basel, Switzerland
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Hugo A Katus
- Department of Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ziya Kaya
- Department of Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
30
|
Stroikova V, Fischer A, Bockstahler M, Müller AM, Katus HA, Kaya Z. Adiponectin deficiency has no effect in murine autoimmune myocarditis. Cytokine 2019; 116:139-149. [PMID: 30716658 DOI: 10.1016/j.cyto.2018.12.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/25/2018] [Accepted: 12/27/2018] [Indexed: 01/21/2023]
Abstract
BACKGROUND Adiponectin is a hormone that together with its receptors modulates a number of metabolic processes including gluconeogenesis and lipid catabolism. It belongs to the C1QTNF (complement C1q tumor necrosis factor-related protein) family, which has a variety of members with high amino acid sequence homology and overlapping functions. Concentration of adiponectin in blood is inversely correlated with body fat percentage and cardiac risk factors like blood pressure and CRP (C-reactive protein) level. Studies have identified the existence of a cardiac adiponectin system. However, little is known about the role of this system in the pathogenesis of autoimmune myocarditis. Thus, we have studied the involvement of adiponectin in the development of this autoimmune disorder in a mouse model of experimental autoimmune myocarditis (EAM). METHODS Adiponectin knockout (ko) and wild type (wt) mice were immunized with cardiac troponin I (cTnI) to induce an EAM. To determine the severity of myocardial damage, inflammation and fibrosis were scored after HE and Afog staining and high sensitivity troponin T (hsTnT) level was measured. To detect if changes in specific inflammatory cell numbers could be observed between the genotypes, we performed immunohistochemical staining to detect T lymphocytes, B lymphocytes and macrophages. The level of the humoral immune response was determined through the measurement of cTnI-specific serum IgG autoantibodies. Relative mRNA expression of different cytokines, C1QTNF family members and adiponectin receptors in the heart tissue was analyzed with qPCR. RESULTS Animals immunized with cTnI developed autoimmune myocarditis with a significant deterioration of cardiac parameters compared to the corresponding control group. The adiponectin ko group immunized with cTnI showed a tendency towards increased inflammation, fibrosis, heart-to-body-weight ratio, infiltration pattern of T lymphocytes, B lymphocytes and macrophages, hsTnT concentration, humoral immune response and mRNA expression of interleukin 6 in the heart tissue and decreased weight gain compared to the wt group immunized with cTnI. However, the difference to the wt group treated with cTnI was not significant. The analysis of cardiac mRNA expression of adiponectin receptors and four C1QTNF family members, most suitable for fulfilling the functions of adiponectin in adiponectin ko mice, did not show any significant differences between adiponectin ko and wt group at all. CONCLUSION Our study reveals that the absence of adiponectin did not lead to a significantly increased impairment of cardiac function and was also unlikely to be compensated by its receptors or other C1QTNF family members in the murine model of EAM. Here, other synergistic or redundant effects might play a role and must be investigated in further studies to understand the role and function of adiponectin in autoimmune myocarditis.
Collapse
Affiliation(s)
- Vera Stroikova
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Andrea Fischer
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | | | - Anna-Maria Müller
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Ziya Kaya
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
31
|
Ding JW, Luo CY, Wang XA, Zhou T, Zheng XX, Zhang ZQ, Yu B, Zhang J, Tong XH. Glycyrrhizin, a High-Mobility Group Box 1 Inhibitor, Improves Lipid Metabolism and Suppresses Vascular Inflammation in Apolipoprotein E Knockout Mice. J Vasc Res 2019; 55:365-377. [DOI: 10.1159/000495310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/09/2018] [Indexed: 11/19/2022] Open
|
32
|
Meyer IS, Goetzke CC, Kespohl M, Sauter M, Heuser A, Eckstein V, Vornlocher HP, Anderson DG, Haas J, Meder B, Katus HA, Klingel K, Beling A, Leuschner F. Silencing the CSF-1 Axis Using Nanoparticle Encapsulated siRNA Mitigates Viral and Autoimmune Myocarditis. Front Immunol 2018; 9:2303. [PMID: 30349538 PMCID: PMC6186826 DOI: 10.3389/fimmu.2018.02303] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022] Open
Abstract
Myocarditis is an inflammatory disease of the heart muscle most commonly caused by viral infection and often maintained by autoimmunity. Virus-induced tissue damage triggers chemokine production and, subsequently, immune cell infiltration with pro-inflammatory and pro-fibrotic cytokine production follows. In patients, the overall inflammatory burden determines the disease outcome. Following the aim to define specific molecules that drive both immunopathology and/or autoimmunity in inflammatory heart disease, here we report on increased expression of colony stimulating factor 1 (CSF-1) in patients with myocarditis. CSF-1 controls monocytes originating from hematopoietic stem cells and subsequent progenitor stages. Both, monocytes and macrophages are centrally involved in mediating tissue damage and fibrotic scarring in the heart. CSF-1 influences monocytes via engagement of CSF-1 receptor, and it is also produced by cells of the mononuclear phagocyte system themselves. Based on this, we sought to modulate the virus-triggered inflammatory response in an experimental model of Coxsackievirus B3-induced myocarditis by silencing the CSF-1 axis in myeloid cells using nanoparticle-encapsulated siRNA. siCSF-1 inverted virus-mediated immunopathology as reflected by lower troponin T levels, a reduction of accumulating myeloid cells in heart tissue and improved cardiac function. Importantly, pathogen control was maintained and the virus was efficiently cleared from heart tissue. Since viral heart disease triggers heart-directed autoimmunity, in a second approach we investigated the influence of CSF-1 upon manifestation of heart tissue inflammation during experimental autoimmune myocarditis (EAM). EAM was induced in Balb/c mice by immunization with a myocarditogenic myosin-heavy chain-derived peptide dissolved in complete Freund's adjuvant. siCSF-1 treatment initiated upon established disease inhibited monocyte infiltration into heart tissue and this suppressed cardiac injury as reflected by diminished cardiac fibrosis and improved cardiac function at later states. Mechanistically, we found that suppression of CSF-1 production arrested both differentiation and maturation of monocytes and their precursors in the bone marrow. In conclusion, during viral and autoimmune myocarditis silencing of the myeloid CSF-1 axis by nanoparticle-encapsulated siRNA is beneficial for preventing inflammatory tissue damage in the heart and preserving cardiac function without compromising innate immunity's critical defense mechanisms.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Carl Christoph Goetzke
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Meike Kespohl
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Martina Sauter
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Arnd Heuser
- Max-Delbrueck-Center for Molecular Medicine Berlin, Berlin, Germany
| | - Volker Eckstein
- Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States.,Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, United States
| | - Jan Haas
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Benjamin Meder
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Hugo Albert Katus
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Antje Beling
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Florian Leuschner
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Heidelberg, Germany
| |
Collapse
|
33
|
Zhang W, Zhang Y, Ding K, Zhang H, Zhao Q, Liu Z, Xu Y. Involvement of JNK1/2-NF-κBp65 in the regulation of HMGB2 in myocardial ischemia/reperfusion-induced apoptosis in human AC16 cardiomyocytes. Biomed Pharmacother 2018; 106:1063-1071. [DOI: 10.1016/j.biopha.2018.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 06/29/2018] [Accepted: 07/01/2018] [Indexed: 12/20/2022] Open
|
34
|
Ding JW, Zhou T, Zheng XX, Wang XA, Tong XH, Luo CY, Zhang ZQ, Yu B. The Effects of High Mobility Group Box-1 Protein on Peripheral Treg/Th17 Balance in Patients with Atherosclerosis. ACTA CARDIOLOGICA SINICA 2018; 34:399-408. [PMID: 30271090 PMCID: PMC6160517 DOI: 10.6515/acs.201809_34(5).20180419a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 04/19/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Atherosclerosis (AS) is defined as chronic inflammation of the vessel wall. The major objective of the this study was to explore the mechanism of Treg/Th17 imbalance and the role of high mobility group box-1 protein (HMGB1) on the balance in AS. METHODS We detected the apoptotic ratios of Treg and Th17 cells in peripheral blood mononuclear cells (PBMCs) from subjects with AS and normal coronary arteries (NCA) by flow cytometry. The effects of recombinant HMGB1 (rHMGB1) on the proportion, apoptosis and differentiation of Treg and Th17 cells were analyzed using flow cytometry, qRT-PCR and ELISA. RESULTS The frequencies of apoptotic Treg cells in the PBMCs from the subjects with AS were significantly higher than in those with NCA (p < 0.01). Stimulation of rHMGB1 obviously increased the level of Th17 cells and acid- related orphan receptor C (RORC) mRNA, and markedly decreased Treg cell frequency and the mRNA expression of factor forkhead family protein 3 (Foxp3) in the PBMCs. rHMGB1 played an obvious role in elevating Treg cell apoptosis ratio (p < 0.01). rHMGB1 treatment significantly decreased Treg cell ratio and IL-10 level, and increased Th17 cell ratio and IL-17A level induced from naïve CD4+ T cells. CONCLUSIONS HMGB1 may modulate Treg/Th17 balance in patients with AS through inducing Treg cell apoptosis and promoting cell differentiation of Th17.
Collapse
Affiliation(s)
- Jia-Wang Ding
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Tian Zhou
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Xia-Xia Zheng
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Xin-An Wang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Xiao-Hong Tong
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Cai-Yun Luo
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Zai-Qiang Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Bin Yu
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| |
Collapse
|
35
|
Aoyagi H, Yamashiro K, Hirata‐Yoshihara C, Ideguchi H, Yamasaki M, Kawamura M, Yamamoto T, Kochi S, Wake H, Nishibori M, Takashiba S. HMGB1‐induced inflammatory response promotes bone healing in murine tooth extraction socket. J Cell Biochem 2018; 119:5481-5490. [DOI: 10.1002/jcb.26710] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/23/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Hiroaki Aoyagi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Keisuke Yamashiro
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Chiaki Hirata‐Yoshihara
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hidetaka Ideguchi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Mutsuyo Yamasaki
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Mari Kawamura
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Tadashi Yamamoto
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Shinsuke Kochi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hidenori Wake
- Department of PharmacologyOkayama University Graduate School of MedicineDentistry and Pharmacological SciencesOkayamaJapan
| | - Masahiro Nishibori
- Department of PharmacologyOkayama University Graduate School of MedicineDentistry and Pharmacological SciencesOkayamaJapan
| | - Shogo Takashiba
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| |
Collapse
|
36
|
Human interstitial cellular model in therapeutics of heart valve calcification. Amino Acids 2017; 49:1981-1997. [DOI: 10.1007/s00726-017-2432-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/27/2017] [Indexed: 12/27/2022]
|
37
|
Basso D, Padoan A, Laufer T, Aneloni V, Moz S, Schroers H, Pelloso M, Saiz A, Krapp M, Fogar P, Cornoldi P, Zambon CF, Rossi E, La Malfa M, Marotti A, Brefort T, Weis TM, Katus HA, Plebani M. Relevance of pre-analytical blood management on the emerging cardiovascular protein biomarkers TWEAK and HMGB1 and on miRNA serum and plasma profiling. Clin Biochem 2016; 50:186-193. [PMID: 27847340 DOI: 10.1016/j.clinbiochem.2016.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND Disease-independent sources of biomarker variability include pre-analytical, analytical and biological variance. The aim of the present study was to evaluate whether the pre-analytical phase has any impact on the emerging heart disease TWEAK and HMGB1 protein markers and miRNA biomarkers, and whether peptidome profiling allows the identification of pre-analytical quality markers. METHODS An assessment was made of sample type (serum, EDTA-Plasma, Citrate-Plasma, ACD-plasma, Heparin-plasma), temperature of sample storage (room temperature or refrigerated), time of sample storage (0.5, 3, 6 and 9h) and centrifugation (one or two-step). Aliquots of all processed samples were immediately frozen (-80°C) before analysis. Proteins were assayed by ELISAs, miRNA expression profile by microarray and peptidome profiling by MALDI-TOF/MS. RESULTS Temperature, time and centrifugation had no impact on TWEAK and HMGB1 results, which were significantly influenced by matrix type, TWEAK levels being significantly higher (F=194.7, p<0.0001), and HMGB1 levels significantly lower (F=36.32, p<0.0001) in serum than in any other plasma type. Unsuitable miRNA results were obtained using Heparin-plasma. Serum miRNA expression profiles depended mainly on temperature, while EDTA-plasma miRNA expression profiles were strongly affected by the centrifugation method used. MALDI-TOF/MS allowed the identification of seven features as indices of pre-analytical serum (m/z at 1206, 1350, 1865 and 2021) or EDTA-plasma (m/z 1897, 2740 and 2917) degradation. CONCLUSIONS Serum and EDTA-plasma allow the analysis of both proteins and miRNA emerging biomarkers of heart diseases. Refrigerated storage prevents an altered miRNA expression profile also in cases of a prolonged time-interval between blood drawing and processing.
Collapse
Affiliation(s)
- Daniela Basso
- Department of Medicine - DIMED, University of Padova, Italy.
| | - Andrea Padoan
- Department of Medicine - DIMED, University of Padova, Italy
| | - Thomas Laufer
- Comprehensive Biomarker Center GmbH, Heidelberg, Germany
| | | | - Stefania Moz
- Department of Medicine - DIMED, University of Padova, Italy
| | | | | | - Anna Saiz
- Comprehensive Biomarker Center GmbH, Heidelberg, Germany
| | - Medea Krapp
- Comprehensive Biomarker Center GmbH, Heidelberg, Germany
| | - Paola Fogar
- Department of Medicine - DIMED, University of Padova, Italy
| | - Paola Cornoldi
- Department of Medicine - DIMED, University of Padova, Italy
| | | | - Elisa Rossi
- Department of Medicine - DIMED, University of Padova, Italy
| | - Marco La Malfa
- Department of Medicine - DIMED, University of Padova, Italy
| | - Alberto Marotti
- UOC Immunotrasfusionale, University-Hospital of Padova, Italy
| | - Thomas Brefort
- Comprehensive Biomarker Center GmbH, Heidelberg, Germany; Eurofins Medigenomix GmbH, Ebersberg, Germany
| | - Tanja M Weis
- Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mario Plebani
- Department of Medicine - DIMED, University of Padova, Italy
| |
Collapse
|
38
|
Jin P, Zhou Q, Song S, Xu J, Zhang M, Zhu M, Kang M, Shi X, Shi J, Lu D, Li J. Elevated preoperative HMGB1 as predictor of myocardial injury post-percutaneous coronary intervention. Medicine (Baltimore) 2016; 95:e5149. [PMID: 27861339 PMCID: PMC5120896 DOI: 10.1097/md.0000000000005149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In this study, we evaluated the impact of preoperative high mobility group box 1 (HMGB1) on myocardial injury post-percutaneous coronary intervention.We evaluated 302 consecutive patients who underwent percutaneous coronary intervention. They were divided into equal tertiles based on their preoperative HMGB1 levels. Creatine kinase-MB and troponin I levels were measured at baseline, 8- and 24-hours after the procedure, while clinical outcomes were followed up for 1 year.The occurrence of post-procedural myocardial injury was significantly higher in the tertile comprising of patients with elevated HMGB1 levels. Moreover, these patients showed significantly higher post-procedural peak values of creatine kinase-MB and troponin I in comparison to patients with lower HMGB1 levels. Event-free survival was significantly associated with HMGB1 levels, with worst event-free survival in patients with elevated HMGB1 levels.Elevated preoperative HMGB1 was a predictor of myocardial injury after percutaneous coronary intervention, and was associated with the worst clinical outcomes during 1-year follow up.
Collapse
Affiliation(s)
- Peng Jin
- Department of Cardiology, China National Petroleum Corporation Central Hospital, Langfang, Hebei, China
| | - Qi Zhou
- Department of Cardiology, China National Petroleum Corporation Central Hospital, Langfang, Hebei, China
| | - Shujiang Song
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Jinpeng Xu
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Minli Zhang
- Department of Cardiology, China National Petroleum Corporation Central Hospital, Langfang, Hebei, China
| | - Ming Zhu
- Department of Cardiology, China National Petroleum Corporation Central Hospital, Langfang, Hebei, China
| | - Meili Kang
- Department of Cardiology, China National Petroleum Corporation Central Hospital, Langfang, Hebei, China
| | - Xiangming Shi
- Department of Cardiology, China National Petroleum Corporation Central Hospital, Langfang, Hebei, China
| | - Junting Shi
- Department of Cardiology, China National Petroleum Corporation Central Hospital, Langfang, Hebei, China
| | - Di Lu
- Department of Cardiology, China National Petroleum Corporation Central Hospital, Langfang, Hebei, China
| | - Jing Li
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| |
Collapse
|
39
|
High Mobility Group Box-1: A Missing Link between Diabetes and Its Complications. Mediators Inflamm 2016; 2016:3896147. [PMID: 27847406 PMCID: PMC5099456 DOI: 10.1155/2016/3896147] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/03/2016] [Indexed: 12/13/2022] Open
Abstract
High mobility group box-1 (HMGB-1), a damage-associated molecular pattern, can be actively or passively released from various cells under different conditions and plays a pivotal role in the pathogenesis of inflammation and angiogenesis-dependent diseases. More and more evidence suggests that inflammation, in addition to its role in progression of diabetes, also promotes initiation and development of diabetic complications. In this review, we focus on the role of HMGB-1 in diabetes-related complications and the therapeutic strategies targeting HMGB-1 in diabetic complications.
Collapse
|
40
|
Poppi NT, Gowdak LHW, Dourado LOC, Adam EL, Leite TNP, Mioto BM, Krieger JE, César LAM, Pereira AC. A prospective study of patients with refractory angina: outcomes and the role of high-sensitivity troponin T. Clin Cardiol 2016; 40:11-17. [PMID: 27754552 DOI: 10.1002/clc.22599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The predictors of cardiovascular events in patients with chronic refractory angina are limited. High-sensitivity cardiac troponin T (hs-cTnT) assays are biomarkers that may be used to determine the prognosis of patients with stable coronary artery disease. HYPOTHESIS Hs-cTnT is a predictor of death and nonfatal myocardial infarction (MI) in patients with refractory angina. METHODS We prospectively enrolled 117 consecutive patients in this study. A heart team ruled out myocardial revascularization feasibility after assessing recent coronary angiograms; evidence of myocardial ischemia served as an inclusion criterion. Optimal medical therapy was encouraged via outpatient visits every 6 months; plasma hs-cTnT levels were determined at baseline. The primary endpoint was the composite incidence of death and nonfatal MI. RESULTS During a median follow-up period of 28.0 months (interquartile range, 18.0-47.5 months), an estimated 28.0-month cumulative event rate of 13.4% was determined via the Kaplan-Meier method. Univariate predictors of the composite endpoint were hs-cTnT levels and LV dysfunction. Following a multivariate analysis, only hs-cTnT was independently associated with the events in question, either as a continuous variable (hazard ratio per unit increase in the natural logarithm: 2.83, 95% confidence interval: 1.62-4.92, P < 0.001) or as a categorical variable (hazard ratio for concentrations above the 99th percentile: 5.14, 95% confidence interval: 2.05-12.91, P < 0.001). CONCLUSIONS In patients with chronic refractory angina, plasma concentration of hs-cTnT is the strongest predictor of death and nonfatal MI. Notably, none of the outcomes in question occurred in patients with baseline plasma levels <5.0 ng/L.
Collapse
Affiliation(s)
- Nilson T Poppi
- Refractory Angina Research Group, Heart Institute (InCor), University of São Paulo, São Paulo, Brazil.,Clinical Unit of Chronic Coronary Heart Disease, InCor, University of São Paulo, São Paulo, Brazil
| | - Luís H W Gowdak
- Refractory Angina Research Group, Heart Institute (InCor), University of São Paulo, São Paulo, Brazil.,Laboratory of Genetics and Molecular Cardiology, InCor, University of São Paulo, São Paulo, Brazil
| | - Luciana O C Dourado
- Refractory Angina Research Group, Heart Institute (InCor), University of São Paulo, São Paulo, Brazil.,Clinical Unit of Chronic Coronary Heart Disease, InCor, University of São Paulo, São Paulo, Brazil
| | - Eduardo L Adam
- Refractory Angina Research Group, Heart Institute (InCor), University of São Paulo, São Paulo, Brazil
| | - Thiago N P Leite
- Refractory Angina Research Group, Heart Institute (InCor), University of São Paulo, São Paulo, Brazil
| | - Bruno M Mioto
- Clinical Unit of Chronic Coronary Heart Disease, InCor, University of São Paulo, São Paulo, Brazil
| | - José E Krieger
- Laboratory of Genetics and Molecular Cardiology, InCor, University of São Paulo, São Paulo, Brazil
| | - Luiz A M César
- Clinical Unit of Chronic Coronary Heart Disease, InCor, University of São Paulo, São Paulo, Brazil
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, InCor, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Non-alcoholic fatty liver disease and cardiovascular risk: Pathophysiological mechanisms and implications. J Hepatol 2016; 65:425-43. [PMID: 27091791 DOI: 10.1016/j.jhep.2016.04.005] [Citation(s) in RCA: 364] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/25/2016] [Accepted: 04/01/2016] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become one of the most frequent chronic liver diseases in the Western society and its prevalence is likely to rise even further. An increasing body of evidence shows that NAFLD is not only a potentially progressive liver disease, but also has systemic consequences. More specifically, evidence points out that NAFLD has to be considered as a significant independent risk factor for subclinical and clinical cardiovascular disease (CVD). Long-term follow-up studies demonstrate cardiovascular mortality to be the most important cause of death in NAFLD patients. Moreover, ample evidence associates NAFLD with endothelial dysfunction, increased pulse wave velocity, increased coronary arterial calcifications and increased carotid intima media thickness, all established markers for CVD. Despite of all this evidence, the mechanisms by which NAFLD causally contributes to CVD are not fully elucidated. Furthermore, an extensive overview of all potential pathophysiological mechanisms and the corresponding current data are lacking. In this review we summarise current knowledge, originating from fundamental and clinical research, that mechanistically links NAFLD to CVD. Subsequently, the impact of CVD on current clinical practice and future research in the area of NALFD are discussed.
Collapse
|
42
|
Ding JW, Zheng XX, Zhou T, Tong XH, Luo CY, Wang XA. HMGB1Modulates the Treg/Th17 Ratio in Atherosclerotic Patients. J Atheroscler Thromb 2016; 23:737-45. [PMID: 26830200 PMCID: PMC7399277 DOI: 10.5551/jat.31088] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 11/16/2015] [Indexed: 01/14/2023] Open
Abstract
AIM Atherosclerosis (AS) characterized as a chronic inflammatory disease. Multiple immune cells and inflammatory cytokines, such as high mobility group protein (HMGB1), regulatory T (Treg) cells, T helper (Th17) cells, and inflammation-related cytokines, play a key role in its pathophysiology. A large number of studies report that HMGB1 and Th17 cells may promote atherosclerosis progression, whereas Treg cells may play a protective role in atherosclerosis; thus, alterations in the Treg/Th17 ratio may exist in atherosclerosis diseases. Up till now, the relationships between HMGB1 levels and the Treg/Th17 ratio remain incompletely understood. The major purpose of this study was to investigate the relationship between HMGB1 levels and the Treg/Th17 ratio in patients with coronary artery atherosclerotic plaques. METHODS We enrolled patients with coronary atherosclerosis and normal coronary artery as the research subjects. Flow cytometry was used to analyze the Treg cells, the Th17 cells frequency, and the Treg/Th17 ratio. Otherwise, real-time polymerase chain reaction was used for assays the mRNA expressions of HMGB1, retinoic acid-related orphan nuclear receptor C (RORC), and forkhead-winged helix transcription factor (Foxp3). Moreover, enzyme-linked immunosorbent assays were used to detect the level of protein and cytokines, such as HMGB1, IL-10, TGF-β1, IL-17A, and IL-23. RESULTS Using flow cytometry, we observed a significantly increased of Th17 cell frequency, whereas Treg cell frequency significantly decreased in atherosclerotic patients. Consistently, the levels of RORC mRNA were significantly increased in coronary atherosclerosis (AS) group compared to normal coronary artery (NCA) group (P<0.01). In contrast, the expression of Foxp3 mRNA was markedly lower in the AS group than in the NCA group (P<0.01). Furthermore, we observed the serum concentrations of HMGB1, IL-17A, and IL-23 were significantly higher in the AS group than in the NCA group (P<0.01, respectively), whereas the concentrations of serum IL-10 and TGF-β1 were significantly lower in the AS group than in the NCA group (P<0.01, respectively). In addition, we also found that HMGB1 levels showed negative correlation with the Treg/Th17 ratio in the two groups (r=-0.6984, P<0.01). CONCLUSIONS The data in our study indicated that HMGB1 may promote atherosclerosis progression via modulating the imbalance in the Treg/Th17 ratio.
Collapse
Affiliation(s)
- Jia-wang Ding
- Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Hubei Province, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei Province, China
| | - Xia-xia Zheng
- Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Hubei Province, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei Province, China
| | - Tian Zhou
- Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Hubei Province, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei Province, China
| | - Xiao-hong Tong
- Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Hubei Province, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei Province, China
| | - Cai-yun Luo
- Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Hubei Province, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei Province, China
| | - Xin-an Wang
- Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Hubei Province, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei Province, China
| |
Collapse
|
43
|
Bangert A, Andrassy M, Müller AM, Bockstahler M, Fischer A, Volz CH, Leib C, Göser S, Korkmaz-Icöz S, Zittrich S, Jungmann A, Lasitschka F, Pfitzer G, Müller OJ, Katus HA, Kaya Z. Critical role of RAGE and HMGB1 in inflammatory heart disease. Proc Natl Acad Sci U S A 2016; 113:E155-64. [PMID: 26715748 PMCID: PMC4720305 DOI: 10.1073/pnas.1522288113] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Autoimmune response to cardiac troponin I (TnI) induces inflammation and fibrosis in the myocardium. High-mobility group box 1 (HMGB1) is a multifunctional protein that exerts proinflammatory activity by mainly binding to receptor for advanced glycation end products (RAGE). The involvement of the HMGB1-RAGE axis in the pathogenesis of inflammatory cardiomyopathy is yet not fully understood. Using the well-established model of TnI-induced experimental autoimmune myocarditis (EAM), we demonstrated that both local and systemic HMGB1 protein expression was elevated in wild-type (wt) mice after TnI immunization. Additionally, pharmacological inhibition of HMGB1 using glycyrrhizin or anti-HMGB1 antibody reduced inflammation in hearts of TnI-immunized wt mice. Furthermore, RAGE knockout (RAGE-ko) mice immunized with TnI showed no structural or physiological signs of cardiac impairment. Moreover, cardiac overexpression of HMGB1 using adeno-associated virus (AAV) vectors induced inflammation in the hearts of both wt and RAGE-ko mice. Finally, patients with myocarditis displayed increased local and systemic HMGB1 and soluble RAGE (sRAGE) expression. Together, our study highlights that HMGB1 and its main receptor, RAGE, appear to be crucial factors in the pathogenesis of TnI-induced EAM, because inhibition of HMGB1 and ablation of RAGE suppressed inflammation in the heart. Moreover, the proinflammatory effect of HMGB1 is not necessarily dependent on RAGE only. Other receptors of HMGB1 such as Toll-like receptors (TLRs) may also be involved in disease pathogenesis. These findings could be confirmed by the clinical relevance of HMGB1 and sRAGE. Therefore, blockage of one of these molecules might represent a novel therapeutic strategy in the treatment of autoimmune myocarditis and inflammatory cardiomyopathy.
Collapse
Affiliation(s)
- Anna Bangert
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Martin Andrassy
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Anna-Maria Müller
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Mariella Bockstahler
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Andrea Fischer
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Christian H Volz
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Christoph Leib
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Göser
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Sevil Korkmaz-Icöz
- Department of Cardiac Surgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Zittrich
- Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Andreas Jungmann
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ziya Kaya
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
44
|
Fan Z, Yang J, Yang J, Yang C, Guo X. HMGB1: A promising therapeutic approach for atherosclerosis. Int J Cardiol 2016; 202:507-508. [PMID: 26440462 DOI: 10.1016/j.ijcard.2015.09.101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/24/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Zhixing Fan
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443000, Hubei Province, China.
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Chaojun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Xin Guo
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443000, Hubei Province, China
| |
Collapse
|
45
|
Cai J, Wen J, Bauer E, Zhong H, Yuan H, Chen AF. The Role of HMGB1 in Cardiovascular Biology: Danger Signals. Antioxid Redox Signal 2015; 23:1351-69. [PMID: 26066838 DOI: 10.1089/ars.2015.6408] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Cardiovascular disease (CVD) is the leading cause of mortality worldwide. Accumulating evidence shows that dysregulated immune response contributes to several types of CVDs such as atherosclerosis and pulmonary hypertension (PH). Vascular intimal impairment and low-density lipoprotein oxidation trigger a complex network of innate immune responses and sterile inflammation. RECENT ADVANCES High-mobility group box 1 (HMGB1), a nuclear DNA-binding protein, was recently discovered to function as a damage-associated molecular pattern molecule (DAMP) that initiates the innate immune responses. These findings lead to the understanding that HMGB1 plays a critical role in the inflammatory response in the pathogenesis of CVD. CRITICAL ISSUES In this review, we highlight the role of extracellular HMGB1 as a proinflammatory mediator as well as a DAMP in coronary artery disease, cerebral artery disease, peripheral artery disease, and PH. FUTURE DIRECTIONS A key focus for future researches on HMGB1 location, structure, modification, and signaling will reveal HMGB1's multiple functions and discover a targeted therapy that can eliminate HMGB1-mediated inflammation without interfering with adaptive immune responses.
Collapse
Affiliation(s)
- Jingjing Cai
- 1 The Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University , Changsha, China
- 2 Department of Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
- 3 Department of Cardiology, The Third Xiangya Hospital, Central South University , Changsha, China
| | - Juan Wen
- 1 The Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University , Changsha, China
- 3 Department of Cardiology, The Third Xiangya Hospital, Central South University , Changsha, China
| | - Eileen Bauer
- 2 Department of Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Hua Zhong
- 1 The Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University , Changsha, China
- 2 Department of Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
- 3 Department of Cardiology, The Third Xiangya Hospital, Central South University , Changsha, China
| | - Hong Yuan
- 1 The Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University , Changsha, China
- 3 Department of Cardiology, The Third Xiangya Hospital, Central South University , Changsha, China
| | - Alex F Chen
- 1 The Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University , Changsha, China
- 2 Department of Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
46
|
Zhong ZX, Li B, Li CR, Zhang QF, Liu ZD, Zhang PF, Gu XF, Luo H, Li MJ, Luo HS, Ye GH, Wen FL. Role of chemokines in promoting instability of coronary atherosclerotic plaques and the underlying molecular mechanism. ACTA ACUST UNITED AC 2014; 48:161-6. [PMID: 25424368 PMCID: PMC4321222 DOI: 10.1590/1414-431x20144195] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/22/2014] [Indexed: 02/01/2023]
Abstract
Our aim was to investigate the role of chemokines in promoting instability of
coronary atherosclerotic plaques and the underlying molecular mechanism. Coronary
angiography and intravascular ultrasound (IVUS) were performed in 60 stable angina
pectoris (SAP) patients and 60 unstable angina pectoris (UAP) patients. The
chemotactic activity of monocytes in the 2 groups of patients was examined in
Transwell chambers. High-sensitivity C-reactive protein (hs-CRP), monocyte
chemoattractant protein-1 (MCP-1), regulated on activation in normal T-cell expressed
and secreted (RANTES), and fractalkine in serum were examined with ELISA kits, and
expression of MCP-1, RANTES, and fractalkine mRNA was examined with real-time PCR. In
the SAP group, 92 plaques were detected with IVUS. In the UAP group, 96 plaques were
detected with IVUS. The plaques in the UAP group were mainly lipid 51.04% (49/96) and
the plaques in the SAP group were mainly fibrous 52.17% (48/92). Compared with the
SAP group, the plaque burden and vascular remodeling index in the UAP group were
significantly greater than in the SAP group (P<0.01). Chemotactic activity and the
number of mobile monocytes in the UAP group were significantly greater than in the
SAP group (P<0.01). Concentrations of hs-CRP, MCP-1, RANTES, and fractalkine in
the serum of the UAP group were significantly higher than in the serum of the SAP
group (P<0.05 or P<0.01), and expression of MCP-1, RANTES, and fractalkine mRNA
was significantly higher than in the SAP group (P<0.05). MCP-1, RANTES, and
fractalkine probably promote instability of coronary atherosclerotic plaque.
Collapse
Affiliation(s)
- Z X Zhong
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - B Li
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - C R Li
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - Q F Zhang
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - Z D Liu
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - P F Zhang
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - X F Gu
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - H Luo
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - M J Li
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - H S Luo
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - G H Ye
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| | - F L Wen
- Department of Cardiology, Meizhou Hospital Affiliated to Zhongshan University, Meizhou, Guangdong, China
| |
Collapse
|
47
|
Low levels of natural IgM antibodies against phosphorylcholine are independently associated with vascular remodeling in patients with coronary artery disease. Clin Res Cardiol 2014; 104:13-22. [PMID: 25103819 DOI: 10.1007/s00392-014-0750-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/31/2014] [Indexed: 01/08/2023]
Abstract
Low anti-phosphorylcholine (PC) IgM plasma levels have been associated with increased incidence of adverse events in coronary artery disease (CAD). The underlying mechanisms are unclear. We hypothesized that atheroprotection mediated by anti-PC IgM antibodies is associated with reduced vascular remodeling and therefore tested whether anti-PC IgM plasma levels independently predict vascular remodeling. In a prospective cross-sectional study, anti-PC IgM plasma levels were measured in 175 consecutive patients with suspected CAD undergoing cardiac computed tomography angiography. Plaque morphology was thoroughly analyzed. Vascular remodeling was defined by a change in the vessel diameter at the plaque site in comparison to the reference segment proximal to the lesion (reference diameter) of ≥10%. Mean age of the patients was 64.8 ± 10.7 years, 48.6% were female. In 98 patients CAD was diagnosed, 57 (58.2%) of which displayed non-obstructive CAD (stenosis <50%), whereas 41 (41.8%) exhibited obstructive CAD (stenosis ≥50%). In 34 of 98 (34.7%) CAD patients vascular remodeling was present. Mean anti-PC IgM levels did not differ between patients with and without CAD (70.8 ± 52.7 vs. 69.1 ± 55.1 U/mL). However, anti-PC IgM levels were significantly lower in CAD patients compared to those without vascular remodeling (46.6 ± 31.6 vs. 73.3 ± 58.5 U/mL, P = 0.024). Using multivariate logistic regression, anti-PC IgM plasma levels independently predicted coronary vascular remodeling (HR 0.322, 95% confidence interval 0.121-0.856, P = 0.023). In conclusion, low anti-PC IgM levels are independently associated with coronary vascular remodeling. These findings may represent the link between in vitro studies demonstrating atheroprotective effects of anti-PC IgM and clinical data demonstrating that low anti-PC IgM levels are associated with adverse outcome in CAD patients.
Collapse
|
48
|
Korosoglou G, Giusca S, Gitsioudis G, Erbel C, Katus HA. Cardiac magnetic resonance and computed tomography angiography for clinical imaging of stable coronary artery disease. Diagnostic classification and risk stratification. Front Physiol 2014; 5:291. [PMID: 25147526 PMCID: PMC4123729 DOI: 10.3389/fphys.2014.00291] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 07/18/2014] [Indexed: 12/18/2022] Open
Abstract
Despite advances in the pharmacologic and interventional treatment of coronary artery disease (CAD), atherosclerosis remains the leading cause of death in Western societies. X-ray coronary angiography has been the modality of choice for diagnosing the presence and extent of CAD. However, this technique is invasive and provides limited information on the composition of atherosclerotic plaque. Coronary computed tomography angiography (CCTA) and cardiac magnetic resonance (CMR) have emerged as promising non-invasive techniques for the clinical imaging of CAD. Hereby, CCTA allows for visualization of coronary calcification, lumen narrowing and atherosclerotic plaque composition. In this regard, data from the CONFIRM Registry recently demonstrated that both atherosclerotic plaque burden and lumen narrowing exhibit incremental value for the prediction of future cardiac events. However, due to technical limitations with CCTA, resulting in false positive or negative results in the presence of severe calcification or motion artifacts, this technique cannot entirely replace invasive angiography at the present time. CMR on the other hand, provides accurate assessment of the myocardial function due to its high spatial and temporal resolution and intrinsic blood-to-tissue contrast. Hereby, regional wall motion and perfusion abnormalities, during dobutamine or vasodilator stress, precede the development of ST-segment depression and anginal symptoms enabling the detection of functionally significant CAD. While CT generally offers better spatial resolution, the versatility of CMR can provide information on myocardial function, perfusion, and viability, all without ionizing radiation for the patients. Technical developments with these 2 non-invasive imaging tools and their current implementation in the clinical imaging of CAD will be presented and discussed herein.
Collapse
|
49
|
Gitsioudis G, Katus HA, Korosoglou G. Assessment of coronary artery disease using coronary computed tomography angiography and biochemical markers. World J Cardiol 2014; 6:663-670. [PMID: 25068026 PMCID: PMC4110614 DOI: 10.4330/wjc.v6.i7.663] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/16/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammatory mechanisms in the arterial wall lead to atherosclerosis, and include endothelial cell damage, inflammation, apoptosis, lipoprotein deposition, calcification and fibrosis. Cardiac computed tomography angiography (CCTA) has been shown to be a promising tool for non-invasive assessment of theses specific compositional and structural changes in coronary arteries. This review focuses on the technical background of CCTA-based quantitative plaque characterization. Furthermore, we discuss the available evidence for CCTA-based plaque characterization and the potential role of CCTA for risk stratification of patients with coronary artery disease.
Collapse
|
50
|
Palmiere C, Augsburger M, Mangin P. High-mobility group box-1 protein determination in postmortem samples. Forensic Sci Int 2014; 239:103-6. [DOI: 10.1016/j.forsciint.2014.03.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/13/2014] [Accepted: 03/24/2014] [Indexed: 12/27/2022]
|