1
|
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
|
2
|
Wojtacha JJ, Morawin B, Wawrzyniak-Gramacka E, Tylutka A, de Freitas AKE, Zembron-Lacny A. Endothelial Dysfunction with Aging: Does Sex Matter? Int J Mol Sci 2024; 25:12203. [PMID: 39596269 PMCID: PMC11594464 DOI: 10.3390/ijms252212203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress and inflammation accompany endothelial dysfunction that results from the excessive or uncontrolled production of reactive oxygen and nitrogen species (RONS) in older adults. This study was designed to assess the usefulness of serum oxi-inflammatory component combinations in vascular disease prediction and prevention with regard to sex. Women (n = 145) and men (n = 50) aged 72.2 ± 7.8 years participated in this project. The females demonstrated the elevated production of hydrogen peroxide (H2O2) and nitric oxide (NO) responsible for intravascular low-density lipoprotein oxidation. NO generation was enhanced in the women, but its bioavailability was reduced, which was expressed by a high 3-nitrotyrosine (3-NitroT) concentration. The relation of NO/3-NitroT (rs = 0.811, p < 0.001) in the women and NO/3-NitroT (rs = -0.611, p < 0.001) in the men showed that sex determines endothelial dysfunction. RONS generation in the women simultaneously promoted endothelial regeneration, as demonstrated by a ~1.5-fold increase in circulating progenitor cells. Inflammation-specific variables, such as the neutrophil-to-lymphocyte ratio, the systemic immune inflammation index, and the neutrophil-to-high-density lipoprotein (HDL) ratio, were reduced in the women and showed their diagnostic utility for clinical prognosis in vascular dysfunction, especially the C-reactive-protein-to-HDL ratio (AUC = 0.980, specificity 94.7%, sensitivity 93.3%, OR = 252, 95% CI 65-967, p < 0.001). This study is the first to have revealed sex-specific changes in the oxi-inflammatory response, which can generate the risk of cardiovascular events at an older age.
Collapse
Affiliation(s)
- Jakub Jozue Wojtacha
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Barbara Morawin
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Edyta Wawrzyniak-Gramacka
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Anna Tylutka
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Ana Karyn Ehrenfried de Freitas
- School of Health Science, Positivo University, 5300 Professor Pedro Viriato Parigot de Souza Street, Campo Comprido, Curitiba 81280-330, PR, Brazil;
- Department of Cardiology, Hospital da Cruz Vermelha Brasileira Filial do Paraná, Av. Vicente Machado, 1280, R. Cap. Souza Franco, 50-Batel, Curitiba 80420-011, PR, Brazil
| | - Agnieszka Zembron-Lacny
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| |
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
|
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
|
5
|
Bagheri B, Khatibiyan Feyzabadi Z, Nouri A, Azadfallah A, Mahdizade Ari M, Hemmati M, Darban M, Alavi Toosi P, Banihashemian SZ. Atherosclerosis and Toll-Like Receptor4 (TLR4), Lectin-Like Oxidized Low-Density Lipoprotein-1 (LOX-1), and Proprotein Convertase Subtilisin/Kexin Type9 (PCSK9). Mediators Inflamm 2024; 2024:5830491. [PMID: 38445291 PMCID: PMC10914434 DOI: 10.1155/2024/5830491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/31/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Atherosclerosis is a leading cause of death in the world. A significant body of evidence suggests that inflammation and various players are implicated and have pivotal roles in the formation of atherosclerotic plaques. Toll-like receptor 4 (TLR4) is linked with different stages of atherosclerosis. This receptor is highly expressed in the endothelial cells (ECs) and atherosclerotic plaques. TLR4 activation can lead to the production of inflammatory cytokines and related responses. Lectin-like oxidized low-density lipoprotein-1 (LOX-1), an integral membrane glycoprotein with widespread expression on the ECs, is involved in atherosclerosis and has some common pathways with TLR4 in atherosclerotic lesions. In addition, proprotein convertase subtilisin/kexin type9 (PCSK9), which is a regulatory enzyme with different roles in cholesterol uptake, is implicated in atherosclerosis. At present, TLR4, PCSK9, and LOX-1 are increasingly acknowledged as key players in the pathogenesis of atherosclerotic cardiovascular diseases. Herein, we presented the current evidence on the structure, functions, and roles of TLR4, PCSK9, and LOX-1 in atherosclerosis.
Collapse
Affiliation(s)
- Bahador Bagheri
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Ahmad Nouri
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Azadfallah
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahyar Mahdizade Ari
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maral Hemmati
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahboubeh Darban
- Department of Internal Medicine, Kowsar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Parisa Alavi Toosi
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | |
Collapse
|
6
|
Yang B, Xiaping Z. The clinical significance of serum HMGB1 in patients with lower extremity arteriosclerosis obliterans after interventional vascular restenosis. Front Surg 2023; 9:1031108. [PMID: 36704514 PMCID: PMC9872960 DOI: 10.3389/fsurg.2022.1031108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/28/2022] [Indexed: 01/08/2023] Open
Abstract
Objective This study explored the correlation between serum HMGB1 levels and postoperative vascular restenosis in patients with lower extremity arteriosclerosis obliterans (LEASO). Methods A total of 362 patients LEASO who received vascular intervention were recruited in this study. Serum HMGB1 levels were measured by enzyme-linked immunosorbent assay. Logistic regression analysis was used to identify the influencing factors associated with vascular restenosis. The R procedure was used to create nomogram model. Receiver operating characteristic (ROC) analysis was used to determine the predictive value of serum HMGB1 and nomogram model for vascular restenosis. Results Of the 362 LEASO patients included, 103 (28.45%) developed restenosis within 6 months of postoperative follow-up. Postoperative HMGB1 levels were significantly higher in patients with restenosis compared to those with non-restenosis. Postoperative HMGB1 levels were significantly and positively correlated with the severity of postoperative restenosis (r = 0.819). The AUC of postoperative HMGB1 for the diagnosis of postoperative restenosis was 0.758 (95% CI: 0.703-0.812), with a sensitivity and specificity of 56.31% and 82.24%, respectively. Multivariate logistic regression analysis showed that diabetes, smoking, regular postoperative medication, increased fibrinogen, decreased red blood cells, increased hs-CRP, and increased postoperative HMGB1 were independently associated with postoperative restenosis in patients with LEASO. The C-index of the nomogram prediction model constructed based on the seven influencing factors mentioned above was 0.918. The nomogram model was significantly more predictive of postoperative restenosis in LEASO patients compared with a single postoperative HMGB1 (AUC: 0.918, 95% CI: 0.757-0.934). Conclusion Postoperative serum HMGB1 is an independent risk factor associated with postoperative vascular restenosis in patients with LEASO, and a novel nomogram model based on postoperative serum HMGB1 combined with clinical characteristics may help to accurately predict the risk of postoperative restenosis in patients with LEASO.
Collapse
Affiliation(s)
- Bo Yang
- Department of Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhang Xiaping
- Department of Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China,Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China,Correspondence: Zhang XiaPing
| |
Collapse
|
7
|
Huo X, Su B, Qin G, Zhao L. HMGB1 promotes Ox-LDL-induced endothelial cell damage by inhibiting PI3K/Akt signaling pathway. BMC Cardiovasc Disord 2022; 22:555. [PMID: 36544080 PMCID: PMC9768960 DOI: 10.1186/s12872-022-03003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Atherosclerosis is the pathological basis of cardio-cerebrovascular diseases. Oxidized low-density lipoprotein (ox-LDL) is an important risk factor for atherosclerosis. Ox-LDL leads to endothelial cell (EC) damage and dysfunction through various processes and promotes the occurrence and deterioration of atherosclerosis. High mobility group box-1 (HMGB1) is a protein associated with cellular damage. In the present study, the effect of HMGB1 on ox-LDL-induced EC damage was determined and the underlying mechanism explored. MATERIALS AND METHODS Human umbilical vein ECs (HUVECs) were exposed to ox-LDL to induce endothelial damage and changes in HMGB1 expression level were detected using western blotting analysis and reverse transcription-quantitative PCR. To observe the effect of HMGB1 on ox-LDL-induced damage, the HMGB1 expression was downregulated with siRNA, and cell viability, cytotoxicity, and apoptosis rate were assessed. HUVECs were pretreated with LY294002, an inhibitor of the PI3K/Akt pathway, to determine whether the effect of HMGB1 on damage is via the PI3K-Akt pathway. RESULTS The results showed that ox-LDL can upregulate HMGB1 expression in HUVECs and downregulation of HMGB1 expression can prevent ox-LDL-induced damage in HUVECs. Furthermore, the effect of HMGB1 on ox-LDL-induced damage could be promoted by inhibiting the PI3K/Akt signaling pathway. CONCLUSION The results indicate HMGB1 may be a promising research target to alleviate ox-LDL-induced EC damage.
Collapse
Affiliation(s)
- Xin Huo
- grid.477425.7Department of Vascular Surgery, Liuzhou People’s Hospital, No. 8 Wenchang Road, Chengzhong District, Liuzhou, 545001 Guangxi China
| | - Boyou Su
- grid.477425.7Department of Vascular Surgery, Liuzhou People’s Hospital, No. 8 Wenchang Road, Chengzhong District, Liuzhou, 545001 Guangxi China
| | - Guoti Qin
- grid.477425.7Department of Vascular Surgery, Liuzhou People’s Hospital, No. 8 Wenchang Road, Chengzhong District, Liuzhou, 545001 Guangxi China
| | - Liming Zhao
- grid.477425.7Department of Vascular Surgery, Liuzhou People’s Hospital, No. 8 Wenchang Road, Chengzhong District, Liuzhou, 545001 Guangxi China
| |
Collapse
|
8
|
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: 4] [Impact Index Per Article: 1.3] [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
|
9
|
Belmadani S, Matrougui K. Role of High Mobility Group Box 1 in Cardiovascular Diseases. Inflammation 2022; 45:1864-1874. [PMID: 35386038 PMCID: PMC11145736 DOI: 10.1007/s10753-022-01668-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/27/2022] [Accepted: 03/28/2022] [Indexed: 11/05/2022]
Abstract
High Mobility Group Box 1 (HMGB1) is a ubiquitous, highly conserved nuclear and cytosolic protein that has diverse biological roles depending on its cellular location and posttranslational modifications. The HMGB1 is localized in the nucleus but can be translocated to the cytoplasm to modulate the intracellular signaling and eventually secreted outside the cells. It is widely established that HMGB1 plays a key role in inflammation; however, the role of HMGB1 in the cardiovascular diseases is not well understood. In this review, we will discuss the latest reports on the pathophysiological link between HMGB1 and cardiovascular complications, with special emphasis on the inflammation. Thus, the understanding of the role of HMGB1 may provide new insights into developing new HMGB1-based therapies.
Collapse
Affiliation(s)
- Souad Belmadani
- Department of Physiological Sciences, EVMS, Norfolk, Virginia, 23501, USA
| | - Khalid Matrougui
- Department of Physiological Sciences, EVMS, Norfolk, Virginia, 23501, USA.
| |
Collapse
|
10
|
A Review with Updated Perspectives on Nutritional and Therapeutic Benefits of Apricot and the Industrial Application of Its Underutilized Parts. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27155016. [PMID: 35956966 PMCID: PMC9370680 DOI: 10.3390/molecules27155016] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022]
Abstract
Fruits maintain the image as the richest sources of vitamins. Focusing on apricots, utilization of apricot species for many applications is possible due to its various benefits. Many research studies demonstrated different perspectives of apricot, especially in medical used as it can act as antioxidant, anti-inflammatory, and antimicrobial agents. Moreover, in the industrial sectors, apricots can be used in the production of biofuels and batteries. All components of the apricot fruit, including seeds and kernels have been found to possess significant interest. This review is to breach the knowledge gap regarding the key nutrients and chemicals of apricot fruit, contributing to its health-promoting properties to emphasize the noble importance of this fruit in the diet and in the management of several diseases. We also cover the application of apricots in the industry that could be developed as a promising and sustainable source.
Collapse
|
11
|
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]
|
12
|
Advanced Glycation End Products: A Sweet Flavor That Embitters Cardiovascular Disease. Int J Mol Sci 2022; 23:ijms23052404. [PMID: 35269546 PMCID: PMC8910157 DOI: 10.3390/ijms23052404] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/24/2022] Open
Abstract
Epidemiological studies demonstrate the role of early and intensive glycemic control in the prevention of micro and macrovascular disease in both type 1 and type 2 diabetes mellitus (DM). Hyperglycemia elicits several pathways related to the etiopathogenesis of cardiovascular disease (CVD), including the generation of advanced glycation end products (AGEs). In this review, we revisit the role played by AGEs in CVD based in clinical trials and experimental evidence. Mechanistic aspects concerning the recognition of AGEs by the advanced glycosylation end product-specific receptor (AGER) and its counterpart, the dolichyl-diphosphooligosaccharide-protein glycosyltransferase (DDOST) and soluble AGER are discussed. A special focus is offered to the AGE-elicited pathways that promote cholesterol accumulation in the arterial wall by enhanced oxidative stress, inflammation, endoplasmic reticulum stress and impairment in the reverse cholesterol transport (RCT).
Collapse
|
13
|
Wang J, Li H, Xia T, Feng J, Zhou R. Pulmonary arterial hypertension and flavonoids: A role in treatment. CHINESE J PHYSIOL 2021; 64:115-124. [PMID: 34169916 DOI: 10.4103/cjp.cjp_25_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a high mortality progressive pulmonary vascular disease that can lead to right heart failure. The use of clinical drugs for the treatment of PAH is limited to a great extent because of its single target and high price. Flavonoids are widely distributed in nature, and have been found in fruits, vegetables, and traditional Chinese medicine. They have diverse biological activities and various pharmacological effects such as antitumor, antioxidation, and anti-inflammatory. This review summarizes the progress in pharmacodynamics and mechanism of flavonoids in the treatment of PAH in recent years, in order to provide some theoretical references for relevant researchers.
Collapse
Affiliation(s)
- Jialing Wang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Hailong Li
- The Third People's Hospital of Ningxia, Yinchuan, China
| | - Tian Xia
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jun Feng
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Ru Zhou
- Department of Pharmacology, College of Pharmacy; Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education; Ningxia Characteristic Traditional Chinese Medicine Modernization Engineering Technology Research Center, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Kim KM, Lee JY, Jeon BH, Quan KT, Na M, Nam KW, Chae S. Extract of Curcuma zedoaria R. prevents atherosclerosis in apolipoprotein E-deficient mice. Nutr Res Pract 2021; 15:319-328. [PMID: 34093973 PMCID: PMC8155225 DOI: 10.4162/nrp.2021.15.3.319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/03/2020] [Accepted: 12/20/2020] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/OBJECTIVES Curcuma zedoaria R. (Zingiberaceae) has been used to treat headache, fever, and hypertension-related symptoms in Asian countries, including Korea, China, and Japan. We investigated whether dietary intake of a C. zedoaria extract (CzE) affected atherosclerosis in vivo. MATERIALS/METHODS Apolipoprotein E-deficient (ApoE−/−) mice (n = 32) were fed a normal diet (ND), a high-cholesterol diet (HCD), an HCD containing CzE (100 mg/kg/day), or an HCD containing simvastatin (10 mg/kg/day) for 12 weeks. The anti-atherosclerotic effects were evaluated by observing changes in fatty streak lesions, immunohistochemical analysis, ex vivo fluorescence imaging, lipid profiles, and western blot analysis. RESULTS The CzE-fed group showed a 41.6% reduction of atherosclerosis. Furthermore, CzE significantly reduced the levels of serum triglyceride, high-density lipoprotein, the chemokine (C-X3-C-motif) ligand 1, the adhesion molecules vascular cell adhesion molecule-1, intracellular adhesion molecule-1, and E-selectin; down-regulation of tumor necrosis factor-α, interleukin-6, high mobility group box-1, and cathepsin levels in the aortic sinuses and aortas of ApoE−/− mice were also observed. CONCLUSIONS The results suggest that the inclusion of a water extract of C. zedoaria in a HCD is closely correlated with reducing the risk of vascular inflammatory diseases in an ApoE mouse model.
Collapse
Affiliation(s)
- Ki Mo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.,Department of Korean Life Science and Technology, University of Science and Technology, Daejeon 34113, Korea
| | - Joo Young Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Byeong Hwa Jeon
- Department of Physiology, School of Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Khong Trong Quan
- Department of Pharmacognosy, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea
| | - MinKyun Na
- Department of Pharmacognosy, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea
| | - Kung-Woo Nam
- Department of Life Science and Biotechnology, Soonchunhyang University, Asan 31538, Korea
| | - Sungwook Chae
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.,Department of Korean Life Science and Technology, University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
17
|
Behl T, Sharma E, Sehgal A, Kaur I, Kumar A, Arora R, Pal G, Kakkar M, Kumar R, Bungau S. Expatiating the molecular approaches of HMGB1 in diabetes mellitus: Highlighting signalling pathways via RAGE and TLRs. Mol Biol Rep 2021; 48:1869-1881. [PMID: 33479829 DOI: 10.1007/s11033-020-06130-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/24/2020] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus (DM) has become one of the major healthcare challenges worldwide in the recent times and inflammation being one of its key pathogenic process/mechanism affect several body parts including the peripheral and central nervous system. High-mobility group box 1 (HMGB1) is one of the major non-histone proteins that plays a key role in triggering the inflammatory response. Upon its release into the extracellular milieu, HMGB1 acts as an "alarmin" for the immune system to initiate tissue repair as a component of the host defense system. Furthermore, HMGB1 along with its downstream receptors like Toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE) serve as the suitable target for DM. The forthcoming research in the field of diabetes would potentially focus on the development of alternative approaches to target the centre of inflammation that is primarily mediated by HMGB1 to improve diabetic-related complications. This review covers the therapeutic actions of HMGB1 protein, which acts by activating the RAGE and TLR molecules to constitute a functional tripod system, in turn activating NF-κB pathway that contributes to the production of mediators for pro-inflammatory cytokines associated with DM. The interaction between TLR2 and TLR4 with ligands present in the host and the activation of RAGE stimulates various immune and metabolic responses that contribute to diabetes. This review emphasizes to elucidate the role of HMGB1 in the initiation and progression of DM and control over the inflammatory tripod as a promising therapeutic approach in the management of DM.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Eshita Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Giridhari Pal
- Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Munish Kakkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ravinder Kumar
- Cardiovascular Research Institute, Icahn School of Medicine, New York, USA
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
18
|
Satish M, Gunasekar P, Asensio JA, Agrawal DK. Vitamin D attenuates HMGB1-mediated neointimal hyperplasia after percutaneous coronary intervention in swine. Mol Cell Biochem 2020; 474:219-228. [PMID: 32737774 DOI: 10.1007/s11010-020-03847-y] [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: 05/06/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
Intracoronary stenting is a common procedure in patients with coronary artery disease (CAD). Stent deployment stretches and denudes the endothelial layer, promoting a local inflammatory response, resulting in neointimal hyperplasia. Vitamin D deficiency associates with CAD. In this study, we examined the association of vitamin D status with high mobility group box 1 (HMGB1)-mediated pathways (HMGB1, receptor for advanced glycation end products [RAGE], and Toll-like receptor-2 and -4 [TLR2 and TLR4]) in neointimal hyperplasia in atherosclerotic swine following bare metal stenting. Yucatan microswine fed with a high-cholesterol diet were stratified to receive vitamin D-deficient (VD-DEF), vitamin D-sufficient (VD-SUF), and vitamin D-supplemented (VD-SUP) diet. After 6 months, PTCA (percutaneous transluminal balloon angioplasty) followed by bare metal stent implantation was performed in the left anterior descending (LAD) artery of each swine. Four months following coronary intervention, angiogram and optical coherence tomography (OCT) were performed and swine euthanized. Histology and immunohistochemistry were performed in excised LAD to evaluate the expression of HMGB1, RAGE, TLR2, and TLR4. OCT analysis revealed the greatest in-stent restenosis area in the LAD of VD-DEF compared to VD-SUF or VD-SUP swine. The protein expression of HMGB1, RAGE, TLR2, and TLR4 was significantly higher in the LAD of VD-DEF compared to VD-SUF or VD-SUP swine. Vitamin D deficiency was associated with both increased in-stent restenosis and increased HMGB1-mediated inflammation noted in coronary arteries following intravascular stenting. Inversely, vitamin D supplementation was associated with both a decrease in this inflammatory profile and in neointimal hyperplasia, warranting further investigation for vitamin D as a potential adjunct therapy following coronary intervention.
Collapse
Affiliation(s)
- Mohan Satish
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Palanikumar Gunasekar
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Juan A Asensio
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
19
|
Hebbel RP, Wei P, Milbauer L, Corban MT, Solovey A, Kiley J, Pattee J, Lerman LO, Pan W, Lerman A. Abnormal Endothelial Gene Expression Associated With Early Coronary Atherosclerosis. J Am Heart Assoc 2020; 9:e016134. [PMID: 32673514 PMCID: PMC7660702 DOI: 10.1161/jaha.120.016134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/29/2020] [Indexed: 12/18/2022]
Abstract
Background We examined feasibility of a unique approach towards gaining insight into heritable risk for early atherosclerosis: surveying gene expression by endothelial cells from living subjects. Methods and Results Subjects aged <50 years (mean age, 37; range, 22-49) without obstructive coronary artery disease underwent coronary reactivity testing that identified them as having normal or abnormal coronary endothelial function. Cultures of Blood Outgrowth Endothelial Cells (BOEC) from 6 normal and 13 abnormal subjects passed rigorous quality control and were used for microarray assessment of gene expression. Of 9 genes differentially expressed at false discovery rate <0.1%, we here focus upon abnormal subjects having elevated expression of HMGB1 (high mobility group box 1) which we unexpectedly found to be linked to low LAMC1 (laminin gamma 1) expression. This linkage was corroborated by 3 of our past studies and confirmed bio-functionally. Compared with normal BOEC, abnormal BOEC released 13±3-fold more HMGB1 in response to lipopolysaccharide; and they deposited one tenth as much LAMC1 into collagen subendothelial matrix during culture. Clinical follow-up data are provided for 4 normal subjects (followed 13.4±0.1 year) and for 12 abnormal subjects (followed 9.1±4.5 years). Conclusions The known pathogenic effects of high-HMGB1 and low-LAMC1 predict that the combination would biologically converge upon the focal adhesion complex, to the detriment of endothelial shear responsiveness. This gene expression pattern may comprise a heritable risk state that promotes early coronary atherosclerosis. If so, the testing could be applied even in childhood, enabling early intervention. This approach offers a way to bridge the information gap between genetics and clinical phenotype.
Collapse
Affiliation(s)
- Robert P. Hebbel
- Division of Hematology‐Oncology‐TransplantationDepartment of Medicine, and Vascular Biology CenterUniversity of Minnesota Medical SchoolMinneapolisMN
| | - Peng Wei
- Division of Hematology‐Oncology‐TransplantationDepartment of Medicine, and Vascular Biology CenterUniversity of Minnesota Medical SchoolMinneapolisMN
- Division of BiostatisticsSchool of Public HealthUniversity of MinnesotaMinneapolisMN
| | - Liming Milbauer
- Division of Hematology‐Oncology‐TransplantationDepartment of Medicine, and Vascular Biology CenterUniversity of Minnesota Medical SchoolMinneapolisMN
| | - Michel T. Corban
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
| | - Anna Solovey
- Division of Hematology‐Oncology‐TransplantationDepartment of Medicine, and Vascular Biology CenterUniversity of Minnesota Medical SchoolMinneapolisMN
| | - James Kiley
- Division of Hematology‐Oncology‐TransplantationDepartment of Medicine, and Vascular Biology CenterUniversity of Minnesota Medical SchoolMinneapolisMN
| | - Jack Pattee
- Division of BiostatisticsSchool of Public HealthUniversity of MinnesotaMinneapolisMN
| | - Lilach O. Lerman
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
- Division of Nephrology and HypertensionDepartment of MedicineMayo Clinic College of Medicine and ScienceRochesterMN
| | - Wei Pan
- Division of BiostatisticsSchool of Public HealthUniversity of MinnesotaMinneapolisMN
| | - Amir Lerman
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
| |
Collapse
|
20
|
Zhu Z, Guo Y, Li X, Teng S, Peng X, Zou P, Zhou S. Glycyrrhizic Acid Attenuates Balloon-Induced Vascular Injury Through Inactivation of RAGE Signaling Pathways. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2020. [DOI: 10.15212/cvia.2019.0577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Percutaneous coronary intervention is a well-established technique used to treat coronary artery disease, but the risk of coronary artery in-stent restenosis following percutaneous coronary intervention is still high. Previous studies revealed that high mobility group protein B1 (HMGB1)
plays a critical role in neointima formation. In this study, we aimed to investigate the role of glycyrrhizic acid (GA), an HMGB1 inhibitor, in the process of neointima formation and the potential mechanisms. We investigated the role of GA in neointima formation through an iliac artery balloon
injury model in rabbits. Proliferation, migration, and phenotype transformation of human vascular smooth muscle cells (VSMCs) were observed. Besides, inflammation and receptor for advanced glycosylation end products (RAGE) signaling pathways were studied. The results indicate that GA attenuated
neointima formation and downregulated HMGB1 expression in injured artery in rabbits. HMGB1 promoted proliferation, migration, and phenotype transformation through the activation of RAGE signaling pathways in VSMCs, and blockade of HMGB1 by GA (1, 10, and 100 μM) could attenuate those processes
and reduce proliferation of human VSMCs. In conclusion, the HMGB1 inhibitor GA might be useful to treat proliferative vascular diseases by downregulating RAGE signaling pathways. Our results indicate a new and promising therapeutic agent for restenosis.
Collapse
Affiliation(s)
- Zhaowei Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanan Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuping Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuai Teng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaofan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pu Zou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
Zhou Y, Little PJ, Downey L, Afroz R, Wu Y, Ta HT, Xu S, Kamato D. The Role of Toll-like Receptors in Atherothrombotic Cardiovascular Disease. ACS Pharmacol Transl Sci 2020; 3:457-471. [PMID: 32566912 PMCID: PMC7296543 DOI: 10.1021/acsptsci.9b00100] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are dominant components of the innate immune system. Activated by both pathogen-associated molecular patterns and damage-associated molecular patterns, TLRs underpin the pathology of numerous inflammation related diseases that include not only immune diseases, but also cardiovascular disease (CVD), diabetes, obesity, and cancers. Growing evidence has demonstrated that TLRs are involved in multiple cardiovascular pathophysiologies, such as atherosclerosis and hypertension. Specifically, a trial called the Canakinumab Anti-inflammatory Thrombosis Outcomes Study showed the use of an antibody that neutralizes interleukin-1β, reduces the recurrence of cardiovascular events, demonstrating inflammation as a therapeutic target and also the research value of targeting the TLR system in CVD. In this review, we provide an update of the interplay between TLR signaling, inflammatory mediators, and atherothrombosis, with an aim to identify new therapeutic targets for atherothrombotic CVD.
Collapse
Affiliation(s)
- Ying Zhou
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Peter J. Little
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| | - Liam Downey
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Rizwana Afroz
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Yuao Wu
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Hang T. Ta
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Suowen Xu
- Aab
Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | - Danielle Kamato
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| |
Collapse
|
22
|
Zhou DM, Ran F, Ni HZ, Sun LL, Xiao L, Li XQ, Li WD. Metformin inhibits high glucose-induced smooth muscle cell proliferation and migration. Aging (Albany NY) 2020; 12:5352-5361. [PMID: 32208365 PMCID: PMC7138554 DOI: 10.18632/aging.102955] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/20/2020] [Indexed: 12/17/2022]
Abstract
We investigated the protective effects and mechanism of action of metformin on high glucose-induced smooth muscle cell proliferation and migration. Vascular smooth muscle cells (VSMCs) were subjected to a series of concentrations (0-10 mM) of metformin. CCK-8, wound healing, and transwell assays were performed. Correlations between metformin concentration and high-mobility group box 1 (HMGB1) and miR-142-3p levels were assessed. In addition, miR-142-3p mimic and siRNA were used to investigate VSMC migration in the presence or absence of metformin. In the high-glucose condition, metformin decreased cell growth and inhibited cell migration. HMGB1 gene expression correlated negatively with metformin concentration, whereas miR-142-3p expression correlated positively with metformin concentration. In addition, mimic-induced miR-142-3p elevation resulted in decreased HMGB1 and LC3II levels and elevated p62 levels in the high-glucose condition, whereas miR-142-3p knockdown had the reverse effects, and metformin abolished those effects. Metformin inhibits high glucose–induced VSMC hyperproliferation and increased migration by inducing miR-142-3p-mediated inhibition of HMGB1 expression via the HMGB1-autophagy related pathway.
Collapse
Affiliation(s)
- Dong-Ming Zhou
- Department of Hematology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Jiangsu, China
| | - Feng Ran
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Jiangsu, China
| | - Hai-Zhen Ni
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Jiangsu, China.,Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Li-Li Sun
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Jiangsu, China
| | - Lun Xiao
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Jiangsu, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Jiangsu, China
| | - Wen-Dong Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Jiangsu, China
| |
Collapse
|
23
|
Glucagon-like peptide-1 receptor agonist dulaglutide prevents ox-LDL-induced adhesion of monocytes to human endothelial cells: An implication in the treatment of atherosclerosis. Mol Immunol 2019; 116:73-79. [PMID: 31630078 DOI: 10.1016/j.molimm.2019.09.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/09/2019] [Accepted: 09/29/2019] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is a common comorbidity of type II diabetes and a leading cause of death worldwide. The presence of oxidized low-density lipoprotein (ox-LDL) drives atherogenesis by inducing oxidative stress, mitochondrial dysfunction, expression of proinflammatory cytokines and chemokines including interleukin (IL)-1β, IL-6, and monocyte chemoattractant protein 1 (MCP-1), adhesion molecules including vascular cellular adhesion molecule 1 (VCAM-1) and E-selectin, and downregulating expression of the Krüppel-like factor 2 (KLF2) transcription factor. Importantly, ox-LDL induced the attachment of THP-1 monocytes to endothelial cells. In the present study, we demonstrate for the first time that the specific glucagon-like peptide 1 receptor (GLP-1R) agonist dulaglutide may prevent these atherosclerotic effects of ox-LDL by preventing suppression of KLF2 by p53 protein in human aortic endothelial cells. KLF2 has been shown to play a major role in protecting vascular endothelial cells from damage induced by ox-LDL and oscillatory shear, and therefore, therapies capable of mediating KLF2 signaling may be an attractive treatment option for preventing the development and progression of atherosclerosis.
Collapse
|
24
|
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
|
25
|
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
|
26
|
Gu HF, Li N, Xu ZQ, Hu L, Li H, Zhang RJ, Chen RM, Zheng XL, Tang YL, Liao DF. Chronic Unpredictable Mild Stress Promotes Atherosclerosis via HMGB1/TLR4-Mediated Downregulation of PPARγ/LXRα/ABCA1 in ApoE -/- Mice. Front Physiol 2019; 10:165. [PMID: 30881312 PMCID: PMC6405526 DOI: 10.3389/fphys.2019.00165] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Although our previous studies have confirmed that the activation of TLR4 is implicated in the development of atherosclerosis induced by chronic unpredicted mild stress (CUMS), the underling mechanism is largely unclear. Here, we hypothesized that CUMS accelerates atherosclerotic development through lowering PPARγ/LXRα-ABCA1 expression via HMGB1/TLR4 signaling. Methods: In present study, CUMS atherosclerotic animal models were established with AopE-/- mice, and CUMS Raw 264.7 macrophage models were mimicked by high corticosterone treatment, These models were treated with Ethyl pyruvate (EP, an inhibitor of HMGB1), TLR4 inhibitor TAK-242, and PPARγ agonist RSG (Rosiglitazone) to test our hypothesis, respectively. Results: Our results indicated that the protein levels of HMGB1, TLR4, and pro-inflammatory cytokines including IL-1β, TNF-α were elevated with the development of atherosclerosis in CUMS mice, while the expressions of PPARγ, LXRα, and ABCA1 declined. Notably, HMGB1 inhibition by EP reversed CUMS-induced atherosclerotic development, pro-inflammatory cytokines upregulation, and PPARγ/LXRα-ABCA1 downregulation. The same trend was observed in the stressed mice treatment with TAK-242. Further experimental evidences indicated that EP, TAK-242, and RSG treatment notably corrected foam cell formation, HMGB1 release, and down-regulation of LXRα and ABCA1 in CUMS Raw 264.7 macrophage model. Conclusion: These results indicate that CUMS exacerbates atherosclerosis is likely via HMGB1-mediated downregulation of PPARγ/LXRα-ABCA1 through TLR4. These data reveal a novel mechanism by which CUMS aggravates atherosclerosis and may offer a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Hong-Feng Gu
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China
| | - Na Li
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China
| | - Zhao-Qian Xu
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China
| | - Lu Hu
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China
| | - Hui Li
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China
| | - Rong-Jie Zhang
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China
| | - Ru-Meng Chen
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China
| | - Xi-Long Zheng
- Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan, Hunan University of Chinese Medicine, Changsha, China
| | - Ya-Ling Tang
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China
| | - Duan-Fang Liao
- Department of Physiology and Institute of Neuroscience, University of South China, Hengyang, China.,Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
27
|
Wu CY, Zhou ZF, Wang B, Ke ZP, Ge ZC, Zhang XJ. MicroRNA-328 ameliorates oxidized low-density lipoprotein-induced endothelial cells injury through targeting HMGB1 in atherosclerosis. J Cell Biochem 2019; 120:1643-1650. [PMID: 30324654 DOI: 10.1002/jcb.27469] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/18/2018] [Indexed: 01/24/2023]
Abstract
Atherosclerosis has been recognized as a chronic inflammatory disease, which can harden the vessel wall and narrow the arteries. MicroRNAs exhibit crucial roles in various diseases including atherosclerosis. However, so far, the role of miR-328 in atherosclerosis remains barely explored. Therefore, our study concentrated on the potential role of miR-328 in vascular endothelial cell injury during atherosclerosis. In our current study, we observed that oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) apoptosis and inhibited cell viability dose-dependently and time-dependently. In addition, indicated dosage of ox-LDL obviously triggered HUVECs inflammation and oxidative stress process. Then, it was found that miR-328 in HUVECs was reduced by ox-LDL. HUVECs apoptosis was greatly repressed and cell survival was significantly upregulated by overexpression of miR-328. Furthermore, mimics of miR-328 rescued cell inflammation and oxidative stress process induced by ox-LDL. Oppositely, inhibitors of miR-328 strongly promoted ox-LDL-induced endothelial cells injury in HUVECs. By using bioinformatics analysis, high-mobility group box-1 (HMGB1) was predicted as a downstream target of miR-328. HMGB1 has been reported to be involved in atherosclerosis development. The correlation between miR-328 and HMGB1 was validated in our current study. Taken these together, it was implied that miR-328 ameliorated ox-LDL-induced endothelial cells injury through targeting HMGB1 in atherosclerosis.
Collapse
Affiliation(s)
- Chun-Yang Wu
- Department of Cardiology, Yancheng Hospital Affiliated to Southeast University School of Medicine, Yancheng, China
| | - Zhao-Feng Zhou
- Department of Cardiology, Yancheng Hospital Affiliated to Southeast University School of Medicine, Yancheng, China
| | - Bin Wang
- Department of Cardiology, Yancheng Hospital Affiliated to Southeast University School of Medicine, Yancheng, China
| | - Zun-Ping Ke
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Zhong-Chun Ge
- Department of Cardiology, People's Hospital of Xuyi, Xuyi, China
| | - Xian-Jin Zhang
- Department of Intensive Care Unit, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
28
|
RAGE and TLRs as Key Targets for Antiatherosclerotic Therapy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7675286. [PMID: 30225265 PMCID: PMC6129363 DOI: 10.1155/2018/7675286] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/08/2018] [Indexed: 02/08/2023]
Abstract
Receptor for advanced glycation end-products (RAGE) and toll-like receptors (TLRs) are the key factors indicating a danger to the organism. They recognize the microbial origin pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). The primary response induced by PAMPs or DAMPs is inflammation. Excessive stimulation of the innate immune system occurs in arterial wall with the participation of effector cells. Persistent adaptive responses can also cause tissue damage and disease. However, inflammation mediated by the molecules innate responses is an important way in which the adaptive immune system protects us from infection. The specific detection of PAMPs and DAMPs by host receptors drives a cascade of signaling that converges at nuclear factor-κB (NF-κB) and interferon regulatory factors (IRFs) and induces the secretion of proinflammatory cytokines, type I interferon (IFN), and chemokines, which promote direct killing of the pathogen. Therefore, signaling of these receptors' pathways also appear to present new avenue for the modulation of inflammatory responses and to serve as potential novel therapeutic targets for antiatherosclerotic therapy.
Collapse
|
29
|
Roh JS, Sohn DH. Damage-Associated Molecular Patterns in Inflammatory Diseases. Immune Netw 2018; 18:e27. [PMID: 30181915 PMCID: PMC6117512 DOI: 10.4110/in.2018.18.e27] [Citation(s) in RCA: 800] [Impact Index Per Article: 114.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/05/2018] [Accepted: 08/05/2018] [Indexed: 12/23/2022] Open
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous danger molecules that are released from damaged or dying cells and activate the innate immune system by interacting with pattern recognition receptors (PRRs). Although DAMPs contribute to the host's defense, they promote pathological inflammatory responses. Recent studies have suggested that various DAMPs, such as high-mobility group box 1 (HMGB1), S100 proteins, and heat shock proteins (HSPs), are increased and considered to have a pathogenic role in inflammatory diseases. Here, we review current research on the role of DAMPs in inflammatory diseases, including rheumatoid arthritis, systemic lupus erythematosus, osteoarthritis, atherosclerosis, Alzheimer's disease, Parkinson's disease, and cancer. We also discuss the possibility of DAMPs as biomarkers and therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Jong Seong Roh
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan 50612, Korea
| |
Collapse
|
30
|
Abstract
Ovalbumin-induced allergic lung inflammation (ALI) is a condition believed to be mediated by cytokines, extracellular matrix remodeling, and redox imbalance. In this study, we evaluated pulmonary function together with inflammatory markers as interleukin-4 (IL-4), myeloperoxidase (MPO), eosinophil cells, and redox markers in the lungs of BALB/c mice after ovalbumin (OVA) sensitization and challenge. Our results showed an increase in bronchial hyperresponsiveness stimulated by methacholine (Mch), inflammatory cell influx, especially eosinophils together with an increase of high mobility group box 1 (HMGB1) and altered lipid peroxidation (LP) and antioxidant defenses in the OVA group compared to the control group (p ≤ 0.5). Thus, we demonstrated that OVA-induced ALI altered redox status concomitantly with impaired lung function, which was associated with HMGB1 expression and proteolytic remodeling. Taken together all results found here, we may suggest HMGB1 is an important therapeutic target for asthma, once orchestrates the redox signaling, inflammation, and remodeling that contribute to the disease development.
Collapse
|
31
|
Coornaert I, Hofmans S, Devisscher L, Augustyns K, Van Der Veken P, De Meyer GRY, Martinet W. Novel drug discovery strategies for atherosclerosis that target necrosis and necroptosis. Expert Opin Drug Discov 2018; 13:477-488. [PMID: 29598451 DOI: 10.1080/17460441.2018.1457644] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Formation and enlargement of a necrotic core play a pivotal role in atherogenesis. Since the discovery of necroptosis, which is a regulated form of necrosis, prevention of necrotic cell death has become an attractive therapeutic goal to reduce plaque formation. Areas covered: This review highlights the triggers and consequences of (unregulated) necrosis and necroptosis in atherosclerosis. The authors discuss different pharmacological strategies to inhibit necrotic cell death in advanced atherosclerotic plaques. Expert opinion: Addition of a necrosis or necroptosis inhibitor to standard statin therapy could be a promising strategy for primary prevention of cardiovascular disease. However, a necrosis inhibitor cannot block all necrosis stimuli in atherosclerotic plaques. A necroptosis inhibitor could be more effective, because necroptosis is mediated by specific proteins, termed receptor-interacting serine/threonine-protein kinases (RIPK) and mixed lineage kinase domain-like pseudokinase (MLKL). Currently, only RIPK1 inhibitors have been successfully used in atherosclerotic mouse models to inhibit necroptosis. However, because RIPK1 is involved in both necroptosis and apoptosis, and also RIPK1-independent necroptosis can occur, we feel that targeting RIPK3 and MLKL could be a more attractive therapeutic approach to inhibit necroptosis. Therefore, future challenges will consist of developing RIPK3 and MLKL inhibitors applicable in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Isabelle Coornaert
- a Laboratory of Physiopharmacology , University of Antwerp , Wilrijk , Belgium
| | - Sam Hofmans
- b Laboratory of Medicinal Chemistry , University of Antwerp , Wilrijk , Belgium
| | - Lars Devisscher
- b Laboratory of Medicinal Chemistry , University of Antwerp , Wilrijk , Belgium
| | - Koen Augustyns
- b Laboratory of Medicinal Chemistry , University of Antwerp , Wilrijk , Belgium
| | | | - Guido R Y De Meyer
- a Laboratory of Physiopharmacology , University of Antwerp , Wilrijk , Belgium
| | - Wim Martinet
- a Laboratory of Physiopharmacology , University of Antwerp , Wilrijk , Belgium
| |
Collapse
|
32
|
Pivotal neuroinflammatory and therapeutic role of high mobility group box 1 in ischemic stroke. Biosci Rep 2017; 37:BSR20171104. [PMID: 29054968 PMCID: PMC5715129 DOI: 10.1042/bsr20171104] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/29/2017] [Accepted: 10/18/2017] [Indexed: 12/27/2022] Open
Abstract
Stroke is a major cause of mortality and disability worldwide. Stroke is a frequent and severe neurovascular disorder. The main cause of stroke is atherosclerosis, and the most common risk factor for atherosclerosis is hypertension. Therefore, prevention and treatment of stroke are crucial issues in humans. High mobility group box 1 (HMGB1) is non-histone nuclear protein that is currently one of the crucial proinflammatory alarmins in ischemic stroke (IS). It is instantly released from necrotic cells in the ischemic core and activates an early inflammatory response. HMGB1 may signal via its putative receptors, such as receptor for advanced glycation end products (RAGE), toll-like receptors (TLRs) as well as matrix metalloproteinase (MMP) enzymes during IS. These receptors are expressed in brain cells. Additionally, brain-released HMGB1 can be redox modified in the circulation and activate peripheral immune cells. The role of HMGB1 may be more complex. HMGB1 possesses beneficial actions, such as endothelial activation, enhancement of neurite outgrowth, and neuronal survival. HMGB1 may also provide a novel link for brain-immune communication leading to post-stroke immunomodulation. Therefore, HMGB1 is new promising therapeutic intervention aimed at promoting neurovascular repair and remodeling after stroke. In this review, we look at the mechanisms of secretion of HMGB1, the role of receptors, MMP enzymes, hypoglycemia, atherosclerosis, edema, angiogenesis as well as neuroimmunological reactions and post-ischemic brain recovery in IS. We also outline therapeutic roles of HMGB1 in IS.
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The present review explores the mechanisms of superficial intimal erosion, a common cause of thrombotic complications of atherosclerosis. RECENT FINDINGS Human coronary artery atheroma that give rise to thrombosis because of erosion differ diametrically from those associated with fibrous cap rupture. Eroded lesions characteristically contain few inflammatory cells, abundant extracellular matrix, and neutrophil extracellular traps (NETs). Innate immune mechanisms such as engagement of Toll-like receptor 2 (TLR2) on cultured endothelial cells can impair their viability, attachment, and ability to recover a wound. Hyaluronan fragments may serve as endogenous TLR2 ligands. Mouse experiments demonstrate that flow disturbance in arteries with neointimas tailored to resemble features of human eroded plaques disturbs endothelial cell barrier function, impairs endothelial cell viability, recruits neutrophils, and provokes endothelial cells desquamation, NET formation, and thrombosis in a TLR2-dependent manner. SUMMARY Mechanisms of erosion have received much less attention than those that provoke plaque rupture. Intensive statin treatment changes the characteristic of plaques that render them less susceptible to rupture. Thus, erosion may contribute importantly to the current residual burden of risk. Understanding the mechanisms of erosion may inform the development and deployment of novel therapies to combat the remaining atherothrombotic risk in the statin era.
Collapse
Affiliation(s)
- Thibaut Quillard
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Grégory Franck
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Thomas Mawson
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Eduardo Folco
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Peter Libby
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
34
|
Liu H, Lin D, Xiang H, Chen W, Zhao S, Peng H, Yang J, Chen P, Chen S, Lu H. The role of tumor necrosis factor-like weak inducer of apoptosis in atherosclerosis via its two different receptors. Exp Ther Med 2017; 14:891-897. [PMID: 28781615 DOI: 10.3892/etm.2017.4600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 03/31/2017] [Indexed: 12/18/2022] Open
Abstract
At present, it is commonly accepted that atherosclerosis is a chronic inflammatory disease characterized by disorder of the arterial wall. As one of the inflammatory cytokines of the tumor necrosis factor superfamily, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) participates in the formation and progression of atherosclerosis. TWEAK, when binding to its initial receptor, fibroblast growth factor inducible molecule 14 (Fn14), exerts adverse biological functions in atherosclerosis, including dysfunction of endothelial cells, phenotypic change of smooth muscle cells and inflammatory responses of monocytes/macrophages. However, accumulating data supports that, besides Fn14, TWEAK also binds to cluster of differentiation (CD)163, an anti-inflammatory cytokine and a scavenger receptor exclusively expressed by monocytes and macrophages. Furthermore, it has been demonstrated that CD163 is able to internalize TWEAK and likely elicits protective effects in atherosclerosis by terminating inflammation induced by TWEAK. In the present study, the role of TWEAK in atherosclerosis was reviewed, with a predominant focus on CD163 and Fn14 receptors.
Collapse
Affiliation(s)
- Hengdao Liu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Dan Lin
- Qingdao Center for Disease Control and Prevention, Qingdao, Shandong 266033, P.R. China
| | - Hong Xiang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Wei Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shaoli Zhao
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hui Peng
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jie Yang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Pan Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shuhua Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hongwei Lu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
35
|
Wang K, Li W, Yu Q, Guo B, Yang B, Zhang C, Li M, Li J, Hu S, Zheng Q, Song Z. High Mobility Group Box 1 Mediates Interferon-γ-Induced Phenotypic Modulation of Vascular Smooth Muscle Cells. J Cell Biochem 2016; 118:518-529. [PMID: 27579780 DOI: 10.1002/jcb.25682] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/26/2016] [Indexed: 02/06/2023]
Abstract
The phenotypic modulation of VSMCs is a key cellular event driving neointimal formation and vascular remodeling. As a multifaceted cytokine of cell-mediated immunity, IFN-γ has been shown to play a critical role in the pathogenesis of vascular proliferative diseases. Although the important function of IFN-γ on regulating VSMC activation is well established, the molecular mechanisms by which elicits VSMC responses are poorly defined. Recent studies have identified HMGB1 as a principal effector to mediate IFN-γ-dependent biological functions in multiple cell types. Moreover, SIRT1 has emerged as a critical regulator of cellular processes through deacetylating multiple substrates, including HMGB1. Thus, we examined the role of IFN-γ on HMGB1 release, SIRT1 expression, and VSMC phenotypic modulation as well as the underlying molecular mechanisms. We show that IFN-γ dose-dependently induces HMGB1 cytoplasmic accumulation and its active release from VSMCs, resulting in enhanced HMGB1 in the medium. Conversely, IFN-γ treatment led to a dramatic decrease in SIRT1 expression. Additionally, pretreatment with resveratrol, a selective SIRT1 activator, abrogated IFN-γ-induced HMGB1 translocation and its release. Moreover, IFN-γ stimulates VSMC phenotypic modulation to an activated synthetic state characterized by the repression of SMC differentiation markers such as SM22α and calponin and the increase in cell motility. In contrast, blocking HMGB1 release or activity by resveratrol and HMGB1-neutralizing antibody prevents IFN-γ-induced phenotypic modulation of VSMCs. Overall, this study provides the first evidence showing that HMGB1 plays a critical role in regulating VSMC phenotypic modulation, suggesting that HMGB1 may be a potential therapeutic target to prevent vascular occlusive diseases. J. Cell. Biochem. 118: 518-529, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kun Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Department of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qihong Yu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Guo
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, Rochester, New York
| | - Bin Yang
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaobo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qichang Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Mesaros C, Blair IA. Mass spectrometry-based approaches to targeted quantitative proteomics in cardiovascular disease. Clin Proteomics 2016; 13:20. [PMID: 27713681 PMCID: PMC5050566 DOI: 10.1186/s12014-016-9121-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/19/2016] [Indexed: 01/11/2023] Open
Abstract
Mass spectrometry-based proteomics methodology has become an important tool in elucidating some of the underlying mechanisms involved in cardiovascular disease. The present review provides details on selected important protein targets where highly selective and specific mass spectrometry-based approaches have led to important new findings and provided new mechanistic information. The role of six proteins involved in the etiology of cardiovascular disease (acetylated platelet cyclooxygenase-1, serum apolipoprotein A1, apolipoprotein C-III, serum C-reactive protein, serum high mobility group box-1 protein, insulin-like growth factor I) and their quantification has been discussed. There are an increasing number of examples where highly selective mass spectrometry-based quantification has provided new important data that could not be obtained with less labor intensive and cheaper immunoassay-based procedures. It is anticipated that these findings will lead to significant advances in a number of important issues related to the role of specific proteins in cardiovascular disease. The availability of a new generation of high-resolution high-sensitivity mass spectrometers will greatly facilitate these studies so that in the future it will be possible to analyze serum proteins of relevance to cardiovascular disease with levels of specificity and/or sensitivity that cannot be attained by immunoassay-based procedures.
Collapse
Affiliation(s)
- Clementina Mesaros
- Penn SRP Center and Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104 USA ; BluePen Biomarkers, 3401 Grays Ferry Avenue, Philadelphia, PA 19146-2799 USA
| | - Ian A Blair
- Penn SRP Center and Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104 USA ; BluePen Biomarkers, 3401 Grays Ferry Avenue, Philadelphia, PA 19146-2799 USA
| |
Collapse
|
37
|
The Role of TLR2, TLR4, and TLR9 in the Pathogenesis of Atherosclerosis. Int J Inflam 2016; 2016:1532832. [PMID: 27795867 PMCID: PMC5067326 DOI: 10.1155/2016/1532832] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/05/2016] [Accepted: 09/15/2016] [Indexed: 02/07/2023] Open
Abstract
Toll-like receptors (TLRs) are key players in the pathogenesis of inflammatory conditions including coronary arterial disease (CAD). They are expressed by a variety of immune cells where they recognize pathogen-associated molecular patterns (PAMPs). TLRs recruit adaptor molecules, including myeloid differentiation primary response protein (MYD88) and TIRF-related adaptor protein (TRAM), to mediate activation of MAPKs and NF-kappa B pathways. They are associated with the development of CAD through various mechanisms. TLR4 is expressed in lipid-rich and atherosclerotic plaques. In TLR2−/− and TLR4−/− mice, atherosclerosis-associated inflammation was diminished. Moreover, TLR2 and TLR4 may induce expression of Wnt5a in advanced staged atheromatous plaque leading to activation of the inflammatory processes. TLR9 is activated by CpG motifs in nucleic acids and have been implicated in macrophage activation and the uptake of oxLDL from the circulation. Furthermore, TLR9 also stimulates interferon-α (INF-α) secretion and increases cytotoxic activity of CD4+ T-cells towards coronary artery tunica media smooth muscle cells. This review outlines the pathophysiological role of TLR2, TLR4, and TLR9 in atherosclerosis, focusing on evidence from animal models of the disease.
Collapse
|
38
|
Kim J, Kim CS, Sohn E, Kim JS. Cytoplasmic translocation of high-mobility group box-1 protein is induced by diabetes and high glucose in retinal pericytes. Mol Med Rep 2016; 14:3655-61. [PMID: 27599553 PMCID: PMC5042730 DOI: 10.3892/mmr.2016.5702] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 07/18/2016] [Indexed: 01/13/2023] Open
Abstract
The aim of the present study was to assess the involvement of the high-mobility group box-1 (HMGB1) protein, receptor for advanced glycation end products (RAGE) and nuclear factor (NF)-κB signaling pathway in the development of diabetic retinopathy. Rat primary retinal pericytes were exposed to 25 mmol/l D-glucose for 48 h. Diabetic retinal vessels were prepared from streptozotocin-induced diabetic rats 12 weeks following the induction of diabetes. The expression of HMGB1 was detected using immunofluorescence staining. The expression of RAGE and the activity of NF-κB were analyzed using western blot and electrophoretic mobility shift assays, respectively. The results showed that HMGB1 was translocated to the cytoplasm of the high glucose-treated pericytes and diabetic retinal pericytes, whereas, in the control cells and the normal retinas, HMGB1 was expressed in the cell nuclei only. The expression of RAGE, a potential receptor for HMGB1, and the activity of NF-κB were also increased in the high glucose-treated pericytes, compared with the normal control cells. In addition, high glucose increased the binding of NF-κB to the RAGE promoter. These findings suggested that the cytoplasmic translocation of HMGB1 may be caused by diabetes and high glucose in retinal pericytes, and that the pathogenic role of HMGB1 may be dependent on the expression of RAGE and activation of NF-κB.
Collapse
Affiliation(s)
- Junghyun Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Chan-Sik Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Eunjin Sohn
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Jin Sook Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| |
Collapse
|
39
|
HMGB1, IL-1α, IL-33 and S100 proteins: dual-function alarmins. Cell Mol Immunol 2016; 14:43-64. [PMID: 27569562 PMCID: PMC5214941 DOI: 10.1038/cmi.2016.34] [Citation(s) in RCA: 347] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/08/2023] Open
Abstract
Our immune system is based on the close collaboration of the innate and adaptive immune systems for the rapid detection of any threats to the host. Recognition of pathogen-derived molecules is entrusted to specific germline-encoded signaling receptors. The same receptors have now also emerged as efficient detectors of misplaced or altered self-molecules that signal tissue damage and cell death following, for example, disruption of the blood supply and subsequent hypoxia. Many types of endogenous molecules have been shown to provoke such sterile inflammatory states when released from dying cells. However, a group of proteins referred to as alarmins have both intracellular and extracellular functions which have been the subject of intense research. Indeed, alarmins can either exert beneficial cell housekeeping functions, leading to tissue repair, or provoke deleterious uncontrolled inflammation. This group of proteins includes the high-mobility group box 1 protein (HMGB1), interleukin (IL)-1α, IL-33 and the Ca2+-binding S100 proteins. These dual-function proteins share conserved regulatory mechanisms, such as secretory routes, post-translational modifications and enzymatic processing, that govern their extracellular functions in time and space. Release of alarmins from mesenchymal cells is a highly relevant mechanism by which immune cells can be alerted of tissue damage, and alarmins play a key role in the development of acute or chronic inflammatory diseases and in cancer development.
Collapse
|
40
|
Sharma AK, Salmon MD, Lu G, Su G, Pope NH, Smith JR, Weiss ML, Upchurch GR. Mesenchymal Stem Cells Attenuate NADPH Oxidase-Dependent High Mobility Group Box 1 Production and Inhibit Abdominal Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2016; 36:908-18. [PMID: 26988591 DOI: 10.1161/atvbaha.116.307373] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/23/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) formation is characterized by inflammation, smooth muscle activation, and matrix degradation. This study tests the hypothesis that macrophage-produced high mobility group box 1 (HMGB1) production is dependent on nicotinamide adenine dinucleotide phosphate oxidase (Nox2), which leads to increase in interleukin (IL)-17 production resulting in AAA formation and that treatment with human mesenchymal stem cells (MSCs) can attenuate this process thereby inhibiting AAA formation. APPROACH AND RESULTS Human aortic tissue demonstrated a significant increase in HMGB1 expression in AAA patients when compared with controls. An elastase-perfusion model of AAA demonstrated a significant increase in HMGB1 production in C57BL/6 (wild-type [WT]) mice, which was attenuated by MSC treatment. Furthermore, anti-HMGB1 antibody treatment of WT mice attenuated AAA formation, IL-17 production, and immune cell infiltration when compared with elastase-perfused WT mice on day 14. Elastase-perfused Nox2(-/y) mice demonstrated a significant attenuation of HMGB1 and IL-17 production, cellular infiltration, matrix metalloproteinase activity, and AAA formation when compared with WT mice on day 14. In vitro studies showed that elastase-treated macrophages from WT mice, but not from Nox2(-/y) mice, produced HMGB1, which was attenuated by MSC treatment. The production of macrophage-dependent HMGB1 involved Nox2 activation and superoxide anion production, which was mitigated by MSC treatment. CONCLUSIONS These results demonstrate that macrophage-produced HMGB1 leads to aortic inflammation and acts as a trigger for CD4(+) T-cell-produced IL-17 during AAA formation. HMGB1 release is dependent on Nox2 activation, which can be inhibited by MSCs leading to attenuation of proinflammatory cytokines, especially IL-17, and protection against AAA formation.
Collapse
Affiliation(s)
- Ashish K Sharma
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Morgan D Salmon
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Guanyi Lu
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Gang Su
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Nicolas H Pope
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Joseph R Smith
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Mark L Weiss
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Gilbert R Upchurch
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.).
| |
Collapse
|
41
|
Rojas A, Delgado-López F, Perez-Castro R, Gonzalez I, Romero J, Rojas I, Araya P, Añazco C, Morales E, Llanos J. HMGB1 enhances the protumoral activities of M2 macrophages by a RAGE-dependent mechanism. Tumour Biol 2016; 37:3321-3329. [PMID: 26440051 DOI: 10.1007/s13277-015-3940-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/17/2015] [Indexed: 02/07/2023] Open
Abstract
The monocyte-macrophage lineage shows a high degree of diversity and plasticity. Once they infiltrate tissues, they may acquire two main functional phenotypes, being known as the classically activated type 1 macrophages (M1) and the alternative activated type 2 macrophages (M2). The M1 phenotype can be induced by bacterial products and interferon-γ and exerts a cytotoxic effect on cancer cells. Conversely, the alternatively activated M2 phenotype is induced by Il-4/IL13 and promotes tumor cell growth and vascularization. Although receptor for advanced glycation end-products (RAGE) engagement in M1 macrophages has been reported by several groups to promote inflammation, nothing is known about the functionality of RAGE in M2 macrophages. In the current study, we demonstrate that RAGE is equally expressed in both macrophage phenotypes and that RAGE activation by high-mobility group protein box1 (HMGB1) promotes protumoral activities of M2 macrophages. MKN45 cells co-cultured with M2 macrophages treated with HMGB1 at different times displayed higher invasive abilities. Additionally, conditioned medium from HMGB1-treated M2 macrophages promotes angiogenesis in vitro. RAGE-targeting knockdown abrogates these activities. Overall, the present findings suggest that HMGB1 may contribute, by a RAGE-dependent mechanism, to the protumoral activities of the M2 phenotype.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, 3605 San Miguel Ave., Talca, Chile.
| | - Fernando Delgado-López
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, 3605 San Miguel Ave., Talca, Chile
| | - Ramón Perez-Castro
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, 3605 San Miguel Ave., Talca, Chile
| | - Ileana Gonzalez
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, 3605 San Miguel Ave., Talca, Chile
| | - Jacqueline Romero
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, 3605 San Miguel Ave., Talca, Chile
| | - Israel Rojas
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, 3605 San Miguel Ave., Talca, Chile
| | - Paulina Araya
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, 3605 San Miguel Ave., Talca, Chile
| | - Carolina Añazco
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, 3605 San Miguel Ave., Talca, Chile
| | | | - Jorge Llanos
- Gastroenterology Unit, Regional Hospital, Talca, Chile
| |
Collapse
|
42
|
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
|
43
|
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
|
44
|
Cai J, Yuan H, Wang Q, Yang H, Al-Abed Y, Hua Z, Wang J, Chen D, Wu J, Lu B, Pribis JP, Jiang W, Yang K, Hackam DJ, Tracey KJ, Billiar TR, Chen AF. HMGB1-Driven Inflammation and Intimal Hyperplasia After Arterial Injury Involves Cell-Specific Actions Mediated by TLR4. Arterioscler Thromb Vasc Biol 2015; 35:2579-93. [PMID: 26515416 DOI: 10.1161/atvbaha.115.305789] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/02/2015] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Endoluminal vascular interventions such as angioplasty initiate a sterile inflammatory response resulting from local tissue damage. This response drives the development of intimal hyperplasia (IH) that, in turn, can lead to arterial occlusion. We hypothesized that the ubiquitous nuclear protein and damage-associated molecular pattern molecule, high-mobility group box 1 (HMGB1), is one of the endogenous mediators that activates processes leading to IH after endoluminal injury to the arterial wall. The aim of this study is to investigate whether approaches that reduce the levels of HMGB1 or inhibit its activity suppresses IH after arterial injury. APPROACH AND RESULTS Here, we show that HMGB1 regulates IH in a mouse carotid wire injury model. Induced genetic deletion or neutralization of HMGB1 prevents IH, monocyte recruitment, and smooth muscle cell growth factor production after endoluminal carotid artery injury. A specific inhibitor of HMGB1 myeloid differentiation factor 2-toll-like receptor 4 (TLR4) interaction, P5779, also significantly inhibits IH. HMGB1 deletion is mimicked in this model by global deletion of TLR4 and partially replicated by myeloid-specific deletion of TLR4 but not TLR2 or receptor for advanced glycation endproducts deletion. The specific HMGB1 isoform known to activate TLR4 signaling (disulfide HMGB1) stimulates smooth muscle cell to migrate and produce monocyte chemotactic protein 1/CCL2) via TLR4. Macrophages produce smooth muscle cell mitogens in response to disulfide HMGB1 also in a TLR4/myeloid differentiation primary response gene (88)/Trif-dependent manner. CONCLUSIONS These findings place HMGB1 and its receptor, TLR4 as critical regulators of the events that drive the inflammation leading to IH after endoluminal arterial injury and identify this pathway as a possible therapeutic target to limit IH to attenuate damage-associated molecular pattern molecule-mediated vascular inflammatory responses.
Collapse
Affiliation(s)
- Jingjing Cai
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Hong Yuan
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Qingde Wang
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Huan Yang
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Yousef Al-Abed
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Zhong Hua
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Jiemei Wang
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Dandan Chen
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Jinze Wu
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Ben Lu
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - John P Pribis
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Weihong Jiang
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Kan Yang
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - David J Hackam
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Kevin J Tracey
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Timothy R Billiar
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.)
| | - Alex F Chen
- From the Center of Clinical Pharmacology of the Third Xiangya Hospital (J.C., H.Y., Q.W., Z.H., J. Wu), the Center of Vascular Disease and Translational Medicine (A.F.C.), Department of Cardiology of the Third Xiangya Hospital (J.C., H.Y., W.J., K.Y.), and Department of Hematology of the Third Xiangya Hospital (B.L.), Central South University, Changsha, China; Department of Surgery, University of Pittsburgh School of Medicine, PA (J.C., Q.W., Z.H., J. Wang, D.C., J. Wu, J.P.P., D.J.H., T.R.B., A.F.C.); and Laboratory of Biomedical Science, the Feinstein Institute for Medical Research, Manhasset, New York (H.Y., Y.A.-A., K.J.T.).
| |
Collapse
|
45
|
Ozkan C, Altinova AE, Cerit ET, Yayla C, Sahinarslan A, Sahin D, Dincel AS, Toruner FB, Akturk M, Arslan M. Markers of early atherosclerosis, oxidative stress and inflammation in patients with acromegaly. Pituitary 2015; 18:621-9. [PMID: 25500981 DOI: 10.1007/s11102-014-0621-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Data regarding atherosclerosis in acromegaly is controversial in literature. We aimed to investigate the markers of early atherosclerosis, oxidative stress, inflammation and their relationships with each other in acromegaly. METHODS Thirty-nine patients with acromegaly and 40 control subjects were enrolled. Patients were classified into two groups; active acromegaly (AA) and controlled acromegaly (CA). Controls were matched by age, gender, body mass index and presence of cardiovascular risk factors. Flow mediated dilatation (FMD), carotid intima media thickness (CIMT), epicardial adipose tissue thickness (EAT) were measured and serum levels of oxidative stress parameters, high mobility group box 1 protein (HMGB1) and high sensitive CRP (hs CRP) were evaluated. RESULTS Significantly decreased FMD, increased CIMT and EAT were found in patients with acromegaly compared to controls (p < 0.01, p < 0.05, p < 0.001, respectively). EAT correlated negatively with FMD (r = -0.24, p = 0.038) and positively with CIMT (r = 0.37, p < 0.01). Presence of acromegaly, hypertension and age were found to be the predictors of early atherosclerosis (p < 0.05). Hs CRP was decreased in AA compared to controls (p = 0.01). There were no significant differences for HMGB1 and oxidized LDL (ox-LDL) cholesterol levels and total antioxidant capacity (TAC) between AA, CA and controls (p > 0.05). CONCLUSION Early atherosclerosis measured with FMD, CIMT and EAT may exist in acromegaly. However, decreased hs CRP and unchanged HMGB1, ox-LDL and TAC levels suggest that inflammation and oxidative stress do not seem to contribute to the development of atherosclerosis in these patients.
Collapse
Affiliation(s)
- Cigdem Ozkan
- Department of Endocrinology and Metabolism, Gazi University Faculty of Medicine, Besevler, 06500, Ankara, Turkey,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ahrens I, Chen YC, Topcic D, Bode M, Haenel D, Hagemeyer CE, Seeba H, Duerschmied D, Bassler N, Jandeleit-Dahm KA, Sweet MJ, Agrotis A, Bobik A, Peter K. HMGB1 binds to activated platelets via the receptor for advanced glycation end products and is present in platelet rich human coronary artery thrombi. Thromb Haemost 2015. [PMID: 26202300 DOI: 10.1160/th14-12-1073] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
High mobility group box 1 (HMGB1) acts as both a nuclear protein that regulates gene expression, as well as a pro-inflammatory alarmin that is released from necrotic or activated cells. Recently, HMGB1-expression in human atherosclerotic plaques was identified. Therapeutic blockade of HMGB1 reduced the development of diet-induced atherosclerosis in ApoE knockout mice. Thus, we hypothesised an interaction between HMGB1 and activated platelets. Binding of recombinant HMGB1 to platelets was assessed by flow cytometry. HMGB1 bound to thrombin-activated human platelets (MFI 2.49 vs 25.01, p=0.0079). Blood from wild-type, TLR4 and RAGE knockout mice was used to determine potential HMGB1 receptors on platelets. HMGB1 bound to platelets from wild type C57Bl6 (MFI 2.64 vs 20.3, p< 0.05), and TLR4-/- mice (MFI 2.11 vs 25.65, p< 0.05) but failed to show binding to platelets from RAGE-/- mice (p > 0.05). RAGE expression on human platelets was detected by RT-PCR with mRNA extracted from highly purified platelets and confirmed by Western blot and immunofluorescence microscopy. Platelet activation increased RAGE surface expression (MFI 4.85 vs 6.74, p< 0.05). Expression of HMGB1 in human coronary artery thrombi was demonstrated by immunohistochemistry and revealed high expression levels. Platelets bind HMGB1 upon thrombin-induced activation. Platelet specific expression of RAGE could be detected at the mRNA and protein level and is involved in the binding of HMGB1. Furthermore, platelet activation up-regulates platelet surface expression of RAGE. HMGB1 is highly expressed in platelet-rich human coronary artery thrombi pointing towards a central role for HMGB1 in atherothrombosis, thereby suggesting the possibility of platelet targeted anti-inflammatory therapies for atherothrombosis.
Collapse
Affiliation(s)
- Ingo Ahrens
- PD Dr. Ingo Ahrens, Heart Center, University of Freiburg, Cardiology and Angiology I, Hugstetter Str. 55, 79106 Freiburg, Germany, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hu H, Zhai C, Qian G, Gu A, Liu J, Ying F, Xu W, Jin D, Wang H, Hu H, Zhang Y, Tang G. Protective effects of tanshinone IIA on myocardial ischemia reperfusion injury by reducing oxidative stress, HMGB1 expression, and inflammatory reaction. PHARMACEUTICAL BIOLOGY 2015; 53:1752-1758. [PMID: 25864557 DOI: 10.3109/13880209.2015.1005753] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Although there were reports on the protective functions of tanshinone IIA (TSA) on rat myocardial ischemia, the exerting mechanism has not been completely clarified. OBJECTIVE An attempt was made to further verify the protective effect of TSA on myocardial ischemia reperfusion injury and elucidate its underlying mechanism. MATERIALS AND METHODS The rats were given TSA (10, 20, and 40 mg/kg bw per day) in intraperitoneal injection for 15 d. Rami anterior descending branch of coronary artery was ligated for 30 min and then re-perfused for 120 min to establish a reperfusion model. Effects of TSA on the infarct area, creatine kinase (CK), aspartate aminotransferase (AST), high mobility group box B1 protein (HMGB1), and inflammation and oxidation were investigated. RESULTS Compared with those in the IR group, infarct size percentages of rats' myocardium in L-TSA, M-TSA, and H-TSA groups were reduced by 1.21, 4.26, and 12.50%, respectively, CK activities by 7.4, 11.2, and 12.5%, respectively, and AST activities also declined (p < 0.05). Furthermore, compared with those in the IR group, SOD and GSH-Px activities increased, and MDA, TNF-α, IL-6, and iNOS levels decreased in L-TSA, M-TSA, and H-TSA groups (p < 0.05). Meanwhile, compared with those in the IR group, HMGB1 expressions in L-TSA, M-TSA, and H-TSA groups were lowered by 21.9, 32.4, and 35.6%, respectively. DISCUSSION AND CONCLUSION The protective function of TSA on myocardial ischemia reperfusion injury may be possibly exerted by inhibiting the increase of ROS caused by the reperfusion to attenuate the expression of HMGB1 and inhibit inflammation.
Collapse
Affiliation(s)
- Huilin Hu
- Department of Cardiology, First Affiliated Hospital of Jiaxing University , Jiaxing, Zhejiang , PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Liu T, Zhang DY, Zhou YH, Han QF, Wang LH, Wu L, Yao HC. Increased serum HMGB1 level may predict the fatal outcomes in patients with chronic heart failure. Int J Cardiol 2015; 184:318-320. [DOI: 10.1016/j.ijcard.2015.02.088] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/24/2015] [Indexed: 01/09/2023]
|
49
|
Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med 2014; 40:1-116. [PMID: 25010388 PMCID: PMC4254084 DOI: 10.1016/j.mam.2014.05.001] [Citation(s) in RCA: 740] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Complex genetic and physiological variations as well as environmental factors that drive emergence of chromosomal instability, development of unscheduled cell death, skewed differentiation, and altered metabolism are central to the pathogenesis of human diseases and disorders. Understanding the molecular bases for these processes is important for the development of new diagnostic biomarkers, and for identifying new therapeutic targets. In 1973, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and termed high-mobility group (HMG) proteins. The HMG proteins include three superfamilies termed HMGB, HMGN, and HMGA. High-mobility group box 1 (HMGB1), the most abundant and well-studied HMG protein, senses and coordinates the cellular stress response and plays a critical role not only inside of the cell as a DNA chaperone, chromosome guardian, autophagy sustainer, and protector from apoptotic cell death, but also outside the cell as the prototypic damage associated molecular pattern molecule (DAMP). This DAMP, in conjunction with other factors, thus has cytokine, chemokine, and growth factor activity, orchestrating the inflammatory and immune response. All of these characteristics make HMGB1 a critical molecular target in multiple human diseases including infectious diseases, ischemia, immune disorders, neurodegenerative diseases, metabolic disorders, and cancer. Indeed, a number of emergent strategies have been used to inhibit HMGB1 expression, release, and activity in vitro and in vivo. These include antibodies, peptide inhibitors, RNAi, anti-coagulants, endogenous hormones, various chemical compounds, HMGB1-receptor and signaling pathway inhibition, artificial DNAs, physical strategies including vagus nerve stimulation and other surgical approaches. Future work further investigating the details of HMGB1 localization, structure, post-translational modification, and identification of additional partners will undoubtedly uncover additional secrets regarding HMGB1's multiple functions.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | - Ruochan Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Qiuhong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Wen Hou
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Sha Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xue-Gong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhengwen Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Experimental Department of Institute of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Herbert J Zeh
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| |
Collapse
|
50
|
Yang PS, Kim DH, Lee YJ, Lee SE, Kang WJ, Chang HJ, Shin JS. Glycyrrhizin, inhibitor of high mobility group box-1, attenuates monocrotaline-induced pulmonary hypertension and vascular remodeling in rats. Respir Res 2014; 15:148. [PMID: 25420924 PMCID: PMC4248446 DOI: 10.1186/s12931-014-0148-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 11/10/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND High mobility group box-1 (HMGB1), a proinflammatory cytokine, plays a pivotal role in tissue remodeling and angiogenesis, both of which are crucial for the pathogenesis of pulmonary arterial hypertension. In this study, we explored the relationship between HMGB1 and pulmonary hypertension and whether glycyrrhizin, an inhibitor of HMGB1, attenuates disease progression in an animal model of pulmonary hypertension induced by monocrotaline sodium (MCT). METHODS After inducing pulmonary hypertension through a single subcutaneous injection of MCT (60 mg/kg) to Sprague-Dawley rats, we administered daily intraperitoneal injections of either glycyrrhizin (GLY, 50 mg/kg), an inhibitor of HMGB1, or saline (control) for either 4 or 6 weeks. RESULTS Expression levels of HMGB1 in serum increased from the second week after MCT injection and remained elevated throughout the experiment periods. Lung tissue levels of HMGB1 assessed by immunohistochemical staining at 4 weeks after MCT injection also increased. Chronic inhibition of HMGB1 by GLY treatment reduced the MCT-induced increase in right ventricular (RV) systolic pressure, RV hypertrophy (ratio of RV to [left ventricle + septum]), and pulmonary inflammation. MCT-induced muscularization of the pulmonary artery was also attenuated in the GLY-treated group. As assessed 6 weeks after MCT injection, the GLY-treated group exhibited increased survival (90% [18 of 20]) when compared with the control group (60% [12 of 20]; p =0.0027). CONCLUSIONS Glycyrrhizin, an inhibitor of HMGB1, attenuates pulmonary hypertension progression and pulmonary vascular remodeling in the MCT-induced pulmonary hypertension rat model. Further studies are needed to confirm the potential of HMGB1 as a novel therapeutic target for pulmonary hypertension.
Collapse
Affiliation(s)
- Pil-Sung Yang
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Dae-Hoon Kim
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Yong Joon Lee
- Departments of Microbiology, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Sang-Eun Lee
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Won Jun Kang
- Departments of Nuclear Medicine, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Hyuk-Jae Chang
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea. .,Severance Biomedical Science Institute, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| | - Jeon-Soo Shin
- Departments of Microbiology, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea. .,Severance Biomedical Science Institute, Yonsei University Health System, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| |
Collapse
|