1
|
Niu J, Ran Y, Chen R, Zhang Y, Zhang Y, Yang Q, Cheng J. Evaluation of Middle Cerebral Artery Culprit Plaque Inflammation in Ischemic Stroke Using CAIPIRINHA-Dixon-TWIST Dynamic Contrast-Enhanced Magnetic Resonance Imaging. J Magn Reson Imaging 2025; 61:2011-2020. [PMID: 39258494 DOI: 10.1002/jmri.29576] [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: 06/06/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Middle cerebral artery (MCA) plaques are a leading cause of ischemic stroke (IS). Plaque inflammation is crucial for plaque stability and urgently needs quantitative detection. PURPOSE To explore the utility of Controlled Aliasing in Parallel Imaging Results in Higher Acceleration (CAIPIRINHA)-Dixon-Time-resolved angiography With Interleaved Stochastic Trajectories (TWIST) (CDT) dynamic contrast-enhanced MRI (DCE-MRI) for evaluating MCA culprit plaque inflammation changes over stroke time and with diabetes mellitus (DM). STUDY TYPE Prospective. POPULATION Ninety-four patients (51.6 ± 12.23 years, 32 females, 23 DM) with acute IS (AIS; N = 43) and non-acute IS (non-AIS; 14 days < stroke time ≤ 3 months; N = 51). FIELD STRENGTH/SEQUENCE 3-T, CDT DCE-MRI and three-dimensional (3D) Sampling Perfection with Application optimized Contrast using different flip angle Evolution (3D-SPACE) T1-weighted imaging (T1WI). ASSESSMENT Stroke time (from initial IS symptoms to MRI) and DM were registered. For 94 MCA culprit plaques, Ktrans from CDT DCE-MRI and enhancement ratio (ER) from 3D-SPACE T1WI were compared between groups with and without AIS and DM. STATISTICAL TESTS Shapiro-Wilk test, Bland-Altman analysis, Passing and Bablok test, independent t-test, Mann-Whitney U test, Chi-squared test, Fisher's exact test, receiver operating characteristics (ROC) with the area under the curve (AUC), DeLong's test, and Spearman rank correlation test with the P-value significance level of 0.05. RESULTS Ktrans and ER of MCA culprit plaques were significantly higher in AIS than non-AIS patients (Ktrans = 0.098 s-1 vs. 0.037 s-1; ER = 0.86 vs. 0.55). Ktrans showed better AUC for distinguishing AIS from non-AIS patients (0.87 vs. 0.75) and stronger negative correlation with stroke time than ER (r = -0.60 vs. -0.34). DM patients had significantly higher Ktrans and ER than non-DM patients in IS and AIS groups. DATA CONCLUSION Imaging by CDT DCE-MRI may allow to quantitatively evaluate MCA culprit plaques over stroke time and DM. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Junxia Niu
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Magnetic Resonance, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuncai Ran
- Department of Magnetic Resonance, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Chen
- Department of Magnetic Resonance, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Zhang
- Department of Magnetic Resonance, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Zhang
- Department of Magnetic Resonance, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jingliang Cheng
- Department of Magnetic Resonance, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Chen Y, Fang C, Zhao J, Jiang S, Xu X, Cui L, Zhao R, Ma X, Yu H, Wei G, Liu Y, Yu B, Dai J, Yang S. Glycemic control and coronary plaque characteristics in patients with acute myocardial infarction. Int J Cardiol 2025; 423:132988. [PMID: 39826579 DOI: 10.1016/j.ijcard.2025.132988] [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: 11/12/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND The impact of glycemic control on the morphological characteristics of non-culprit lesions (NCLs) in patients with acute myocardial infarction (AMI) remains unclear. METHODS AND RESULTS A total of 800 AMI patients who underwent 3-vessel OCT were divided into three groups based on their serum glycated hemoglobin (HbA1c) levels: poorly controlled diabetes mellitus (DM) (HbA1c ≥8.0 %, n = 79), well controlled DM (6.5 % ≤ HbA1c < 8.0 %, n = 105), and non-DM (HbA1c <6.5 %, n = 616). OCT imaging was performed on all NCLs and plaque characteristics were assessed at both the patient and lesion level, including high-risk features. Separate covariate-adjusted multivariate models were performed to determine whether glycemic control was associated with high-risk plaque features in NCLs. Patients with poorly controlled DM had higher NCL vulnerability compared to those with well controlled DM and non-DM, both at the patient level [thin-cap fibroatheroma (TCFA): 63.3 % vs. 32.4 % vs. 27.1 %] and the lesion level [TCFA: 22.1 % vs. 11.6 % vs. 9.5 %; non-culprit plaque rupture (PR): 8.4 % vs. 4.6 % vs. 4.6 %]. Vulnerable plaque features, including TCFA, non-culprit PR, macrophages, microchannels, cholesterol crystals, layered plaque and calcification, were more prevalent with increasing HbA1c levels. Multivariate analysis revealed that at the patient level, poorly controlled DM was an independent predictor of TCFA and microchannels, whereas at the lesion level it independently predicted TCFA and non-culprit PR. CONCLUSIONS In AMI patients, poor glycemic control is associated with increased vulnerability of NCLs. As HbA1c levels increase, there is a worsening of plaque characteristics, including greater plaque burden and more advanced features of vulnerability.
Collapse
Affiliation(s)
- Yuzhu Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Chao Fang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Jiawei Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Senqing Jiang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Xueming Xu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Lina Cui
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Rui Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Xianqin Ma
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Huai Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Guo Wei
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Yanchao Liu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin 150086, China
| | - Jiannan Dai
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin 150086, China.
| | - Shuang Yang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China, Harbin 150086, China.
| |
Collapse
|
3
|
Li W, Xu G, Chai GW, Ball A, Zhang Q, Kutryk MJB. The MiR-139-5p and CXCR4 axis may play a role in high glucose-induced inflammation by regulating monocyte migration. Sci Rep 2025; 15:6738. [PMID: 40000897 PMCID: PMC11861593 DOI: 10.1038/s41598-025-91100-1] [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: 10/03/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
MicroRNAs, a class of small non-coding RNA molecules that regulate gene expression post-transcriptionally, are implicated in various pathological conditions including diabetes mellitus (DM). DM has been increasingly recognized as an inflammatory disease and monocytes play a key role in propagating inflammation under hyperglycemic conditions. We hypothesize that high glucose dysregulates microRNAs to promote monocyte inflammatory activity, which may contribute to the pathogenesis of DM. THP-1 monocytes were cultured in normal (5 mM) and high (25 mM) glucose conditions. RT-qPCR and Western blotting were performed to assay microRNAs and proteins, respectively. Monocytes were transfected with microRNA mimics using Lipofectamine RNAiMAX reagent. THP-1 monocyte growth was assessed using Calcein-AM dye and a Boyden chamber assay was applied to measure monocyte migration. The results showed that high glucose downregulated miR-139-5p associated with increased protein expression of CXCR4, an experimentally validated target of miR-139-5p. Correspondingly, treatment with high glucose resulted in a significant increase in THP-1 cell migration towards SDF-1, a cognate ligand for CXCR4. MiR-139-5p overexpression inhibited high glucose-induced CXCR4 expression, leading to reduced cell migration towards SDF-1. High glucose did not affect THP-1 monocyte growth. In conclusion, the miR-139-5p-CXCR4 axis may play a role in high glucose-induced inflammation by regulating monocyte migration.
Collapse
Affiliation(s)
- Weifang Li
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Gengchen Xu
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Gregory W Chai
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Alexander Ball
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Qiuwang Zhang
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada.
| | - Michael J B Kutryk
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Viggiano D, Joshi R, Borriello G, Cacciola G, Gonnella A, Gigliotti A, Nigro M, Gigliotti G. SGLT2 Inhibitors: The First Endothelial-Protector for Diabetic Nephropathy. J Clin Med 2025; 14:1241. [PMID: 40004772 PMCID: PMC11856817 DOI: 10.3390/jcm14041241] [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: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Sodium-glucose co-transporter type 2 inhibitors (SGLT2i) have emerged as a class of agents relevant for managing diabetic nephropathy and cardiopathy. In a previous report, we noticed that these drugs share, with other drugs with "nephroprotective" effects, the ability to reduce the glomerular filtration rate (GFR), thus suggesting the kidney hemodynamic effect as a proxy for optimal drug dosage. We also noticed that all known nephroprotective drugs exert cardioprotective functions, suggesting the possibility of activities not mediated by the kidney. Finally, we observe that nephroprotective drugs can be grouped according to their effects on hemoglobin levels, thus suggesting their mechanism of action. While the primary mechanism of SGLT2i involves glycosuria and natriuria, growing evidence suggests broader therapeutic effects beyond hemodynamic modulation. Specifically, the evidence that SGLT2 can be expressed in several atypical regions under pathological conditions, supports the possibility that its inhibition has several extratubular effects. Evidence supports the hypothesis that SGLT2i influence mitochondrial function in various cell types affected by diabetes, particularly in the context of diabetic nephropathy. Notably, in SGLT2i-treated patients, the extent of albumin-creatinine ratio (ACR) reduction post-treatment may be correlated with mitochondrial staining intensity in glomerular endothelial cells. This implies that the anti-proteinuric effects of SGLT2i could involve direct actions on glomerular endothelial cell. Our investigation into the role of SGLT2 inhibitors (SGLT2i) in endothelial function suggests that the aberrant expression of SGLT2 in endothelial cells in T2DM would lead to intracellular accumulation of glucose; therefore, SGLT2i are the first type of endothelial protective drugs available today, with potential implications for ageing-related kidney disease. The review reveals two major novel findings: SGLT2 inhibitors are the first known class of endothelial-protective drugs, due to their ability to prevent glucose accumulation in endothelial cells where SGLT2 is aberrantly expressed in Type 2 Diabetes. Additionally, the research demonstrates that SGLT2 inhibitors share a GFR-reducing effect with other nephroprotective drugs, suggesting both a mechanism for optimal drug dosing and potential broader applications in ageing-related kidney disease through their effects on mitochondrial function and glomerular endothelial cells.
Collapse
Affiliation(s)
- Davide Viggiano
- Department Translational Medical Sciences, University of Campania, 80138 Naples, Italy; (R.J.); (G.B.); (G.C.)
| | - Rashmi Joshi
- Department Translational Medical Sciences, University of Campania, 80138 Naples, Italy; (R.J.); (G.B.); (G.C.)
| | - Gianmarco Borriello
- Department Translational Medical Sciences, University of Campania, 80138 Naples, Italy; (R.J.); (G.B.); (G.C.)
| | - Giovanna Cacciola
- Department Translational Medical Sciences, University of Campania, 80138 Naples, Italy; (R.J.); (G.B.); (G.C.)
| | - Annalisa Gonnella
- Department Nephrology, Eboli Hospital, 84025 Eboli, Italy; (A.G.); (A.G.); (M.N.); (G.G.)
| | - Andrea Gigliotti
- Department Nephrology, Eboli Hospital, 84025 Eboli, Italy; (A.G.); (A.G.); (M.N.); (G.G.)
| | - Michelangelo Nigro
- Department Nephrology, Eboli Hospital, 84025 Eboli, Italy; (A.G.); (A.G.); (M.N.); (G.G.)
| | - Giuseppe Gigliotti
- Department Nephrology, Eboli Hospital, 84025 Eboli, Italy; (A.G.); (A.G.); (M.N.); (G.G.)
| |
Collapse
|
5
|
Gebert ZM, Kwiecinski J, Weir-McCall JR, Adamson PD, Mills NL, Roditi G, van Beek EJR, Nicol ED, Berman DS, Slomka PJ, Dweck MR, Dey D, Newby DE, Williams MC. Impact of diabetes mellitus on coronary artery plaque characteristics and outcomes in the SCOT-HEART trial. J Cardiovasc Comput Tomogr 2025:S1934-5925(24)00583-5. [PMID: 39788814 DOI: 10.1016/j.jcct.2024.12.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Diabetes mellitus is an established cardiovascular risk factor. We assessed the impact of diabetes mellitus on quantitative plaque and long-term outcomes in patients with and without diabetes mellitus in the Scottish COmputed Tomography of the HEART (SCOT-HEART) trial. METHODS Coronary artery calcium (CAC) was assessed on non-contrast computed tomography (CT). Coronary stenoses, visually assessed adverse plaque characteristics and quantitative plaque burdens (calcified, non-calcified, low attenuation and total, Autoplaque 2.5) were assessed on coronary CT angiography. Multivariable and survival analyses were performed. RESULTS Images of 1769 patients were assessed (56 % male, 58 ± 9 years). Diabetes mellitus was present in 196 (11 %) patients. Patients with diabetes mellitus had higher 10-year cardiovascular risk score (29 [interquartile range 21, 40] versus 15 [9, 21] %, p < 0.001) and CAC score (71 [1, 447] versus 17 [0, 209] Agatston units, p < 0.001), but were not more likely to have obstructive disease or visually assessed adverse plaque characteristics. Patients with diabetes mellitus had higher quantitatively assessed calcified, non-calcified, low attenuation and total plaque burdens. After adjustment for age and sex, diabetes mellitus was an independent predictor of calcified plaque burden (p = 0.009), but not the other plaque types. During 8.7 [IQR 8, 9.6] years follow-up, diabetes mellitus was associated with an increased risk of fatal or non-fatal myocardial infarction, adjusted for age and sex (hazard ratio 1.85, 95 % confidence interval 1.09 to 3.17, p = 0.024). CONCLUSION Diabetes mellitus was an independent predictor of quantitatively assessed plaque burden, particularly calcified plaque, and was associated with an increased risk of myocardial infarction.
Collapse
Affiliation(s)
- Zuzanna M Gebert
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
| | - Jacek Kwiecinski
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom; Department of Interventional Cardiology and Angiology, Institute of Cardiology, Warsaw, Poland
| | | | - Philip D Adamson
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom; Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Nicholas L Mills
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
| | - Giles Roditi
- Institute of Clinical Sciences, University of Glasgow, United Kingdom
| | - Edwin J R van Beek
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom; Edinburgh Imaging, University of Edinburgh, United Kingdom
| | - Edward D Nicol
- Royal Brompton Hospital, London, United Kingdom; School of Biomedical Engineering and Imaging Sciences, Kings College London, United Kingdom
| | - Daniel S Berman
- School of Biomedical Engineering and Imaging Sciences, Kings College London, United Kingdom
| | | | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
| | - Damini Dey
- Cedars-Sinai Medical Centre, Los Angeles, CA, USA
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom; Edinburgh Imaging, University of Edinburgh, United Kingdom
| | - Michelle C Williams
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom; Edinburgh Imaging, University of Edinburgh, United Kingdom.
| |
Collapse
|
6
|
Lu Y, Wu S, Zhu S, Shen J, Liu C, Zhao C, Su S, Ma H, Xiang M, Xie Y. Integrated Single-Cell Analysis Revealed Novel Subpopulations of Foamy Macrophages in Human Atherosclerotic Plaques. Biomolecules 2024; 14:1606. [PMID: 39766313 PMCID: PMC11675068 DOI: 10.3390/biom14121606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Foam cell formation is a hallmark of atherosclerosis, yet the cellular complexity within foam cells in human plaques remains unexplored. Here, we integrate published single-cell RNA-sequencing, spatial transcriptomic, and chromatin accessibility sequencing datasets of human atherosclerotic lesions across eight distinct studies. Through this large-scale integration of patient-derived information, we identified foamy macrophages enriched for genes characteristic of the foamy signature. We further re-clustered the foamy macrophages into five unique subsets with distinct potential functions: (i) pro-foamy macrophages, exhibiting relatively high inflammatory and adhesive properties; (ii) phagocytic foamy macrophages, specialized in efferocytosis; (iii) high-efflux foamy macrophages marked by high NR1H3 expression; (iv) mature foamy macrophages prone to programmed cell death; and (v) synthetic subset. Trajectory analysis elucidated a bifurcated differentiation cell fate from pro-foam macrophages toward either the programmed death (iv) or synthetic (v) phenotype. The existence of these foamy macrophage subsets was validated by immunostaining. Moreover, these foamy macrophage subsets exhibited strong potential ligand-receptor interactions. Finally, we conducted Mendelian randomization analyses to identify a possible causal relationship between key regulatory genes along the programmed death pathway in foamy macrophages and atherosclerotic diseases. This study provides a high-resolution map of foam cell diversity and a set of potential key regulatory genes in atherosclerotic plaques, offering novel insights into the multifaceted pathophysiology underlying human atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Meixiang Xiang
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (Y.L.); (S.W.); (S.Z.); (J.S.); (C.L.); (C.Z.); (S.S.); (H.M.)
| | - Yao Xie
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (Y.L.); (S.W.); (S.Z.); (J.S.); (C.L.); (C.Z.); (S.S.); (H.M.)
| |
Collapse
|
7
|
Li Q, Fu J, Park K, Shah H, Li Q, Wu IH, King GL. Insulin receptors in vascular smooth muscle cells regulate plaque stability of atherosclerosis. Cardiovasc Res 2024; 120:2017-2030. [PMID: 39197028 PMCID: PMC11646105 DOI: 10.1093/cvr/cvae193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/09/2024] [Accepted: 07/05/2024] [Indexed: 08/30/2024] Open
Abstract
AIMS Increased prevalence of acute myocardial infarction related to diabetes and insulin resistance is associated with an elevated risk of unstable atherosclerotic plaques, which are characterized by reduced vascular smooth muscle cells (VSMCs) and extracellular matrix (ECM) and increased inflammation. Thus, insulin resistance may reduce plaque stability, as deleting insulin receptors (IRs) in VSMCs decreases their proliferation and enhances apoptosis. METHODS AND RESULTS Direct effects of insulin on VSMCs to alter plaque composition were studied using mice with double knockout of ApoE and IR genes in VSMCs with SMIRKO/ApoE-/-, Myh11-CreERT2EYFP+/ApoE-/-, and Myh11-CreERT2EYFP+IRKO/ApoE-/- mice, which were also used for lineage tracing studies. Compared with ApoE-/- mice, SMIRKO/ApoE-/- mice exhibited more atherosclerotic plaques, which contained less VSMCs and collagen but increased levels of VSMC apoptosis and necrotic areas. Lineage tracing studies showed that Icam1+ Vcam1+ VSMC was inflammatory, which increased in the aortas of Myh11-CreERT2EYFP+IRKO/ApoE-/- mice compared with control mice. Isolated VSMCs lacking IRs expressed higher inflammatory cytokines than cells with IRs. Cell-based studies indicated that insulin's anti-apoptotic and pro-proliferative effects in VSMCs were mediated via activation of the IR/Akt pathway, which were decreased in VSMCs from SMIRKO or high-fat diet mice. An analysis of the IR targets that regulated inflammatory cytokines in VSMCs showed that thrombospondin 1 (Thbs1) and Mmp2 were consistently increased with a loss of IRs. Insulin inhibited Thbs1 expression, but not Mmp2 expression, through p-Akt/p-FoxO1 pathways in VSMCs from ApoE-/- mice, and was impaired in cells from SMIRKO/ApoE-/- mice. Thbs1 further induced Icam1 and Mmp2 expressions in VSMCs. CONCLUSION Insulin via IRs has significant actions in VSMCs to decrease inflammation, apoptosis, and ECM turnover via the activation of Akt and FoxO1 pathways. The inhibition of insulin actions and related pathways related to insulin resistance and diabetes may contribute to the formation of unstable atherosclerotic plaques.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Plaque, Atherosclerotic
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Apoptosis
- Mice, Knockout, ApoE
- Disease Models, Animal
- Signal Transduction
- Receptor, Insulin/metabolism
- Receptor, Insulin/genetics
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Aortic Diseases/genetics
- Aortic Diseases/prevention & control
- Mice, Inbred C57BL
- Cell Proliferation
- Insulin/metabolism
- Male
- Cells, Cultured
- Rupture, Spontaneous
- Insulin Resistance
- Proto-Oncogene Proteins c-akt/metabolism
- Thrombospondin 1/metabolism
- Thrombospondin 1/genetics
- Aorta/pathology
- Aorta/metabolism
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 2/genetics
- Inflammation Mediators/metabolism
- Necrosis
- Apolipoproteins E/genetics
- Apolipoproteins E/deficiency
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Mice
- Receptor, IGF Type 1
Collapse
Affiliation(s)
- Qian Li
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
| | - Jialin Fu
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
| | - Kyoungmin Park
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
| | - Hetal Shah
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
| | - Qin Li
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
| | - I Hsien Wu
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
| | - George L King
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
| |
Collapse
|
8
|
Singh P, Sun J, Cavalera M, Al-Sharify D, Matthes F, Barghouth M, Tengryd C, Dunér P, Persson A, Sundius L, Nitulescu M, Bengtsson E, Rattik S, Engelbertsen D, Orho-Melander M, Nilsson J, Monaco C, Goncalves I, Edsfeldt A. Dysregulation of MMP2-dependent TGF-ß2 activation impairs fibrous cap formation in type 2 diabetes-associated atherosclerosis. Nat Commun 2024; 15:10464. [PMID: 39653743 PMCID: PMC11628557 DOI: 10.1038/s41467-024-50753-8] [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: 06/23/2023] [Accepted: 07/18/2024] [Indexed: 12/12/2024] Open
Abstract
Type 2 diabetes is associated with cardiovascular disease, possibly due to impaired vascular fibrous repair. Yet, the mechanisms are elusive. Here, we investigate alterations in the fibrous repair processes in type 2 diabetes atherosclerotic plaque extracellular matrix by combining multi-omics from the human Carotid Plaque Imaging Project cohort and functional studies. Plaques from type 2 diabetes patients have less collagen. Interestingly, lower levels of transforming growth factor-ß distinguish type 2 diabetes plaques and, in these patients, lower levels of fibrous repair markers are associated with cardiovascular events. Transforming growth factor-ß2 originates mostly from contractile vascular smooth muscle cells that interact with synthetic vascular smooth muscle cells in the cap, leading to collagen formation and vascular smooth muscle cell differentiation. This is regulated by free transforming growth factor-ß2 which is affected by hyperglycemia. Our findings underscore the importance of transforming growth factor-ß2-driven fibrous repair in type 2 diabetes as an area for future therapeutic strategies.
Collapse
MESH Headings
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/pathology
- Humans
- Transforming Growth Factor beta2/metabolism
- Matrix Metalloproteinase 2/metabolism
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Female
- Animals
- Middle Aged
- Aged
- Collagen/metabolism
- Extracellular Matrix/metabolism
- Mice
- Cell Differentiation
Collapse
Affiliation(s)
- Pratibha Singh
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Jiangming Sun
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Michele Cavalera
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Dania Al-Sharify
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Frank Matthes
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Mohammad Barghouth
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Christoffer Tengryd
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Pontus Dunér
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Ana Persson
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Lena Sundius
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Mihaela Nitulescu
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Department of Biomedical Science, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Sara Rattik
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | | | | | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Isabel Goncalves
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
- Department of Cardiology, University Hospital of Skåne, Lund/Malmö, Sweden
| | - Andreas Edsfeldt
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden.
- Department of Cardiology, University Hospital of Skåne, Lund/Malmö, Sweden.
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
9
|
Cross K, Vetter SW, Alam Y, Hasan MZ, Nath AD, Leclerc E. Role of the Receptor for Advanced Glycation End Products (RAGE) and Its Ligands in Inflammatory Responses. Biomolecules 2024; 14:1550. [PMID: 39766257 PMCID: PMC11673996 DOI: 10.3390/biom14121550] [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: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Since its discovery in 1992, the receptor for advanced glycation end products (RAGE) has emerged as a key receptor in many pathological conditions, especially in inflammatory conditions. RAGE is expressed by most, if not all, immune cells and can be activated by many ligands. One characteristic of RAGE is that its ligands are structurally very diverse and belong to different classes of molecules, making RAGE a promiscuous receptor. Many of RAGE ligands are damaged associated molecular patterns (DAMPs) that are released by cells under inflammatory conditions. Although RAGE has been at the center of a lot of research in the past three decades, a clear understanding of the mechanisms of RAGE activation by its ligands is still missing. In this review, we summarize the current knowledge of the role of RAGE and its ligands in inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (K.C.); (S.W.V.); (Y.A.); (M.Z.H.); (A.D.N.)
| |
Collapse
|
10
|
Zhu B, Liu Y, Peng D. The double-edged role and therapeutic potential of TREM2 in atherosclerosis. Biomark Res 2024; 12:131. [PMID: 39497214 PMCID: PMC11533605 DOI: 10.1186/s40364-024-00675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/18/2024] [Indexed: 11/07/2024] Open
Abstract
Atherosclerosis is a chronic lipid-driven inflammatory disease characterized by infiltration of large numbers of macrophages. The progression of the disease is closely related to the status of macrophages in atherosclerotic plaques. Recent advances in plaque analysis have revealed a subpopulation of macrophages that express high levels of triggering receptor expressed on myeloid cells 2 (TREM2). Although TREM2 is known to play a critical role in inflammation, lipid metabolism, and tissue repair, its role in atherosclerosis is still not fully understood. Recent studies have shown that TREM2 promotes macrophage cholesterol uptake and efflux, enhances efferocytosis function, regulates inflammation and metabolism, and promotes cell survival, all of which are significant functions in atherosclerosis. In early plaques TREM2 promotes lipid uptake and increases lesion size. In advanced plaques TREM2 promotes macrophage survival and increases plaque stability. The dualistic nature of TREM2 in atherosclerosis, where it can exert both protective effect and a side effect of increased lesion size, presents a complex but crucial area of study. Understanding these dual roles could help in the development of new therapeutic strategies to modulate TREM2 activity and utilize its atheroprotective function while mitigating its deleterious effects. In this review, we discuss the roles and mechanisms of TREM2 during different stages of atherosclerotic plaques, as well as the potential applications of TREM2 in the diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Botao Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Yuxuan Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| |
Collapse
|
11
|
Koga JI, Umezu R, Kondo Y, Shirouzu T, Orkhonselenge N, Ueno H, Katsuki S, Matoba T, Nishimura Y, Kataoka M. Cyclophilin D induces necrotic core formation by mediating mitochondria-associated macrophage death in advanced atherosclerotic lesions. Atherosclerosis 2024; 396:118524. [PMID: 38972156 DOI: 10.1016/j.atherosclerosis.2024.118524] [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: 10/17/2023] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND AND AIMS In advanced atherosclerotic lesions, macrophage deaths result in necrotic core formation and plaque vulnerability. Cyclophilin D (CypD) is a mitochondria-specific cyclophilin involved in the process of cell death after organ ischemia-reperfusion. However, the role of CypD in atherosclerosis, especially in necrotic core formation, is unknown. Therefore, this experiment aims to clarify the role of CypD in necrotic core formation. METHODS To clarify the specific role of CypD, encoded by Ppif in mice, apolipoprotein-E/CypD-double knockout (Apoe-/-Ppif-/-) mice were generated. These mice were fed a high-fat diet containing 0.15 % cholesterol for 24 weeks to accelerate atherosclerotic lesion development. RESULTS Deletion of CypD decreased the necrotic core size, accompanied by a reduction of macrophage apoptosis compared to control Apoe-/- mice. In RAW264.7 cells, siRNA-mediated knockdown of CypD attenuated the release of cytochrome c from the mitochondria to the cytosol induced by endoplasmic reticulum stress inducer thapsigargin. In addition, necroptosis, induced by TNF-α and caspase inhibitor, was attenuated by knockdown of CypD. Ly-6Chigh inflammatory monocytes in peripheral blood leukocytes and mRNA expression of Il1b in the aorta were decreased by deletion of CypD. In contrast, siRNA-mediated knockdown of CypD did not significantly decrease Il1b nor Ccl2 mRNA expression in RAW264.7 cells treated with LPS and IFN-γ, suggesting that inhibition of inflammation in vivo is likely due to decreased cell death in the atherosclerotic lesions rather than a direct action of CypD deletion on the macrophage. CONCLUSIONS These results indicate that CypD induces macrophage death and mediates necrotic core formation in advanced atherosclerotic lesions. CypD could be a novel therapeutic target for treating atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Jun-Ichiro Koga
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Ryuta Umezu
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki Kondo
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; The Department of Cardiovascular Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tomohiro Shirouzu
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Nasanbadrakh Orkhonselenge
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiromichi Ueno
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shunsuke Katsuki
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Matoba
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yosuke Nishimura
- The Department of Cardiovascular Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masaharu Kataoka
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
12
|
Aslan EI, Ozkara G, Kilicarslan O, Ser OS, Bostan C, Yildiz A, Diren Borekcioglu A, Ozturk O, Kucukhuseyin O, Yilmaz Aydogan H. Receptor for advanced glycation end products polymorphisms in coronary artery ectasia. Gene 2024; 916:148450. [PMID: 38588932 DOI: 10.1016/j.gene.2024.148450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND Although the implication of receptor of advanced glycation endproducts (RAGE) has been reported in coronary artery disease, its roles in coronary artery ectasia (CAE) have remained undetermined. Furthermore, the effect of RAGE polymorfisms were not well-defined in scope of soluble RAGE (sRAGE) levels. Thus, we aimed to investigate the influence of the functional polymorphisms of RAGE -374T > A (rs1800624) and G82S (rs2070600) in CAE development. METHODS This prospective observational study was conducted in 2 groups selected of 2452 patients who underwent elective coronary angiography (CAG) for evaluation after positive noninvasive heart tests. Group-I included 98 patients with non-obstructive coronary artery disease and CAE, and Group-II (control) included 100 patients with normal coronary arteries. SNPs were genotyped by real-time PCR using Taqman® genotyping assay. Serum sRAGE and soluble lectin-like oxidized receptor-1 (sOLR1) were assayed by ELISA and serum lipids were measured enzymatically. RESULTS The frequencies of the RAGE -374A allele and -374AA genotype were significantly higher in CAE patients compared to controls (p < 0.001). sRAGE levels were not different between study groups, while sOLR1 levels were elevated in CAE (p = 0.004). In controls without systemic disease, -374A allele was associated with low sRAGE levels (p < 0.05), but this association was not significant in controls with HT. Similarly, sRAGE levels of CAE patients with both HT and T2DM were higher than those no systemic disease (p = 0.02). The -374A allele was also associated with younger patient age and higher platelet count in the CAE group in both total and subgroup analyses. In the correlation analyses, the -374A allele was also negatively correlated with age and positively correlated with Plt in all of these CAE groups. In the total CAE group, sRAGE levels also showed a positive correlation with age and a negative correlation with HDL-cholesterol levels. On the other hand, a negative correlation was observed between sRAGE and Plt in the total, hypertensive and no systemic disease control subgroups. Multivariate logistic regression analysis confirmed that the -374A allele (p < 0.001), hyperlipidemia (p < 0.05), and high sOLR1 level (p < 0.05) are risk factors for CAE. ROC curve analysis shows that RAGE -374A allele has AUC of 0.713 (sensitivity: 83.7 %, specificity: 59.0 %), which is higher than HLD (sensitivity: 59.2 %, specificity: 69.0 %), HT (sensitivity: 62.4 %, specificity: 61.1 %) and high sOLR1 level (≥0.67 ng/ml)) (sensitivity: 59.8 %, specificity: 58.5 %). CONCLUSION Beside the demonstration of the relationship between -374A allele and increased risk of CAE for the first time, our results indicate that antihypertensive and antidiabetic treatment in CAE patients causes an increase in sRAGE levels. The lack of an association between the expected -374A allele and low sRAGE levels in total CAE group was attributed to the high proportion of hypertensive patients and hence to antihypertensive treatment. Moreover, the RAGE -374A allele is associated with younger age at CAE and higher Plt, suggesting that -374A may also be associated with platelet activation, which plays a role in the pathogenesis of CAE. However, our data need to be confirmed in a large study for definitive conclusions.
Collapse
Affiliation(s)
- Ezgi Irmak Aslan
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Department of Medical Biochemistry, Faculty of Medicine, Istanbul Nisantasi University, Istanbul, Turkey.
| | - Gulcin Ozkara
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Department of Medical Biology, Bezmialem Vakıf University, Istanbul, Turkey.
| | - Onur Kilicarslan
- Department of Cardiology, Institute of Cardiology, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Ozgur Selim Ser
- Department of Cardiology, Institute of Cardiology, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Cem Bostan
- Department of Cardiology, Institute of Cardiology, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Ahmet Yildiz
- Department of Cardiology, Institute of Cardiology, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Ayca Diren Borekcioglu
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| | - Oguz Ozturk
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| | - Ozlem Kucukhuseyin
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| | - Hulya Yilmaz Aydogan
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
13
|
Luciani L, Pedrelli M, Parini P. Modification of lipoprotein metabolism and function driving atherogenesis in diabetes. Atherosclerosis 2024; 394:117545. [PMID: 38688749 DOI: 10.1016/j.atherosclerosis.2024.117545] [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: 12/06/2023] [Revised: 03/18/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease, characterized by raised blood glucose levels and impaired lipid metabolism resulting from insulin resistance and relative insulin deficiency. In diabetes, the peculiar plasma lipoprotein phenotype, consisting in higher levels of apolipoprotein B-containing lipoproteins, hypertriglyceridemia, low levels of HDL cholesterol, elevated number of small, dense LDL, and increased non-HDL cholesterol, results from an increased synthesis and impaired clearance of triglyceride rich lipoproteins. This condition accelerates the development of the atherosclerotic cardiovascular disease (ASCVD), the most common cause of death in T2DM patients. Here, we review the alteration of structure, functions, and distribution of circulating lipoproteins and the pathophysiological mechanisms that induce these modifications in T2DM. The review analyzes the influence of diabetes-associated metabolic imbalances throughout the entire process of the atherosclerotic plaque formation, from lipoprotein synthesis to potential plaque destabilization. Addressing the different pathophysiological mechanisms, we suggest improved approaches for assessing the risk of adverse cardiovascular events and clinical strategies to reduce cardiovascular risk in T2DM and cardiometabolic diseases.
Collapse
Affiliation(s)
- Lorenzo Luciani
- Cardio Metabolic Unit, Department of Laboratory Medicine, and Department of Medicine at Huddinge, Karolinska Institutet, Stockholm, Sweden; Interdisciplinary Center for Health Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Matteo Pedrelli
- Cardio Metabolic Unit, Department of Laboratory Medicine, and Department of Medicine at Huddinge, Karolinska Institutet, Stockholm, Sweden; Medicine Unit of Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Paolo Parini
- Cardio Metabolic Unit, Department of Laboratory Medicine, and Department of Medicine at Huddinge, Karolinska Institutet, Stockholm, Sweden; Medicine Unit of Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
14
|
Balgobin S, Basak S, Teoh CW, Noone D. Hypertension in diabetes. Pediatr Nephrol 2024; 39:1739-1758. [PMID: 37831122 DOI: 10.1007/s00467-023-06163-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/01/2023] [Accepted: 09/01/2023] [Indexed: 10/14/2023]
Abstract
Diabetes mellitus, a disease that affects hundreds of millions of people worldwide, is increasing in prevalence in all age groups, including children and adolescents. Much of the morbidity and mortality associated with diabetes is closely related to hypertension, often coincident with diabetes. Comorbid hypertension and diabetes often worsen the outcomes of each other, likely rooted in some overlapping pathogenic mechanisms. In this educational review, we will discuss the shared pathophysiology of diabetes and hypertension, particularly in regard to inflammation and oxidative stress, the sympathetic nervous system, vascular remodeling, and the renin-angiotensin-aldosterone system (RAAS). We will also review current hypertension diagnosis and management guidelines from many international jurisdictions for both adult and paediatric populations in the setting of diabetes. Many of these guidelines highlight the use and utility of RAAS blockers in this clinical scenario; however, on review of the evidence for their use, several meta-analyses and systematic reviews fail to demonstrate superiority of RAAS blockers over other anti-hypertensive medications. Finally, we discuss several new anti-hypertensive medications, review their mechanisms of action, and highlight some of the evidence for their use in the setting of hypertension and diabetes.
Collapse
Affiliation(s)
- Steve Balgobin
- Division of Paediatric Nephrology, The Hospital for Sick Children, 555 University Avenue, Toronto, M5G 1X8, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Sanjukta Basak
- Pediatric Endocrinologist, BC Children's Hospital, Vancouver, BC, Canada
- Division of Endocrinology & Metabolism, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Chia Wei Teoh
- Division of Paediatric Nephrology, The Hospital for Sick Children, 555 University Avenue, Toronto, M5G 1X8, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Damien Noone
- Division of Paediatric Nephrology, The Hospital for Sick Children, 555 University Avenue, Toronto, M5G 1X8, Canada.
- Department of Paediatrics, University of Toronto, Toronto, Canada.
| |
Collapse
|
15
|
Zhao J, Wu T, Tan J, Chen Y, Xu X, Guo Y, Jin C, Xiu L, Zhao R, Sun S, Peng C, Li S, Yu H, Liu Y, Wei G, Li L, Wang Y, Hou J, Dai J, Fang C, Yu B. Pancoronary plaque characteristics in STEMI patients with rapid plaque progression: An optical coherence tomography study. Int J Cardiol 2024; 400:131821. [PMID: 38301829 DOI: 10.1016/j.ijcard.2024.131821] [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: 11/19/2023] [Revised: 01/16/2024] [Accepted: 01/28/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Non-culprit plaque progression is associated with recurrent cardiac ischemic events and worse clinical outcomes. Given that atherosclerosis is a systemic disease, the pancoronary characteristics of patients with rapid plaque progression are unknown. This study aims to identify pancoronary plaque features in patients with ST-segment elevation myocardial infarction (STEMI) with and without rapid plaque progression, focused on the patient level. METHODS AND RESULTS From January 2017 to July 2019, 291 patients underwent 3-vessel optical coherence tomography imaging at the time of the primary procedure and a follow-up angiography interval of 12 months. The final analysis included 237 patients. Overall, 308 non-culprit lesions were found in 78 STEMI patients with rapid plaque progression, and 465 non-culprit plaques were found in 159 STEMI patients without rapid plaque progression. These patients had a higher pancoronary vulnerability (CLIMA-defined high-risk plaque: 47.4% vs. 33.3%; non-culprit plaque rupture: 25.6% vs. 14.5%) and a significantly higher prevalence of other vulnerable plaque characteristics (i.e., lipid-rich plaque, cholesterol crystal, microchannels, calcification, spotty calcification, and thrombus) at baseline versus those without rapid plaque progression. Lesions with rapid progression were highly distributed at the LAD, tending to be near the bifurcation. In multivariate analysis, age ≥ 65 years was an independent predictor of subsequent rapid lesion progression at the patient level, whereas microchannel, spotty calcification, and cholesterol crystal were independent predictors for STEMI patients ≥65 years old. CONCLUSIONS STEMI patients with subsequent rapid plaque progression had higher pancoronary vulnerability and commonly presented vulnerable plaque morphology. Aging was the only predictor of subsequent rapid plaque progression.
Collapse
Affiliation(s)
- Jiawei Zhao
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Tianyu Wu
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Jinfeng Tan
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Yuzhu Chen
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Xueming Xu
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Yibo Guo
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Chengmei Jin
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Lili Xiu
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Rui Zhao
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Sibo Sun
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Cong Peng
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Shuang Li
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Huai Yu
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Yanchao Liu
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Guo Wei
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Lulu Li
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Yini Wang
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Jingbo Hou
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Jiannan Dai
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China
| | - Chao Fang
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China.
| | - Bo Yu
- Department of Cardiology, The 2(nd) Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, 246 Xuefu Road, Harbin 150086, China.
| |
Collapse
|
16
|
He W, Fang T, Fu X, Lao M, Xiao X. Risk factors and the CCTA application in patients with vulnerable coronary plaque in type 2 diabetes: a retrospective study. BMC Cardiovasc Disord 2024; 24:89. [PMID: 38311736 PMCID: PMC10840286 DOI: 10.1186/s12872-024-03717-1] [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: 07/26/2023] [Accepted: 01/06/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Diabetes is an independent risk factor for cardiovascular disease. The purpose of this study was to identify the risk factors for vulnerable coronary plaques (VCPs), which are associated with adverse cardiovascular events, and to determine the value of coronary CT angiography (CCTA) in patients with type 2 diabetes mellitus (T2DM) and VCPs. METHODS Ninety-eight T2DM patients who underwent CCTA and intravascular ultrasound (IVUS) were retrospectively included and analyzed. The patients were grouped and analyzed according to the presence or absence of VCPs. RESULTS Among the patients with T2DM, time in range [TIR {the percentage of time blood glucose levels were in the target range}] (OR = 0.93, 95% CI = 0.89-0.96; P < 0.001) and the high-density lipoprotein-cholesterol (HDL-C) concentration (OR = 0.24, 95% CI = 0.09-0.63; P = 0.04) were correlated with a lower risk of VCP, but the triglycerides (TG) concentration was correlated with a higher risk of VCP (OR = 1.79, 95% CI = 1.01-3.18; P = 0.045). The area under the receiver operator characteristic curve (AUC) of TIR, and HDL-C and TG concentrations were 0.76, 0.73, and 0.65, respectively. The combined predicted AUC of TIR, and HDL-C and TG concentrations was 0.83 (P < 0.05). The CCTA sensitivity, specificity, false-negative, and false-positive values for the diagnosis of VCP were 95.74%, 94.12%, 4.26%, and 5.88%, respectively. The identification of VCP by CCTA was positively correlated with IVUS (intraclass correlation coefficient [ICC] = 0.90). CONCLUSIONS The TIR and HDL-C concentration are related with lower risk of VCP and the TG concentration was related with higher risk of VCP in patients with T2DM. In clinical practice, TIR, HDL-C and TG need special attention in patients with T2DM. The ability of CCTA to identify VCP is highly related to IVUS findings.
Collapse
Affiliation(s)
- Weihong He
- Department of Radiology, Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Foshan, China.
| | - Tingsong Fang
- Department of Radiology, Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Foshan, China
| | - Xi Fu
- Department of Radiology, Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Foshan, China
| | - Meiling Lao
- Department of Endocrinology, Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Foshan, China
| | - Xiuyun Xiao
- Department of Radiology, Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Foshan, China
| |
Collapse
|
17
|
Dobrucki IT, Miskalis A, Nelappana M, Applegate C, Wozniak M, Czerwinski A, Kalinowski L, Dobrucki LW. Receptor for advanced glycation end-products: Biological significance and imaging applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1935. [PMID: 37926944 DOI: 10.1002/wnan.1935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
The receptor for advanced glycation end-products (RAGE or AGER) is a transmembrane, immunoglobulin-like receptor that, due to its multiple isoform structures, binds to a diverse range of endo- and exogenous ligands. RAGE activation caused by the ligand binding initiates a cascade of complex pathways associated with producing free radicals, such as reactive nitric oxide and oxygen species, cell proliferation, and immunoinflammatory processes. The involvement of RAGE in the pathogenesis of disorders such as diabetes, inflammation, tumor progression, and endothelial dysfunction is dictated by the accumulation of advanced glycation end-products (AGEs) at pathologic states leading to sustained RAGE upregulation. The involvement of RAGE and its ligands in numerous pathologies and diseases makes RAGE an interesting target for therapy focused on the modulation of both RAGE expression or activation and the production or exogenous administration of AGEs. Despite the known role that the RAGE/AGE axis plays in multiple disease states, there remains an urgent need to develop noninvasive, molecular imaging approaches that can accurately quantify RAGE levels in vivo that will aid in the validation of RAGE and its ligands as biomarkers and therapeutic targets. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Iwona T Dobrucki
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Academy of Medical and Social Applied Sciences, Elblag, Poland
| | - Angelo Miskalis
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Michael Nelappana
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
| | - Catherine Applegate
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Cancer Center at Illinois, Urbana, Illinois, USA
| | - Marcin Wozniak
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Division of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
| | - Andrzej Czerwinski
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
| | - Leszek Kalinowski
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Division of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, Gdansk, Poland
| | - Lawrence W Dobrucki
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA
- Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, Urbana, Illinois, USA
- Division of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
18
|
Jiang S, Xia N, Buonfiglio F, Böhm EW, Tang Q, Pfeiffer N, Olinger D, Li H, Gericke A. High-fat diet causes endothelial dysfunction in the mouse ophthalmic artery. Exp Eye Res 2024; 238:109727. [PMID: 37972749 DOI: 10.1016/j.exer.2023.109727] [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: 07/13/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Obesity is a significant health concern that leads to impaired vascular function and subsequent abnormalities in various organs. The impact of obesity on ocular blood vessels, however, remains largely unclear. In this study, we examined the hypothesis that obesity induced by high-fat diet produces vascular endothelial dysfunction in the ophthalmic artery. Mice were subjected to a high-fat diet for 20 weeks, while age-matched controls were maintained on a standard diet. Reactivity of isolated ophthalmic artery segments was assessed in vitro. Reactive oxygen species (ROS) were quantified in cryosections by dihydroethidium (DHE) staining. Redox gene expression was determined in ophthalmic artery explants by real-time PCR. Furthermore, the expression of nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2), the receptor for advanced glycation end products (RAGE), and of the lectin-like oxidized low-density-lipoprotein receptor-1 (LOX-1) was determined in cryosections using immunofluorescence microscopy. Ophthalmic artery segments from mice on a high-fat diet exhibited impaired vasodilation responses to the endothelium-dependent vasodilator acetylcholine, while endothelium-independent responses to nitroprusside remained preserved. DHE staining intensity in the vascular wall was notably stronger in mice on a high-fat diet. Messenger RNA expression for NOX2 was elevated in the ophthalmic artery of mice subjected to high fat diet. Likewise, immunostainings revealed increased expression of NOX2 and of RAGE, but not of LOX-1. These findings suggest that a high-fat diet triggers endothelial dysfunction by inducing oxidative stress in the ophthalmic artery via involvement of RAGE and NOX2.
Collapse
Affiliation(s)
- Subao Jiang
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Elsa W Böhm
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Qi Tang
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Dominik Olinger
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Huige Li
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
19
|
Suzuki K, Niida T, Yuki H, Kinoshita D, Fujimoto D, Lee H, McNulty I, Takano M, Nakamura S, Kakuta T, Mizuno K, Jang I. Coronary Plaque Characteristics and Underlying Mechanism of Acute Coronary Syndromes in Different Age Groups of Patients With Diabetes. J Am Heart Assoc 2023; 12:e031474. [PMID: 38014673 PMCID: PMC10727321 DOI: 10.1161/jaha.123.031474] [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: 06/20/2023] [Accepted: 10/17/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND High cardiovascular mortality has been reported in young patients with diabetes. However, the underlying pathology in different age groups of patients with diabetes has not been studied. METHODS AND RESULTS The aim of this study was to investigate the plaque characteristics and underlying pathology of acute coronary syndrome in different age groups of patients with or without diabetes in a large cohort. Patients who presented with acute coronary syndrome and underwent preintervention optical coherence tomography imaging were included. Culprit plaque was classified as plaque rupture, plaque erosion, or calcified plaque and stratified into 5 age groups. Plaque characteristics including features of vulnerability were examined by optical coherence tomography. Among 1394 patients, 482 (34.6%) had diabetes. Patients with diabetes, compared with patients without diabetes, had a higher prevalence of lipid-rich plaque (71.2% versus 64.8%, P=0.016), macrophage (72.0% versus 62.6%, P<0.001), and cholesterol crystal (27.6% versus 19.7%, P<0.001). Both diabetes and nondiabetes groups showed a decreasing trend in plaque erosion with age (patients with diabetes, P=0.020; patients without diabetes, P<0.001). Patients without diabetes showed an increasing trend with age in plaque rupture (P=0.004) and lipid-rich plaque (P=0.018), whereas patients with diabetes had a high prevalence of these vulnerable features at an early age that remained high across age groups. CONCLUSIONS Patients without diabetes showed an increasing trend with age in plaque rupture and lipid-rich plaque, whereas patients with diabetes had a high prevalence of these vulnerable features at an early age. These results suggest that atherosclerotic vascular changes with increased vulnerability start at a younger age in patients with diabetes. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifiers: NCT04523194, NCT03479723. URL: https://www.umin.ac.jp/ctr/. Unique identifier: UMIN000041692.
Collapse
Affiliation(s)
- Keishi Suzuki
- Cardiology DivisionMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Takayuki Niida
- Cardiology DivisionMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Haruhito Yuki
- Cardiology DivisionMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Daisuke Kinoshita
- Cardiology DivisionMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Daichi Fujimoto
- Cardiology DivisionMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Hang Lee
- Biostatistics CenterMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Iris McNulty
- Cardiology DivisionMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Masamichi Takano
- Cardiovascular CenterNippon Medical School Chiba Hokusoh HospitalInzai, ChibaJapan
| | - Sunao Nakamura
- Interventional Cardiology UnitNew Tokyo HospitalChibaJapan
| | - Tsunekazu Kakuta
- Department of CardiologyTsuchiura Kyodo General Hospital, TsuchiuraIbarakiJapan
| | | | - Ik‐Kyung Jang
- Cardiology DivisionMassachusetts General HospitalHarvard Medical SchoolBostonMA
| |
Collapse
|
20
|
Viggiano D. Mechanisms of Diabetic Nephropathy Not Mediated by Hyperglycemia. J Clin Med 2023; 12:6848. [PMID: 37959313 PMCID: PMC10650633 DOI: 10.3390/jcm12216848] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Diabetes mellitus (DM) is characterized by the appearance of progressive kidney damage, which may progress to end-stage kidney disease. The control of hyperglycemia is usually not sufficient to halt this progression. The kidney damage is quantitatively and qualitatively different in the two forms of diabetes; the typical nodular fibrosis (Kimmelstiel Wilson nodules) appears mostly in type 1 DM, whereas glomerulomegaly is primarily present in type 2 obese DM. An analysis of the different metabolites and hormones in type 1 and type 2 DM and their differential pharmacological treatments might be helpful to advance the hypotheses on the different histopathological patterns of the kidneys and their responses to sodium/glucose transporter type 2 inhibitors (SGLT2i).
Collapse
Affiliation(s)
- Davide Viggiano
- Department of Translational Medical Sciences, University of Campania, 80131 Naples, Italy
| |
Collapse
|
21
|
Thielemans R, Speeckaert R, Delrue C, De Bruyne S, Oyaert M, Speeckaert MM. Unveiling the Hidden Power of Uromodulin: A Promising Potential Biomarker for Kidney Diseases. Diagnostics (Basel) 2023; 13:3077. [PMID: 37835820 PMCID: PMC10572911 DOI: 10.3390/diagnostics13193077] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Uromodulin, also known as Tamm-Horsfall protein, represents the predominant urinary protein in healthy individuals. Over the years, studies have revealed compelling associations between urinary and serum concentrations of uromodulin and various parameters, encompassing kidney function, graft survival, cardiovascular disease, glucose metabolism, and overall mortality. Consequently, there has been a growing interest in uromodulin as a novel and effective biomarker with potential applications in diverse clinical settings. Reduced urinary uromodulin levels have been linked to an elevated risk of acute kidney injury (AKI) following cardiac surgery. In the context of chronic kidney disease (CKD) of different etiologies, urinary uromodulin levels tend to decrease significantly and are strongly correlated with variations in estimated glomerular filtration rate. The presence of uromodulin in the serum, attributable to basolateral epithelial cell leakage in the thick ascending limb, has been observed. This serum uromodulin level is closely associated with kidney function and histological severity, suggesting its potential as a biomarker capable of reflecting disease severity across a spectrum of kidney disorders. The UMOD gene has emerged as a prominent locus linked to kidney function parameters and CKD risk within the general population. Extensive research in multiple disciplines has underscored the biological significance of the top UMOD gene variants, which have also been associated with hypertension and kidney stones, thus highlighting the diverse and significant impact of uromodulin on kidney-related conditions. UMOD gene mutations are implicated in uromodulin-associated kidney disease, while polymorphisms in the UMOD gene show a significant association with CKD. In conclusion, uromodulin holds great promise as an informative biomarker, providing valuable insights into kidney function and disease progression in various clinical scenarios. The identification of UMOD gene variants further strengthens its relevance as a potential target for better understanding kidney-related pathologies and devising novel therapeutic strategies. Future investigations into the roles of uromodulin and regulatory mechanisms are likely to yield even more profound implications for kidney disease diagnosis, risk assessment, and management.
Collapse
Affiliation(s)
- Raïsa Thielemans
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (R.T.); (C.D.)
| | | | - Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (R.T.); (C.D.)
| | - Sander De Bruyne
- Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (S.D.B.); (M.O.)
| | - Matthijs Oyaert
- Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (S.D.B.); (M.O.)
| | - Marijn M. Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium; (R.T.); (C.D.)
- Research Foundation Flanders, 1000 Brussels, Belgium
| |
Collapse
|
22
|
Ménégaut L, Laubriet A, Crespy V, Leleu D, Pilot T, Van Dongen K, de Barros JPP, Gautier T, Petit JM, Thomas C, Nguyen M, Steinmetz E, Masson D. Inflammation and oxidative stress markers in type 2 diabetes patients with Advanced Carotid atherosclerosis. Cardiovasc Diabetol 2023; 22:248. [PMID: 37710315 PMCID: PMC10503074 DOI: 10.1186/s12933-023-01979-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/03/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a major global health issue and a significant risk factor for atherosclerosis. Atherosclerosis in T2DM patients has been associated with inflammation, insulin resistance, hyperglycemia, dyslipidemia, and oxidative stress. Identifying molecular features of atherosclerotic plaques in T2DM patients could provide valuable insights into the pathogenesis of the disease. METHODS The MASCADI (Arachidonic Acid Metabolism in Carotid Stenosis Plaque in Diabetic Patients) study aimed to investigate the increase of 2-arachidonoyl-lysophatidylcholine (2-AA-LPC) in carotid plaques from T2DM and control patients and to explore its association with plaque vulnerability as well as with blood and intra-plaque biomarkers altered during diabetes. RESULTS In a population of elderly, polymedicated patients with advanced stage of atherosclerosis, we found that T2DM patients had higher systemic inflammation markers, such as high-sensitivity C-reactive protein (hsCRP) and IL-1β, higher levels of oxysterols, increased triglyceride levels, and decreased HDL levels as compared to control patients. Furthermore, 2-AA-LPC was significantly enriched in plaques from diabetic patients, suggesting its potential role in diabetic atherosclerosis. Interestingly, 2-AA-LPC was not associated with systemic markers related to diabetes, such as hsCRP, triglycerides, or HDL cholesterol. However, it was significantly correlated with the levels of inflammatory markers within the plaques such as lysophospholipids and 25-hydroxycholesterol, strengthening the link between local inflammation, arachidonic acid metabolism and diabetes. CONCLUSION Our study is in line with a key role for inflammation in the pathogenesis of diabetic atherosclerosis and highlights the involvement of 2-AA-LPC. Further research is needed to better understand the local processes involved in the alteration of plaque composition in T2DM and to identify potential therapeutic targets. TRIAL REGISTRATION The MASCADI was registered on ClinicalTrials.gov (clinical registration number: NCT03202823).
Collapse
Affiliation(s)
- Louise Ménégaut
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
- CHU Dijon, Laboratory of Clinical Chemistry, Dijon, France
| | - Aline Laubriet
- Department of Cardiovascular and Thoracic Surgery, CHU Dijon, Dijon, France
| | - Valentin Crespy
- Department of Cardiovascular and Thoracic Surgery, CHU Dijon, Dijon, France
| | - Damien Leleu
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
- CHU Dijon, Laboratory of Clinical Chemistry, Dijon, France
| | - Thomas Pilot
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Kevin Van Dongen
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Jean-Paul Pais de Barros
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
- Lipidomic Analytic Platform, Université Bourgogne Franche-Comté, Dijon, France
| | - Thomas Gautier
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Jean-Michel Petit
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
- Department of Endocrinology and metabolic diseases, CHU Dijon, Dijon, France
| | - Charles Thomas
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Maxime Nguyen
- Université Bourgogne, LNC UMR1231, Dijon, France
- INSERM, UMR1231, Dijon, France
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
- CHU Dijon Department of Anesthesiology and Intensive Care, Dijon, France
| | - Eric Steinmetz
- Department of Cardiovascular and Thoracic Surgery, CHU Dijon, Dijon, France
| | - David Masson
- Université Bourgogne, LNC UMR1231, Dijon, France.
- INSERM, UMR1231, Dijon, France.
- Université Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France.
- CHU Dijon, Laboratory of Clinical Chemistry, Dijon, France.
- UFR des sciences de santé, Bvd Jeanne d'Arc, Dijon, 21000, France.
| |
Collapse
|
23
|
Çakır MO, Gören MT. Comparison of Atherosclerotic Plaque Compositions in Diabetic and Non-diabetic Patients. Cureus 2023; 15:e45721. [PMID: 37745746 PMCID: PMC10513476 DOI: 10.7759/cureus.45721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 09/26/2023] Open
Abstract
INTRODUCTION Diabetes mellitus is one of the major risk factors for coronary artery disease. Intravascular ultrasound (IVUS) imaging has an important role in the evaluation of atherosclerotic coronary artery disease. The aim of the study was to investigate the potential link between diabetes mellitus and plaque vulnerability in patients with coronary artery disease. METHODS In total, 26 patients with acute coronary syndrome (eight with diabetes mellitus) and 34 with stable angina pectoris (16 with diabetes mellitus) constituted the study population. Patients underwent IVUS ultrasound and virtual histology (VH)-IVUS imaging during routine diagnostic catheterization procedures. A total of 70 plaques in 60 patients were examined. RESULTS Patients with diabetes mellitus had a significantly greater percentage of fibrofatty components in the minimal lumen area (MLA) (17 ± 12 in diabetics; 12 ± 6 in non-diabetics; p=0.06). Thin-cap fibroatheromas were more frequent in patients with diabetes mellitus (72% versus 45%; p=0.012). There was a positive correlation between the presence of attenuated plaque and hemoglobin A1C (HbA1c) levels as well (7.09 ± 1.66 versus 6.02 ± 1.00; p=0.011). Patients with HbA1C ≥7.5% also had the highest prevalence of attenuated plaque. CONCLUSION As shown by VH-IVUS, the prevalence of vulnerable plaques in patients with diabetes mellitus was much higher than that in non-diabetic patients. The presence of attenuated plaque detected in grayscale intravascular ultrasonography was associated with high HbA1C levels in diabetic patients. Diabetes mellitus may cause cardiovascular vulnerability by changing the plaque morphology.
Collapse
Affiliation(s)
- Mustafa Ozan Çakır
- Department of Cardiology, Bulent Ecevit University Faculty of Medicine, Zonguldak, TUR
| | - Mustafa Taner Gören
- Department of Cardiology, Istanbul University School of Medicine, Istanbul, TUR
| |
Collapse
|
24
|
Takata K, Imaizumi S, Iwata A, Zhang B, Kawachi E, Miura SI, Ogawa M. Associations of High-Density Lipoprotein Functionality with Coronary Plaque Characteristics in Diabetic Patients with Coronary Artery Disease: Integrated Backscatter Intravascular Ultrasound Analysis. Biomolecules 2023; 13:1278. [PMID: 37759677 PMCID: PMC10526738 DOI: 10.3390/biom13091278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
High-density lipoprotein (HDL) functionality has been reported to be associated with coronary artery disease (CAD). However, little is known about the impact of HDL functionality on coronary atherosclerosis. Thirty-eight type 2 diabetic patients with CAD who underwent percutaneous coronary intervention were examined. Coronary atheroma burden and plaque composition of the culprit lesions were assessed using conventional gray-scale and integrated backscatter intravascular ultrasound. HDL-mediated cholesterol efflux capacity (HDL-CEC) and HDL antioxidant capacity, estimated as HDL inflammatory index (HII), were examined. The associations between HDL functionality and coronary plaques were analyzed using multivariate data analysis, including principal components analysis and orthogonal partial least squares (OPLS) models. Percent atheroma volume was correlated with HDL-CEC (r = 0.34, p = 0.04) but not with HII (p = 0.65). The OPLS model demonstrated that the percentage lipid volume was significantly associated with HDL functionality [coefficient (95% confidence interval); HDL-CEC: -0.26 (-0.49, -0.04); HII: 0.34 (0.08, 2.60), respectively]. HII exhibited the highest variable importance in projection score, indicating the greatest contribution. HDL functionality was associated with coronary plaque composition, a key component of plaque vulnerability. Our findings highlight the potential importance of HDL functionality for coronary plaque stabilization.
Collapse
Affiliation(s)
- Kohei Takata
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (S.-i.M.); (M.O.)
- Department of Clinical Laboratory and Transfusion, Fukuoka University Hospital, Fukuoka 814-0180, Japan
| | - Satoshi Imaizumi
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (S.-i.M.); (M.O.)
- Department of Bioethics and Medical Ethics, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Atsushi Iwata
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (S.-i.M.); (M.O.)
- Fukuoka University Health Care Center, Fukuoka 814-0180, Japan
| | - Bo Zhang
- Information Technology Center, Fukuoka University, Fukuoka 814-0180, Japan
- Department of Biochemistry, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Emi Kawachi
- Department of Bioethics and Medical Ethics, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Shin-ichiro Miura
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (S.-i.M.); (M.O.)
| | - Masahiro Ogawa
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (S.-i.M.); (M.O.)
- Department of Clinical Laboratory and Transfusion, Fukuoka University Hospital, Fukuoka 814-0180, Japan
| |
Collapse
|
25
|
Zernecke A, Erhard F, Weinberger T, Schulz C, Ley K, Saliba AE, Cochain C. Integrated single-cell analysis-based classification of vascular mononuclear phagocytes in mouse and human atherosclerosis. Cardiovasc Res 2023; 119:1676-1689. [PMID: 36190844 PMCID: PMC10325698 DOI: 10.1093/cvr/cvac161] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/09/2022] [Accepted: 09/24/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Accumulation of mononuclear phagocytes [monocytes, macrophages, and dendritic cells (DCs)] in the vessel wall is a hallmark of atherosclerosis. Using integrated single-cell analysis of mouse and human atherosclerosis, we here aimed to refine the nomenclature of mononuclear phagocytes in atherosclerotic vessels and to compare their transcriptomic profiles in mouse and human disease. METHODS AND RESULTS We integrated 12 single-cell RNA-sequencing (scRNA-seq) datasets of immune cells isolated from healthy or atherosclerotic mouse aortas, and data from 11 patients (n = 4 coronary vessels, n = 7 carotid endarterectomy specimens) from two studies. Integration of mouse data identified subpopulations with discrete transcriptomic signatures within previously described populations of aortic resident (Lyve1), inflammatory (Il1b), as well as foamy (Trem2hi) macrophages. We identified unique transcriptomic features distinguishing aortic intimal resident macrophages from atherosclerosis-associated Trem2hi macrophages. Also, populations of Xcr1+ Type 1 classical DCs (cDC1), Cd209a+ cDC2, and mature DCs (Ccr7, Fscn1) with a 'mreg-DC' signature were detected. In humans, we uncovered macrophage and DC populations with gene expression patterns similar to those observed in mice. In particular, core transcripts of the foamy/Trem2hi signature (TREM2, SPP1, GPNMB, CD9) mapped to a specific population of macrophages in human lesions. Comparison of mouse and human data and direct cross-species data integration suggested transcriptionally similar macrophage and DC populations in mice and humans. CONCLUSIONS We refined the nomenclature of mononuclear phagocytes in mouse atherosclerotic vessels, and show conserved transcriptomic features of macrophages and DCs in atherosclerosis in mice and humans, emphasizing the relevance of mouse models to study mononuclear phagocytes in atherosclerosis.
Collapse
Affiliation(s)
- Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Josef Schneider Str. 2, 97080 Würzburg, Germany
| | - Florian Erhard
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacher Straße 7, 97078 Würzburg, Germany
| | - Tobias Weinberger
- Medizinische Klinik und Poliklinik I, Klinikum der Universität, Ludwig-Maximilians-Universität, Campus Großhadern Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität, Ludwig-Maximilians-Universität, Campus Großhadern Marchioninistraße 15, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Klaus Ley
- La Jolla Institute for Immunology, 9420 Athena Circle La Jolla, CA 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Josef Schneider Str. 2, 97080 Würzburg, Germany
| | - Clément Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Josef Schneider Str. 2, 97080 Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
| |
Collapse
|
26
|
Hsu CC, Fidler TP, Kanter JE, Kothari V, Kramer F, Tang J, Tall AR, Bornfeldt KE. Hematopoietic NLRP3 and AIM2 Inflammasomes Promote Diabetes-Accelerated Atherosclerosis, but Increased Necrosis Is Independent of Pyroptosis. Diabetes 2023; 72:999-1011. [PMID: 37083999 PMCID: PMC10281813 DOI: 10.2337/db22-0962] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/15/2023] [Indexed: 04/22/2023]
Abstract
Serum apolipoprotein C3 (APOC3) predicts incident cardiovascular events in people with type 1 diabetes, and silencing of APOC3 prevents both lesion initiation and advanced lesion necrotic core expansion in a mouse model of type 1 diabetes. APOC3 acts by slowing the clearance of triglyceride-rich lipoproteins, but lipid-free APOC3 has recently been reported to activate an inflammasome pathway in monocytes. We therefore investigated the contribution of hematopoietic inflammasome pathways to atherosclerosis in mouse models of type 1 diabetes. LDL receptor-deficient diabetes mouse models were transplanted with bone marrow from donors deficient in NOD, LRR and pyrin domain-containing protein 3 (NLRP3), absent in melanoma 2 (AIM2) or gasdermin D (GSDMD), an inflammasome-induced executor of pyroptotic cell death. Mice with diabetes exhibited inflammasome activation and consistently, increased plasma interleukin-1β (IL-1β) and IL-18. Hematopoietic deletions of NLRP3, AIM2, or GSDMD caused smaller atherosclerotic lesions in diabetic mice. The increased lesion necrotic core size in diabetic mice was independent of macrophage pyroptosis because hematopoietic GSDMD deficiency failed to prevent necrotic core expansion in advanced lesions. Our findings demonstrate that AIM2 and NLRP3 inflammasomes contribute to atherogenesis in diabetes and suggest that necrotic core expansion is independent of macrophage pyroptosis. ARTICLE HIGHLIGHTS The contribution of hematopoietic cell inflammasome activation to atherosclerosis associated with type 1 diabetes is unknown. The goal of this study was to address whether hematopoietic NOD, LRR, and pyrin domain-containing protein 3 (NLRP3), absent in melanoma 2 (AIM2) inflammasomes, or the pyroptosis executioner gasdermin D (GSDMD) contributes to atherosclerosis in mouse models of type 1 diabetes. Diabetic mice exhibited increased inflammasome activation, with hematopoietic deletions of NLRP3, AIM2, or GSDMD causing smaller atherosclerotic lesions in diabetic mice, but the increased lesion necrotic core size in diabetic mice was independent of macrophage pyroptosis. Further studies on whether inflammasome activation contributes to cardiovascular complications in people with type 1 diabetes are warranted.
Collapse
Affiliation(s)
- Cheng-Chieh Hsu
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Trevor P. Fidler
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Jenny E. Kanter
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Vishal Kothari
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Farah Kramer
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jingjing Tang
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Alan R. Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Karin E. Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
27
|
Sanjurjo L, Castelblanco E, Julve J, Villalmanzo N, Téllez É, Ramirez-Morros A, Alonso N, Mauricio D, Sarrias MR. Contribution of Elevated Glucose and Oxidized LDL to Macrophage Inflammation: A Role for PRAS40/Akt-Dependent Shedding of Soluble CD14. Antioxidants (Basel) 2023; 12:antiox12051083. [PMID: 37237950 DOI: 10.3390/antiox12051083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Atherosclerosis, a process in which macrophages play a key role, is accelerated in diabetes. Elevated concentrations of serum-oxidized low-density lipoproteins (oxLDL) represent a common feature of both conditions. The main goal of this study was to determine the contribution of oxLDL to the inflammatory response of macrophages exposed to diabetic-mimicking conditions. THP1 cells and peripheral blood monocytes purified from non-diabetic healthy donors were cultured under normal (5 mM) or high glucose (HG) (15 mM) with oxLDL. Then, foam cell formation, expression of CD80, HLADR, CD23, CD206, and CD163, as well as toll-like receptor 4 (TLR4) and co-receptors CD36 and CD14 (both at the cell surface and soluble (sCD14)), and inflammatory mediators' production were measured by flow cytometry, RT-qPCR, or ELISA. Additionally, serum sCD14 was determined in subjects with subclinical atherosclerosis with and without diabetes by ELISA. Our results showed that oxLDL-mediated intracellular lipid accumulation via CD36 increased under HG and that HG + oxLDL enhanced TNF, IL1B, and IL8, and decreased IL10. Moreover, TLR4 was upregulated in macrophages under HG and monocytes of subjects with diabetes and atherosclerosis. Interestingly, HG-oxLDL upregulated CD14 gene expression, although its total cellular protein abundance remained unaltered. sCD14 shedding via PRAS40/Akt-dependent mechanisms, with pro-inflammatory activity, was significantly increased in cultured macrophages and plasma from subjects with diabetes and subclinical atherosclerosis or hypercholesterolemia. Our data support an enhanced synergistic pro-inflammatory effect induced by HG and oxLDL in cultured human macrophages, possibly explained by increased sCD14 shedding.
Collapse
Affiliation(s)
- Lucía Sanjurjo
- Innate Immunity Group, Health Sciences Research Institute Germans Trias i Pujol (IGTP), 08916 Badalona, Spain
| | - Esmeralda Castelblanco
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Unitat de Suport a la Recerca Barcelona, Institut Universitari d'Investigació en Atenció Primària Jordi Gol i Gurina, 08007 Barcelona, Spain
| | - Josep Julve
- Endocrinology, Diabetes and Nutrition Group, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau (IRHSCSP), 08041 Barcelona, Spain
- Centre for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Nuria Villalmanzo
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
| | - Érica Téllez
- Innate Immunity Group, Health Sciences Research Institute Germans Trias i Pujol (IGTP), 08916 Badalona, Spain
| | - Anna Ramirez-Morros
- Unitat de Suport a la Recerca Barcelona, Institut Universitari d'Investigació en Atenció Primària Jordi Gol i Gurina, 08007 Barcelona, Spain
- Gerència Territorial de la Catalunya Central, Institut Català de la Salut, 08272 Sant Fruitós de Bages, Spain
| | - Núria Alonso
- Centre for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), ISCIII, 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Health Sciences Research Institute and University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
| | - Dídac Mauricio
- Unitat de Suport a la Recerca Barcelona, Institut Universitari d'Investigació en Atenció Primària Jordi Gol i Gurina, 08007 Barcelona, Spain
- Centre for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), ISCIII, 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau and Sant Pau Biomedical Research Institute, 08041 Barcelona, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia, 08500 Vic, Spain
| | - Maria-Rosa Sarrias
- Innate Immunity Group, Health Sciences Research Institute Germans Trias i Pujol (IGTP), 08916 Badalona, Spain
- Centre for Biomedical Research on Liver and Digestive Diseases (CIBEREHD), ISCIII, 28029 Madrid, Spain
| |
Collapse
|
28
|
Ren Y, Zhang H. Emerging role of exosomes in vascular diseases. Front Cardiovasc Med 2023; 10:1090909. [PMID: 36937921 PMCID: PMC10017462 DOI: 10.3389/fcvm.2023.1090909] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/11/2023] [Indexed: 03/06/2023] Open
Abstract
Exosomes are biological small spherical lipid bilayer vesicles secreted by most cells in the body. Their contents include nucleic acids, proteins, and lipids. Exosomes can transfer material molecules between cells and consequently have a variety of biological functions, participating in disease development while exhibiting potential value as biomarkers and therapeutics. Growing evidence suggests that exosomes are vital mediators of vascular remodeling. Endothelial cells (ECs), vascular smooth muscle cells (VSMCs), inflammatory cells, and adventitial fibroblasts (AFs) can communicate through exosomes; such communication is associated with inflammatory responses, cell migration and proliferation, and cell metabolism, leading to changes in vascular function and structure. Essential hypertension (EH), atherosclerosis (AS), and pulmonary arterial hypertension (PAH) are the most common vascular diseases and are associated with significant vascular remodeling. This paper reviews the latest research progress on the involvement of exosomes in vascular remodeling through intercellular information exchange and provides new ideas for understanding related diseases.
Collapse
Affiliation(s)
- Yi Ren
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Honggang Zhang
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
29
|
Dorenkamp M, Nasiry M, Semo D, Koch S, Löffler I, Wolf G, Reinecke H, Godfrey R. Pharmacological Targeting of the RAGE-NFκB Signalling Axis Impedes Monocyte Activation under Diabetic Conditions through the Repression of SHP-2 Tyrosine Phosphatase Function. Cells 2023; 12:cells12030513. [PMID: 36766855 PMCID: PMC9914555 DOI: 10.3390/cells12030513] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 02/09/2023] Open
Abstract
Monocytes play a vital role in the development of cardiovascular diseases. Type 2 diabetes mellitus (T2DM) is a major CVD risk factor, and T2DM-induced aberrant activation and enhanced migration of monocytes is a vital pathomechanism that leads to atherogenesis. We recently reported the upregulation of SHP-2 phosphatase expression in mediating the VEGF resistance of T2DM patient-derived monocytes or methylglyoxal- (MG, a glucose metabolite and advanced glycation end product (AGE) precursor) treated monocytes. However, the exact mechanisms leading to SHP-2 upregulation in hyperglycemic monocytes are unknown. Since inflammation and accumulation of AGEs is a hallmark of T2DM, we hypothesise that inflammation and AGE-RAGE (Receptor-for-AGEs) signalling drive SHP-2 expression in monocytes and blockade of these pathways will repress SHP-2 function. Indeed, monocytes from T2DM patients revealed an elevated SHP-2 expression. Under normoglycemic conditions, the serum from T2DM patients strongly induced SHP-2 expression, indicating that the T2DM serum contains critical factors that directly regulate SHP-2 expression. Activation of pro-inflammatory TNFα signalling cascade drove SHP-2 expression in monocytes. In line with this, linear regression analysis revealed a significant positive correlation between TNFα expression and SHP-2 transcript levels in T2DM monocytes. Monocytes exposed to MG or AGE mimetic AGE-BSA, revealed an elevated SHP-2 expression and co-treatment with an NFκB inhibitor or genetic inhibition of p65 reversed it. The pharmacological inhibition of RAGE was sufficient to block MG- or AGE-BSA-induced SHP-2 expression and activity. Confirming the importance of RAGE-NFκB signalling in regulating SHP-2 expression, the elevated binding of NFκB to the SHP-2 promoter-induced by MG or AGE-BSA-was reversed by RAGE and NFκB inhibition. Besides, we detected elevated RAGE levels in human and murine T2DM monocytes and monocytes exposed to MG or AGE-BSA. Importantly, MG and AGE-BSA treatment of non-T2DM monocytes phenocopied the aberrant pro-migratory phenotype of T2DM monocytes, which was reversed entirely by either SHP-2- or RAGE inhibition. In conclusion, these findings suggest a new therapeutic approach to prevent accelerated atherosclerosis in T2DM patients since inhibiting the RAGE-NFκB-SHP-2 axis impeded the T2DM-driven, SHP-2-dependent monocyte activation.
Collapse
Affiliation(s)
- Marc Dorenkamp
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Madina Nasiry
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Dilvin Semo
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Sybille Koch
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Ivonne Löffler
- Department of Internal Medicine III, University Hospital Jena, 07743 Jena, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, 07743 Jena, Germany
| | - Holger Reinecke
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
| | - Rinesh Godfrey
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I—Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany
- Correspondence: ; Tel.: +49-251-83-57089; Fax: +49-251-83-55747
| |
Collapse
|
30
|
Hadzi-Petrushev N, Angelovski M, Mladenov M. Advanced Glycation End Products and Diabetes. CONTEMPORARY ENDOCRINOLOGY 2023:99-127. [DOI: 10.1007/978-3-031-39721-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
31
|
Bill O, Mazya MV, Michel P, Prazeres Moreira T, Lambrou D, Meyer IA, Hirt L. Intima-Media Thickness and Pulsatility Index of Common Carotid Arteries in Acute Ischaemic Stroke Patients with Diabetes Mellitus. J Clin Med 2022; 12:jcm12010246. [PMID: 36615047 PMCID: PMC9821495 DOI: 10.3390/jcm12010246] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022] Open
Abstract
Ultrasonographic parameters such as the common carotid artery (CCA) pulsatility index (PI) and CCA intima-media thickness (IMT) have been associated with an increased mortality and risk of recurrent stroke, respectively. We hypothesized that these ultrasonographic parameters may be useful for monitoring diabetic patients after an acute stroke. We analysed retrospective data of consecutive acute ischaemic stroke patients from the ASTRAL registry who underwent pre-cerebral ultrasonographic evaluation within 7 days of symptom onset. We compared clinical, demographic, radiological and ultrasonographic parameters in diabetic versus non-diabetic patients (univariable and multivariable analyses) and the association of these parameters with CCA PI and CCA IMT. We analysed 1507 carotid duplex ultrasound examinations from patients with a median age of 74 years. Cardiovascular co-morbidities, including hypertension, hypercholesterolemia, obstructive sleep apnoea syndrome, higher body-mass index (BMI) and peripheral artery disease, were associated with diabetes mellitus (DM). Diabetics were more often under antiplatelet therapy and had atrial fibrillation at admission. Diabetic patients showed an increased CCA PI and IMT in line with more atherosclerotic changes on acute CTA compared to non-diabetic patients. Taking IMT as the dependent variable in a second analysis, DM, higher age, hypertension, smoking and CCA PI were associated with higher IMT. Taking CCA PI as the dependent variable in a third analysis, DM, higher age and higher NIHSS at admission were associated with higher CCA PI values. Increased IMT was also associated with higher PI. We show that CCA PI and IMT are higher in diabetic patients in the first week after an initial stroke.
Collapse
Affiliation(s)
- Olivier Bill
- Department of Neurology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
- Correspondence: (O.B.); (L.H.)
| | - Michael V. Mazya
- Department of Neurology, Karolinska University Hospital, 171 77 Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Patrik Michel
- Department of Neurology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Tiago Prazeres Moreira
- Department of Neurology, Karolinska University Hospital, 171 77 Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Dimitris Lambrou
- Department of Neurology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Ivo A. Meyer
- Department of Neurology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
- Department of Old Age Psychiatry and Psychotherapy, University of Bern, 3012 Bern, Switzerland
| | - Lorenz Hirt
- Department of Neurology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
- Correspondence: (O.B.); (L.H.)
| |
Collapse
|
32
|
Gellen B, Thorin‐Trescases N, Thorin E, Gand E, Ragot S, Montaigne D, Pucheu Y, Mohammedi K, Gatault P, Potier L, Liuu E, Hadjadj S, Saulnier P. Increased serum S100A12 levels are associated with higher risk of acute heart failure in patients with type 2 diabetes. ESC Heart Fail 2022; 9:3909-3919. [PMID: 36637406 PMCID: PMC9773733 DOI: 10.1002/ehf2.14036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/09/2022] [Accepted: 06/08/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS The hyperglycaemic stress induces the release of inflammatory proteins such as S100A12, one of the endogenous ligands of the receptors for advanced glycation end products (RAGE). Chronic activation of RAGE has multiple deleterious effects in target tissues such as the heart and the vessels by promoting oxidative stress, inflammation by the release of cytokines, macrophages infiltration, and vascular cell migration and proliferation, causing ultimately endothelial cell and cardiomyocyte dysfunction. The aim of our study was to investigate the prognostic value of circulating S100A12 beyond established cardiovascular risk factors (CVRF) for heart failure (HF) and major adverse cardiovascular events (MACE) in a cohort of patients with type 2 diabetes. METHODS AND RESULTS Serum S100A12 concentrations were measured at baseline in 1345 type 2 diabetes patients (58% men, 64 ± 11 years) recruited in the SURDIAGENE prospective cohort. Endpoints were the occurrence of acute HF requiring hospitalization (HHF) and MACE. We used a proportional hazard model adjusted for established CVRF (age, sex, duration of diabetes, estimated glomerular filtration rate, albumin/creatinine ratio, history of coronary artery disease) and serum S100A12. During the median follow-up of 84 months, 210 (16%) and 505 (38%) patients developed HHF and MACE, respectively. Baseline serum S100A12 concentrations were associated with an increased risk of HHF [hazard ratio (HR) (95% confidence interval) 1.28 (1.01-1.62)], but not MACE [1.04 (0.90-1.20)]. After adjustment for CVRF, S100A12 concentrations remained significantly associated with an increased risk of HHF [1.29 (1.01-1.65)]. In a sub-analysis, patients with high probability of pre-existing HF [N terminal pro brain natriuretic peptide (NT-proBNP) >1000 pg/mL, n = 87] were excluded. In the remaining 1258 patients, the association of serum S100A12 with the risk of HHF tended to be more pronounced [1.39 (1.06-1.83)]. When including the gold standard HF marker NT-proBNP in the model, the prognostic value of S100A12 for HHF did not reach significance. Youden method performed at 7 years for HHF prediction yielded an optimal cut-off for S100A12 concentration of 49 ng/mL (sensitivity 53.3, specificity 52.2). Compared with those with S100A12 ≤ 49 ng/mL, patients with S100A12 > 49 ng/mL had a significantly increased risk of HHF in the univariate model [HR = 1.58 (1.19-2.09), P = 0.0015] but also in the multivariate model [HR = 1.63 (1.23-2.16), P = 0.0008]. After addition of NT-proBNP to the multivariate model, S100A12 > 49 ng/mL remained associated with an increased risk of HHF [HR = 1.42 (1.07-1.90), P = 0.0160]. However, the addition of S100A12 categories on top of multivariate model enriched by NT-pro BNP did not improve the ability of the model to predict HHF (relative integrated discrimination improvement = 1.9%, P = 0.1500). CONCLUSIONS In patients with type 2 diabetes, increased serum S100A12 concentration is independently associated with risk of HHF, but not with risk of MACE. Compared with NT-proBNP, the potential clinical interest of S100A12 for the prediction of HF events remains limited. However, S100A12 could be a candidate for a multimarker approach for HF risk assessment in diabetic patients.
Collapse
Affiliation(s)
- Barnabas Gellen
- ELSAN—Polyclinique de Poitiers1 Rue de la ProvidenceF‐86000PoitiersFrance
| | | | - Eric Thorin
- Montreal Heart Institute, Research CenterMontrealQuebecCanada
- Department of Surgery, Faculty of MedicineUniversity of Montréal, Montreal Heart InstituteMontrealQuebecCanada
| | - Elise Gand
- Centre d'Investigation Clinique CIC1402Université de Poitiers, CHU de Poitiers, INSERMPoitiersFrance
| | - Stephanie Ragot
- Centre d'Investigation Clinique CIC1402Université de Poitiers, CHU de Poitiers, INSERMPoitiersFrance
| | - David Montaigne
- Department of Clinical Physiology—EchocardiographyCHU LilleLilleFrance
- INSERMU1011, EGID, Institut Pasteur de LilleUniversity of LilleLilleFrance
| | - Yann Pucheu
- Department of CardiologyCHU de BordeauxPessacFrance
| | - Kamel Mohammedi
- Hôpital Haut‐Lévêque, Department of Endocrinology, Diabetes and Nutrition; University of Bordeaux, Faculty of Medicine; INSERM unit 1034, Biology of Cardiovascular DiseasesBordeaux University HospitalBordeauxFrance
| | | | - Louis Potier
- Department of DiabetologyHôpital Bichat—Claude‐Bernard, APHP, Université de ParisParisFrance
- Cordeliers Research Centre, ImMeDiab team, INSERMParisFrance
| | - Evelyne Liuu
- Centre d'Investigation Clinique CIC1402Université de Poitiers, CHU de Poitiers, INSERMPoitiersFrance
- Department of GeriatricsCHU de PoitiersPoitiersFrance
| | - Samy Hadjadj
- L'institut du ThoraxINSERM, CNRS, UNIV Nantes, CHU NantesNantesFrance
| | - Pierre‐Jean Saulnier
- Centre d'Investigation Clinique CIC1402Université de Poitiers, CHU de Poitiers, INSERMPoitiersFrance
| | | |
Collapse
|
33
|
Bangar NS, Gvalani A, Ahmad S, Khan MS, Tupe RS. Understanding the role of glycation in the pathology of various non-communicable diseases along with novel therapeutic strategies. Glycobiology 2022; 32:1068-1088. [PMID: 36074518 DOI: 10.1093/glycob/cwac060] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/10/2022] [Accepted: 09/02/2022] [Indexed: 01/07/2023] Open
Abstract
Glycation refers to carbonyl group condensation of the reducing sugar with the free amino group of protein, which forms Amadori products and advanced glycation end products (AGEs). These AGEs alter protein structure and function by configuring a negative charge on the positively charged arginine and lysine residues. Glycation plays a vital role in the pathogenesis of metabolic diseases, brain disorders, aging, and gut microbiome dysregulation with the aid of 3 mechanisms: (i) formation of highly reactive metabolic pathway-derived intermediates, which directly affect protein function in cells, (ii) the interaction of AGEs with its associated receptors to create oxidative stress causing the activation of transcription factor NF-κB, and (iii) production of extracellular AGEs hinders interactions between cellular and matrix molecules affecting vascular and neural genesis. Therapeutic strategies are thus required to inhibit glycation at different steps, such as blocking amino and carbonyl groups, Amadori products, AGEs-RAGE interactions, chelating transition metals, scavenging free radicals, and breaking crosslinks formed by AGEs. The present review focused on explicitly elaborating the impact of glycation-influenced molecular mechanisms in developing and treating noncommunicable diseases.
Collapse
Affiliation(s)
- Nilima S Bangar
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Armaan Gvalani
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, University of Hail, Hail City 2440, Saudi Arabia
| | - Mohd S Khan
- Department of Biochemistry, Protein Research Chair, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| |
Collapse
|
34
|
Monocyte-Lymphocyte Ratio and Dysglycemia: A Retrospective, Cross-Sectional Study of the Saudi Population. Healthcare (Basel) 2022; 10:healthcare10112289. [PMID: 36421613 PMCID: PMC9690849 DOI: 10.3390/healthcare10112289] [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: 11/01/2022] [Revised: 11/10/2022] [Accepted: 11/13/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Abnormalities in fasting blood glucose (FBG) resulting in hypoglycemia (OG), impaired fasting glycemia (IFG), or hyperglycemia (HG) arise from disordered metabolic regulation caused in part by inflammation. To date, there is a dearth of evidence regarding the clinical utility of the monocyte−lymphocyte ratio (MLR), an emerging inflammatory index, in the management of dysglycemia. Methods: This retrospective, cross-sectional study explored MLR fluctuations as a function of glycemic control in 14,173 Saudi subjects. Data collected from 11 August 2014 to 18 July 2020 were retrieved from Al-Borg Medical Laboratories. Medians were compared by Mann−Whitney U or Kruskal−Wallis tests and the prevalence, relative risk (RR), and odds ratio (OR) were calculated. Results: MLR was significantly elevated in IFG (p < 0.0001) and HG (p < 0.05) groups compared to the normoglycemia (NG) group, and individuals with elevated MLR (>0.191) had significantly increased FBG (p < 0.001). The risk of IFG (RR = 1.12, 95% CI: 1.06−1.19, p < 0.0002) and HG (RR = 1.10, 95% CI: 1.01−1.20, p < 0.0216) was significantly increased if MLR was elevated, and individuals with elevated MLR were 1.17 times more likely to have IFG (OR = 1.17, 95% CI: 1.08−1.26, p < 0.0002) and 1.13 times more likely to have HG (OR = 1.13, 95% CI: 1.02−1.24, p < 0.0216). Conclusion: Elevated MLR is correlated with and carries a greater risk for IFG and HG. However, large prospective cohort studies are needed to establish the temporal relationship between MLR and FBG and to examine the prognostic value of this novel marker.
Collapse
|
35
|
Inggriani MP, Musthafa A, Puspitawati I, Fachiroh J, Dewi FST, Hartopo AB. Increased endothelin-1 levels in coronary artery disease with diabetes mellitus in an Indonesian population. Can J Physiol Pharmacol 2022; 100:1097-1105. [DOI: 10.1139/cjpp-2022-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus (DM) increases risk of coronary artery disease (CAD). Endothelin-1 (ET-1) is a potential biomarker of endothelial dysfunction. This study aimed to evaluate ET-1 level in CAD patients and its relationship with DM. The cross-sectional design included subjects with angiographically proven CAD and controls among Indonesian. DM was defined by medical history and anti-diabetics use. Serum ET-1 level was measured in both subject groups. We recruited 305 subjects, 183 CAD patients and 122 controls. CAD subjects had higher percentage of males, DM, hypertension, dyslipidemia, smoking, family history of cardiovascular disease, and obesity. ET-1 level was significantly higher in CAD than in controls (2.44 ± 1.49 pg/mL vs. 1.76 ± 0.83 pg/mL; p < 0.001). Increased ET-1 level was significantly associated with DM and dyslipidemia. The highest ET-1 level was observed in CAD with DM, followed by CAD non-DM (2.79 ± 1.63 pg/mL vs. 2.29 ± 1.40 pg/mL; p = 0.023). Among controls, ET-1 level was the lowest in non-DM subjects. Female CAD had higher proportion of DM; however, ET-1 level was similar to male CAD with DM. In conclusion, an increased ET-1 level was significantly associated with DM in patients with CAD. Further research should investigate the potential role of ET-1 receptor antagonists in the secondary prevention of CAD with DM.
Collapse
Affiliation(s)
- Maria Patricia Inggriani
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada–Dr. Sardjito Hospital, Yogyakarta55281, Indonesia
| | - Ahmad Musthafa
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada–Dr. Sardjito Hospital, Yogyakarta55281, Indonesia
| | - Ira Puspitawati
- Department of Clinical Pathology and Laboratory Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada–Dr. Sardjito Hospital, Yogyakarta55281, Indonesia
| | - Jajah Fachiroh
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing—Biobank Unit, Universitas Gadjah Mada, Yogyakarta55281, Indonesia
| | - Fatwa Sari Tetra Dewi
- Department of Health Behaviour, Environment and Social Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta55281, Indonesia
| | - Anggoro Budi Hartopo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada–Dr. Sardjito Hospital, Yogyakarta55281, Indonesia
| |
Collapse
|
36
|
Jang EJ, Kim H, Baek SE, Jeon EY, Kim JW, Kim JY, Kim CD. HMGB1 increases RAGE expression in vascular smooth muscle cells via ERK and p-38 MAPK-dependent pathways. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:389-396. [PMID: 36039739 PMCID: PMC9437367 DOI: 10.4196/kjpp.2022.26.5.389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/15/2022]
Abstract
The increased expression of receptors for advanced glycation end-product (RAGE) is known as a key player in the progression of vascular remodeling. However, the precise signal pathways regulating RAGE expression in vascular smooth muscle cells (VSMCs) in the injured vasculatures are unclear. Given the importance of mitogen-activated protein kinase (MAPK) signaling in cell proliferation, we investigated the importance of MAPK signaling in high-mobility group box 1 (HMGB1)-induced RAGE expression in VSMCs. In HMGB1 (100 ng/ml)-stimulated human VSMCs, the expression of RAGE mRNA and protein was increased in association with an increase in AGE-induced VSMC proliferation. The HMGB1-induced RAGE expression was attenuated in cells pretreated with inhibitors for ERK (PD98059, 10 μM) and p38 MAPK (SB203580, 10 μM) as well as in cells deficient in ERK and p38 MAPK using siRNAs, but not in cells deficient of JNK signaling. In cells stimulated with HMGB1, the phosphorylation of ERK, JNK, and p38 MAPK was increased. This increase in ERK and p38 MAPK phosphorylation was inhibited by p38 MAPK and ERK inhibitors, respectively, but not by JNK inhibitor. Moreover, AGE-induced VSMC proliferation in HMGB1-stimulated cells was attenuated in cells treated with ERK and p38 MAPK inhibitors. Taken together, our results indicate that ERK and p38 MAPK signaling are involved in RAGE expression in HMGB1-stimulated VSMCs. Thus, the ERK/p38 MAPK-RAGE signaling axis in VSMCs was suggested as a potential therapeutic target for vascular remodeling in the injured vasculatures.
Collapse
Affiliation(s)
- Eun Jeong Jang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Heejeong Kim
- Department of Laboratory Medicine, Pusan National University Hospital, Busan 49241, Korea
| | - Seung Eun Baek
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Eun Yeong Jeon
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Ji Won Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Ju Yeon Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
37
|
Ménégaut L, Laubriet A, Crespy V, Nguyen M, Petit JM, Tarris G, Pilot T, Varin A, Choubley H, Bergas V, de Barros JPP, Thomas C, Steinmetz E, Masson D. Profiling of lipid mediators in atherosclerotic carotid plaques from type 2 diabetic and non-diabetic patients. Prostaglandins Leukot Essent Fatty Acids 2022; 184:102477. [PMID: 35952424 DOI: 10.1016/j.plefa.2022.102477] [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: 12/24/2021] [Revised: 07/18/2022] [Accepted: 08/03/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS Diabetes is associated with an accelerated development of atherosclerosis. Specific mechanisms related to diabetes and hyperglycemia may play a role in this process. In particular, alterations of arachidonic acid (AA) metabolism have been reported. Our main goal was to investigate for differences in the concentration of LTB4 and RvD1 as well as selected cyclooxygenase-derived mediators in carotid plaques from diabetic and non-diabetic patients. We also aimed to analyze the relationship between omega 6 and omega 3 Poly-Unsaturated Fatty acids (PUFAs) content in the plaques and the concentrations of these lipid mediators. METHODS 29 type 2 diabetic patients and 30 control patients admitted for surgical treatment of carotid stenosis were enrolled in the present study. Carotid plaques were harvested for in-depth lipidomic profiling. RESULTS No differences for LTB4 or other lipid mediators were observed between diabetic and non-diabetic patients. RvD1 levels were below the threshold of quantification in most of the samples. A significant correlation was found between LTB4 and 5(S)-HETE levels. Omega 3 enrichment was not significantly different between control and diabetic plaques. There was a negative correlation between DHA/AA ratio and the level of 5(S)-HETE while there was a positive association with TXB2 and PGD2 concentrations. CONCLUSION-PERSPECTIVES Our results does not support the hypothesis of a specific involvement of LTB4 or COX-derived mediators in diabetic atherosclerosis. The relationship between DHA enrichment and the concentrations of specific inflammatory mediators within the plaque is of interest and will need to be confirmed in larger studies.
Collapse
Affiliation(s)
- Louise Ménégaut
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; CHU Dijon, Laboratory of Clinical Chemistry, Dijon, France
| | - Aline Laubriet
- CHU Dijon, Department of Cardiovascular Surgery, Dijon, France
| | - Valentin Crespy
- CHU Dijon, Department of Cardiovascular Surgery, Dijon, France
| | - Maxime Nguyen
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; CHU Dijon Department of Anesthesiology and Intensive Care, Dijon, France
| | - Jean-Michel Petit
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; CHU Dijon, Department of Endocrinology and metabolic diseases, Dijon, France
| | | | - Thomas Pilot
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Alexis Varin
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Lipidomic Analytic Platform, Université Bourgogne Franche-Comté, Dijon, France
| | - Hélène Choubley
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Lipidomic Analytic Platform, Université Bourgogne Franche-Comté, Dijon, France
| | - Victoria Bergas
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Lipidomic Analytic Platform, Université Bourgogne Franche-Comté, Dijon, France
| | - Jean-Paul Pais de Barros
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Lipidomic Analytic Platform, Université Bourgogne Franche-Comté, Dijon, France
| | - Charles Thomas
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Eric Steinmetz
- CHU Dijon, Department of Cardiovascular Surgery, Dijon, France
| | - David Masson
- Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France; INSERM, UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; CHU Dijon, Laboratory of Clinical Chemistry, Dijon, France.
| |
Collapse
|
38
|
The Burden of Impaired Serum Albumin Antioxidant Properties and Glyco-Oxidation in Coronary Heart Disease Patients with and without Type 2 Diabetes Mellitus. Antioxidants (Basel) 2022; 11:antiox11081501. [PMID: 36009220 PMCID: PMC9404962 DOI: 10.3390/antiox11081501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 01/27/2023] Open
Abstract
Human serum albumin (HSA) has an important antioxidant activity due to the presence of the reduced cysteine at position 34, which represents the most abundant free thiol in the plasma. In oxidative-based diseases, HSA undergoes S-thiolation (THIO-HSA) with changes in the antioxidant function of albumin that could contribute to the progression of the disease. The aim of this study was to verify, for the first time, the different burdens of THIO-HSA, glycated HSA (GLY-HSA), and advanced glycation end products (AGE) accumulation both in type 2 diabetes mellitus (T2DM) patients and in non-diabetic patients, with or without coronary heart disease (CHD). In this study, we assessed the presence of modified forms of HSA, THIO-HSA, and GLY-HSA by means of mass spectrometry in 33 patients with both T2DM and CHD, in 31 patients with T2DM and without CHD, in 30 patients without diabetes with a history of CHD, and 27 subjects without diabetes and CHD. All the patients’ anthropometric and clinical data were recorded including age, sex, duration of diabetes, body mass index (BMI), blood pressure, and history of CHD defined with anamnestic data. Metabolic parameters, such as fasting plasma glucose (FPG), glycated hemoglobin (HbA1c), lipids, pentosidine, AGE, receptor for advanced glycation end-products (RAGE) and its soluble form (sRAGE), were measured. AGE and pentosidine are significantly higher in T2DM patients with and without CHD with respect to non-diabetic patients with CHD and control subjects. RAGE levels are significantly higher in T2DM patients with respect to non-diabetic patients, and among T2DM patients, the group with CHD showed significantly higher RAGE levels than those without CHD (217 ± 171 pg/mL and 140 ± 61 pg/mL, respectively). Albumin isoforms discriminate between non-diabetic patients with CHD and T2DM patients with and without CHD and control subjects, with GLY-HSA levels higher in T2DM with and without CHD, and THIO-HSA higher in CHD patients without T2DM. Finally, we demonstrated that the oxidized forms of HSA can increase the expression of the inflammatory cytokine Tumor Necrosis Factor-alpha (TNFα) in monocytic cells. In patients with CHD, GLY-HSA and THIO-HSA have a different prevalent distribution, the first one prevailing in patients with T2DM and the second one in patients without T2DM. These findings suggest that albumin quality and homeostasis balance between glyco-oxidation and thiolation might have an impact on the antioxidant defense system in cardiovascular diseases.
Collapse
|
39
|
Dong H, Zhang Y, Huang Y, Deng H. Pathophysiology of RAGE in inflammatory diseases. Front Immunol 2022; 13:931473. [PMID: 35967420 PMCID: PMC9373849 DOI: 10.3389/fimmu.2022.931473] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a non-specific multi-ligand pattern recognition receptor capable of binding to a range of structurally diverse ligands, expressed on a variety of cell types, and performing different functions. The ligand-RAGE axis can trigger a range of signaling events that are associated with diabetes and its complications, neurological disorders, cancer, inflammation and other diseases. Since RAGE is involved in the pathophysiological processes of many diseases, targeting RAGE may be an effective strategy to block RAGE signaling.
Collapse
|
40
|
Physiological responses and adaptations to high methane production in Japanese Black cattle. Sci Rep 2022; 12:11154. [PMID: 35778422 PMCID: PMC9249741 DOI: 10.1038/s41598-022-15146-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022] Open
Abstract
In this study, using enteric methane emissions, we investigated the metabolic characteristics of Japanese Black cattle. Their methane emissions were measured at early (age 13 months), middle (20 months), and late fattening phases (28 months). Cattle with the highest and lowest methane emissions were selected based on the residual methane emission values, and their liver transcriptome, blood metabolites, hormones, and rumen fermentation characteristics were analyzed. Blood β-hydroxybutyric acid and insulin levels were high, whereas blood amino acid levels were low in cattle with high methane emissions. Further, propionate and butyrate levels differed depending on the enteric methane emissions. Hepatic genes, such as SERPINI2, SLC7A5, ATP6, and RRAD, which were related to amino acid transport and glucose metabolism, were upregulated or downregulated during the late fattening phase. The above mentioned metabolites and liver transcriptomes could be used to evaluate enteric methanogenesis in Japanese Black cattle.
Collapse
|
41
|
Dongliang Y, Yang R, Peng S, Deng J, Huo Y, Deng Z, Yau Y, Liu J, Liao D, Cheng C. Guanxin Xiaoban capsules could treat atherosclerosis by affecting the gut microbiome and inhibiting the AGE-RAGE signalling pathway. J Med Microbiol 2022; 71. [PMID: 35580023 DOI: 10.1099/jmm.0.001530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Atherosclerosis is a chronic disorder in which plaque builds up in the arteries and is associated with several cardiovascular and cerebrovascular diseases such as coronary artery disease, cerebral infarction and cerebral haemorrhage. Therefore, there is an urgent need to discover new medications to treat or prevent atherosclerosis.Hypothesis/Gap Statement. The active components of Guanxin Xiaoban capsules may have an effect on the gut microbiome of patients with atherosclerosis and have a role in their therapeutic targets.Aim. The aim of this study was to identify genes and pathways targeted by active ingredients in Guanxin Xiaoban capsules for the treatment of atherosclerosis based on network pharmacology and analysis of changes to the gut microbiome.Methods. Mice were treated with Guanxin Xiaoban capsules. The 16S rDNA genome sequence of all microorganisms from each group of faecal samples was used to evaluate potential structural changes in the gut microbiota after treatment with Guanxin Xiaoban capsules. Western blotting and real-time quantitative PCR were used to detect gene targets in aortic and liver tissues. Haematoxylin and eosin staining was used to observe improvements in mouse arterial plaques.Results. The gut microbiota of atherosclerotic mice is disturbed. After Guanxin Xiaoban treatment, the abundance of bacteria in the mice improved, with an increase in the proportion of Akkermansia and a significant decrease in the proportion of Faecalibaculum. The main ingredients of Guanxin Xiaoban capsules are calycosin, liquiritin, ferulic acid, ammonium glycyrrhizate, aloe emodin, rhein and emodin. The core genes of this network were determined to be glutathione S-transferase mu 1 (GSTM1), vascular endothelial growth factor A (VEGFA) and cyclin-dependent kinase inhibitor 1A (CDKN1A). The compound-target gene network revealed an interaction between multiple components and targets and contributed to a better understanding of the potential therapeutic effects of the capsules on atherosclerosis. In addition, expression of the AGE-receptor for the AGE (RAGE) pathway was significantly inhibited and the mice showed signs of arterial plaque reduction. Guanxin Xiaoban capsules may improve atherosclerosis and reduce the plaque area by inhibiting the AGE-RAGE signalling pathway to delay the development of atherosclerosis. This mechanism appears to involve changes in the gut microbiota. Therefore, Guanxin Xiaoban capsules have potential value as a treatment for atherosclerosis.
Collapse
Affiliation(s)
- Yin Dongliang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, PR China.,Department of Rehabilitation Medicine, Third Xiangya Hospital, Central South University, Changsha, PR China
| | - Rong Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, PR China
| | - Sha Peng
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, PR China
| | - Jing Deng
- Hunan Key Laboratory for Quality Evaluation of Bulk Herbs, Hunan University of Chinese Medicine, Changsha, PR China
| | - Yanjie Huo
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, PR China
| | - Zhe Deng
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| | - Yuenming Yau
- School of Medicine, Xiamen University, Xiamen, PR China
| | - Jianhe Liu
- Department of Cardiovascular Diseases, The First Hospital of Hunan University of Chinese Medicine, Changsha, PR China
| | - Duanfang Liao
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, PR China
| | - Choufu Cheng
- Department of Cardiovascular Diseases, The First Hospital of Hunan University of Chinese Medicine, Changsha, PR China
| |
Collapse
|
42
|
Singh S, Siva BV, Ravichandiran V. Advanced Glycation End Products: key player of the pathogenesis of atherosclerosis. Glycoconj J 2022; 39:547-563. [PMID: 35579827 DOI: 10.1007/s10719-022-10063-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 01/08/2023]
Abstract
Atherosclerosis is the most common type of cardiovascular disease, and it causes intima thickening, plaque development, and ultimate blockage of the artery lumen. Advanced glycation end products (AGEs) are thought to have a role in the development and progression of atherosclerosis. there is developing an enthusiasm for AGEs as a potential remedial target. AGES mainly induce arterial damage and exacerbate the development of atherosclerotic plaques by triggering cell receptor-dependent signalling. The interplay of AGEs with RAGE, a transmembrane signalling receptor present across all cells important to atherosclerosis, changes cell activity, boosts expression of genes, and increases the outflow of inflammatory compounds, resulting in arterial wall injury and plaque formation. Here in this review, function of AGEs in the genesis, progression, and instability of atherosclerosis is discussed. In endothelial and smooth muscle cells, as well as platelets, the interaction of AGEs with their transmembrane cell receptor, RAGE, triggers intracellular signalling, resulting in endothelial damage, vascular smooth muscle cell function modification, and changed platelet activity.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, 844102, Dist:Vaishali, Hajipur, Bihar, India.
| | - Boddu Veerabadra Siva
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, 844102, Dist:Vaishali, Hajipur, Bihar, India
| | - V Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, 844102, Dist:Vaishali, Hajipur, Bihar, India
| |
Collapse
|
43
|
Increased frequency of proangiogenic tunica intima endothelial kinase 2 (Tie2) expressing monocytes in individuals with type 2 diabetes mellitus. Cardiovasc Diabetol 2022; 21:72. [PMID: 35549955 PMCID: PMC9102255 DOI: 10.1186/s12933-022-01497-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/18/2022] [Indexed: 12/02/2022] Open
Abstract
Background Individuals with type 2 diabetes mellitus (T2DM) have an increased risk for developing macrovascular disease (MVD) manifested by atherosclerosis. Phenotypically and functionally different monocyte subsets (classical; CD14++CD16−, non-classical; CD14+CD16++, and intermediate; CD14++CD16+) including pro-angiogenic monocytes expressing Tie2 (TEMs) can be identified. Here we investigated monocyte heterogeneity and its association with T2DM and MVD. Methods Individuals with (N = 51) and without (N = 56) T2DM were recruited and allocated to "non-MVD" or "with MVD" (i.e., peripheral or coronary artery disease) subgroups. Blood monocyte subsets were quantified based on CD14, CD16 and Tie2 expression levels. Plasma levels of Tie2-ligands angiopoietin-1 and angiopoietin-2 were determined using ELISA. Carotid endarterectomy samples from individuals with (N = 24) and without (N = 22) T2DM were stained for intraplaque CD68+ macrophages (inflammation) and CD34+ (angiogenesis), as plaque vulnerability markers. Results Monocyte counts were similar between individuals with T2DM and healthy controls (non-diabetic, non-MVD). Non-classical monocytes were reduced (p < 0.05) in T2DM, whereas the percentage of TEMs within the intermediate subset was increased (p < 0.05). T2DM was associated with increased angiopoietin-1 (p < 0.05) and angiopoietin-2 (p = 0.0001) levels. Angiopoietin-2 levels were higher in T2DM individuals with MVD compared with non-MVD (p < 0.01). Endarterectomized plaques showed no differences in macrophage influx and microvessel number between individuals with and without T2DM. Conclusions Monocyte subset distribution is altered in T2DM with reduced non-classical monocytes and increased TEM percentage in the intermediate monocyte subset. Increased angiopoietin-2 levels together with increased frequency of TEMs might promote plaque vulnerability in T2DM which could however not be confirmed at tissue level in advanced atherosclerotic lesions.
Collapse
|
44
|
Lu YK, Dong J, Li YL, Liu YH, Hu LK, Chu X, Yan YX. Association between insulin resistance and incidence of carotid atherosclerotic plaque: A cohort study. Nutr Metab Cardiovasc Dis 2022; 32:981-993. [PMID: 35168827 DOI: 10.1016/j.numecd.2022.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/23/2021] [Accepted: 01/10/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS There is limited evidence on the association between insulin resistance (IR) and carotid plaque was reported in prospective study. We aimed to exploit the relationship between IR and carotid plaque in a prospective cohort study. METHODS AND RESULTS The study was performed in a functional community cohort in urban Beijing. In 2015, a total of 7061 individuals without intima-media thickness (IMT) thickening and carotid artery plaque were recruited and followed up until 2019. Restricted cubic spline was conducted to exploit the dose-response relationship between carotid plaque and baseline HOMA-IR or TyG index as continuous variables. Logistic regression was used to analyze the associations between carotid plaque and HOMA-IR or TyG index. During the average 4 years follow-up, 589 subjects developed carotid plaque. Both HOMA-IR and TyG index showed significant linear dose-response relationship on carotid plaque (p < 0.001). The RRs (95%CI) for subjects with baseline HOMA-IR in quartile 2, quartile 3 and quartile 4 were 1.52 (1.14-2.04), 1.86 (1.40-2.46), and 2.55 (1.94-3.35) compared to quartile 1, respectively. Compared to the first quartile of TyG, the RRs (95%CI) for subjects in quartile 2, quartile 3 and quartile 4 were 1.43 (1.08-1.90), 1.59 (1.20-2.12), and 1.69 (1.26-2.25), respectively. In total population, the predictive ability of HOMA-IR for carotid plaque was significantly better than that of TyG index (p = 0.025). CONCLUSION IR is an independent risk factor of carotid plaque. Both HOMA-IR and TyG has significant predictive ability for carotid plaque.
Collapse
Affiliation(s)
- Ya-Ke Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Jing Dong
- Physical Examination Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yan-Ling Li
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yu-Hong Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Li-Kun Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xi Chu
- Physical Examination Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing 100069, China.
| |
Collapse
|
45
|
Abachi S, Pilon G, Marette A, Bazinet L, Beaulieu L. Beneficial effects of fish and fish peptides on main metabolic syndrome associated risk factors: Diabetes, obesity and lipemia. Crit Rev Food Sci Nutr 2022; 63:7896-7944. [PMID: 35297701 DOI: 10.1080/10408398.2022.2052261] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The definition of metabolic syndrome (MetS) fairly varies from one to another guideline and health organization. Per description of world health organization, occurrence of hyperinsulinemia or hyperglycemia in addition to two or more factors of dyslipidemia, hypoalphalipoproteinemia, hypertension and or large waist circumference factors would be defined as MetS. Conventional therapies and drugs, commonly with adverse effects, are used to treat these conditions and diseases. Nonetheless, in the recent decades scientific community has focused on the discovery of natural compounds to diminish the side effects of these medications. Among many available bioactives, biologically active peptides have notable beneficial effects on the management of diabetes, obesity, hypercholesterolemia, and hypertension. Marine inclusive of fish peptides have exerted significant bioactivities in different experimental in-vitro, in-vivo and clinical settings. This review exclusively focuses on studies from the recent decade investigating hypoglycemic, hypolipidemic, hypercholesterolemic and anti-obesogenic fish and fish peptides. Related extraction, isolation, and purification methodologies of anti-MetS fish biopeptides are reviewed herein for comparison purposes only. Moreover, performance of biopeptides in simulated gastrointestinal environment and structure-activity relationship along with absorption, distribution, metabolism, and excretion properties of selected oligopeptides have been discussed, in brief, to broaden the knowledge of readers on the design and discovery trends of anti-MetS compounds.Supplemental data for this article is available online at https://doi.org/10.1080/10408398.2022.2052261 .
Collapse
Affiliation(s)
- Soheila Abachi
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada
- Department of Food Science, Faculty of Agricultural and Food Sciences, Université Laval, Quebec, Quebec, Canada
| | - Geneviève Pilon
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Quebec Heart and Lung Institute, Quebec, Quebec, Canada
| | - André Marette
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Quebec Heart and Lung Institute, Quebec, Quebec, Canada
| | - Laurent Bazinet
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada
- Department of Food Science, Faculty of Agricultural and Food Sciences, Université Laval, Quebec, Quebec, Canada
- Laboratory of Food Processing and ElectroMembrane Processes (LTAPEM), Université Laval, Quebec, Quebec, Canada
| | - Lucie Beaulieu
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada
- Department of Food Science, Faculty of Agricultural and Food Sciences, Université Laval, Quebec, Quebec, Canada
| |
Collapse
|
46
|
Pan J, Bao X, Gonçalves I, Jujić A, Engström G. Skin autofluorescence, a measure of tissue accumulation of advanced glycation end products, is associated with subclinical atherosclerosis in coronary and carotid arteries. Atherosclerosis 2022; 345:26-32. [DOI: 10.1016/j.atherosclerosis.2022.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/02/2022]
|
47
|
Yu T, Xu B, Bao M, Gao Y, Zhang Q, Zhang X, Liu R. Identification of potential biomarkers and pathways associated with carotid atherosclerotic plaques in type 2 diabetes mellitus: A transcriptomics study. Front Endocrinol (Lausanne) 2022; 13:981100. [PMID: 36187128 PMCID: PMC9523108 DOI: 10.3389/fendo.2022.981100] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) affects the formation of carotid atherosclerotic plaques (CAPs) and patients are prone to plaque instability. It is crucial to clarify transcriptomics profiles and identify biomarkers related to the progression of T2DM complicated by CAPs. Ten human CAP samples were obtained, and whole transcriptome sequencing (RNA-seq) was performed. Samples were divided into two groups: diabetes mellitus (DM) versus non-DM groups and unstable versus stable groups. The Limma package in R was used to identify lncRNAs, circRNAs, and mRNAs. Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, protein-protein interaction (PPI) network creation, and module generation were performed for differentially expressed mRNAs. Cytoscape was used to create a transcription factor (TF)-mRNA regulatory network, lncRNA/circRNA-mRNA co-expression network, and a competitive endogenous RNA (ceRNA) network. The GSE118481 dataset and RT-qPCR were used to verify potential mRNAs.The regulatory network was constructed based on the verified core genes and the relationships were extracted from the above network. In total, 180 differentially expressed lncRNAs, 343 circRNAs, and 1092 mRNAs were identified in the DM versus non-DM group; 240 differentially expressed lncRNAs, 390 circRNAs, and 677 mRNAs were identified in the unstable versus stable group. Five circRNAs, 14 lncRNAs, and 171 mRNAs that were common among all four groups changed in the same direction. GO/KEGG functional enrichment analysis showed that 171 mRNAs were mainly related to biological processes, such as immune responses, inflammatory responses, and cell adhesion. Five circRNAs, 14 lncRNAs, 46 miRNAs, and 54 mRNAs in the ceRNA network formed a regulatory relationship. C22orf34-hsa-miR-6785-5p-RAB37, hsacirc_013887-hsa-miR-6785-5p/hsa-miR-4763-5p/hsa-miR-30b-3p-RAB37, MIR4435-1HG-hsa-miR-30b-3p-RAB37, and GAS5-hsa-miR-30b-3p-RAB37 may be potential RNA regulatory pathways. Seven upregulated mRNAs were verified using the GSE118481 dataset and RT-qPCR. The regulatory network included seven mRNAs, five circRNAs, six lncRNAs, and 14 TFs. We propose five circRNAs (hsacirc_028744, hsacirc_037219, hsacirc_006308, hsacirc_013887, and hsacirc_045622), six lncRNAs (EPB41L4A-AS1, LINC00969, GAS5, MIR4435-1HG, MIR503HG, and SNHG16), and seven mRNAs (RAB37, CCR7, CD3D, TRAT1, VWF, ICAM2, and TMEM244) as potential biomarkers related to the progression of T2DM complicated with CAP. The constructed ceRNA network has important implications for potential RNA regulatory pathways.
Collapse
Affiliation(s)
- Tian Yu
- Department of Very Important People (VIP) Unit, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Baofeng Xu
- Department of Stroke Center, First Hospital of Jilin University, Changchun, China
- School of Stomatology, Changsha Medical University, Changsha, China
| | - Meihua Bao
- School of Stomatology, Changsha Medical University, Changsha, China
| | - Yuanyuan Gao
- Department of Very Important People (VIP) Unit, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qiujuan Zhang
- Department of Very Important People (VIP) Unit, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuejiao Zhang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Rui Liu
- Department of Very Important People (VIP) Unit, China-Japan Union Hospital of Jilin University, Changchun, China
- School of Stomatology, Changsha Medical University, Changsha, China
- *Correspondence: Rui Liu,
| |
Collapse
|
48
|
Abstract
Inflammation is intimately involved at all stages of atherosclerosis and remains a substantial residual cardiovascular risk factor in optimally treated patients. The proof of concept that targeting inflammation reduces cardiovascular events in patients with a history of myocardial infarction has highlighted the urgent need to identify new immunotherapies to treat patients with atherosclerotic cardiovascular disease. Importantly, emerging data from new clinical trials show that successful immunotherapies for atherosclerosis need to be tailored to the specific immune alterations in distinct groups of patients. In this Review, we discuss how single-cell technologies - such as single-cell mass cytometry, single-cell RNA sequencing and cellular indexing of transcriptomes and epitopes by sequencing - are ideal for mapping the cellular and molecular composition of human atherosclerotic plaques and how these data can aid in the discovery of new precise immunotherapies. We also argue that single-cell data from studies in humans need to be rigorously validated in relevant experimental models, including rapidly emerging single-cell CRISPR screening technologies and mouse models of atherosclerosis. Finally, we discuss the importance of implementing single-cell immune monitoring tools in early phases of drug development to aid in the precise selection of the target patient population for data-driven translation into randomized clinical trials and the successful translation of new immunotherapies into the clinic.
Collapse
Affiliation(s)
- Dawn M Fernandez
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Giannarelli
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- New York University Cardiovascular Research Center, New York University Langone Health, New York, NY, USA.
- Department of Pathology, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
49
|
Yu Y, Ding X, Yu L, Dai X, Wang Y, Zhang J. Increased coronary pericoronary adipose tissue attenuation in diabetic patients compared to non-diabetic controls: a propensity score matching analysis. J Cardiovasc Comput Tomogr 2022; 16:327-335. [DOI: 10.1016/j.jcct.2022.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/28/2022]
|
50
|
Zhang X, Cheng M, Gao N, Li Y, Yan C, Tian X, Liu D, Qiu M, Wang X, Luan B, Deng J, Wang S, Tian H, Wang G, Ma X, Stone GW, Han Y. Utility of S100A12 as an Early Biomarker in Patients With ST-Segment Elevation Myocardial Infarction. Front Cardiovasc Med 2021; 8:747511. [PMID: 34977174 PMCID: PMC8718434 DOI: 10.3389/fcvm.2021.747511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/22/2021] [Indexed: 01/16/2023] Open
Abstract
Importance: S100A12 is a calcium binding protein which is involved in inflammation and progression of atherosclerosis.Objective: We sought to investigate the utility of S100A12 as a biomarker for the early diagnosis and prognostication of patients presenting with ST-segment elevation myocardial infarction (STEMI).Design, Setting, and Participants: S100A12 was measured in 1023 patients presenting to the emergency department with acute chest pain between June 2012 and November 2015. An independent cohort of 398 patients enrolled at 3 different hospitals served as a validation cohort.Main Outcomes and Measures: The primary clinical endpoint of interest was major adverse cardiac and cerebral events (MACCE) defined as a composite of all-cause death, MI, stroke, or hospitalization for heart failure.Results: A total of 438/1023 patients (42.8%) in the diagnosis cohort were adjudicated as STEMI, among whom plasma S100A12 levels increased within 30 min and peaked 1–2 h after symptom onset. Compared with high-sensitivity cardiac troponin T and creatine kinase-MB isoenzyme, S100A12 more accurately identified STEMI, especially within the first 2 h after symptom onset (area under the curve 0.963 compared with 0.860 for hscTnT and 0.711 for CK-MB, both P < 0.05). These results were consistent in the 243-patient validation cohort. The 1-year rate of MACCE was greatest in patients in the highest peak S100A12 tertile, intermediate in the middle tertile and least in the lowest tertile (9.3 vs. 5.7 vs. 3.0% respectively, Ptrend = 0.0006). By multivariable analysis the peak plasma concentration of S100A12 was an independent predictor of MACCE within 1 year after STEMI (HR, 1.001, 95%CI, 1.000–1.002; P = 0.0104).Conclusions and Relevance: S100A12 rapidly identified patients with STEMI, more accurately than other cardiac biomarkers, especially within the first 2 h after symptom onset. The peak plasma S100A12 level was a strong predictor of 1-year prognosis after STEMI.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Minghui Cheng
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Naijing Gao
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Yi Li
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Miaohan Qiu
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaozeng Wang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Bo Luan
- Department of Cardiology, Liaoning Provincial People's Hospital, Shenyang, China
| | - Jie Deng
- Department of Cardiology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shouli Wang
- Department of Cardiology, General Hospital of the Strategic Support Force of the Chinese People's Liberation Army, Beijing, China
| | - Hongyan Tian
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Geng Wang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Gregg W. Stone
- Icahn School of Medicine at Mount Sinai, Mount Sinai Heart and the Cardiovascular Research Foundation, New York, NY, United States
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
- *Correspondence: Yaling Han
| |
Collapse
|